1
|
Leonardi R, Pellino G, Floridia E, Lo Bianco M, Ruggieri M, Cho SY, Pavone V, Pavone P, Polizzi A. Polydactyly and syndactyly linked to GLI3 and TBX5 mutations: A pediatric case report. Glob Med Genet 2025; 12:100033. [PMID: 39925448 PMCID: PMC11800310 DOI: 10.1016/j.gmg.2024.100033] [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: 11/04/2024] [Revised: 11/25/2024] [Accepted: 11/28/2024] [Indexed: 02/11/2025] Open
Abstract
Background Polydactyly and syndactyly, which are commonly encountered congenital limb deformities, rarely occur together and are linked with significant genetic mutations. This report sheds light on a unique co-presentation involving mutations in both the GLI3 and TBX5 genes, offering a deeper understanding of the genetic interactions that may influence limb development. This case report is important to increase our knowledge on genetic bases of limb malformations. Case presentation We report the case of an 8-month-old boy, born to non-consanguineous parents, presenting with both polydactyly and syndactyly in his limbs, in particular, complete syndactyly between the third to fifth fingers and post-axial polydactyly of the feet. His father showed a similar phenotype. Genetic testing identified a pathogenic heterozygous variant in the GLI3 gene (c .3762 T > A, p.(Tyr1254 *)) and a variant of uncertain significance in the TBX5 gene (c .1063 C>T, p.(Arg355Cys)). Conclusions This case highlights the complex nature of diagnosing and managing congenital limb deformities driven by genetic factors. It underscores the critical importance of comprehensive genetic testing in determining the etiology of limb malformations. The GLI3 variant, classified according to ACMG guidelines as a class IV mutation, likely results in a truncated protein due to a premature stop codon, confirmed by family segregation analysis indicating its paternal origin, suggesting autosomal dominant inheritance. Notably, the TBX5 gene variant, often associated with Holt-Oram syndrome-which is characterized by only hand skeletal anomalies and early-onset atrial fibrillation-suggests a risk of developing cardiac issues that are not currently present but may emerge as the child grows. This potential for evolving clinical manifestations necessitates vigilant long-term monitoring and may influence future medical management and therapeutic approaches.
Collapse
Affiliation(s)
- R. Leonardi
- Postgraduate Training Program in Pediatrics, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - G. Pellino
- Postgraduate Training Program in Genetics, Department of Clinical and Molecular Biomedicine Ingrassia, University of Catania, Italy
| | - E. Floridia
- School of Medicine, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - M. Lo Bianco
- Postgraduate Training Program in Pediatrics, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - M. Ruggieri
- Unit of Clinical Pediatrics, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - SY. Cho
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - V. Pavone
- Department of General Surgery and Medical Surgical Specialties, Section of Orthopedics and Traumatology, A.O.U. Policlinico, University of Catania, Via Santa Sofia 78, Catania 95123, Italy
| | - P. Pavone
- Unit of Clinical Pediatrics, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
- Unit of Catania, Institute for Biomedical Research and Innovation, National Council of Research, Catania, Italy
| | - A. Polizzi
- Unit of Clinical Pediatrics, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
- Department of Educational Science, University of Catania, Catania 95100, Italy
| |
Collapse
|
2
|
Jin Y, Pan Z, Zhou J, Wang K, Zhu P, Wang Y, Xu X, Zhang J, Hao C. Hedgehog signaling pathway regulates Th17 cell differentiation in asthma via IL-6/STAT3 signaling. Int Immunopharmacol 2024; 139:112771. [PMID: 39074418 DOI: 10.1016/j.intimp.2024.112771] [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: 04/23/2024] [Revised: 07/10/2024] [Accepted: 07/22/2024] [Indexed: 07/31/2024]
Abstract
Asthma is the most prevalent chronic inflammatory disease of the airways in children. The most prevalent phenotype of asthma is eosinophilic asthma, which is driven by a Th2 immune response and can be effectively managed by inhaled corticosteroid therapy. However, there are phenotypes of asthma with Th17 immune response that are insensitive to corticosteroid therapy and manifest a more severe phenotype. The treatment of this corticosteroid-insensitive asthma is currently immature and requires further attention. The objective of this study is to elucidate the regulation of the Hedgehog signaling pathway in Th17 cell differentiation in asthma. The study demonstrated that both Smo and Gli3, key components of the Hedgehog signaling pathway, were upregulated in Th17 polarization in vitro and in a Th17-dominant asthma model in vivo. Inhibiting Smo with a small molecule inhibitor or genetically knocking down Gli3 was found to suppress Th17 polarization. Smo was found to increase in Th1, Th2, Th17 and Treg polarization, while Gli3 specifically increased in Th17 polarization. ChIP-qPCR analyses indicated that Gli3 can directly interact with IL-6 in T cells, inducing STAT3 phosphorylation and promoting Th17 cell differentiation. Furthermore, the study demonstrated a correlation between elevated Gli3 expression and IL-17A and IL-6 expression in children with asthma. In conclusion, the study demonstrated that the Hedgehog signaling pathway plays an important role in the pathogenesis of asthma, as it regulates the differentiation of Th17 cells through the IL-6/STAT3 signaling. This may provide a potential therapeutic target for corticosteroid-insensitive asthma driven by Th17 cells.
Collapse
Affiliation(s)
- Yuting Jin
- Department of Respiratory Medicine, Children's Hospital of Soochow University, Suzhou, China; Department of Pediatrics, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Zhenzhen Pan
- Department of Respiration, Wuxi Children's Hospital, Wuxi, China
| | - Ji Zhou
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Kai Wang
- Department of Pediatrics, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Peijie Zhu
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Yufeng Wang
- Department of Respiratory Medicine, Children's Hospital of Soochow University, Suzhou, China
| | - Xuena Xu
- Department of Respiratory Medicine, Children's Hospital of Soochow University, Suzhou, China
| | - Jinping Zhang
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China.
| | - Chuangli Hao
- Department of Respiratory Medicine, Children's Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
3
|
Rouault P, Guimbal S, Cornuault L, Bourguignon C, Foussard N, Alzieu P, Choveau F, Benoist D, Chapouly C, Gadeau AP, Couffinhal T, Renault MA. Thrombosis in the Coronary Microvasculature Impairs Cardiac Relaxation and Induces Diastolic Dysfunction. Arterioscler Thromb Vasc Biol 2024; 44:e1-e18. [PMID: 38031839 DOI: 10.1161/atvbaha.123.320040] [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: 08/22/2023] [Accepted: 11/14/2023] [Indexed: 12/01/2023]
Abstract
BACKGROUND Heart failure with preserved ejection fraction is proposed to be caused by endothelial dysfunction in cardiac microvessels. Our goal was to identify molecular and cellular mechanisms underlying the development of cardiac microvessel disease and diastolic dysfunction in the setting of type 2 diabetes. METHODS We used Leprdb/db (leptin receptor-deficient) female mice as a model of type 2 diabetes and heart failure with preserved ejection fraction and identified Hhipl1 (hedgehog interacting protein-like 1), which encodes for a decoy receptor for HH (hedgehog) ligands as a gene upregulated in the cardiac vascular fraction of diseased mice. RESULTS We then used Dhh (desert HH)-deficient mice to investigate the functional consequences of impaired HH signaling in the adult heart. We found that Dhh-deficient mice displayed increased end-diastolic pressure while left ventricular ejection fraction was comparable to that of control mice. This phenotype was associated with a reduced exercise tolerance in the treadmill test, suggesting that Dhh-deficient mice do present heart failure. At molecular and cellular levels, impaired cardiac relaxation in DhhECKO mice was associated with a significantly decreased PLN (phospholamban) phosphorylation on Thr17 (threonine 17) and an alteration of sarcomeric shortening ex vivo. Besides, as expected, Dhh-deficient mice exhibited phenotypic changes in their cardiac microvessels including a prominent prothrombotic phenotype. Importantly, aspirin therapy prevented the occurrence of both diastolic dysfunction and exercise intolerance in these mice. To confirm the critical role of thrombosis in the pathophysiology of diastolic dysfunction, we verified Leprdb/db also displays increased cardiac microvessel thrombosis. Moreover, consistently, with Dhh-deficient mice, we found that aspirin treatment decreased end-diastolic pressure and improved exercise tolerance in Leprdb/db mice. CONCLUSIONS Altogether, these results demonstrate that microvessel thrombosis may participate in the pathophysiology of heart failure with preserved ejection fraction.
Collapse
Affiliation(s)
- Paul Rouault
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1034, Biology of Cardiovascular Diseases, University of Bordeaux, Pessac, France (P.R., S.G., L.C., C.B., N.F., P.A., C.C., A.-P.G., T.C., M.-A.R.)
| | - Sarah Guimbal
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1034, Biology of Cardiovascular Diseases, University of Bordeaux, Pessac, France (P.R., S.G., L.C., C.B., N.F., P.A., C.C., A.-P.G., T.C., M.-A.R.)
| | - Lauriane Cornuault
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1034, Biology of Cardiovascular Diseases, University of Bordeaux, Pessac, France (P.R., S.G., L.C., C.B., N.F., P.A., C.C., A.-P.G., T.C., M.-A.R.)
| | - Célia Bourguignon
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1034, Biology of Cardiovascular Diseases, University of Bordeaux, Pessac, France (P.R., S.G., L.C., C.B., N.F., P.A., C.C., A.-P.G., T.C., M.-A.R.)
| | - Ninon Foussard
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1034, Biology of Cardiovascular Diseases, University of Bordeaux, Pessac, France (P.R., S.G., L.C., C.B., N.F., P.A., C.C., A.-P.G., T.C., M.-A.R.)
| | - Philippe Alzieu
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1034, Biology of Cardiovascular Diseases, University of Bordeaux, Pessac, France (P.R., S.G., L.C., C.B., N.F., P.A., C.C., A.-P.G., T.C., M.-A.R.)
| | - Frank Choveau
- INSERM U1045, CRCTB (Centre de recherche cardio-thoracique de Bordeaux), IHU Liryc (Institut Hospitalo Universitaire des maladies du rythme cardiaque), University of Bordeaux, France (F.C., D.B.)
| | - David Benoist
- INSERM U1045, CRCTB (Centre de recherche cardio-thoracique de Bordeaux), IHU Liryc (Institut Hospitalo Universitaire des maladies du rythme cardiaque), University of Bordeaux, France (F.C., D.B.)
| | - Candice Chapouly
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1034, Biology of Cardiovascular Diseases, University of Bordeaux, Pessac, France (P.R., S.G., L.C., C.B., N.F., P.A., C.C., A.-P.G., T.C., M.-A.R.)
| | - Alain-Pierre Gadeau
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1034, Biology of Cardiovascular Diseases, University of Bordeaux, Pessac, France (P.R., S.G., L.C., C.B., N.F., P.A., C.C., A.-P.G., T.C., M.-A.R.)
| | - Thierry Couffinhal
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1034, Biology of Cardiovascular Diseases, University of Bordeaux, Pessac, France (P.R., S.G., L.C., C.B., N.F., P.A., C.C., A.-P.G., T.C., M.-A.R.)
| | - Marie-Ange Renault
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1034, Biology of Cardiovascular Diseases, University of Bordeaux, Pessac, France (P.R., S.G., L.C., C.B., N.F., P.A., C.C., A.-P.G., T.C., M.-A.R.)
| |
Collapse
|
4
|
Mannan A, Dhiamn S, Garg N, Singh TG. Pharmacological modulation of Sonic Hedgehog signaling pathways in Angiogenesis: A mechanistic perspective. Dev Biol 2023; 504:58-74. [PMID: 37739118 DOI: 10.1016/j.ydbio.2023.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 09/13/2023] [Accepted: 09/18/2023] [Indexed: 09/24/2023]
Abstract
The Sonic hedgehog (SHh) signaling pathway is an imperative operating network that helps in regulates the critical events during the development processes like multicellular embryo growth and patterning. Disruptions in SHh pathway regulation can have severe consequences, including congenital disabilities, stem cell renewal, tissue regeneration, and cancer/tumor growth. Activation of the SHh signal occurs when SHh binds to the receptor complex of Patch (Ptc)-mediated Smoothened (Smo) (Ptc-smo), initiating downstream signaling. This review explores how pharmacological modulation of the SHh pathway affects angiogenesis through canonical and non-canonical pathways. The canonical pathway for angiogenesis involves the activation of angiogenic cytokines such as fibroblast growth factor (FGF), vascular endothelial growth factor (VEGF), placental growth factor (PGF), hepatocyte growth factor (HGF), platelet-derived growth factor (PDGF), stromal cell-derived factor 1α, transforming growth factor-β1 (TGF-β1), and angiopoietins (Ang-1 and Ang-2), which facilitate the process of angiogenesis. The Non-canonical pathway includes indirect activation of certain pathways like iNOS/Netrin-1/PKC, RhoA/Rock, ERK/MAPK, PI3K/Akt, Wnt/β-catenin, Notch signaling pathway, and so on. This review will provide a better grasp of the mechanistic approach of SHh in mediating angiogenesis, which can aid in the suppression of certain cancer and tumor growths.
Collapse
Affiliation(s)
- Ashi Mannan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India.
| | - Sonia Dhiamn
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India.
| | - Nikhil Garg
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India.
