1
|
Volk-Draper L, Athaiya S, Espinosa Gonzalez M, Bhattarai N, Wilber A, Ran S. Tumor microenvironment restricts IL-10 induced multipotent progenitors to myeloid-lymphatic phenotype. PLoS One 2024; 19:e0298465. [PMID: 38640116 PMCID: PMC11029653 DOI: 10.1371/journal.pone.0298465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 01/24/2024] [Indexed: 04/21/2024] Open
Abstract
Lymphangiogenesis is induced by local pro-lymphatic growth factors and bone marrow (BM)-derived myeloid-lymphatic endothelial cell progenitors (M-LECP). We previously showed that M-LECP play a significant role in lymphangiogenesis and lymph node metastasis in clinical breast cancer (BC) and experimental BC models. We also showed that differentiation of mouse and human M-LECP can be induced through sequential activation of colony stimulating factor-1 (CSF-1) and Toll-like receptor-4 (TLR4) pathways. This treatment activates the autocrine interleukin-10 (IL-10) pathway that, in turn, induces myeloid immunosuppressive M2 phenotype along with lymphatic-specific proteins. Because IL-10 is implicated in differentiation of numerous lineages, we sought to determine whether this pathway specifically promotes the lymphatic phenotype or multipotent progenitors that can give rise to M-LECP among other lineages. Analyses of BM cells activated either by CSF-1/TLR4 ligands in vitro or orthotopic breast tumors in vivo showed expansion of stem/progenitor population and coincident upregulation of markers for at least four lineages including M2-macrophage, lymphatic endothelial, erythroid, and T-cells. Induction of cell plasticity and multipotency was IL-10 dependent as indicated by significant reduction of stem cell markers and those for multiple lineages in differentiated cells treated with anti-IL-10 receptor (IL-10R) antibody or derived from IL-10R knockout mice. However, multipotent CD11b+/Lyve-1+/Ter-119+/CD3e+ progenitors detected in BM appeared to split into a predominant myeloid-lymphatic fraction and minor subsets expressing erythroid and T-cell markers upon establishing tumor residence. Each sub-population was detected at a distinct intratumoral site. This study provides direct evidence for differences in maturation status between the BM progenitors and those reaching tumor destination. The study results suggest preferential tumor bias towards expansion of myeloid-lymphatic cells while underscoring the role of IL-10 in early BM production of multipotent progenitors that give rise to both hematopoietic and endothelial lineages.
Collapse
Affiliation(s)
- Lisa Volk-Draper
- Department of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, United States of America
| | - Shaswati Athaiya
- Department of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, United States of America
| | - Maria Espinosa Gonzalez
- Department of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, United States of America
| | - Nihit Bhattarai
- Department of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, United States of America
| | - Andrew Wilber
- Department of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, United States of America
- Simmons Cancer Institute, Southern Illinois University School of Medicine, Springfield, IL, United States of America
| | - Sophia Ran
- Department of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, United States of America
- Simmons Cancer Institute, Southern Illinois University School of Medicine, Springfield, IL, United States of America
| |
Collapse
|
2
|
Guan L, Fan P, Wang Y, Liu X, Liu R, Ma W, Bai H. Lymphangiogenic responses of lymphatic endothelial cells to steady direct-current electric fields. Cell Adh Migr 2023; 17:1-14. [PMID: 37889090 PMCID: PMC10761046 DOI: 10.1080/19336918.2023.2271260] [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: 09/28/2022] [Accepted: 10/12/2023] [Indexed: 10/28/2023] Open
Abstract
Lymphangiogenesis plays pivotal roles in diverse physiological and pathological conditions. Steady direct-current electric fields (DC EFs) induce vascular endothelial behaviors related to angiogenesis have been observed. This study investigated the effects of DC EFs on the lymphangiogenic response of lymphatic endothelial cells (LECs). We demonstrated that EFs stimulation induced directional migration, reorientation, and elongation of human LECs in culture. These lymphangiogenic responses required VEGF receptor 3 (VEGFR-3) activation and were mediated through the PI3K-Akt, Erk1/2, and p38 MAPK signaling pathways in relation to the reorganization of the actin cytoskeleton. Our results indicate that endogenous EFs may play a role in lymphangiogenesis in vivo, and VEGFR-3 signaling activation may be involved in the cellular function of LECs driven by EFs.
Collapse
Affiliation(s)
- Linbo Guan
- Laboratory of Genetic Disease and Perinatal Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children of the Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Ping Fan
- Laboratory of Genetic Disease and Perinatal Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children of the Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yufeng Wang
- Laboratory of Genetic Disease and Perinatal Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children of the Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Xinghui Liu
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Rui Liu
- Division of Peptides Related with Human Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Wandi Ma
- Laboratory of Genetic Disease and Perinatal Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children of the Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Huai Bai
- Laboratory of Genetic Disease and Perinatal Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children of the Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
3
|
Saveh-Shemshaki N, Barajaa MA, Otsuka T, Mirdamadi ES, Nair LS, Laurencin CT. Electroconductivity, a regenerative engineering approach to reverse rotator cuff muscle degeneration. Regen Biomater 2023; 10:rbad099. [PMID: 38020235 PMCID: PMC10676522 DOI: 10.1093/rb/rbad099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 10/25/2023] [Accepted: 10/28/2023] [Indexed: 12/01/2023] Open
Abstract
Muscle degeneration is one the main factors that lead to the high rate of retear after a successful repair of rotator cuff (RC) tears. The current surgical practices have failed to treat patients with chronic massive rotator cuff tears (RCTs). Therefore, regenerative engineering approaches are being studied to address the challenges. Recent studies showed the promising outcomes of electroactive materials (EAMs) on the regeneration of electrically excitable tissues such as skeletal muscle. Here, we review the most important biological mechanism of RC muscle degeneration. Further, the review covers the recent studies on EAMs for muscle regeneration including RC muscle. Finally, we will discuss the future direction toward the application of EAMs for the augmentation of RCTs.