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India.
| |
Collapse
|
5
|
Lee Q, Chan WC, Qu X, Sun Y, Abdelkarim H, Le J, Saqib U, Sun MY, Kruse K, Banerjee A, Hitchinson B, Geyer M, Huang F, Guaiquil V, Mutso AA, Sanders M, Rosenblatt MI, Maienschein-Cline M, Lawrence MS, Gaponenko V, Malik AB, Komarova YA. End binding-3 inhibitor activates regenerative program in age-related macular degeneration. Cell Rep Med 2023; 4:101223. [PMID: 37794584 PMCID: PMC10591057 DOI: 10.1016/j.xcrm.2023.101223] [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: 08/22/2020] [Revised: 07/19/2023] [Accepted: 09/12/2023] [Indexed: 10/06/2023]
Abstract
Wet age-related macular degeneration (AMD), characterized by leaky neovessels emanating from the choroid, is a main cause of blindness. As current treatments for wet AMD require regular intravitreal injections of anti-vascular endothelial growth factor (VEGF) biologics, there is a need for the development of less invasive treatments. Here, we designed an allosteric inhibitor of end binding-3 (EB3) protein, termed EBIN, which reduces the effects of environmental stresses on endothelial cells by limiting pathological calcium signaling. Delivery of EBIN via eye drops in mouse and non-human primate (NHP) models of wet AMD prevents both neovascular leakage and choroidal neovascularization. EBIN reverses the epigenetic changes induced by environmental stresses, allowing an activation of a regenerative program within metabolic-active endothelial cells comprising choroidal neovascularization (CNV) lesions. These results suggest the therapeutic potential of EBIN in preventing the degenerative processes underlying wet AMD.
Collapse
Affiliation(s)
- Quinn Lee
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Wan Ching Chan
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Xinyan Qu
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Ying Sun
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | | | - Jonathan Le
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Uzma Saqib
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Mitchell Y Sun
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Kevin Kruse
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Avik Banerjee
- Department of Chemistry, The University of Illinois, Chicago, IL 60612, USA
| | - Ben Hitchinson
- Department of Biochemistry and Molecular Genetics, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Melissa Geyer
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Fei Huang
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Victor Guaiquil
- Department of Ophthalmology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Amelia A Mutso
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | | | - Mark I Rosenblatt
- Department of Ophthalmology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | | | | | - Vadim Gaponenko
- Department of Biochemistry and Molecular Genetics, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Asrar B Malik
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Yulia A Komarova
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA.
| |
Collapse
|
6
|
Foussard N, Rouault P, Cornuault L, Reynaud A, Buys ES, Chapouly C, Gadeau AP, Couffinhal T, Mohammedi K, Renault MA. Praliciguat Promotes Ischemic Leg Reperfusion in Leptin Receptor-Deficient Mice. Circ Res 2023; 132:34-48. [PMID: 36448444 DOI: 10.1161/circresaha.122.322033] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
BACKGROUND Lower-limb peripheral artery disease is one of the major complications of diabetes. Peripheral artery disease is associated with poor limb and cardiovascular prognoses, along with a dramatic decrease in life expectancy. Despite major medical advances in the treatment of diabetes, a substantial therapeutic gap remains in the peripheral artery disease population. Praliciguat is an orally available sGC (soluble guanylate cyclase) stimulator that has been reported both preclinically and in early stage clinical trials to have favorable effects in metabolic and hemodynamic outcomes, suggesting that it may have a potential beneficial effect in peripheral artery disease. METHODS We evaluated the effect of praliciguat on hind limb ischemia recovery in a mouse model of type 2 diabetes. Hind limb ischemia was induced in leptin receptor-deficient (Leprdb/db) mice by ligation and excision of the left femoral artery. Praliciguat (10 mg/kg/day) was administered in the diet starting 3 days before surgery. RESULTS Twenty-eight days after surgery, ischemic foot perfusion and function parameters were better in praliciguat-treated mice than in vehicle controls. Improved ischemic foot perfusion was not associated with either improved traditional cardiovascular risk factors (ie, weight, glycemia) or increased angiogenesis. However, treatment with praliciguat significantly increased arteriole diameter, decreased ICAM1 (intercellular adhesion molecule 1) expression, and prevented the accumulation of oxidative proangiogenic and proinflammatory muscle fibers. While investigating the mechanism underlying the beneficial effects of praliciguat therapy, we found that praliciguat significantly downregulated Myh2 and Cxcl12 mRNA expression in cultured myoblasts and that conditioned medium form praliciguat-treated myoblast decreased ICAM1 mRNA expression in endothelial cells. These results suggest that praliciguat therapy may decrease ICAM1 expression in endothelial cells by downregulating Cxcl12 in myocytes. CONCLUSIONS Our results demonstrated that praliciguat promotes blood flow recovery in the ischemic muscle of mice with type 2 diabetes, at least in part by increasing arteriole diameter and by downregulating ICAM1 expression.
Collapse
Affiliation(s)
- Ninon Foussard
- Univ. Bordeaux, Inserm, Biology of Cardiovascular Diseases, Pessac, France (N.F., P.R., L.C., A.R., C.C., A.-P.G., T.C., K.M., M.-A.R.)
| | - Paul Rouault
- Univ. Bordeaux, Inserm, Biology of Cardiovascular Diseases, Pessac, France (N.F., P.R., L.C., A.R., C.C., A.-P.G., T.C., K.M., M.-A.R.)
| | - Lauriane Cornuault
- Univ. Bordeaux, Inserm, Biology of Cardiovascular Diseases, Pessac, France (N.F., P.R., L.C., A.R., C.C., A.-P.G., T.C., K.M., M.-A.R.)
| | - Annabel Reynaud
- Univ. Bordeaux, Inserm, Biology of Cardiovascular Diseases, Pessac, France (N.F., P.R., L.C., A.R., C.C., A.-P.G., T.C., K.M., M.-A.R.)
| | | | - Candice Chapouly
- Univ. Bordeaux, Inserm, Biology of Cardiovascular Diseases, Pessac, France (N.F., P.R., L.C., A.R., C.C., A.-P.G., T.C., K.M., M.-A.R.)
| | - Alain-Pierre Gadeau
- Univ. Bordeaux, Inserm, Biology of Cardiovascular Diseases, Pessac, France (N.F., P.R., L.C., A.R., C.C., A.-P.G., T.C., K.M., M.-A.R.)
| | - Thierry Couffinhal
- Univ. Bordeaux, Inserm, Biology of Cardiovascular Diseases, Pessac, France (N.F., P.R., L.C., A.R., C.C., A.-P.G., T.C., K.M., M.-A.R.)
| | - Kamel Mohammedi
- Univ. Bordeaux, Inserm, Biology of Cardiovascular Diseases, Pessac, France (N.F., P.R., L.C., A.R., C.C., A.-P.G., T.C., K.M., M.-A.R.)
| | - Marie-Ange Renault
- Univ. Bordeaux, Inserm, Biology of Cardiovascular Diseases, Pessac, France (N.F., P.R., L.C., A.R., C.C., A.-P.G., T.C., K.M., M.-A.R.)
| |
Collapse
|
7
|
|
8
|
Aavani F, Biazar E, Kheilnezhad B, Amjad F. 3D Bio-printing For Skin Tissue Regeneration: Hopes and Hurdles. Curr Stem Cell Res Ther 2022; 17:415-439. [DOI: 10.2174/1574888x17666220204144544] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 11/10/2021] [Accepted: 12/03/2021] [Indexed: 11/22/2022]
Abstract
Abstract:
For many years, discovering the appropriate methods for the treatment of skin irritation has been challenging for specialists and researchers. Bio-printing can be extensively applied to address the demand for proper skin substitutes to improve skin damage. Nowadays, to make more effective bio-mimicking of natural skin, many research teams have developed cell-seeded bio-inks for bioprinting of skin substitutes. These loaded cells can be single or co-cultured in these structures. The present review gives a comprehensive overview of the methods, substantial parameters of skin bioprinting, examples of in vitro and in vivo studies, and current advances and challenges for skin tissue engineering.
Collapse
Affiliation(s)
- Farzaneh. Aavani
- Biomedical Engineering Faculty, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Esmaeil Biazar
- Tissue Engineering Group, Department of Biomedical Engineering, Tonekabon Branch, Islamic Azad University, Tonekabon, Iran
| | - Bahareh Kheilnezhad
- Biomedical Engineering Faculty, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Fatemeh Amjad
- Biomedical Engineering Faculty, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| |
Collapse
|
9
|
Bist D, Pawde AM, Amarpal, Kinjavdekar P, Mukherjee R, Singh KP, Verma MR, Sharun K, Kumar A, Dubey PK, Mohan D, Verma A, Sharma GT. Evaluation of canine bone marrow-derived mesenchymal stem cells for experimental full-thickness cutaneous wounds in a diabetic rat model. Expert Opin Biol Ther 2021; 21:1655-1664. [PMID: 34620044 DOI: 10.1080/14712598.2022.1990260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
BACKGROUND The wound healing potential of canine bone marrow-derived mesenchymal stem cells (BMSCs) was evaluated in the excisional wound of streptozotocin-induced diabetic rats. RESEARCH DESIGN AND METHODS Xenogenic BMSCs were collected aseptically from the iliac crest of healthy canine donors under general anesthesia. Full-thickness experimental wounds (20 × 20 mm2) on the dorsum of forty-eight adult healthy Wistar white rats. The wounds were assigned randomly to three treatment groups: PBS (Group A) or BMSCs (Group B) injected into the wound margins on days 0, 7, and 14 or BMSCs (Group C) injected into the wound margins on days 7, 14, and 21 post-wounding. The degree of wound healing was evaluated based on macroscopical, hemato-biochemical, histopathological, and histochemical parameters. RESULTS The results indicated granulation tissue formation with reduced exudation and peripheral swelling in the treatment groups compared to the control group A. Similarly, the degree of wound contraction was significantly higher in groups B and C animals than group A on days 14 and 21 post-wounding. The transplantation of BMSCs resulted in early drying of wounds, granulation tissue appearance, and enhanced cosmetic appearance. CONCLUSION The histopathological, histochemical, and gross findings suggested the therapeutic potential of xenogeneic mesenchymal stem cell therapy in managing diabetic wounds. ABBREVIATIONS BMSCs-bone marrow-derived mesenchymal stem cells, PBS-phosphate-buffered saline, MSCs-mesenchymal stem cells, FBS-fetal bovine serum, ECM-extracellular matrix.
Collapse
Affiliation(s)
- Deepika Bist
- Division of Surgery, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - A M Pawde
- Division of Surgery, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Amarpal
- Division of Surgery, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Prakash Kinjavdekar
- Division of Surgery, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Reena Mukherjee
- Division of Medicine, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - K P Singh
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Med Ram Verma
- Division of Livestock Economics, Statistics and Information Technology, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Khan Sharun
- Division of Surgery, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Amit Kumar
- Animal Genetics Division, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Pawan K Dubey
- Centre for Genetic Disorders, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Divya Mohan
- Veterinary Surgeon, Animal Husbandry Department, Government of Kerala, India
| | - Amit Verma
- Veterinary Officer, Veterinary Hospital, Sirmour, Himachal Pradesh, India
| | - G Taru Sharma
- Division of Physiology & Climatology, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| |
Collapse
|
10
|
Zhang T, Chen J, Zhang J, Guo YT, Zhou X, Li MW, Zheng ZZ, Zhang TZ, Murphy RW, Nevo E, Shi P. Phenotypic and genomic adaptations to the extremely high elevation in plateau zokor (Myospalax baileyi). Mol Ecol 2021; 30:5765-5779. [PMID: 34510615 DOI: 10.1111/mec.16174] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 08/07/2021] [Accepted: 08/31/2021] [Indexed: 12/31/2022]
Abstract
The evolutionary outcomes of high elevation adaptation have been extensively described. However, whether widely distributed high elevation endemic animals adopt uniform mechanisms during adaptation to different elevational environments remains unknown, especially with respect to extreme high elevation environments. To explore this, we analysed the phenotypic and genomic data of seven populations of plateau zokor (Myospalax baileyi) along elevations ranging from 2,700 to 4,300 m. Based on whole-genome sequencing data and demographic reconstruction of the evolutionary history, we show that two populations of plateau zokor living at elevations exceeding 3,700 m diverged from other populations nearly 10,000 years ago. Further, phenotypic comparisons reveal stress-dependent adaptation, as two populations living at elevations exceeding 3,700 m have elevated ratios of heart mass to body mass relative to other populations, and the highest population (4,300 m) displays alterations in erythrocytes. Correspondingly, genomic analysis of selective sweeps indicates that positive selection might contribute to the observed phenotypic alterations in these two extremely high elevation populations, with the adaptive cardiovascular phenotypes of both populations possibly evolving under the functional constrains of their common ancestral population. Taken together, phenotypic and genomic evidence demonstrates that heterogeneous stressors impact adaptations to extreme elevations and reveals stress-dependent and genetically constrained adaptation to hypoxia, collectively providing new insights into the high elevation adaptation.
Collapse
Affiliation(s)
- Tao Zhang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China
| | - Jie Chen
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China
| | - Jia Zhang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Yuan-Ting Guo
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China
| | - Xin Zhou
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China
| | - Meng-Wen Li
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Zhi-Zhong Zheng
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China
| | - Tong-Zuo Zhang
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, China
| | - Robert W Murphy
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,Centre for Biodiversity and Conservation Biology, Royal Ontario Museum, Toronto, ON, Canada
| | - Eviatar Nevo
- Institute of Evolution, University of Haifa, Haifa, Israel
| | - Peng Shi
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,School of Future Technology, University of Chinese Academy of Sciences, Beijing, China.,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, China
| |
Collapse
|
11
|
Henao-Restrepo J, Caro-Urrego YA, Barrera-Arenas LM, Arango-Viana JC, Bermudez-Munoz M. Expression of activator proteins of SHH/GLI and PI3K/Akt/mTORC1 signaling pathways in human gliomas is associated with high grade tumors. Exp Mol Pathol 2021; 122:104673. [PMID: 34371011 DOI: 10.1016/j.yexmp.2021.104673] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 07/25/2021] [Accepted: 08/03/2021] [Indexed: 01/15/2023]
Abstract
Recent findings have demonstrated a synergic crosstalk between SHH/GLI and PI3K/Akt/mTORC1 signaling in glioblastoma progression cells in vitro and in tumors in mice, but it is not known if this also occurs in human gliomas. We then aimed to investigate the expression of key proteins of these pathways in different human gliomas. The expression of PTEN, phospho-Akt (Ser473), phospho-S6K1 (Thr389), SHH, GLI1, GLI2 and GLI3 was assessed by immunohistochemistry in gliomas and in control brain tissues. The pattern of expression of each protein was established according to glioma type, glioma grade and to cell type; the relative expression of each protein was used to perform statistical analyses. We found that the expression of proteins of both signaling pathways differs between normal brain and glioma tissues. For instance, normal astrocytes had a different protein expression pattern compared with reactive and tumoral astrocytes. Interestingly, we detected a recurrent pattern of expression of GLI3 in oligodendrocytes and of phospho-S6K1 in mitotic neoplastic cells. We also identified differences of cell signaling according to glioma type: oligodendrogliomas and ependymomas are related with the expression of SHH/GLI proteins. Finally, we detected that high grade gliomas statistically correlate with the expression of GLI1 and GLI2, and that GLI1, GLI2, phospho-Akt and phospho-S6K1 are more expressed in patients with less survival, suggesting that activation of these cell signaling influences glioma outcome and patient survival. In summary, our results show that proteins of PI3K/Akt/mTORC1 and SHH/GLI pathways are differentially expressed in human gliomas according to tumor type and grade, and suggest that the activation of these signaling networks is associated with glioma progression.
Collapse
Affiliation(s)
- Julián Henao-Restrepo
- Institute of Biology, Faculty of Exact and Natural Sciences, University of Antioquia, Calle 67 #53-108, 050010 Medellín, Colombia.
| | - Yudys Anggelly Caro-Urrego
- Department of Pathology, Faculty of Medicine, University of Antioquia, Cra. 51d #62-29, 050010 Medellín, Colombia
| | - Lina Marcela Barrera-Arenas
- Grupo de Investigaciones Biomédicas, Health Sciences Faculty, University Corporation Remington, Calle 51 #51-27, Medellín, Colombia.
| | - Juan Carlos Arango-Viana
- Department of Pathology, Faculty of Medicine, University of Antioquia, Cra. 51d #62-29, 050010 Medellín, Colombia.
| | - Maria Bermudez-Munoz
- Institute of Biology, Faculty of Exact and Natural Sciences, University of Antioquia, Calle 67 #53-108, 050010 Medellín, Colombia.
| |
Collapse
|
12
|
Guimbal S, Cornuault L, Rouault P, Hollier PL, Chapouly C, Bats ML, Imbault J, Gadeau AP, Couffinhal T, Renault MA. Mast Cells Are the Trigger of Small Vessel Disease and Diastolic Dysfunction in Diabetic Obese Mice. Arterioscler Thromb Vasc Biol 2021; 41:e193-e207. [PMID: 33567863 DOI: 10.1161/atvbaha.121.315900] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
[Figure: see text].
Collapse
MESH Headings
- Animals
- Cell Degranulation
- Cells, Cultured
- Coronary Vessels/immunology
- Coronary Vessels/metabolism
- Coronary Vessels/pathology
- Diabetes Mellitus/genetics
- Diabetes Mellitus/immunology
- Diabetes Mellitus/metabolism
- Diastole
- Disease Models, Animal
- Female
- Heart Failure/etiology
- Heart Failure/immunology
- Heart Failure/metabolism
- Heart Failure/physiopathology
- Histamine Release
- Humans
- Mast Cells/immunology
- Mast Cells/metabolism
- Mast Cells/pathology
- Mice, Knockout
- Myocardium/immunology
- Myocardium/metabolism
- Myocardium/pathology
- Obesity/complications
- Obesity/immunology
- Obesity/metabolism
- Receptors, Leptin/deficiency
- Receptors, Leptin/genetics
- Ventricular Dysfunction, Left/etiology
- Ventricular Dysfunction, Left/immunology
- Ventricular Dysfunction, Left/metabolism
- Ventricular Dysfunction, Left/physiopathology
- Ventricular Function, Left
- Mice
Collapse
Affiliation(s)
- Sarah Guimbal
- University of Bordeaux, Inserm, Biology of Cardiovascular Diseases, U1034, CHU de Bordeaux, Pessac, France
| | - Lauriane Cornuault
- University of Bordeaux, Inserm, Biology of Cardiovascular Diseases, U1034, CHU de Bordeaux, Pessac, France
| | - Paul Rouault
- University of Bordeaux, Inserm, Biology of Cardiovascular Diseases, U1034, CHU de Bordeaux, Pessac, France
| | - Pierre-Louis Hollier
- University of Bordeaux, Inserm, Biology of Cardiovascular Diseases, U1034, CHU de Bordeaux, Pessac, France
| | - Candice Chapouly
- University of Bordeaux, Inserm, Biology of Cardiovascular Diseases, U1034, CHU de Bordeaux, Pessac, France
| | - Marie-Lise Bats
- University of Bordeaux, Inserm, Biology of Cardiovascular Diseases, U1034, CHU de Bordeaux, Pessac, France
| | - Julien Imbault
- University of Bordeaux, Inserm, Biology of Cardiovascular Diseases, U1034, CHU de Bordeaux, Pessac, France
| | - Alain-Pierre Gadeau
- University of Bordeaux, Inserm, Biology of Cardiovascular Diseases, U1034, CHU de Bordeaux, Pessac, France
| | - Thierry Couffinhal
- University of Bordeaux, Inserm, Biology of Cardiovascular Diseases, U1034, CHU de Bordeaux, Pessac, France
| | - Marie-Ange Renault
- University of Bordeaux, Inserm, Biology of Cardiovascular Diseases, U1034, CHU de Bordeaux, Pessac, France
| |
Collapse
|
13
|
miR-125b acts as anti-fibrotic therapeutic target through regulating Gli3 in vivo and in vitro. Ann Hepatol 2020; 18:825-832. [PMID: 31548167 DOI: 10.1016/j.aohep.2019.06.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Revised: 06/03/2019] [Accepted: 03/26/2019] [Indexed: 02/04/2023]
Abstract
INTRODUCTION AND OBJECTIVES Liver fibrosis is a major characteristic of most chronic liver diseases which leads to accumulation of extracellular matrix (ECM) proteins. Hedgehog (Hh) pathway activated by Gli genes participated in the pathogenesis of liver fibrosis. However, the regulatory role of miR-125b in liver fibrosis via targeting Gli genes remains unknown. MATERIALS AND METHODS RT-qPCR and western blot were employed to the expression levels of mRNA and protein, respectively. The fibrosis level of liver tissue was determined by Masson's trichrome staining. The interaction between miR-125b and Gli3 was tested by luciferase reporter assay. In addition, LX2 cells were activated and CCl4-induced rat model was used in this study. RESULTS miR-125b was significantly declined in serum samples of the clinical liver fibrosis patient, activated LX2 cells and the liver tissues of the CCl4-induced rat model. Furthermore, in cellular level, the alpha-smooth muscle actin (α-SMA) and Albumin expressions were ascending and descending in LX2 cells, respectively, with the decline of miR-125b. However, when transfecting with miR-125b mimic, the expressions of α-SMA and Albumin was reversed and Gli3 expression was notably repressed in LX2 cells. The target interaction between miR-125b and Gli3 was determined by dual-luciferase assays. It was further discovered that the changes of α-SMA, Albumin, and Gli3 were similar to the expression trend in LX2 cells with miR-125b mimic transfection. CONCLUSION These results suggested that miR-125b might be protective against liver fibrosis via regulating Gli3 and it might be a promising target in the development of novel therapies to treat pathological fibrotic disorders.
Collapse
|
14
|
Matissek SJ, Elsawa SF. GLI3: a mediator of genetic diseases, development and cancer. Cell Commun Signal 2020; 18:54. [PMID: 32245491 PMCID: PMC7119169 DOI: 10.1186/s12964-020-00540-x] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 02/27/2020] [Indexed: 02/07/2023] Open
Abstract
The transcription factor GLI3 is a member of the Hedgehog (Hh/HH) signaling pathway that can exist as a full length (Gli3-FL/GLI3-FL) or repressor (Gli3-R/GLI3-R) form. In response to HH activation, GLI3-FL regulates HH genes by targeting the GLI1 promoter. In the absence of HH signaling, GLI3 is phosphorylated leading to its partial degradation and the generation of GLI3-R which represses HH functions. GLI3 is also involved in tissue development, immune cell development and cancer. The absence of Gli3 in mice impaired brain and lung development and GLI3 mutations in humans are the cause of Greig cephalopolysyndactyly (GCPS) and Pallister Hall syndromes (PHS). In the immune system GLI3 regulates B, T and NK-cells and may be involved in LPS-TLR4 signaling. In addition, GLI3 was found to be upregulated in multiple cancers and was found to positively regulate cancerous behavior such as anchorage-independent growth, angiogenesis, proliferation and migration with the exception in acute myeloid leukemia (AML) and medulloblastoma where GLI plays an anti-cancerous role. Finally, GLI3 is a target of microRNA. Here, we will review the biological significance of GLI3 and discuss gaps in our understanding of this molecule. Video Abstract.
Collapse
Affiliation(s)
- Stephan J. Matissek
- Department of Molecular, Cellular and Biomedical Sciences, University of New Hampshire, 46 College Rd Rudman 291, Durham, NH 03824 USA
| | - Sherine F. Elsawa
- Department of Molecular, Cellular and Biomedical Sciences, University of New Hampshire, 46 College Rd Rudman 291, Durham, NH 03824 USA
| |
Collapse
|
15
|
Giarretta I, Gaetani E, Bigossi M, Tondi P, Asahara T, Pola R. The Hedgehog Signaling Pathway in Ischemic Tissues. Int J Mol Sci 2019; 20:ijms20215270. [PMID: 31652910 PMCID: PMC6862352 DOI: 10.3390/ijms20215270] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 10/22/2019] [Indexed: 12/19/2022] Open
Abstract
Hedgehog (Hh) proteins are prototypical morphogens known to regulate epithelial/mesenchymal interactions during embryonic development. In addition to its pivotal role in embryogenesis, the Hh signaling pathway may be recapitulated in post-natal life in a number of physiological and pathological conditions, including ischemia. This review highlights the involvement of Hh signaling in ischemic tissue regeneration and angiogenesis, with particular attention to the heart, the brain, and the skeletal muscle. Updated information on the potential role of the Hh pathway as a therapeutic target in the ischemic condition is also presented.
Collapse
Affiliation(s)
- Igor Giarretta
- Department of Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| | - Eleonora Gaetani
- Department of Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| | - Margherita Bigossi
- Department of Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| | - Paolo Tondi
- Department of Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| | - Takayuki Asahara
- Department of Regenerative Medicine Science, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa 259-1193, Japan.
| | - Roberto Pola
- Department of Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| |
Collapse
|
16
|
Role of Hedgehog Signaling in Vasculature Development, Differentiation, and Maintenance. Int J Mol Sci 2019; 20:ijms20123076. [PMID: 31238510 PMCID: PMC6627637 DOI: 10.3390/ijms20123076] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 06/17/2019] [Accepted: 06/20/2019] [Indexed: 12/16/2022] Open
Abstract
The role of Hedgehog (Hh) signaling in vascular biology has first been highlighted in embryos by Pepicelli et al. in 1998 and Rowitch et al. in 1999. Since then, the proangiogenic role of the Hh ligands has been confirmed in adults, especially under pathologic conditions. More recently, the Hh signaling has been proposed to improve vascular integrity especially at the blood–brain barrier (BBB). However, molecular and cellular mechanisms underlying the role of the Hh signaling in vascular biology remain poorly understood and conflicting results have been reported. As a matter of fact, in several settings, it is currently not clear whether Hh ligands promote vessel integrity and quiescence or destabilize vessels to promote angiogenesis. The present review relates the current knowledge regarding the role of the Hh signaling in vasculature development, maturation and maintenance, discusses the underlying proposed mechanisms and highlights controversial data which may serve as a guideline for future research. Most importantly, fully understanding such mechanisms is critical for the development of safe and efficient therapies to target the Hh signaling in both cancer and cardiovascular/cerebrovascular diseases.