Collapse
Affiliation(s)
- Nikoo Saveh-Shemshaki
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, Farmington, CT 06030, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Mohammed A Barajaa
- Department of Biomedical Engineering, Imam Abdulrahman Bin Faisal University, Dammam 31451, Saudi Arabia
| | - Takayoshi Otsuka
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, Farmington, CT 06030, USA
| | - Elnaz S Mirdamadi
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, Farmington, CT 06030, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Lakshmi S Nair
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, Farmington, CT 06030, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
- Department of Orthopedic Surgery, University of Connecticut Health Center, Farmington, CT 06030, USA
- Department of Materials Science and Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Cato T Laurencin
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, Farmington, CT 06030, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
- Department of Orthopedic Surgery, University of Connecticut Health Center, Farmington, CT 06030, USA
- Department of Materials Science and Engineering, University of Connecticut, Storrs, CT 06269, USA
- Department of Chemical & Biomolecular Engineering, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
4
|
Kaur P, Khan H, Grewal AK, Dua K, Singh TG. Therapeutic potential of NOX inhibitors in neuropsychiatric disorders. Psychopharmacology (Berl) 2023; 240:1825-1840. [PMID: 37507462 DOI: 10.1007/s00213-023-06424-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023]
Abstract
RATIONALE Neuropsychiatric disorders encompass a broad category of medical conditions that include both neurology as well as psychiatry such as major depressive disorder, autism spectrum disorder, bipolar disorder, schizophrenia as well as psychosis. OBJECTIVE NADPH-oxidase (NOX), which is the free radical generator, plays a substantial part in oxidative stress in neuropsychiatric disorders. It is thought that elevated oxidative stress as well as neuroinflammation plays a part in the emergence of neuropsychiatric disorders. Including two linked with membranes and four with subunits of cytosol, NOX is a complex of multiple subunits. NOX has been linked to a significant source of reactive oxygen species in the brain. NOX has been shown to control memory processing and neural signaling. However, excessive NOX production has been linked to cardiovascular disorders, CNS degeneration, and neurotoxicity. The increase in NOX leads to the progression of neuropsychiatric disorders. RESULT Our review mainly emphasized the characteristics of NOX and its various mechanisms, the modulation of NOX in various neuropsychiatric disorders, and various studies supporting the fact that NOX might be the potential therapeutic target for neuropsychiatric disorders. CONCLUSION Here, we summarizes various pharmacological studies involving NOX inhibitors in neuropsychiatric disorders.
Collapse
Affiliation(s)
- Parneet Kaur
- Chitkara College of Pharmacy, Chitkara University, Punjab, 140401, India
| | - Heena Khan
- Chitkara College of Pharmacy, Chitkara University, Punjab, 140401, India
| | | | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW, 2007, Australia
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | | |
Collapse
|
5
|
Higginbottom SL, Tomaskovic-Crook E, Crook JM. Considerations for modelling diffuse high-grade gliomas and developing clinically relevant therapies. Cancer Metastasis Rev 2023; 42:507-541. [PMID: 37004686 PMCID: PMC10348989 DOI: 10.1007/s10555-023-10100-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 03/16/2023] [Indexed: 04/04/2023]
Abstract
Diffuse high-grade gliomas contain some of the most dangerous human cancers that lack curative treatment options. The recent molecular stratification of gliomas by the World Health Organisation in 2021 is expected to improve outcomes for patients in neuro-oncology through the development of treatments targeted to specific tumour types. Despite this promise, research is hindered by the lack of preclinical modelling platforms capable of recapitulating the heterogeneity and cellular phenotypes of tumours residing in their native human brain microenvironment. The microenvironment provides cues to subsets of glioma cells that influence proliferation, survival, and gene expression, thus altering susceptibility to therapeutic intervention. As such, conventional in vitro cellular models poorly reflect the varied responses to chemotherapy and radiotherapy seen in these diverse cellular states that differ in transcriptional profile and differentiation status. In an effort to improve the relevance of traditional modelling platforms, recent attention has focused on human pluripotent stem cell-based and tissue engineering techniques, such as three-dimensional (3D) bioprinting and microfluidic devices. The proper application of these exciting new technologies with consideration of tumour heterogeneity and microenvironmental interactions holds potential to develop more applicable models and clinically relevant therapies. In doing so, we will have a better chance of translating preclinical research findings to patient populations, thereby addressing the current derisory oncology clinical trial success rate.
Collapse
Affiliation(s)
- Sarah L Higginbottom
- Intelligent Polymer Research Institute, AIIM Facility, Innovation Campus, University of Wollongong, Fairy Meadow, NSW, 2519, Australia
- Arto Hardy Family Biomedical Innovation Hub, Chris O'Brien Lifehouse, Camperdown, NSW, 2050, Australia
| | - Eva Tomaskovic-Crook
- Intelligent Polymer Research Institute, AIIM Facility, Innovation Campus, University of Wollongong, Fairy Meadow, NSW, 2519, Australia.
- Arto Hardy Family Biomedical Innovation Hub, Chris O'Brien Lifehouse, Camperdown, NSW, 2050, Australia.
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, 2006, Australia.
| | - Jeremy M Crook
- Intelligent Polymer Research Institute, AIIM Facility, Innovation Campus, University of Wollongong, Fairy Meadow, NSW, 2519, Australia.
- Arto Hardy Family Biomedical Innovation Hub, Chris O'Brien Lifehouse, Camperdown, NSW, 2050, Australia.
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, 2006, Australia.
| |
Collapse
|
6
|
Heng BC, Bai Y, Li X, Lim LW, Li W, Ge Z, Zhang X, Deng X. Electroactive Biomaterials for Facilitating Bone Defect Repair under Pathological Conditions. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204502. [PMID: 36453574 PMCID: PMC9839869 DOI: 10.1002/advs.202204502] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 10/24/2022] [Indexed: 06/02/2023]
Abstract
Bone degeneration associated with various diseases is increasing due to rapid aging, sedentary lifestyles, and unhealthy diets. Living bone tissue has bioelectric properties critical to bone remodeling, and bone degeneration under various pathological conditions results in significant changes to these bioelectric properties. There is growing interest in utilizing biomimetic electroactive biomaterials that recapitulate the natural electrophysiological microenvironment of healthy bone tissue to promote bone repair. This review first summarizes the etiology of degenerative bone conditions associated with various diseases such as type II diabetes, osteoporosis, periodontitis, osteoarthritis, rheumatoid arthritis, osteomyelitis, and metastatic osteolysis. Next, the diverse array of natural and synthetic electroactive biomaterials with therapeutic potential are discussed. Putative mechanistic pathways by which electroactive biomaterials can mitigate bone degeneration are critically examined, including the enhancement of osteogenesis and angiogenesis, suppression of inflammation and osteoclastogenesis, as well as their anti-bacterial effects. Finally, the limited research on utilization of electroactive biomaterials in the treatment of bone degeneration associated with the aforementioned diseases are examined. Previous studies have mostly focused on using electroactive biomaterials to treat bone traumatic injuries. It is hoped that this review will encourage more research efforts on the use of electroactive biomaterials for treating degenerative bone conditions.