Collapse
|
17
|
Li L, Zhang L, Zhao X, Cao J, Li J, Chu G. Downregulation of miR-152 contributes to the progression of liver fibrosis via targeting Gli3 in vivo and in vitro. Exp Ther Med 2019; 18:425-434. [PMID: 31258681 PMCID: PMC6566101 DOI: 10.3892/etm.2019.7595] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 01/24/2019] [Indexed: 12/13/2022] Open
Abstract
The Gli family is known to be required for the activation of hedgehog signalling, which participates in the pathogenesis of liver fibrosis. The aim of the present study was to identify the association between microRNA (miR)-152 and GLI family zinc finger 3 (Gli3) and their roles in liver fibrosis. In a carbon tetrachloride (CCl4)-treated rat model, fibrogenesis-associated indexes, including hydroxyproline content, collagen deposition, and α-smooth muscle actin (α-SMA) and albumin expression, were examined in in vivo and in vitro models. The expression of miR-152 and Gli3 in cells and tissues was determined by reverse transcription quantitative polymerase chain reaction and western blot analysis. The interaction of Gli3 and miR-152 was evaluated by bioinformatical analysis and a dual-luciferase reporter assay. The results demonstrated that miR-152 was significantly downregulated in serum samples from clinical patients, liver tissues from CCl4-treated rats and activated LX2 cells. Furthermore, at the cellular level, the mRNA and protein expression levels of α-SMA and albumin were increased and decreased, respectively, in LX2 cells. Nevertheless, following transfection with an miR-152 mimic, the expression levels of α-SMA and albumin were reversed, and Gli3 expression was notably decreased in LX2 cells. Additionally, the target interaction between miR-152 and Gli3 was demonstrated. Finally, an miR-152 mimic was introduced into the rat model and additionally demonstrated that the changes in α-SMA, albumin and Gli3 expression levels were similar to the expression pattern in LX2 cells following miR-152 mimic transfection. These data provided insight into the potential function of miR-152 as an anti-fibrotic therapy through the modulation of Gli3.
Collapse
Affiliation(s)
- Li Li
- Department of Hepatobiliary Surgery, First People's Hospital of Kunming City, Kunming, Yunnan 650034, P.R. China
| | - Lei Zhang
- Department of Hepatobiliary Surgery, First People's Hospital of Kunming City, Kunming, Yunnan 650034, P.R. China
| | - Xiongqi Zhao
- Department of Hepatobiliary Surgery, First People's Hospital of Kunming City, Kunming, Yunnan 650034, P.R. China
| | - Jun Cao
- Department of Hepatobiliary Surgery, First People's Hospital of Kunming City, Kunming, Yunnan 650034, P.R. China
| | - Jingfeng Li
- Department of Hepatobiliary Surgery, First People's Hospital of Kunming City, Kunming, Yunnan 650034, P.R. China
| | - Guang Chu
- Department of Hepatobiliary Surgery, First People's Hospital of Kunming City, Kunming, Yunnan 650034, P.R. China
| |
Collapse
|
18
|
Niu C, Chen Z, Kim KT, Sun J, Xue M, Chen G, Li S, Shen Y, Zhu Z, Wang X, Liang J, Jiang C, Cong W, Jin L, Li X. Metformin alleviates hyperglycemia-induced endothelial impairment by downregulating autophagy via the Hedgehog pathway. Autophagy 2019; 15:843-870. [PMID: 30653446 PMCID: PMC6526809 DOI: 10.1080/15548627.2019.1569913] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Studies regarding macroautophagic/autophagic regulation in endothelial cells (ECs) under diabetic conditions are very limited. Clinical evidence establishes an endothelial protective effect of metformin, but the underlying mechanisms remain unclear. We aimed to investigate whether metformin exerts its protective role against hyperglycemia-induced endothelial impairment through the autophagy machinery. db/db mice were treated with intravitreal metformin injections. Human umbilical vein endothelial cells (HUVECs) were cultured either in normal glucose (NG, 5.5 mM) or high glucose (HG, 33 mM) medium in the presence or absence of metformin for 72 h. We observed an obvious inhibition of hyperglycemia-triggered autophagosome synthesis in both the diabetic retinal vasculature and cultured HUVECs by metformin, along with restoration of hyperglycemia-impaired Hedgehog (Hh) pathway activity. Specifically, deletion of ATG7 in retinal vascular ECs of db/db mice and cultured HUVECs indicated a detrimental role of autophagy in hyperglycemia-induced endothelial dysfunction. Pretreatment with GANT61, a Hh pathway inhibitor, abolished the metformin-mediated downregulation of autophagy and endothelial protective action. Furthermore, GLI-family (transcription factors of the Hh pathway) knockdown in HUVECs and retinal vasculature revealed that downregulation of hyperglycemia-activated autophagy by the metformin-mediated Hh pathway activation was GLI1 dependent. Mechanistically, GLI1 knockdown-triggered autophagy was related to upregulation of BNIP3, which subsequently disrupted the association of BECN1/Beclin 1 and BCL2. The role of BNIP3 in BECN1 dissociation from BCL2 was further confirmed by BNIP3 overexpression or BNIP3 RNAi. Taken together, the endothelial protective effect of metformin under hyperglycemia conditions could be partly attributed to its role in downregulating autophagy via Hh pathway activation. Abbreviations: 3-MA = 3-methyladenine; 8×GLI BS-FL = 8×GLI-binding site firefly luciferase; AAV = adeno-associated virus; AAV-Cdh5-sh-Atg7 = AAV vectors carrying shRNA against murine Atg7 under control of murine Cdh5 promoter; AAV-Cdh5-sh-Gli1 = AAV vectors carrying shRNA against murine Gli1 under control of murine Cdh5 promoter; AAV-Cdh5-Gli1 = AAV vectors carrying murine Gli1 cDNA under the control of murine Cdh5 core promoter; ACAC = acetyl-CoA carboxylase; Ad-BNIP3 = adenoviruses harboring human BNIP3`; Ad-GLI1 = adenoviruses harboring human GLI1; Ad-sh-ATG7 = adenoviruses harboring shRNA against human ATG7; Ad-sh-BNIP3 = adenoviruses harboring shRNA against human BNIP3; Ad-sh-GLI = adenoviruses harboring shRNA against human GLI; AGEs = advanced glycation end products; ATG = autophagy-related; atg7flox/flox mice = mice bearing an Atg7flox allele, in which exon 14 of the Atg7 gene is flanked by 2 loxP sites; BafA1 = bafilomycin A1; BECN1 = beclin 1; CDH5/VE-cadherin = cadherin 5; CASP3 = caspase 3; CASP8 = caspase 8; CASP9 = caspase 9; ECs = endothelial cells; GAPDH = glyceraldehyde-3-phosphate dehydrogenase; GCL = ganglion cell layer; GFP-LC3B = green fluorescent protein labelled LC3B; HG = high glucose; Hh = Hedgehog; HHIP = hedgehog interacting protein; HUVECs = human umbilical vein endothelial cells; IB4 = isolectin B4; INL = inner nuclear layer; i.p. = intraperitoneal; MAP1LC3/LC3 = microtubule-associated protein 1 light chain 3; MAN = mannitol; MET = metformin; NG = normal glucose; ONL = outer nuclear layer; p-ACAC = phosphorylated acetyl-CoA carboxylase; PECAM1/CD31= platelet/endothelial cell adhesion molecule 1; PRKAA1/2 = protein kinase AMP-activated catalytic subunits alpha 1/2; p-PRKAA1/2 = phosphorylated PRKAA1/2; PTCH1 = patched 1; RAPA = rapamycin; RL = Renilla luciferase; SHH = sonic hedgehog; shRNA = short hairpin RNA; sh-PRKAA1/2 = short hairpin RNA against human PRKAA1/2; scrambled shRNA = the scrambled short hairpin RNA serves as a negative control for the target-specific short hairpin RNA, which has the same nucleotide composition as the input sequence and has no match with any mRNA of the selected organism database; SMO = smoothened, frizzled class receptor; sqRT-PCR = semi-quantitative RT-PCR; TEK/Tie2 = TEK receptor tyrosine kinase; Tek-Cre (+) mice = a mouse strain expressing Cre recombinase under the control of the promoter/enhancer of Tek, in a pan-endothelial fashion; TUNEL = terminal deoxynucleotidyl transferase dUTP-mediated nick-end labeling.
Collapse
Affiliation(s)
- Chao Niu
- Pediatric Research Institute, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, P.R. China,School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, P.R. China
| | - Zhiwei Chen
- Pediatric Research Institute, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, P.R. China,School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, P.R. China
| | - Kyoung Tae Kim
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, P.R. China
| | - Jia Sun
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, P.R. China
| | - Mei Xue
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, P.R. China
| | - Gen Chen
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, P.R. China
| | - Santie Li
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, P.R. China
| | - Yingjie Shen
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, P.R. China
| | - Zhongxin Zhu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, P.R. China
| | - Xu Wang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, P.R. China
| | - Jiaojiao Liang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, P.R. China
| | - Chao Jiang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, P.R. China,CONTACT Litai Jin ; Weitao Cong ; Chao Jiang School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, P.R. China
| | - Weitao Cong
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, P.R. China,CONTACT Litai Jin ; Weitao Cong ; Chao Jiang School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, P.R. China
| | - Litai Jin
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, P.R. China,CONTACT Litai Jin ; Weitao Cong ; Chao Jiang School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, P.R. China
| | - Xiaokun Li
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, P.R. China
| |
Collapse
|
19
|
Targeting GLI Transcription Factors in Cancer. Molecules 2018; 23:molecules23051003. [PMID: 29695137 PMCID: PMC6100584 DOI: 10.3390/molecules23051003] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 04/19/2018] [Accepted: 04/20/2018] [Indexed: 12/22/2022] Open
Abstract
Aberrant activation of hedgehog (Hh) signaling has been observed in a wide variety of tumors and accounts for more than 25% of human cancer deaths. Inhibitors targeting the Hh signal transducer Smoothened (SMO) are widely used and display a good initial efficacy in patients suffering from basal cell carcinoma (BCC); however, a large number of patients relapse. Though SMO mutations may explain acquired therapy resistance, a growing body of evidence suggests that the non-canonical, SMO-independent activation of the Hh pathway in BCC patients can also account for this adverse effect. In this review, we highlight the importance of glioma-associated oncogene (GLI) transcription factors (the main downstream effectors of the canonical and the non-canonical Hh cascade) and their putative role in the regulation of multiple oncogenic signaling pathways. Moreover, we discuss the contribution of the Hh signaling to malignant transformation and propose GLIs as central hubs in tumor signaling networks and thus attractive molecular targets in anti-cancer therapies.
Collapse
|
20
|
Guerra AD, Rose WE, Hematti P, Kao WJ. Minocycline modulates NFκB phosphorylation and enhances antimicrobial activity against Staphylococcus aureus in mesenchymal stromal/stem cells. Stem Cell Res Ther 2017; 8:171. [PMID: 28732530 PMCID: PMC5521110 DOI: 10.1186/s13287-017-0623-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 06/05/2017] [Accepted: 06/28/2017] [Indexed: 02/06/2023] Open
Abstract
Background Mesenchymal stromal/stem cells (MSCs) have demonstrated pro-healing properties due to their anti-inflammatory, angiogenic, and even antibacterial properties. We have shown previously that minocycline enhances the wound healing phenotype of MSCs, and MSCs encapsulated in poly(ethylene glycol) and gelatin-based hydrogels with minocycline have antibacterial properties against Staphylococcus aureus (SA). Here, we investigated the signaling pathway that minocycline modulates in MSCs which results in their enhanced wound healing phenotype and determined whether preconditioning MSCs with minocycline has an effect on antimicrobial activity. We further investigated the in-vivo antimicrobial efficacy of MSC and antibiotic-loaded hydrogels in inoculated full-thickness cutaneous wounds. Methods Modulation of cell signaling pathways in MSCs with minocycline was analyzed via western blot, immunofluorescence, and ELISA. Antimicrobial efficacy of MSCs pretreated with minocycline was determined by direct and transwell coculture with SA. MSC viability after SA coculture was determined via a LIVE/DEAD® stain. Internalization of SA by MSCs pretreated with minocycline was determined via confocal imaging. All protein and cytokine analysis was done via ELISA. The in-vivo antimicrobial efficacy of MSC and antibiotic-loaded hydrogels was determined in Sprague–Dawley rats inoculated with SA. Two-way ANOVA for multiple comparisons was used with Bonferroni test assessment and an unpaired two-tailed Student’s t test was used to determine p values for all assays with multiple or two conditions, respectively. Results Minocycline leads to the phosphorylation of transcriptional nuclear factor-κB (NFκB), but not c-Jun NH2-terminal kinase (JNK) or mitogen-activated protein kinase (ERK). Inhibition of NFκB activation prevented the minocycline-induced increase in VEGF secretion. Preconditioning of MSCs with minocycline led to a reduced production of the antimicrobial peptide LL-37, but enhanced antimicrobial activity against SA via an increased production of IL-6 and SA internalization. MSC and antibiotic-loaded hydrogels reduced SA bioburden in inoculated wounds over 3 days and accelerated reepithelialization. Conclusions Minocycline modulates the NFκB pathway in MSCs that leads to an enhanced production of IL-6 and internalization of SA. This mechanism may have contributed to the in-vivo antibacterial efficacy of MSC and antibiotic-loaded hydrogels. Electronic supplementary material The online version of this article (doi:10.1186/s13287-017-0623-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Alberto Daniel Guerra
- School of Pharmacy, Division of Pharmaceutical Sciences, Pharmacy Practice Division, University of Wisconsin-Madison, 777 Highland Avenue, 7123 Rennebohm Hall, Madison, WI, 53705, USA
| | - Warren E Rose
- School of Pharmacy, Division of Pharmaceutical Sciences, Pharmacy Practice Division, University of Wisconsin-Madison, 777 Highland Avenue, 7123 Rennebohm Hall, Madison, WI, 53705, USA
| | - Peiman Hematti
- School of Medicine and Public Health, Department of Medicine, Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, 1685 Highland Avenue, Madison, WI, 53705, USA
| | - W John Kao
- School of Pharmacy, Division of Pharmaceutical Sciences, Pharmacy Practice Division, University of Wisconsin-Madison, 777 Highland Avenue, 7123 Rennebohm Hall, Madison, WI, 53705, USA. .,College of Engineering, Department of Biomedical Engineering, University of Wisconsin-Madison, 1415 Engineering Drive, Madison, WI, 53706, USA. .,School of Medicine and Public Health, Department of Surgery, University of Wisconsin-Madison, 1685 Highland Avenue, Madison, WI, 53705, USA. .,Present Address: 10/F Knowles Building, Pokfulam, Hong Kong.