Collapse
Affiliation(s)
- Boon Chin Heng
- Central LaboratoryPeking University School and Hospital of StomatologyBeijing100081P. R. China
- School of Medical and Life SciencesSunway UniversityDarul EhsanSelangor47500Malaysia
| | - Yunyang Bai
- Department of Geriatric DentistryPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Xiaochan Li
- Department of Geriatric DentistryPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Lee Wei Lim
- Neuromodulation LaboratorySchool of Biomedical SciencesLi Ka Shing Faculty of MedicineThe University of Hong KongPokfulamHong KongP. R. China
| | - Wang Li
- Department of Biomedical EngineeringPeking UniversityBeijing100871P. R. China
| | - Zigang Ge
- Department of Biomedical EngineeringPeking UniversityBeijing100871P. R. China
| | - Xuehui Zhang
- Department of Dental Materials & Dental Medical Devices Testing CenterPeking University School and Hospital of StomatologyBeijing100081P. R. China
- National Engineering Research Center of Oral Biomaterials and Digital Medical DevicesNMPA Key Laboratory for Dental MaterialsBeijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital StomatologyPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Xuliang Deng
- Department of Geriatric DentistryPeking University School and Hospital of StomatologyBeijing100081P. R. China
- Department of Dental Materials & Dental Medical Devices Testing CenterPeking University School and Hospital of StomatologyBeijing100081P. R. China
- National Engineering Research Center of Oral Biomaterials and Digital Medical DevicesNMPA Key Laboratory for Dental MaterialsBeijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital StomatologyPeking University School and Hospital of StomatologyBeijing100081P. R. China
| |
Collapse
|
7
|
Lou T, Chen K, Luo Q, Liu C, Yuan Y, Fan C. Periosteum-inspired in situ CaP generated nanocomposite hydrogels with strong bone adhesion and superior stretchability for accelerated distraction osteogenesis. Biomater Res 2022; 26:91. [PMID: 36581951 PMCID: PMC9801553 DOI: 10.1186/s40824-022-00330-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 11/27/2022] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Distraction osteogenesis (DO) is an efficacious but lengthy procedure to reconstruct segmental bone defects under the principle of tension-stress, during which the periosteum-mediated mechanical stimulation plays a pivotal role. Inspired by the dynamic process of DO and the mechanical stimulation of periosteum, a new design of bionic periosteum was developed to simulate the mechanical transduction of natural periosteum for the application in DO procedure. METHODS In this study, an injectable organic-inorganic hybrid hydrogel was developed based on a novel combination of the PEGylated poly (glycerol sebacate) (PEGS) polymer network and in situ formed CaP nanoparticles (ICPNs). Rat bone marrow mesenchymal stem cells (rBMSCs) and human umbilical vein endothelial cells (HUVECs) were cultured and tested in vitro to evaluate biocompatibility, cell adhesion, proliferation, and pro-osteogenic and pro-angiogenic activity. In vivo experiments were conducted in the rat tibial model of distraction osteogenesis. RESULTS The developed nanocomposite hydrogels exhibited excellent injectability, robust bone adhesion, superior stretchability, and enhanced osteogenic activity. The results of in vitro and in vivo studies showed that PEGS/ICPN hydrogels could promote new bone formation and mineralization during the dynamic distraction process through the synergistic effects of angiogenesis and osteogenesis. CONCLUSIONS This periosteum-inspired nanocomposite hydrogel represents a mechanobiology approach for effectively restoring large bone defects through the dynamic DO process.
Collapse
Affiliation(s)
- Tengfei Lou
- grid.412528.80000 0004 1798 5117Orthopaedic Department, Shanghai Sixth People’s Hospital, Shanghai, 200233 People’s Republic of China
| | - Kai Chen
- grid.28056.390000 0001 2163 4895Key Laboratory for Ultrafine Materials of Ministry of Education, and School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237 People’s Republic of China ,grid.28056.390000 0001 2163 4895Frontiers Science Center for Materiobiology and Dynamic Chemistry, and Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237 People’s Republic of China
| | - Qiyu Luo
- grid.412528.80000 0004 1798 5117Orthopaedic Department, Shanghai Sixth People’s Hospital, Shanghai, 200233 People’s Republic of China
| | - Changsheng Liu
- grid.28056.390000 0001 2163 4895Key Laboratory for Ultrafine Materials of Ministry of Education, and School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237 People’s Republic of China ,grid.28056.390000 0001 2163 4895Frontiers Science Center for Materiobiology and Dynamic Chemistry, and Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237 People’s Republic of China
| | - Yuan Yuan
- grid.28056.390000 0001 2163 4895Key Laboratory for Ultrafine Materials of Ministry of Education, and School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237 People’s Republic of China ,grid.28056.390000 0001 2163 4895Frontiers Science Center for Materiobiology and Dynamic Chemistry, and Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237 People’s Republic of China
| | - Cunyi Fan
- grid.412528.80000 0004 1798 5117Orthopaedic Department, Shanghai Sixth People’s Hospital, Shanghai, 200233 People’s Republic of China
| |
Collapse
|
8
|
Šafaříková E, Ehlich J, Stříteský S, Vala M, Weiter M, Pacherník J, Kubala L, Víteček J. Conductive Polymer PEDOT:PSS-Based Platform for Embryonic Stem-Cell Differentiation. Int J Mol Sci 2022; 23:ijms23031107. [PMID: 35163031 PMCID: PMC8835127 DOI: 10.3390/ijms23031107] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/13/2022] [Accepted: 01/17/2022] [Indexed: 01/12/2023] Open
Abstract
Organic semiconductors are constantly gaining interest in regenerative medicine. Their tunable physico-chemical properties, including electrical conductivity, are very promising for the control of stem-cell differentiation. However, their use for combined material-based and electrical stimulation remains largely underexplored. Therefore, we carried out a study on whether a platform based on the conductive polymer poly(3,4-ethylenedioxythiophene):polystyrene sulfonate (PEDOT:PSS) can be beneficial to the differentiation of mouse embryonic stem cells (mESCs). The platform was prepared using the layout of a standard 24-well cell-culture plate. Polyethylene naphthalate foil served as the substrate for the preparation of interdigitated gold electrodes by physical vapor deposition. The PEDOT:PSS pattern was fabricated by precise screen printing over the gold electrodes. The PEDOT:PSS platform was able to produce higher electrical current with the pulsed-direct-current (DC) electrostimulation mode (1 Hz, 200 mV/mm, 100 ms pulse duration) compared to plain gold electrodes. There was a dominant capacitive component. In proof-of-concept experiments, mESCs were able to respond to such electrostimulation by membrane depolarization and elevation of cytosolic calcium. Further, the PEDOT:PSS platform was able to upregulate cardiomyogenesis and potentially inhibit early neurogenesis per se with minor contribution of electrostimulation. Hence, the present work highlights the large potential of PEDOT:PSS in regenerative medicine.
Collapse
Affiliation(s)
- Eva Šafaříková
- Institute of Biophysics of the Czech Academy of Sciences, Královopolská 135, 612 65 Brno, Czech Republic; (E.Š.); (L.K.)
- Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic;
| | - Jiří Ehlich
- Faculty of Chemistry, Brno University of Technology, Purkyňova 118, 612 00 Brno, Czech Republic; (J.E.); (S.S.); (M.V.); (M.W.)
| | - Stanislav Stříteský
- Faculty of Chemistry, Brno University of Technology, Purkyňova 118, 612 00 Brno, Czech Republic; (J.E.); (S.S.); (M.V.); (M.W.)
| | - Martin Vala
- Faculty of Chemistry, Brno University of Technology, Purkyňova 118, 612 00 Brno, Czech Republic; (J.E.); (S.S.); (M.V.); (M.W.)
| | - Martin Weiter
- Faculty of Chemistry, Brno University of Technology, Purkyňova 118, 612 00 Brno, Czech Republic; (J.E.); (S.S.); (M.V.); (M.W.)
| | - Jiří Pacherník
- Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic;
| | - Lukáš Kubala
- Institute of Biophysics of the Czech Academy of Sciences, Královopolská 135, 612 65 Brno, Czech Republic; (E.Š.); (L.K.)
- Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic;
- International Clinical Research Center, St. Anne’s University Hospital Brno, Pekařská 53, 656 91 Brno, Czech Republic
| | - Jan Víteček
- Institute of Biophysics of the Czech Academy of Sciences, Královopolská 135, 612 65 Brno, Czech Republic; (E.Š.); (L.K.)
- Correspondence: ; Tel./Fax: +420-541-517104; Fax: +420-541-517104
| |
Collapse
|
9
|
Guan L, Fan P, Liu X, Liu R, Liu Y, Bai H. Migration of Human Renal Tubular Epithelial Cells in Response to Physiological Electric Signals. Front Cell Dev Biol 2021; 9:724012. [PMID: 34595174 PMCID: PMC8476913 DOI: 10.3389/fcell.2021.724012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 08/27/2021] [Indexed: 02/05/2023] Open
Abstract
Restoration of proximal tubular cell integrity and function after ischemic injury involves cell migration and proliferation. Endogenous fields are present during embryonic development and wound healing. Electric field (EF)-induced effects on cell migration have been observed in many cell types. This study investigated the effect of physiological direct current EF (dc EF) on the motility of renal epithelial cells. Human renal tubular epithelial (HK-2) and human-derived renal epithelial (HEK-293) cells were exposed to dc EF at physiological magnitude. Cell images were recorded and analyzed using an image analyzer. Cell lysates were used to detect protein expression by western blot. Scratch wounds were created in monolayers of HK-2 cells, and wound areas of cells were measured in response to EF exposure. Cells migrated significantly faster in the presence of an EF and toward the cathode. Application of an EF led to activation of the Erk1/2, p38 MAPK, and Akt signaling pathways. Pharmacological inhibition of Erk1/2, p38 MAPK, and Akt impaired EF-induced migratory responses, such as motility rate and directedness. In addition, exposure of the monolayers to EF enhanced EF-induced HK-2 wound healing. Our results suggest that EFs augment the rate of single renal epithelium migration and induce cell cathodal migration through activation of Erk1/2, p38 MAPK, and Akt signaling. Moreover, exposure of the renal epithelium to EF facilitated closure of in vitro small wounds by enhancing cell migration.
Collapse
Affiliation(s)
- Linbo Guan
- Laboratory of Genetic Disease and Perinatal Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children of the Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Ping Fan
- Laboratory of Genetic Disease and Perinatal Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children of the Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Xinghui Liu
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Rui Liu
- Division of Peptides Related with Human Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Yu Liu
- Department of Biochemistry and Molecular Biology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, China
| | - Huai Bai
- Laboratory of Genetic Disease and Perinatal Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children of the Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
10
|
Ma L, Wang X, Zhou Y, Ji X, Cheng S, Bian D, Fan L, Zhou L, Ning C, Zhang Y. Biomimetic Ti-6Al-4V alloy/gelatin methacrylate hybrid scaffold with enhanced osteogenic and angiogenic capabilities for large bone defect restoration. Bioact Mater 2021; 6:3437-3448. [PMID: 33817419 PMCID: PMC7988351 DOI: 10.1016/j.bioactmat.2021.03.010] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/01/2021] [Accepted: 03/02/2021] [Indexed: 02/07/2023] Open
Abstract
Titanium-based scaffolds are widely used implant materials for bone defect treatment. However, the unmatched biomechanics and poor bioactivities of conventional titanium-based implants usually lead to insufficient bone integration. To tackle these challenges, it is critical to develop novel titanium-based scaffolds that meet the bioadaptive requirements for load-bearing critical bone defects. Herein, inspired by the microstructure and mechanical properties of natural bone tissue, we developed a Ti–6Al–4V alloy (TC4)/gelatin methacrylate (GelMA) hybrid scaffold with dual bionic features (GMPT) for bone defect repair. GMPT is composed of a hard 3D-printed porous TC4 metal scaffold (PT) backbone, which mimics the microstructure and mechanical properties of natural cancellous bone, and a soft GelMA hydrogel matrix infiltrated into the pores of PT that mimics the microenvironment of the extracellular matrix. Ascribed to the unique dual bionic design, the resultant GMPT demonstrates better osteogenic and angiogenic capabilities than PT, as confirmed by the in vitro and rabbit radius bone defect experimental results. Moreover, controlling the concentration of GelMA (10%) in GMPT can further improve the osteogenesis and angiogenesis of GMPT. The fundamental mechanisms were revealed by RNA-Seq analysis, which showed that the concentration of GelMA significantly influenced the expression of osteogenesis- and angiogenesis-related genes via the Pi3K/Akt/mTOR pathway. The results of this work indicate that our dual bionic implant design represents a promising strategy for the restoration of large bone defects. A novel TC4/GelMA hybrid scaffold (GMPT) was designed to mimic natural bone microstructure and mechanical property. The GMPT with 10 wt% of GelMA showed best capability for promoting osteogenesis and angiogenesis. A bioactive soft surface with suitable stiffness can activate focal adhesion pathway and the downstream PI3K/AKT pathway.
Collapse
Affiliation(s)
- Limin Ma
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510080, China
| | - Xiaolan Wang
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510080, China.,School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, China
| | - Ye Zhou
- Laboratory of Basic Medicine, General Hospital of Southern Theatre Command of the PLA, Guangzhou, 510010, China
| | - Xiongfa Ji
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510080, China
| | - Shi Cheng
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510080, China
| | - Dong Bian
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510080, China
| | - Lei Fan
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, China
| | - Lei Zhou
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, China
| | - Chengyun Ning
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, China
| | - Yu Zhang
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510080, China
| |
Collapse
|
11
|
Hahner F, Moll F, Schröder K. NADPH oxidases in the differentiation of endothelial cells. Cardiovasc Res 2020; 116:262-268. [PMID: 31393561 DOI: 10.1093/cvr/cvz213] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 05/10/2019] [Accepted: 08/08/2019] [Indexed: 12/14/2022] Open
Abstract
The differentiation of stem cells into endothelial cells involves the modulation of highly interconnected metabolic and epigenetic processes. Therefore, the differentiation of endothelial cells is a tightly controlled process, which is adjusted at multiple levels, meaning that even the smallest variation can result in major consequences. Reactive oxygen species (ROS) represent a group of second messengers that can interfere with both metabolic and epigenetic processes. Besides their generation by mitochondria, ROS are produced in a controlled manner by the family of NADPH oxidases. The different members of the NADPH oxidase family produce superoxide anions or hydrogen peroxide. Due to the specific sub-cellular localization of the different NADPH oxidases, ROS are produced at diverse sites in the cell, such as the plasma membrane or the endoplasmic reticulum. Once produced, ROS interfere with proteins, lipids, and DNA to modulate intracellular signal cascades. Accordingly, ROS represent a group of readily available and specifically localized modulators of the highly sophisticated signalling network that eventually leads to the differentiation of stem cells into endothelial cells. This review focuses on the role of NADPH oxidases in the differentiation of stem cells into endothelial cells.