| |
Collapse
|
21
|
Hh signaling in regeneration of the ischemic heart. Cell Mol Life Sci 2017; 74:3481-3490. [PMID: 28523343 PMCID: PMC5589787 DOI: 10.1007/s00018-017-2534-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2016] [Revised: 04/10/2017] [Accepted: 05/02/2017] [Indexed: 12/23/2022]
Abstract
Myocardial infarction (MI) is caused by the occlusion of a coronary artery due to underlying atherosclerosis complicated by localized thrombosis. The blockage of blood flow leads to cardiomyocyte (CM) death in the infarcted area. Adult mammalian cardiomyocytes have little capacity to proliferate in response to injury; however, some pathways active during embryogenesis and silent during adult life are recruited in response to tissue injury. One such example is hedgehog (Hh) signaling. Hh is involved in the embryonic development of the heart and coronary vascular system. Pathological conditions including ischemia activate Hh signaling in adult tissues. This review highlights the involvement of Hh signaling in ischemic tissue regeneration with a particular emphasis on heart regeneration and discusses its potential role as a therapeutic agent.
Collapse
|
22
|
Guerra AD, Rose WE, Hematti P, Kao WJ. Minocycline enhances the mesenchymal stromal/stem cell pro-healing phenotype in triple antimicrobial-loaded hydrogels. Acta Biomater 2017; 51:184-196. [PMID: 28069512 PMCID: PMC5704963 DOI: 10.1016/j.actbio.2017.01.021] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 01/04/2017] [Accepted: 01/05/2017] [Indexed: 01/12/2023]
Abstract
Mesenchymal stromal/stem cells (MSCs) have demonstrated pro-healing properties including an anti-inflammatory cytokine profile and the promotion of angiogenesis via expression of growth factors in pre-clinical models. MSCs encapsulated in poly(ethylene glycol) diacrylate (PEGdA) and thiolated gelatin poly(ethylene glycol) (Gel-PEG-Cys) crosslinked hydrogels have led to controlled cellular presentation at wound sites with favorable wound healing outcomes. However, the therapeutic potential of MSC-loaded hydrogels may be limited by non-specific protein adsorption on the delivery matrix that could facilitate the initial adhesion of microorganisms and subsequent virulent biofilm formation. Antimicrobials loaded concurrently in the hydrogels with MSCs could reduce microbial bioburden and promote healing, but the antimicrobial effect on the MSC wound healing capacity and the antibacterial efficacy of the hydrogels is unknown. We demonstrate that minocycline specifically induces a favorable change in MSC migration capacity, proliferation, gene expression, extracellular matrix (ECM) attachment, and adhesion molecule and growth factor release with subsequent increased angiogenesis. We then demonstrate that hydrogels loaded with MSCs, minocycline, vancomycin, and linezolid can significantly decrease bacterial bioburden. Our study suggests that minocycline can serve as a dual mechanism for the regenerative capacity of MSCs and the reduction of bioburden in triple antimicrobial-loaded hydrogels. STATEMENT OF SIGNIFICANCE Wound healing is a complex biological process that can be hindered by bacterial infection, excessive inflammation, and inadequate microvasculature. In this study, we develop a new formulation of poly(ethylene glycol) diacrylate and thiolated gelatin poly(ethylene glycol) crosslinked hydrogels loaded with minocycline, vancomycin, linezolid, and mesenchymal stromal/stem cells that induces a favorable wound healing phenotype in mesenchymal stromal/stem cells and prevents bacterial bioburden on the hydrogel. This combinatorial approach to biomaterial development has the potential to impact wound healing for contaminated full thickness cutaneous wounds.
Collapse
Affiliation(s)
- Alberto Daniel Guerra
- School of Pharmacy, Division of Pharmaceutical Sciences, Pharmacy Practice Division, University of Wisconsin-Madison, 777 Highland Avenue, Madison, WI 53705, USA.
| | - Warren E Rose
- School of Pharmacy, Division of Pharmaceutical Sciences, Pharmacy Practice Division, University of Wisconsin-Madison, 777 Highland Avenue, Madison, WI 53705, USA.
| | - Peiman Hematti
- School of Medicine and Public Health, Department of Medicine, Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, 1685 Highland Avenue, Madison, WI 53705, USA.
| | - W John Kao
- School of Pharmacy, Division of Pharmaceutical Sciences, Pharmacy Practice Division, University of Wisconsin-Madison, 777 Highland Avenue, Madison, WI 53705, USA; College of Engineering, Department of Biomedical Engineering, University of Wisconsin-Madison, 1415 Engineering Drive, Madison, WI 53706, USA; School of Medicine and Public Health, Department of Surgery, University of Wisconsin-Madison, 1685 Highland Avenue, Madison, WI 53705, USA.
| |
Collapse
|
23
|
Lisovsky A, Sefton MV. Shh pathway in wounds in non-diabetic Shh-Cre-eGFP/Ptch1-LacZ mice treated with MAA beads. Biomaterials 2016; 102:198-208. [DOI: 10.1016/j.biomaterials.2016.06.027] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 06/06/2016] [Accepted: 06/12/2016] [Indexed: 01/20/2023]
|
24
|
Lisovsky A, Zhang DKY, Sefton MV. Effect of methacrylic acid beads on the sonic hedgehog signaling pathway and macrophage polarization in a subcutaneous injection mouse model. Biomaterials 2016; 98:203-14. [PMID: 27264502 DOI: 10.1016/j.biomaterials.2016.04.033] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 04/14/2016] [Accepted: 04/20/2016] [Indexed: 12/23/2022]
Abstract
Poly(methacrylic acid-co-methyl methacrylate) (MAA) beads promote a vascular regenerative response when used in diabetic wound healing. Previous studies reported that MAA beads modulated the expression of sonic hedgehog (Shh) and inflammation related genes in diabetic wounds. The aim of this work was to follow up on these observations in a subcutaneous injection model to study the host response in the absence of the confounding factors of diabetic wound healing. In this model, MAA beads improved vascularization in healthy mice of both sexes compared to control poly(methyl methacrylate) (MM) beads, with a stronger effect seen in males than females. MAA-induced vessels were perfusable, as evidenced from the CLARITY-processed images. In Shh-Cre-eGFP/Ptch1-LacZ non-diabetic transgenic mice, the increased vessel formation was accompanied by a higher density of cells expressing GFP (Shh) and β-Gal (patched 1, Ptch1) suggesting MAA enhanced the activation of the Shh pathway. Ptch1 is the Shh receptor and a target of the pathway. MAA beads also modulated the inflammatory cell infiltrate in CD1 mice: more neutrophils and more macrophages were noted with MAA relative to MM beads at days 1 and 7, respectively. In addition, MAA beads biased macrophages towards a MHCII-CD206+ ("M2") polarization state. This study suggests that the Shh pathway and an altered inflammatory response are two elements of the complex mechanism whereby MAA-based biomaterials effect vascular regeneration.
Collapse
Affiliation(s)
- Alexandra Lisovsky
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Suite 407, Toronto, Ontario, Canada M5S 3G9
| | - David K Y Zhang
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Suite 407, Toronto, Ontario, Canada M5S 3G9
| | - Michael V Sefton
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Suite 407, Toronto, Ontario, Canada M5S 3G9; Department of Chemical Engineering and Applied Chemistry, University of Toronto, 164 College Street, Suite 407, Toronto, Ontario, Canada M5S 3G9.
| |
Collapse
|
25
|
Lisovsky A, Chamberlain MD, Wells LA, Sefton MV. Cell Interactions with Vascular Regenerative MAA-Based Materials in the Context of Wound Healing. Adv Healthc Mater 2015; 4:2375-87. [PMID: 26010569 DOI: 10.1002/adhm.201500192] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 04/13/2015] [Indexed: 12/19/2022]
Abstract
In diabetic patients the development of chronic non-healing wounds is a common complication. A methacrylic acid-based biomaterial is a vascular regenerative material that enhances diabetic healing without the use of cells or growth factors. The bioactive nature of this material is thought to be associated with its anionic charge or surface chemistry. Contact between the methacrylic acid-based biomaterial and tissue begins with protein (including complement) adsorption and is followed by interaction of the biomaterial with resident and infiltrating cells in the wound bed (e.g., macrophages and endothelial cells). This results in changes to their surface receptors to activate phosphorylation cascades that lead to differential activation of signalling pathways such as those involving osteopontin and sonic hedgehog. These changes modulate the phenotype of the cells in the wound bed, eventually improving vessel formation and wound healing. Understanding the molecular and cellular mechanisms will have broad implications for biomaterials, not just the methacrylic acid-based material, and will facilitate the advancement of regenerative biomaterials for diverse applications.
Collapse
Affiliation(s)
- Alexandra Lisovsky
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; M5S 3G9 Canada
| | | | - Laura Anne Wells
- Department of Chemical Engineering; Queen's University; K7L 3N6 Canada
| | - Michael Vivian Sefton
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; M5S 3G9 Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; 164 College Street, Suite 407 Toronto Ontario M5S 3G9 Canada
| |
Collapse
|
26
|
Shin EH, Caterson EJ, Jackson WM, Nesti LJ. Quality of healing: defining, quantifying, and enhancing skeletal muscle healing. Wound Repair Regen 2015; 22 Suppl 1:18-24. [PMID: 24813360 DOI: 10.1111/wrr.12163] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Accepted: 01/24/2014] [Indexed: 12/12/2022]
Abstract
Skeletal muscle injury is common in everyday physical activity and athletics, as well as in orthopedic trauma and disease. The overall functional disability resulting from muscle injury is directly related to the intrinsic healing properties of muscle and extrinsic treatment options designed to maximize repair and/or regeneration of muscle tissue all while minimizing pathologic healing pathways. It is important to understand the injury and repair pathways in order to improve the speed and quality of recovery. Recent military conflicts in Iraq and Afghanistan have highlighted the importance of successfully addressing muscular injury and showed the need for novel treatment options that will maximize functional regeneration of the damaged tissue. These severe, wartime injuries, when juxtaposed to peacetime, sports-related injuries, provide us with interesting case examples of the two extreme forms of muscular damage. Comparing and contrasting the differences in these healing pathways will likely provide helpful cues that will help physicians recapitulate the near complete repair and regeneration in less traumatic injuries in addition to more severe cases.
Collapse
Affiliation(s)
- Emily H Shin
- Department of Orthopaedic Surgery, Walter Reed National Military Medical Center, Bethesda, Maryland; Clinical and Experimental Orthopaedics group, National Institutes of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland
| | | | | | | |
Collapse
|
27
|
Guerra AD, Cantu DA, Vecchi JT, Rose WE, Hematti P, Kao WJ. Mesenchymal Stromal/Stem Cell and Minocycline-Loaded Hydrogels Inhibit the Growth of Staphylococcus aureus that Evades Immunomodulation of Blood-Derived Leukocytes. AAPS JOURNAL 2015; 17:620-30. [PMID: 25716147 DOI: 10.1208/s12248-015-9728-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 01/28/2015] [Indexed: 01/03/2023]
Abstract
Mesenchymal stromal/stem cells (MSCs) have demonstrated favorable wound healing properties in addition to their differentiation capacity. MSCs encapsulated in biomaterials such as gelatin and polyethylene glycol (PEG) composite hydrogels have displayed an immunophenotype change that leads to the release of cytokines and growth factors to accelerate wound healing. However, therapeutic potential of implanted MSC-loaded hydrogels may be limited by non-specific protein adsorption that facilitates adhesion of bacterial pathogens such as planktonic Staphylococcus aureus (SA) to the surface with subsequent biofilm formation resistant to immune cell recognition and antibiotic activity. In this study, we demonstrate that blood-derived primary leukocytes and bone marrow-derived MSCs cannot inhibit colony-forming abilities of planktonic or biofilm-associated SA. However, we show that hydrogels loaded with MSCs and minocycline significantly inhibit colony-forming abilities of planktonic SA while maintaining MSC viability and multipotency. Our results suggest that minocycline and MSC-loaded hydrogels may decrease the bioburden of SA at implant sites in wounds, and may improve the wound healing capabilities of MSC-loaded hydrogels.