Collapse
Affiliation(s)
- Fabian Hahner
- Institute for Cardiovascular Physiology, Goethe-University, Theodor-Stern Kai 7, 60590 Frankfurt, Germany
| | - Franziska Moll
- Institute for Cardiovascular Physiology, Goethe-University, Theodor-Stern Kai 7, 60590 Frankfurt, Germany
| | - Katrin Schröder
- Institute for Cardiovascular Physiology, Goethe-University, Theodor-Stern Kai 7, 60590 Frankfurt, Germany
| |
Collapse
|
12
|
Godoy-Parejo C, Deng C, Zhang Y, Liu W, Chen G. Roles of vitamins in stem cells. Cell Mol Life Sci 2020; 77:1771-1791. [PMID: 31676963 PMCID: PMC11104807 DOI: 10.1007/s00018-019-03352-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 10/12/2019] [Accepted: 10/21/2019] [Indexed: 12/13/2022]
Abstract
Stem cells can differentiate to diverse cell types in our body, and they hold great promises in both basic research and clinical therapies. For specific stem cell types, distinctive nutritional and signaling components are required to maintain the proliferation capacity and differentiation potential in cell culture. Various vitamins play essential roles in stem cell culture to modulate cell survival, proliferation and differentiation. Besides their common nutritional functions, specific vitamins are recently shown to modulate signal transduction and epigenetics. In this article, we will first review classical vitamin functions in both somatic and stem cell cultures. We will then focus on how stem cells could be modulated by vitamins beyond their nutritional roles. We believe that a better understanding of vitamin functions will significantly benefit stem cell research, and help realize their potentials in regenerative medicine.
Collapse
Affiliation(s)
- Carlos Godoy-Parejo
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Chunhao Deng
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Yumeng Zhang
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Weiwei Liu
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
- Bioimaging and Stem Cell Core Facility, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Guokai Chen
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China.
- Bioimaging and Stem Cell Core Facility, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China.
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China.
| |
Collapse
|
13
|
Jing W, Zhang Y, Cai Q, Chen G, Wang L, Yang X, Zhong W. Study of Electrical Stimulation with Different Electric-Field Intensities in the Regulation of the Differentiation of PC12 Cells. ACS Chem Neurosci 2019; 10:348-357. [PMID: 30212623 DOI: 10.1021/acschemneuro.8b00286] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The strategy of using electrical stimulation (ES) to promote the neural differentiation and regeneration of injured nerves is proven feasible. Study of the possible molecular mechanisms in relation to this ES promotion effect should be helpful for understanding the phenomenon. In this study, it was identified that the neuronal differentiation of PC12 cells was enhanced when the electric field intensity was in the range of 30-80 mV/mm, and a lower or higher electric-field intensity displayed inferior effects. Under ES, however, levels of intracellular reactive oxygen species (ROS), intracellular Ca2+ dynamics, and expression of TREK-1 were measured as being gradually increasing alongside higher electric-field intensity. In trying to understand the relationship between the ES enhancement on differentiation and these variations in cell activities, parallel experiments were conducted by introducing exogeneous H2O2 into culture systems at different concentrations. Similarly, the effects of H2O2 concentration on the neuronal differentiation of PC12 cells, intracellular ROS and Ca2+ levels, and TREK-1 expression were systematically characterized. In comparative studies, it was found in two cases that ES of 50 mV/mm for 2 h/day and H2O2 of 5 μM in culture medium shared comparable results for intracellular ROS and Ca2+ levels and TREK-1 expression. Higher H2O2 concentrations (e.g., 10 and 20 μM) demonstrated adverse effects on cell differentiation and caused DNA damage. A stronger ES (e.g., 100 mV/mm), being associated with a higher intracellular ROS level, also resulted in weaker enhancement of the neuronal differentiation of PC12 cells. These facts suggested that the intracellular ROS generated under ES might be an intermediate signal transducer involved in cascade reactions relative to cell differentiation.
Collapse
Affiliation(s)
- Wei Jing
- State Key Laboratory of Organic−Inorganic Composites; Beijing Laboratory of Biomedical Materials; Beijing University of Chemical Technology, Beijing 100029, PR China
| | - Yifan Zhang
- State Key Laboratory of Organic−Inorganic Composites; Beijing Laboratory of Biomedical Materials; Beijing University of Chemical Technology, Beijing 100029, PR China
| | - Qing Cai
- State Key Laboratory of Organic−Inorganic Composites; Beijing Laboratory of Biomedical Materials; Beijing University of Chemical Technology, Beijing 100029, PR China
| | - Guoqiang Chen
- Department of Neurosurgery, Aviation General Hospital of China Medical University, Beijing 100012, PR China
| | - Lin Wang
- Department of Neurosurgery, Aviation General Hospital of China Medical University, Beijing 100012, PR China
| | - Xiaoping Yang
- State Key Laboratory of Organic−Inorganic Composites; Beijing Laboratory of Biomedical Materials; Beijing University of Chemical Technology, Beijing 100029, PR China
| | - Weihong Zhong
- School of Mechanical and Materials Engineering, Washington State University, Pullman, Washington 99164, United States
| |
Collapse
|
14
|
Nugud A, Sandeep D, El-Serafi AT. Two faces of the coin: Minireview for dissecting the role of reactive oxygen species in stem cell potency and lineage commitment. J Adv Res 2018; 14:73-79. [PMID: 30023134 PMCID: PMC6047483 DOI: 10.1016/j.jare.2018.05.012] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 05/30/2018] [Accepted: 05/31/2018] [Indexed: 12/29/2022] Open
Abstract
Reactive oxygen species (ROS) are produced as by-products of several intracellular metabolic pathways and are reduced to more stable molecules by several protective pathways. The presence of high levels of ROS can be associated with disturbance of cell function and could lead to apoptosis. The presence of ROS within the physiological range has many effects on several signalling pathways. In stem cells, this role can range between keeping the potency of the naive stem cells to differentiation towards a certain lineage. In addition, the level of certain ROS would change according to the differentiation stage. For example, the presence of ROS can be associated with increasing the proliferation of mesenchymal stem cells, decreasing the potency of embryonic stem cells and adding to the genomic stability of induced pluripotent stem cells. ROS can enhance the differentiation of stem cells into cardiomyocytes, adipocytes, endothelial cells, keratinocytes and neurons. In the meantime, ROS inhibits osteogenesis and enhances the differentiation of cartilage to the hypertrophic stage, which is associated with chondrocyte death. Thus, ROS may form a link between naïve stem cells in the body and the environment. In addition, monitoring of ROS levels in vitro may help in tissue regeneration studies.
Collapse
Affiliation(s)
- Ahmed Nugud
- Sharjah Institute for Medical and Health Research, University of Sharjah, United Arab Emirates
| | - Divyasree Sandeep
- Sharjah Institute for Medical and Health Research, University of Sharjah, United Arab Emirates
| | - Ahmed T. El-Serafi
- Sharjah Institute for Medical and Health Research, University of Sharjah, United Arab Emirates
- Faculty of Medicine, Suez Canal University, Egypt
- Department of Clinical and Experimental Medicine, Linköping University, Sweden
| |
Collapse
|
15
|
Chen Y, Ye L, Guan L, Fan P, Liu R, Liu H, Chen J, Zhu Y, Wei X, Liu Y, Bai H. Physiological electric field works via the VEGF receptor to stimulate neovessel formation of vascular endothelial cells in a 3D environment. Biol Open 2018; 7:7/9/bio035204. [PMID: 30232195 PMCID: PMC6176943 DOI: 10.1242/bio.035204] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Electrical stimulation induces significant neovessel formation in vivo We have shown that electrical stimulation of endothelial cells functions as an important contributor to angiogenesis in monolayer culture. Because angiogenesis occurs in a three-dimensional (3D) environment, in this study we investigated the effects of a direct current (DC) electrical field (EF) on endothelial neovessel formation in 3D culture. There was a significant increase in tube formation when endothelial cells were stimulated with EF for 4 h. The lengths of the tube-like structures were augmented further by the continued EF exposure. The lengths of the tubes also increased dose-dependently in the EF-treated cultures in the field strengths of 50 mV/mm∼200 mV/mm for 6 h. Electrical fields of small physiological magnitude enhanced VEGF expression by endothelial cells in 3D culture. EF treatment also resulted in activation of VEGFR2, Akt, extracellular regulated kinase 1,2 (Erk1/2), as well as the c-Jun NH2-terminal kinase (JNK). The tyrosine kinase inhibitor SU1498 that blocks VEGFR2 activity exhibited a potent inhibition of tube growth, and the Akt inhibitor MK-2206 2HCl, the Erk1/2 inhibitor U0126 and the JNK inhibitor SB203580 significantly reduced EF-stimulated tubulogenesis. These results suggest the importance of the VEGFR2 signaling pathway during EF-induced angiogenesis. The results of this study provide novel evidence that endogenous EFs may promote blood vessel formation of endothelial cells by activating the VEGF receptor signaling pathway.
Collapse
Affiliation(s)
- Yihong Chen
- Laboratory of Genetic Disease and Perinatal Medicine and Key Laboratory of Birth Defects and Related Diseases of Women and Children of the Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, Sichuan, P.R. China.,Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu 610041, Sichuan, P.R. China
| | - Liyan Ye
- Laboratory of Genetic Disease and Perinatal Medicine and Key Laboratory of Birth Defects and Related Diseases of Women and Children of the Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, Sichuan, P.R. China
| | - Linbo Guan
- Laboratory of Genetic Disease and Perinatal Medicine and Key Laboratory of Birth Defects and Related Diseases of Women and Children of the Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, Sichuan, P.R. China
| | - Ping Fan
- Laboratory of Genetic Disease and Perinatal Medicine and Key Laboratory of Birth Defects and Related Diseases of Women and Children of the Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, Sichuan, P.R. China
| | - Rui Liu
- Division of peptides related with human disease, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Hao Liu
- Department of Biochemistry and Molecular Biology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu 610041, Sichuan, P.R. China
| | - Jinxin Chen
- Laboratory of Genetic Disease and Perinatal Medicine and Key Laboratory of Birth Defects and Related Diseases of Women and Children of the Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, Sichuan, P.R. China
| | - Yue Zhu
- Laboratory of Genetic Disease and Perinatal Medicine and Key Laboratory of Birth Defects and Related Diseases of Women and Children of the Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, Sichuan, P.R. China
| | - Xing Wei
- Laboratory of Genetic Disease and Perinatal Medicine and Key Laboratory of Birth Defects and Related Diseases of Women and Children of the Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, Sichuan, P.R. China
| | - Yu Liu
- Department of Biochemistry and Molecular Biology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu 610041, Sichuan, P.R. China
| | - Huai Bai
- Laboratory of Genetic Disease and Perinatal Medicine and Key Laboratory of Birth Defects and Related Diseases of Women and Children of the Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, Sichuan, P.R. China
| |
Collapse
|
16
|
Ye L, Guan L, Fan P, Liu Y, Xiong W, Liu R, Wei X, Zhu Y, Liu Y, Bai H. Effect of a Small Physiological Electric Field on Angiogenic Activity in First-Trimester Extravillous Trophoblast Cells. Reprod Sci 2018; 26:745-756. [PMID: 30111245 DOI: 10.1177/1933719118792102] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Electrical stimulation induces significant angiogenesis in vivo. We have shown that electrical stimulation of trophoblast cells has important functions in aspects of angiogenesis. In this study, we investigated the effects of a direct current electrical field on trophoblast angiogenic tube formation. A 6-hour exposure to electric fields ranging from 50 to 150 mV/mm dose dependently increased tube growth and network formation. Additionally, the effect was time dependent, with increased tube formation occurring between 4 and 8 hours, indicating stimulation of trophoblast cell angiogenesis. Electrical fields of small physiological magnitude stimulated vascular endothelial growth factor expression by trophoblast cells in the culture. Electric field treatment also resulted in activation of Akt, while the activity of extracellular-regulated kinase 1/2, p38, and c-Jun NH2-terminal kinase was not significantly changed. Pretreatment with the vascular endothelial growth factor receptor (VEGFR)-2 inhibitor, SU1498, resulted in potent inhibition of tube growth, and the Akt inhibitor, MK-2206 2HCl, significantly reduced electric field-stimulated tubulogenesis. These data suggest the importance of the VEGFR-2 signaling pathway during electric field-induced trophoblastic angiogenesis. This novel evidence indicates that endogenous electrical fields may promote angiogenesis of trophoblast cells by stimulating the VEGFR signaling pathway.
Collapse
Affiliation(s)
- Liyan Ye
- Laboratory of Genetic Disease and Perinatal Medicine and Key Laboratory of Birth Defects and Related Diseases of Women and Children of the Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China
| | - Linbo Guan
- Laboratory of Genetic Disease and Perinatal Medicine and Key Laboratory of Birth Defects and Related Diseases of Women and Children of the Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China
| | - Ping Fan
- Laboratory of Genetic Disease and Perinatal Medicine and Key Laboratory of Birth Defects and Related Diseases of Women and Children of the Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China
| | - Yinghui Liu
- Department of Biochemistry and Molecular Biology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Wei Xiong
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Rui Liu
- Division of Peptides Related with Human Disease, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Xing Wei
- Laboratory of Genetic Disease and Perinatal Medicine and Key Laboratory of Birth Defects and Related Diseases of Women and Children of the Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China
| | - Yue Zhu
- Laboratory of Genetic Disease and Perinatal Medicine and Key Laboratory of Birth Defects and Related Diseases of Women and Children of the Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China
| | - Yu Liu
- Department of Biochemistry and Molecular Biology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Huai Bai
- Laboratory of Genetic Disease and Perinatal Medicine and Key Laboratory of Birth Defects and Related Diseases of Women and Children of the Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China.