Collapse
Affiliation(s)
- Alberto Daniel Guerra
- School of Pharmacy, Division of Pharmaceutical Sciences, Pharmacy Practice Division, University of Wisconsin-Madison, 777 Highland Avenue, 7123 Rennebohm Hall, Madison, WI, 53705, USA
| | | | | | | | | | | |
Collapse
|
28
|
Wen SY, Lin Y, Yu YQ, Cao SJ, Zhang R, Yang XM, Li J, Zhang YL, Wang YH, Ma MZ, Sun WW, Lou XL, Wang JH, Teng YC, Zhang ZG. miR-506 acts as a tumor suppressor by directly targeting the hedgehog pathway transcription factor Gli3 in human cervical cancer. Oncogene 2015; 34:717-725. [PMID: 24608427 DOI: 10.1038/onc.2014.9] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Revised: 02/02/2014] [Accepted: 02/03/2014] [Indexed: 02/07/2023]
Abstract
Although significant advances have recently been made in the diagnosis and treatment of cervical carcinoma, the long-term survival rate for advanced cervical cancer remains low. Therefore, an urgent need exists to both uncover the molecular mechanisms and identify potential therapeutic targets for the treatment of cervical cancer. MicroRNAs (miRNAs) have important roles in cancer progression and could be used as either potential therapeutic agents or targets. miR-506 is a component of an X chromosome-linked miRNA cluster. The biological functions of miR-506 have not been well established. In this study, we found that miR-506 expression was downregulated in approximately 80% of the cervical cancer samples examined and inversely correlated with the expression of Ki-67, a marker of cell proliferation. Gain-of-function and loss-of-function studies in human cervical cancer, Caski and SiHa cells, demonstrated that miR-506 acts as a tumor suppressor by inhibiting cervical cancer growth in vitro and in vivo. Further studies showed that miR-506 induced cell cycle arrest at the G1/S transition, and enhanced apoptosis and chemosensitivity of cervical cancer cell. We subsequently identified Gli3, a hedgehog pathway transcription factor, as a direct target of miR-506 in cervical cancer. Furthermore, Gli3 silencing recapitulated the effects of miR-506, and reintroduction of Gli3 abrogated miR-506-induced cell growth arrest and apoptosis. Taken together, we conclude that miR-506 exerts its anti-proliferative function by directly targeting Gli3. This newly identified miR-506/Gli3 axis provides further insight into the pathogenesis of cervical cancer and indicates a potential novel therapeutic agent for the treatment of cervical cancer.
Collapse
Affiliation(s)
- S-Y Wen
- 1] Department of Obstetrics & Gynecology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, PR China [2] Department of Obstetrics & Gynecology, International Peace Maternity & Child Health Hospital of the China Welfare Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China [3] State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Y Lin
- Department of Obstetrics & Gynecology, International Peace Maternity & Child Health Hospital of the China Welfare Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Y-Q Yu
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, PR China
| | - S-J Cao
- Department of Obstetrics and Gynecology, Shanghai Songjiang District Central Hospital, Shanghai, PR China
| | - R Zhang
- Department of Obstetrics and Gynecology, Shanghai Fengxian District Central Hospital, Shanghai, PR China
| | - X-M Yang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - J Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Y-L Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Y-H Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - M-Z Ma
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - W-W Sun
- Department of Obstetrics and Gynecology, Shanghai Songjiang District Central Hospital, Shanghai, PR China
| | - X-L Lou
- Department of Obstetrics and Gynecology, Shanghai Songjiang District Central Hospital, Shanghai, PR China
| | - J-H Wang
- Department of Obstetrics and Gynecology, Southern Medical University, Guangzhou, PR China
| | - Y-C Teng
- Department of Obstetrics & Gynecology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, PR China
| | - Z-G Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| |
Collapse
|
29
|
Abstract
The extracellular matrix (ECM) is best known for its function as a structural scaffold for the tissue and more recently as a microenvironment to sequester growth factors and cytokines allowing for rapid and localized changes in their activity in the absence of new protein synthesis. In this review, we explore this and additional new aspects of ECM function in mediating cell-to-cell communications. Fibrillar and nonfibrillar components of ECM can limit and facilitate the transport of molecules through the extracellular space while also regulating interstitial hydrostatic pressure. In turn, transmembrane communications via molecules, such as ECM metalloproteinase inducer, thrombospondins, and integrins, can further mediate cell response to extracellular cues and affect ECM composition and tissue remodeling. Other means of cell-to-cell communication include extracellular microRNA transport and its contribution to gene expression in target cells and the nanotube formation between distant cells, which has recently emerged as a novel conduit for intercellular organelle sharing thereby influencing cell survival and function. The information summarized and discussed here are not limited to the cardiovascular ECM but encompass ECM in general with specific references to the cardiovascular system.
Collapse
Affiliation(s)
- Dong Fan
- From the Department of Physiology, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada (D.F., Z.K.); and Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands (E.E.C.)
| | | | | |
Collapse
|
30
|
Mesenchymal stem cells for chronic wounds therapy. Cell Tissue Bank 2014; 16:19-26. [PMID: 24651970 DOI: 10.1007/s10561-014-9440-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2014] [Accepted: 03/14/2014] [Indexed: 12/13/2022]
Abstract
Wound healing is a complex process that involves interaction of soluble mediators, extracellular matrix and infiltrating blood cells. Chronic and non-healing skin defects contribute significantly to morbidity and mortality of many patients. Recently, despite the current medical progress, the chronic and non-healing wounds still represent a serious medical problem. In many cases, conventional therapeutic approaches, such as dermal substitutes and growth factor therapy failed and do not produce the expected results, patients are exposed to a high risk of infection, sepsis or amputation. For that reason clinicians and researchers are forced to searching for alternative methods to induce healing process which may result into complete wound closure. Mesenchymal stem cells (MSCs) represent a unique tool of tissue engineering and regenerative medicine and a promising therapeutic strategy. Due to their unique biological properties, MSCs seem to be the perspective modality method for these patients. Many preclinical and clinical studies suggest the possibility of using these cells in tissue regeneration, healing acute and chronic wounds and scar remodelling. The objective of the present review is to summarize the current information and preclinical data about MSCs, their biological characteristics and mode of action during regenerative and healing processes, as well as their clinical application in chronic wounds treatment.
Collapse
|
31
|
Renault MA, Robbesyn F, Chapouly C, Yao Q, Vandierdonck S, Reynaud A, Belloc I, Traiffort E, Ruat M, Desgranges C, Gadeau AP. Hedgehog-dependent regulation of angiogenesis and myogenesis is impaired in aged mice. Arterioscler Thromb Vasc Biol 2013; 33:2858-66. [PMID: 24135022 DOI: 10.1161/atvbaha.113.302494] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The purpose of this study is to further document alteration of signal transduction pathways, more particularly of hedgehog (Hh) signaling, causing impaired ischemic muscle repair in old mice. APPROACH AND RESULTS We used 12-week-old (young mice) and 20- to 24-month-old C57BL/6 mice (old mice) to investigate the activity of Hh signaling in the setting of hindlimb ischemia-induced angiogenesis and skeletal muscle repair. In this model, delayed ischemic muscle repair observed in old mice was associated with an impaired upregulation of Gli1. Sonic Hh expression was not different in old mice compared with young mice, whereas desert Hh (Dhh) expression was downregulated in the skeletal muscle of old mice both in healthy and ischemic conditions. The rescue of Dhh expression by gene therapy in old mice promoted ischemia-induced angiogenesis and increased nerve density; nevertheless, it failed to promote myogenesis or to increase Gli1 mRNA expression. After further investigation, we found that, in addition to Dhh, smoothened expression was significantly downregulated in old mice. We used smoothened haploinsufficient mice to demonstrate that smoothened knockdown by 50% is sufficient to impair activation of Hh signaling and ischemia-induced muscle repair. CONCLUSIONS The present study demonstrates that Hh signaling is impaired in aged mice because of Dhh and smoothened downregulation. Moreover, it shows that hegdehog-dependent regulation of angiogenesis and myogenesis involves distinct mechanisms.
Collapse
Affiliation(s)
- Marie-Ange Renault
- From the University of Bordeaux, Adaptation cardiovasculaire à l'ischémie, UMR1034, Pessac, France (M.-A.R., F.R., C.C., Q.Y., S.V., A.R., I.B., C.D., A.-P.G.); INSERM, Adaptation cardiovasculaire à l'ischémie, U1034, Pessac, France (M.-A.R., F.R., C.C., Q.Y., S.V., A.R., I.B., C.D., A.-P.G.); CHU de Bordeaux, Pharmacie de l'Hôpital Haut-Lévêque, Bordeaux, France (C.C., S.V.); and CNRS, UPR-3294, Laboratoire de Neurobiologie et Développement, Institut de Neurobiologie Alfred Fessard IFR2118, Gif-sur-Yvette, France (E.T., M.R.)
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Renault MA, Vandierdonck S, Chapouly C, Yu Y, Qin G, Metras A, Couffinhal T, Losordo DW, Yao Q, Reynaud A, Jaspard-Vinassa B, Belloc I, Desgranges C, Gadeau AP. Gli3 regulation of myogenesis is necessary for ischemia-induced angiogenesis. Circ Res 2013; 113:1148-58. [PMID: 24044950 DOI: 10.1161/circresaha.113.301546] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
RATIONALE A better understanding of the mechanism underlying skeletal muscle repair is required to develop therapies that promote tissue regeneration in adults. Hedgehog signaling has been shown previously to be involved in myogenesis and angiogenesis: 2 crucial processes for muscle development and regeneration. OBJECTIVE The objective of this study was to identify the role of the hedgehog transcription factor Gli3 in the cross-talk between angiogenesis and myogenesis in adults. METHODS AND RESULTS Using conditional knockout mice, we found that Gli3 deficiency in endothelial cells did not affect ischemic muscle repair, whereas in myocytes, Gli3 deficiency resulted in severely delayed ischemia-induced myogenesis. Moreover, angiogenesis was also significantly impaired in HSA-Cre(ERT2); Gli3(Flox/Flox) mice, demonstrating that impaired myogenesis indirectly affects ischemia-induced angiogenesis. The role of Gli3 in myocytes was then further investigated. We found that Gli3 promotes myoblast differentiation through myogenic factor 5 regulation. In addition, we found that Gli3 regulates several proangiogenic factors, including thymidine phosphorylase and angiopoietin-1 both in vitro and in vivo, which indirectly promote endothelial cell proliferation and arteriole formation. In addition, we found that Gli3 is upregulated in proliferating myoblasts by the cell cycle-associated transcription factor E2F1. CONCLUSIONS This study shows for the first time that Gli3-regulated postnatal myogenesis is necessary for muscle repair-associated angiogenesis. Most importantly, it implies that myogenesis drives angiogenesis in the setting of skeletal muscle repair and identifies Gli3 as a potential target for regenerative medicine.
Collapse
Affiliation(s)
- Marie-Ange Renault
- From the Université de Bordeaux, Adaptation cardiovasculaire à l'ischémie, U1034, Pessac, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Wells LA, Valic MS, Lisovsky A, Sefton MV. Angiogenic Biomaterials to Promote Tissue Vascularization and Integration. Isr J Chem 2013. [DOI: 10.1002/ijch.201300053] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
34
|
Xie G, Choi SS, Syn WK, Michelotti GA, Swiderska-Syn M, Karaca G, Chan IS, Chen Y, Diehl AM. Hedgehog signalling regulates liver sinusoidal endothelial cell capillarisation. Gut 2013; 62:299-309. [PMID: 22362915 PMCID: PMC3595101 DOI: 10.1136/gutjnl-2011-301494] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Vascular remodelling during liver damage involves loss of healthy liver sinusoidal endothelial cell (LSEC) phenotype via capillarisation. Hedgehog (Hh) signalling regulates vascular development and increases during liver injury. This study therefore examined its role in capillarisation. DESIGN Primary LSEC were cultured for 5 days to induce capillarisation. Pharmacological, antibody-mediated and genetic approaches were used to manipulate Hh signalling. Effects on mRNA and protein expression of Hh-regulated genes and capillarisation markers were evaluated by quantitative reverse transcription PCR and immunoblot. Changes in LSEC function were assessed by migration and tube forming assay, and gain/loss of fenestrae was examined by electron microscopy. Mice with acute or chronic liver injury were treated with Hh inhibitors; effects on capillarisation were assessed by immunohistochemistry. RESULTS Freshly isolated LSEC expressed Hh ligands, Hh receptors and Hh ligand antagonist Hhip. Capillarisation was accompanied by repression of Hhip and increased expression of Hh-regulated genes. Treatment with Hh agonist further induced expression of Hh ligands and Hh-regulated genes, and upregulated capillarisation-associated genes; whereas Hh signalling antagonist or Hh ligand neutralising antibody each repressed expression of Hh target genes and capillarisation markers. LSEC isolated from Smo(loxP/loxP) transgenic mice that had been infected with adenovirus expressing Cre-recombinase to delete Smoothened showed over 75% knockdown of Smoothened. During culture, Smoothened-deficient LSEC had inhibited Hh signalling, less induction of capillarisation-associated genes and retention of fenestrae. In mice with injured livers, inhibiting Hh signalling prevented capillarisation. CONCLUSIONS LSEC produce and respond to Hh ligands, and use Hh signalling to regulate complex phenotypic changes that occur during capillarisation.