| |
Collapse
|
17
|
Bacakova L, Zarubova J, Travnickova M, Musilkova J, Pajorova J, Slepicka P, Kasalkova NS, Svorcik V, Kolska Z, Motarjemi H, Molitor M. Stem cells: their source, potency and use in regenerative therapies with focus on adipose-derived stem cells - a review. Biotechnol Adv 2018; 36:1111-1126. [PMID: 29563048 DOI: 10.1016/j.biotechadv.2018.03.011] [Citation(s) in RCA: 372] [Impact Index Per Article: 53.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Revised: 03/12/2018] [Accepted: 03/15/2018] [Indexed: 02/08/2023]
Abstract
Stem cells can be defined as units of biological organization that are responsible for the development and the regeneration of organ and tissue systems. They are able to renew their populations and to differentiate into multiple cell lineages. Therefore, these cells have great potential in advanced tissue engineering and cell therapies. When seeded on synthetic or nature-derived scaffolds in vitro, stem cells can be differentiated towards the desired phenotype by an appropriate composition, by an appropriate architecture, and by appropriate physicochemical and mechanical properties of the scaffolds, particularly if the scaffold properties are combined with a suitable composition of cell culture media, and with suitable mechanical, electrical or magnetic stimulation. For cell therapy, stem cells can be injected directly into damaged tissues and organs in vivo. Since the regenerative effect of stem cells is based mainly on the autocrine production of growth factors, immunomodulators and other bioactive molecules stored in extracellular vesicles, these structures can be isolated and used instead of cells for a novel therapeutic approach called "stem cell-based cell-free therapy". There are four main sources of stem cells, i.e. embryonic tissues, fetal tissues, adult tissues and differentiated somatic cells after they have been genetically reprogrammed, which are referred to as induced pluripotent stem cells (iPSCs). Although adult stem cells have lower potency than the other three stem cell types, i.e. they are capable of differentiating into only a limited quantity of specific cell types, these cells are able to overcome the ethical and legal issues accompanying the application of embryonic and fetal stem cells and the mutational effects associated with iPSCs. Moreover, adult stem cells can be used in autogenous form. These cells are present in practically all tissues in the organism. However, adipose tissue seems to be the most advantageous tissue from which to isolate them, because of its abundancy, its subcutaneous location, and the need for less invasive techniques. Adipose tissue-derived stem cells (ASCs) are therefore considered highly promising in present-day regenerative medicine.
Collapse
Affiliation(s)
- Lucie Bacakova
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14220 Prague, 4-Krc, Czech Republic.
| | - Jana Zarubova
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14220 Prague, 4-Krc, Czech Republic
| | - Martina Travnickova
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14220 Prague, 4-Krc, Czech Republic
| | - Jana Musilkova
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14220 Prague, 4-Krc, Czech Republic
| | - Julia Pajorova
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14220 Prague, 4-Krc, Czech Republic
| | - Petr Slepicka
- Department of Solid State Engineering, University of Chemistry and Technology Prague, Technicka 5, 166 28 Prague, 6-Dejvice, Czech Republic
| | - Nikola Slepickova Kasalkova
- Department of Solid State Engineering, University of Chemistry and Technology Prague, Technicka 5, 166 28 Prague, 6-Dejvice, Czech Republic
| | - Vaclav Svorcik
- Department of Solid State Engineering, University of Chemistry and Technology Prague, Technicka 5, 166 28 Prague, 6-Dejvice, Czech Republic
| | - Zdenka Kolska
- Faculty of Science, J.E. Purkyne University, Ceske mladeze 8, 400 96 Usti nad Labem, Czech Republic
| | - Hooman Motarjemi
- Clinic of Plastic Surgery, Faculty Hospital Na Bulovce, Budinova 67/2, 180 81 Prague, 8-Liben, Czech Republic
| | - Martin Molitor
- Clinic of Plastic Surgery, Faculty Hospital Na Bulovce, Budinova 67/2, 180 81 Prague, 8-Liben, Czech Republic
| |
Collapse
|
18
|
Unraveling the mechanistic effects of electric field stimulation towards directing stem cell fate and function: A tissue engineering perspective. Biomaterials 2017; 150:60-86. [PMID: 29032331 DOI: 10.1016/j.biomaterials.2017.10.003] [Citation(s) in RCA: 230] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 09/27/2017] [Accepted: 10/02/2017] [Indexed: 02/06/2023]
Abstract
Electric field (EF) stimulation can play a vital role in eliciting appropriate stem cell response. Such an approach is recently being established to guide stem cell differentiation through osteogenesis/neurogenesis/cardiomyogenesis. Despite significant recent efforts, the biophysical mechanisms by which stem cells sense, interpret and transform electrical cues into biochemical and biological signals still remain unclear. The present review critically analyses the variety of EF stimulation approaches that can be employed to evoke appropriate stem cell response and also makes an attempt to summarize the underlying concepts of this notion, placing special emphasis on stem cell based tissue engineering and regenerative medicine. This review also discusses the major signaling pathways and cellular responses that are elicited by electric stimulation, including the participation of reactive oxygen species and heat shock proteins, modulation of intracellular calcium ion concentration, ATP production and numerous other events involving the clustering or reassembling of cell surface receptors, cytoskeletal remodeling and so on. The specific advantages of using external electric stimulation in different modalities to regulate stem cell fate processes are highlighted with explicit examples, in vitro and in vivo.
Collapse
|
19
|
Glushkova OV, Khrenov MO, Vinogradova EV, Lunin SM, Fesenko EE, Novoselova EG. The role of p38 protein kinase in mouse responses to low-intensity electromagnetic radiation of the centimeter range. Biophysics (Nagoya-shi) 2016. [DOI: 10.1134/s0006350916040114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
20
|
Bekhite MM, Finkensieper A, Abou-Zaid FA, El-Shourbagy IK, El-Fiky NK, Omar KM, Sauer H, Wartenberg M. Differential effects of high and low strength magnetic fields on mouse embryonic development and vasculogenesis of embryonic stem cells. Reprod Toxicol 2016; 65:46-58. [PMID: 27346840 DOI: 10.1016/j.reprotox.2016.06.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 06/01/2016] [Accepted: 06/22/2016] [Indexed: 01/24/2023]
Abstract
Man-made magnetic fields (MFs) may exert adverse effects on mammalian embryonic development. Herein, we analysed the effect of 10mT 50Hz sinusoidal (AC) or static (DC) MFs versus 1mT MFs on embryonic development of mice. Exposure for 20days during gestation to 10mT MFs increased resorptions and dead fetuses, decreased crown-rump length and fresh weight, reduced blood vessel differentiation and caused histological changes, accompanied with diminished vascular endothelial growth factor (VEGF) protein expression in several organs. In embryonic stem (ES) cell-derived embryoid bodies exposure towards 10mT MFs increased reactive oxygen species (ROS), decreased vascular marker as well as VEGF expression and enhanced apoptosis. In conclusion, our combined data from in vivo and in vitro experiments identified VEGF as an important mediator during embryonic development that can be influenced by high strength MFs, which in consequence leads to severe abnormalities in fetus organs and blood vessel formation.