Collapse
Affiliation(s)
- Guanhua Xie
- Division of Gastroenterology, Duke University Medical Center, Durham, NC
| | - Steve S. Choi
- Division of Gastroenterology, Duke University Medical Center, Durham, NC
,Section of Gastroenterology, Department of Medicine, Durham Veteran Affairs Medical Center, Durham, NC
| | - Wing-Kin Syn
- Division of Gastroenterology, Duke University Medical Center, Durham, NC
,Section of Regeneration and Repair, Institute of Hepatology,, London, UK
| | | | | | - Gamze Karaca
- Division of Gastroenterology, Duke University Medical Center, Durham, NC
| | - Isaac S. Chan
- Division of Gastroenterology, Duke University Medical Center, Durham, NC
| | - Yuping Chen
- Division of Gastroenterology, Duke University Medical Center, Durham, NC
| | - Anna Mae Diehl
- Division of Gastroenterology, Duke University Medical Center, Durham, NC
| |
Collapse
|
35
|
Renault MA, Chapouly C, Yao Q, Larrieu-Lahargue F, Vandierdonck S, Reynaud A, Petit M, Jaspard-Vinassa B, Belloc I, Traiffort E, Ruat M, Duplàa C, Couffinhal T, Desgranges C, Gadeau AP. Desert hedgehog promotes ischemia-induced angiogenesis by ensuring peripheral nerve survival. Circ Res 2013; 112:762-70. [PMID: 23343527 DOI: 10.1161/circresaha.113.300871] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
RATIONALE Blood vessel growth and patterning have been shown to be regulated by nerve-derived signals. Desert hedgehog (Dhh), one of the Hedgehog family members, is expressed by Schwann cells of peripheral nerves. OBJECTIVE The purpose of this study was to investigate the contribution of Dhh to angiogenesis in the setting of ischemia. METHODS AND RESULTS We induced hindlimb ischemia in wild-type and Dhh(-/-) mice. First, we found that limb perfusion is significantly impaired in the absence of Dhh. This effect is associated with a significant decrease in capillary and artery density in Dhh(-/-). By using mice in which the Hedgehog signaling pathway effector Smoothened was specifically invalidated in endothelial cells, we demonstrated that Dhh does not promote angiogenesis by a direct activation of endothelial cells. On the contrary, we found that Dhh promotes peripheral nerve survival in the ischemic muscle and, by doing so, maintains the pool of nerve-derived proangiogenic factors. Consistently, we found that denervation of the leg, immediately after the onset of ischemia, severely impairs ischemia-induced angiogenesis and decreases expression of vascular endothelial growth factor A, angiopoietin 1, and neurotrophin 3 in the ischemic muscle. CONCLUSIONS This study demonstrates the crucial roles of nerves and factors regulating nerve physiology in the setting of ischemia-induced angiogenesis.
Collapse
Affiliation(s)
- Marie-Ange Renault
- Institut National de la Santé et de la Recherche Médicale, 1 Avenue de Magellan, Pessac, France.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Jackson WM, Nesti LJ, Tuan RS. Mesenchymal stem cell therapy for attenuation of scar formation during wound healing. Stem Cell Res Ther 2012; 3:20. [PMID: 22668751 PMCID: PMC3392767 DOI: 10.1186/scrt111] [Citation(s) in RCA: 196] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Scars are a consequence of cutaneous wound healing that can be both unsightly and detrimental to the function of the tissue. Scar tissue is generated by excessive deposition of extracellular matrix tissue by wound healing fibroblasts and myofibroblasts, and although it is inferior to the uninjured skin, it is able to restore integrity to the boundary between the body and its environment. Scarring is not a necessary process to repair the dermal tissues. Rather, scar tissue forms due to specific mechanisms that occur during the adult wound healing process and are modulated primarily by the inflammatory response at the site of injury. Adult tissue-derived mesenchymal stem cells, which participate in normal wound healing, are trophic mediators of tissue repair. These cells participate in attenuating inflammation in the wound and reprogramming the resident immune and wound healing cells to favor tissue regeneration and inhibit fibrotic tissue formation. As a result, these cells have been considered and tested as a likely candidate for a cellular therapy to promote scar-less wound healing. This review identifies specific mechanisms by which mesenchymal stem cells can limit tissue fibrosis and summarizes recent in vivo studies where these cells have been used successfully to limit scar formation.
Collapse
Affiliation(s)
- Wesley M Jackson
- Clinical and Experimental Orthopaedics Laboratory, Department of SurgeryUniformed Services University, Bethesda, MD 20814, USA
| | | | | |
Collapse
|
37
|
Fitzpatrick LE, Lisovsky A, Sefton MV. The expression of sonic hedgehog in diabetic wounds following treatment with poly(methacrylic acid-co-methyl methacrylate) beads. Biomaterials 2012; 33:5297-307. [PMID: 22541537 DOI: 10.1016/j.biomaterials.2012.04.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2012] [Accepted: 04/01/2012] [Indexed: 12/15/2022]
Abstract
The expression of native sonic hedgehog (Shh) was significantly increased in poly(methacrylic acid-co-methyl methacrylate) bead (MAA) treated wounds at day 4 compared to both poly(methyl methacrylate) bead (PMMA) treated and untreated wounds in diabetic db/db mice. MAA beads also increased the expression of the Shh transcription factor Gli3 at day 4. Previously, topical application of MAA beads (45 mol % methacrylic acid) improved wound closure and blood vessel density in excisional wounds in these mice, while PMMA beads did not. Gene expression within the granulation tissue of healing wounds was studied to provide insight into the mechanism of vessel formation and wound healing in the presence of MAA beads. In addition to the increased expression of Shh, MAA-treated wounds had increased expression of osteopontin (OPN), IL-1β and TNF-α, (at day 7) similar to the previously reported MAA response of macrophage-like and endothelial cells in vitro.
Collapse
Affiliation(s)
- Lindsay E Fitzpatrick
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada.
| | | | | |
Collapse
|
38
|
Jackson WM, Nesti LJ, Tuan RS. Concise review: clinical translation of wound healing therapies based on mesenchymal stem cells. Stem Cells Transl Med 2011. [PMID: 23197639 DOI: 10.5966/sctm.2011-0024] [Citation(s) in RCA: 193] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
There is enormous worldwide demand for therapies to promote the efficient resolution of hard-to-heal wounds with minimal appearance of scarring. Recent in vitro studies with mesenchymal stem cells (MSCs) have identified numerous mechanisms by which these cells can promote the process of wound healing, and there is significant interest in the clinical translation of an MSC-based therapy to promote dermal regeneration. This review provides a systematic analysis of recent preclinical and clinical research to evaluate the use of MSCs in wound healing applications. These in vivo studies provide overwhelming evidence that MSCs can accelerate wound closure by modulating the inflammatory environment, promoting the formation of a well-vascularized granulation matrix, encouraging the migration of keratinocytes, and inhibiting apoptosis of wound healing cells. The trophic effects of MSC therapy also appear to augment wound healing in diabetic tissues, thereby preventing the formation of nonhealing ulcers. Finally, a number of delivery systems have been evaluated and indicate that MSCs could be the basis of a versatile therapy to fulfill the clinical needs for dermal regeneration. However, despite the apparent advantages of MSC-based therapies, there have been only limited clinical investigations of this type of therapy in humans. Thus, our review concludes with a discussion of the translational barriers that are limiting the widespread clinical use of MSCs to enhance wound healing.
Collapse
Affiliation(s)
- Wesley M Jackson
- Clinical and Experimental Orthopaedics Laboratory, Department of Surgery, Uniformed Services University, Bethesda, Maryland, USA
| | | | | |
Collapse
|
39
|
Lee MY, Racine V, Jagadpramana P, Sun L, Yu W, Du T, Spencer-Dene B, Rubin N, Le L, Ndiaye D, Bellusci S, Kratochwil K, Veltmaat JM. Ectodermal influx and cell hypertrophy provide early growth for all murine mammary rudiments, and are differentially regulated among them by Gli3. PLoS One 2011; 6:e26242. [PMID: 22046263 PMCID: PMC3203106 DOI: 10.1371/journal.pone.0026242] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2011] [Accepted: 09/22/2011] [Indexed: 11/29/2022] Open
Abstract
Mammary gland development starts in utero with one or several pairs of mammary rudiments (MRs) budding from the surface ectodermal component of the mammalian embryonic skin. Mice develop five pairs, numbered MR1 to MR5 from pectoral to inguinal position. We have previously shown that Gli3Xt-J/Xt-J mutant embryos, which lack the transcription factor Gli3, do not form MR3 and MR5. We show here that two days after the MRs emerge, Gli3Xt-J/Xt-J MR1 is 20% smaller, and Gli3Xt-J/Xt-J MR2 and MR4 are 50% smaller than their wild type (wt) counterparts. Moreover, while wt MRs sink into the underlying dermis, Gli3Xt-J/Xt-J MR4 and MR2 protrude outwardly, to different extents. To understand why each of these five pairs of functionally identical organs has its own, distinct response to the absence of Gli3, we determined which cellular mechanisms regulate growth of the individual MRs, and whether and how Gli3 regulates these mechanisms. We found a 5.5 to 10.7-fold lower cell proliferation rate in wt MRs compared to their adjacent surface ectoderm, indicating that MRs do not emerge or grow via locally enhanced cell proliferation. Cell-tracing experiments showed that surface ectodermal cells are recruited toward the positions where MRs emerge, and contribute to MR growth during at least two days. During the second day of MR development, peripheral cells within the MRs undergo hypertrophy, which also contributes to MR growth. Limited apoptotic cell death counterbalances MR growth. The relative contribution of each of these processes varies among the five MRs. Furthermore, each of these processes is impaired in the absence of Gli3, but to different extents in each MR. This differential involvement of Gli3 explains the variation in phenotype among Gli3Xt-J/Xt-J MRs, and may help to understand the variation in numbers and positions of mammary glands among mammals.
Collapse
Affiliation(s)
- May Yin Lee
- Institute of Molecular and Cell Biology, A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
- Graduate School for Integrative Sciences and Engineering, National University of Singapore, Centre for Life Sciences (CeLS), Singapore, Singapore
| | - Victor Racine
- Institute of Molecular and Cell Biology, A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Peter Jagadpramana
- Institute of Molecular and Cell Biology, A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Li Sun
- Institute of Molecular and Cell Biology, A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Weimiao Yu
- Institute of Molecular and Cell Biology, A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Tiehua Du
- Bio-Informatics Institute, A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Bradley Spencer-Dene
- Experimental Histopathology Laboratory, Cancer Research UK London Research Institute, London, United Kingdom
| | - Nicole Rubin
- Developmental Biology Program, The Saban Research Institute of Children's Hospital Los Angeles, Los Angeles, California, United States of America
| | - Lendy Le
- Developmental Biology Program, The Saban Research Institute of Children's Hospital Los Angeles, Los Angeles, California, United States of America
| | - Delphine Ndiaye
- Institut Curie/CNRS-UMR144, Equipe de Morphogenèse Cellulaire et Progression Tumorale, Paris, France
| | - Saverio Bellusci
- Developmental Biology Program, The Saban Research Institute of Children's Hospital Los Angeles, Los Angeles, California, United States of America
| | | | - Jacqueline M. Veltmaat
- Institute of Molecular and Cell Biology, A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
- Department of Anatomy, Yong Loo Lin School of Medicine, National University Health System, Singapore, Singapore
- * E-mail:
| |
Collapse
|
40
|
Abstract
Numerous signaling pathways are misregulated in pancreatic ductal adenocarcinoma (PDAC), a highly malignant type of cancer. One of these is the Hedgehog (HH) pathway, which is normally involved in patterning processes in the developing embryo. Expression of the main ligand Sonic Hedgehog is an early event in carcinogenesis and correlates with the mutation of the KRAS oncogene, the cardinal molecular feature of pancreatic cancer. Recent data establish a functional role for HH signaling primarily in the tumor microenvironment, where it is involved in myofibroblast differentiation and the induction of stroma-derived growth promoting molecules. Given the protumorigenic functions of the abundant stromal desmoplasia typically associated with pancreatic cancer, targeting the HH pathway might prove beneficial in the treatment of the disease. First data using small molecule antagonists of HH signaling in mouse models of pancreatic cancer are promising and reveal a substantial, yet transient, effect on the myofibroblastic stroma. In this review, we try to give an outline on the current knowledge about HH signaling in pancreatic cancer including a perspective of using pharmacological inhibitors of this pathway in the clinic.
Collapse
Affiliation(s)
- Matthias Lauth
- Institute of Molecular Biology and Tumor Research (IMT), Philipps University, Marburg, Germany
| | | |
Collapse
|
41
|
Wu R, Smeele KM, Wyatt E, Ichikawa Y, Eerbeek O, Sun L, Chawla K, Hollmann MW, Nagpal V, Heikkinen S, Laakso M, Jujo K, Wasserstrom JA, Zuurbier CJ, Ardehali H. Reduction in hexokinase II levels results in decreased cardiac function and altered remodeling after ischemia/reperfusion injury. Circ Res 2011; 108:60-69. [PMID: 21071708 PMCID: PMC3017633 DOI: 10.1161/circresaha.110.223115] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2010] [Accepted: 10/29/2010] [Indexed: 01/10/2023]
Abstract
RATIONALE Cardiomyocytes switch substrate utilization from fatty acid to glucose under ischemic conditions; however, it is unknown how perturbations in glycolytic enzymes affect cardiac response to ischemia/reperfusion (I/R). Hexokinase (HK)II is a HK isoform that is expressed in the heart and can bind to the mitochondrial outer membrane. OBJECTIVE We sought to define how HKII and its binding to mitochondria play a role in cardiac response and remodeling after I/R. METHODS AND RESULTS We first showed that HKII levels and its binding to mitochondria are reduced 2 days after I/R. We then subjected the hearts of wild-type and heterozygote HKII knockout (HKII(+/)⁻) mice to I/R by coronary ligation. At baseline, HKII(+/)⁻ mice have normal cardiac function; however, they display lower systolic function after I/R compared to wild-type animals. The mechanism appears to be through an increase in cardiomyocyte death and fibrosis and a reduction in angiogenesis; the latter is through a decrease in hypoxia-inducible factor-dependent pathway signaling in cardiomyocytes. HKII mitochondrial binding is also critical for cardiomyocyte survival, because its displacement in tissue culture with a synthetic peptide increases cell death. Our results also suggest that HKII may be important for the remodeling of the viable cardiac tissue because its modulation in vitro alters cellular energy levels, O₂ consumption, and contractility. CONCLUSIONS These results suggest that reduction in HKII levels causes altered remodeling of the heart in I/R by increasing cell death and fibrosis and reducing angiogenesis and that mitochondrial binding is needed for protection of cardiomyocytes.
Collapse
Affiliation(s)
| | | | - Eugene Wyatt
- Division of Cardiology, Department of Medicine, Northwestern University School of Medicine, Chicago, IL (R.W., E.W., L.S., V.P., V.N., K.J., J.A.W., H.A.) Department of Anesthesiology, Laboratory of Experimental Intensive Care and Anesthesiology (K.M.S., M.W.H., C.J.Z.), and Department of Physiology (O.E.), Academic Medical Center, Amsterdam, The Netherlands. Department of Medicine, University of Kuopio and Kuopio University Hospital, 70210 Kuopio, Finland (S.H., M.L.)
| | - Yoshihiko Ichikawa
- Division of Cardiology, Department of Medicine, Northwestern University School of Medicine, Chicago, IL (R.W., E.W., L.S., V.P., V.N., K.J., J.A.W., H.A.) Department of Anesthesiology, Laboratory of Experimental Intensive Care and Anesthesiology (K.M.S., M.W.H., C.J.Z.), and Department of Physiology (O.E.), Academic Medical Center, Amsterdam, The Netherlands. Department of Medicine, University of Kuopio and Kuopio University Hospital, 70210 Kuopio, Finland (S.H., M.L.)
| | - Otto Eerbeek
- Division of Cardiology, Department of Medicine, Northwestern University School of Medicine, Chicago, IL (R.W., E.W., L.S., V.P., V.N., K.J., J.A.W., H.A.) Department of Anesthesiology, Laboratory of Experimental Intensive Care and Anesthesiology (K.M.S., M.W.H., C.J.Z.), and Department of Physiology (O.E.), Academic Medical Center, Amsterdam, The Netherlands. Department of Medicine, University of Kuopio and Kuopio University Hospital, 70210 Kuopio, Finland (S.H., M.L.)
| | - Lin Sun
- Division of Cardiology, Department of Medicine, Northwestern University School of Medicine, Chicago, IL (R.W., E.W., L.S., V.P., V.N., K.J., J.A.W., H.A.) Department of Anesthesiology, Laboratory of Experimental Intensive Care and Anesthesiology (K.M.S., M.W.H., C.J.Z.), and Department of Physiology (O.E.), Academic Medical Center, Amsterdam, The Netherlands. Department of Medicine, University of Kuopio and Kuopio University Hospital, 70210 Kuopio, Finland (S.H., M.L.)
| | - Kusum Chawla
- Division of Cardiology, Department of Medicine, Northwestern University School of Medicine, Chicago, IL (R.W., E.W., L.S., V.P., V.N., K.J., J.A.W., H.A.) Department of Anesthesiology, Laboratory of Experimental Intensive Care and Anesthesiology (K.M.S., M.W.H., C.J.Z.), and Department of Physiology (O.E.), Academic Medical Center, Amsterdam, The Netherlands. Department of Medicine, University of Kuopio and Kuopio University Hospital, 70210 Kuopio, Finland (S.H., M.L.)
| | - Markus W. Hollmann
- Division of Cardiology, Department of Medicine, Northwestern University School of Medicine, Chicago, IL (R.W., E.W., L.S., V.P., V.N., K.J., J.A.W., H.A.) Department of Anesthesiology, Laboratory of Experimental Intensive Care and Anesthesiology (K.M.S., M.W.H., C.J.Z.), and Department of Physiology (O.E.), Academic Medical Center, Amsterdam, The Netherlands. Department of Medicine, University of Kuopio and Kuopio University Hospital, 70210 Kuopio, Finland (S.H., M.L.)
| | - Varun Nagpal
- Division of Cardiology, Department of Medicine, Northwestern University School of Medicine, Chicago, IL (R.W., E.W., L.S., V.P., V.N., K.J., J.A.W., H.A.) Department of Anesthesiology, Laboratory of Experimental Intensive Care and Anesthesiology (K.M.S., M.W.H., C.J.Z.), and Department of Physiology (O.E.), Academic Medical Center, Amsterdam, The Netherlands. Department of Medicine, University of Kuopio and Kuopio University Hospital, 70210 Kuopio, Finland (S.H., M.L.)
| | - Sami Heikkinen
- Division of Cardiology, Department of Medicine, Northwestern University School of Medicine, Chicago, IL (R.W., E.W., L.S., V.P., V.N., K.J., J.A.W., H.A.) Department of Anesthesiology, Laboratory of Experimental Intensive Care and Anesthesiology (K.M.S., M.W.H., C.J.Z.), and Department of Physiology (O.E.), Academic Medical Center, Amsterdam, The Netherlands. Department of Medicine, University of Kuopio and Kuopio University Hospital, 70210 Kuopio, Finland (S.H., M.L.)
| | - Markku Laakso
- Division of Cardiology, Department of Medicine, Northwestern University School of Medicine, Chicago, IL (R.W., E.W., L.S., V.P., V.N., K.J., J.A.W., H.A.) Department of Anesthesiology, Laboratory of Experimental Intensive Care and Anesthesiology (K.M.S., M.W.H., C.J.Z.), and Department of Physiology (O.E.), Academic Medical Center, Amsterdam, The Netherlands. Department of Medicine, University of Kuopio and Kuopio University Hospital, 70210 Kuopio, Finland (S.H., M.L.)
| | - Kentaro Jujo
- Division of Cardiology, Department of Medicine, Northwestern University School of Medicine, Chicago, IL (R.W., E.W., L.S., V.P., V.N., K.J., J.A.W., H.A.) Department of Anesthesiology, Laboratory of Experimental Intensive Care and Anesthesiology (K.M.S., M.W.H., C.J.Z.), and Department of Physiology (O.E.), Academic Medical Center, Amsterdam, The Netherlands. Department of Medicine, University of Kuopio and Kuopio University Hospital, 70210 Kuopio, Finland (S.H., M.L.)
| | - J. Andrew Wasserstrom
- Division of Cardiology, Department of Medicine, Northwestern University School of Medicine, Chicago, IL (R.W., E.W., L.S., V.P., V.N., K.J., J.A.W., H.A.) Department of Anesthesiology, Laboratory of Experimental Intensive Care and Anesthesiology (K.M.S., M.W.H., C.J.Z.), and Department of Physiology (O.E.), Academic Medical Center, Amsterdam, The Netherlands. Department of Medicine, University of Kuopio and Kuopio University Hospital, 70210 Kuopio, Finland (S.H., M.L.)
| | | | | |
Collapse
|
42
|
Reffelmann T, Kloner RA. Blood supply of the graft after cellular cardiomyoplasty. Regen Med 2010; 5:777-86. [PMID: 20868332 DOI: 10.2217/rme.10.40] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Cellular cardiomyoplasty is under extensive investigation as a potential therapeutic strategy after myocardial infarction, in congestive heart failure and chronic ischemic heart disease. Various cell sources and techniques for transplantation have been studied in animal models of cardiac disease. The initial goal of replacing myocardial scar tissue by vital myocardial cells, integrated into the host, simultaneously beating and contributing to systolic force, has not yet been accomplished. However, most experimental models provided evidence for enhanced vascularization after cell transplantation. In some investigations, neovascularization was also shown to be accompanied by increased myocardial perfusion. Mechanisms by which vascularization occurs have not been fully elucidated: either the transplanted cells provide an angiogenic stimulus, involving various paracrine or hormone-like factors, which induces the formation of a new vasculature or, depending on the source of transplanted cells, the cells incorporate into the vascular network after proliferation and differentiation. This review summarizes research that specifically studied the occurrence, magnitude and mechanisms of enhanced myocardial blood supply after cellular cardiomyoplasty.
Collapse
Affiliation(s)
- Thorsten Reffelmann
- The Heart Institute, Good Samaritan Hospital, Division of Cardiology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90017-2395, USA.
| | | |
Collapse
|
43
|
Mimeault M, Batra SK. Frequent deregulations in the hedgehog signaling network and cross-talks with the epidermal growth factor receptor pathway involved in cancer progression and targeted therapies. Pharmacol Rev 2010; 62:497-524. [PMID: 20716670 PMCID: PMC2964899 DOI: 10.1124/pr.109.002329] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The hedgehog (Hh)/glioma-associated oncogene (GLI) signaling network is among the most important and fascinating signal transduction systems that provide critical functions in the regulation of many developmental and physiological processes. The coordinated spatiotemporal interplay of the Hh ligands and other growth factors is necessary for the stringent control of the behavior of diverse types of tissue-resident stem/progenitor cells and their progenies. The activation of the Hh cascade might promote the tissue regeneration and repair after severe injury in numerous organs, insulin production in pancreatic beta-cells, and neovascularization. Consequently, the stimulation of the Hh pathway constitutes a potential therapeutic strategy to treat diverse human disorders, including severe tissue injuries; diabetes mellitus; and brain, skin, and cardiovascular disorders. In counterbalance, a deregulation of the Hh signaling network might lead to major tissular disorders and the development of a wide variety of aggressive and metastatic cancers. The target gene products induced through the persistent Hh activation can contribute to the self-renewal, survival, migration, and metastasis of cancer stem/progenitor cells and their progenies. Moreover, the pivotal role mediated through the Hh/GLI cascade during cancer progression also implicates the cooperation with other oncogenic products, such as mutated K-RAS and complex cross-talk with different growth factor pathways, including tyrosine kinase receptors, such as epidermal growth factor receptor (EGFR), Wnt/beta-catenin, and transforming growth factor-beta (TGF-beta)/TGF-beta receptors. Therefore, the molecular targeting of distinct deregulated gene products, including Hh and EGFR signaling components and other signaling elements that are frequently deregulated in highly tumorigenic cancer-initiating cells and their progenies, might constitute a potential therapeutic strategy to eradicate the total cancer cell mass. Of clinical interest is that these multitargeted approaches offer great promise as adjuvant treatments for improving the current antihormonal therapies, radiotherapies, and/or chemotherapies against locally advanced and metastatic cancers, thereby preventing disease relapse and the death of patients with cancer.
Collapse
Affiliation(s)
- Murielle Mimeault
- Department of Biochemistry and Molecular Biology, Eppley Institute for Research in Cancer, and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | | |
Collapse
|
44
|
Roncalli J, Tongers J, Losordo DW. Update on gene therapy for myocardial ischaemia and left ventricular systolic dysfunction or heart failure. Arch Cardiovasc Dis 2010; 103:469-76. [PMID: 21074126 DOI: 10.1016/j.acvd.2010.04.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2010] [Revised: 04/01/2010] [Accepted: 04/02/2010] [Indexed: 11/30/2022]
Abstract
Despite considerable advances in pharmacological, surgical and technology-based cardiovascular therapy, left ventricular dysfunction and heart failure are increasingly prevalent health problems. Recent studies suggest that angiogenic gene therapy can restore perfusion in ischaemic myocardial tissue, and that the transfer of nonangiogenic genes may correct defects in calcium handling that contribute to abnormal contractile function in patients with heart failure; however, large clinical trials of gene therapy for treatment of left ventricular dysfunction and heart failure have yet to be completed, and only a small number of genes have been evaluated in patients. Researchers continue to investigate new genes, combinations of genes and approaches that combine gene and cell therapy, and to develop novel expression vectors and delivery systems; collectively, these refinements promise to improve both patient response and safety.
Collapse
Affiliation(s)
- Jerome Roncalli
- Feinberg Cardiovascular Research Institute, Northwestern University School of Medicine and Northwestern Memorial Hospital, Chicago, IL, USA.
| | | | | |
Collapse
|
45
|
Bijlsma MF, Spek CA. The Hedgehog morphogen in myocardial ischemia-reperfusion injury. Exp Biol Med (Maywood) 2010; 235:447-54. [PMID: 20407076 DOI: 10.1258/ebm.2009.009303] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The developmental Hedgehog (Hh) protein family is known to be pivotal in many embryonic patterning events and the number of processes in which Hh plays an essential role is expanding persistently. Recently, it has become clear that the Hh pathway is not only active in the developing embryo but also in the adult organism. For example, Hh has been suggested to salvage ischemia-induced tissue damage although endogenous Hh might be deleterious during the early phase of myocardial ischemia-reperfusion. The current review provides an overview of the history of Hh biology and discusses some novel insights on Hh cell biology. Hh function in pathophysiology as well as recent findings concerning Hh signaling in ischemia models, especially in light of cardiovascular disease, is discussed in more detail and future perspectives are proposed.
Collapse
Affiliation(s)
- Maarten F Bijlsma
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | | |
Collapse
|