Collapse
Affiliation(s)
- Mohamed M Bekhite
- University Heart Center, Clinic of Internal Medicine I, Department of Cardiology, Jena University Hospital, Jena, Germany; Department of Zoology, Faculty of Science, Tanta University, Tanta 31527, Egypt.
| | - Andreas Finkensieper
- University Heart Center, Clinic of Internal Medicine I, Department of Cardiology, Jena University Hospital, Jena, Germany
| | - Fouad A Abou-Zaid
- Department of Zoology, Faculty of Science, Tanta University, Tanta 31527, Egypt
| | | | - Nabil K El-Fiky
- Department of Zoology, Faculty of Science, Tanta University, Tanta 31527, Egypt
| | - Khaled M Omar
- Physics Department, Faculty of Science, Tanta University, 31527, Egypt
| | - Heinrich Sauer
- Department of Physiology, Justus Liebig University Giessen, Germany
| | - Maria Wartenberg
- University Heart Center, Clinic of Internal Medicine I, Department of Cardiology, Jena University Hospital, Jena, Germany
| |
Collapse
|
21
|
Huang YH, Sharifpanah F, Becker S, Wartenberg M, Sauer H. Impact of Arachidonic Acid and the Leukotriene Signaling Pathway on Vasculogenesis of Mouse Embryonic Stem Cells. Cells Tissues Organs 2016; 201:319-32. [DOI: 10.1159/000445680] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/21/2016] [Indexed: 11/19/2022] Open
Abstract
Embryonic stem (ES) cells can differentiate into various kinds of cells, such as endothelial and hematopoietic cells. In addition, some evidence suggests that inflammatory mediators such as leukotrienes (LTs), which include the 5-lipoxygenase (LOX) family, can regulate endothelial cell differentiation. In the present study, the eicosanoid precursor arachidonic acid (AA) stimulated vasculogenesis of ES cells by increasing the number of fetal liver kinase-1+ vascular progenitor cells as well as vascular structures positive for platelet endothelial cell adhesion protein-1 and vascular endothelial cadherin. The stimulation of vasculogenesis and expression of the rate-limiting enzyme in the LT signaling pathway, 5-LOX-activating protein (FLAP), was blunted upon treatment with the FLAP inhibitors AM643 and REV5901. Vasculogenesis was significantly restored upon exogenous addition of LTs. Downstream of FLAP, the LTB4 receptor (BLT1) blocker U75302, the BLT2 receptor blocker LY255283 as well as the cysteinyl LT blocker BAY-u9773 inhibited vasculogenesis of ES cells. AA treatment of differentiating ES cells increased reactive oxygen species (ROS) generation, which was not affected upon either FLAP or cyclooxygenase-2 inhibition. Prevention of ROS generation by either the free radical scavengers vitamin E and N-(2-mercaptopropionyl)glycine or the NADPH oxidase inhibitor VAS2870 downregulated vasculogenesis of ES cells and blunted the provasculogenic effect of AA. In summary, our data demonstrate that proinflammatory AA stimulates vasculogenesis of ES cells via the LT pathway by mechanisms involving ROS generation.
Collapse
|
22
|
Sharifpanah F, De Silva S, Bekhite MM, Hurtado-Oliveros J, Preissner KT, Wartenberg M, Sauer H. Stimulation of vasculogenesis and leukopoiesis of embryonic stem cells by extracellular transfer RNA and ribosomal RNA. Free Radic Biol Med 2015; 89:1203-17. [PMID: 26524400 DOI: 10.1016/j.freeradbiomed.2015.10.423] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 10/27/2015] [Accepted: 10/28/2015] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Cell injury releases nucleic acids supporting inflammation and stem cell activation. Here, the impact of extracellular ribonucleic acid, especially transfer RNA (ex-tRNA), on vasculogenesis and leukopoiesis of mouse embryonic stem (ES) cells was investigated. APPROACH AND RESULTS ex-tRNA, whole cell RNA and ribosomal RNA (ex-rRNA) but not DNA increased CD31-positive vascular structures in embryoid bodies. Ex-tRNA and ex-rRNA increased numbers of VEGFR2(+), CD31(+) and VE-cadherin(+) vascular cells as well as CD18(+), CD45(+) and CD68(+) cells, indicating leukocyte/macrophage differentiation. This was paralleled by mRNA and protein expression of hypoxia-inducible factor-1α (HIF-1α), vascular endothelial growth factor-165 (VEGF165) and neuropilin 1 (NRP1), phosphorylation of phosphatidyl inositol 3-kinase (PI3K) and VEGF receptor 2 (VEGFR2) as well as mRNA expression of α-smooth muscle actin (α-SMA). ex-tRNA was taken up by endosomes, increased expression of the pro-angiogenic semaphorin B4 receptor plexin B1 as well as the ephrin-type B receptor 4 (EphB4) and ephrinB2 ligand and enhanced cell migration, which was inhibited by the VEGFR2 antagonist SU5614 and the PI3K inhibitor LY294002. This likewise abolished the effects of ex-tRNA on vasculogenesis and leukopoiesis of ES cells. Ex-tRNA increased NOX1, NOX2, NOX4 and DUOX2 mRNA and boosted the generation of superoxide and hydrogen peroxide which was inhibited by radical scavengers, the NADPH oxidase inhibitors apocynin, VAS2870, ML171, and plumbagin as well as shRNA silencing of NOX1 and NOX4. CONCLUSIONS Our findings indicate that ex-tRNA treatment induces vasculogenesis and leukopoiesis of ES cells via superoxide/hydrogen peroxide generated by NADPH oxidase and activation of VEGFR2 and PI3K.
Collapse
Affiliation(s)
- Fatemeh Sharifpanah
- Department of Physiology, Medical School, Justus Liebig University, Giessen, Germany
| | - Sepali De Silva
- Department of Physiology, Medical School, Justus Liebig University, Giessen, Germany
| | - Mohamed M Bekhite
- Clinic of Internal Medicine I, Cardiology Division, Friedrich Schiller University, Jena, Germany; Department of Zoology, Faculty of Science, Tanta University, Tanta, Egypt
| | | | - Klaus T Preissner
- Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| | - Maria Wartenberg
- Clinic of Internal Medicine I, Cardiology Division, Friedrich Schiller University, Jena, Germany
| | - Heinrich Sauer
- Department of Physiology, Medical School, Justus Liebig University, Giessen, Germany.
| |
Collapse
|
23
|
Rehman AA, Elmore KB, Mattei TA. The effects of alternating electric fields in glioblastoma: current evidence on therapeutic mechanisms and clinical outcomes. Neurosurg Focus 2015; 38:E14. [PMID: 25727223 DOI: 10.3171/2015.1.focus14742] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Glioblastoma is both the most common and most lethal primary CNS malignancy in adults, accounting for 45.6% of all malignant CNS tumors, with a 5-year survival rate of only 5.0%, despite the utilization of multimodal therapy including resection, chemotherapy, and radiation. Currently available treatment options for glioblastoma often remain limited, offering brief periods of improved survival, but with substantial side effects. As such, improvements in current treatment strategies or, more likely, the implementation of novel strategies altogether are warranted. In this topic review, the authors provide a comprehensive review on the potential of alternating electric fields (AEFs) in the treatment of glioblastoma. Alternating electric fields-also known as tumor-treating fields (TTFs)-represent an entirely original therapeutic modality with preliminary studies suggesting comparable, and at times improved, efficacy to standard chemotherapeutic agents in the treatment of recurrent glioblastoma. A recent multicenter, Phase III, randomized clinical trial comparing NovoTTF-100A monotherapy to physician's best choice chemotherapy in patients with recurrent glioblastoma revealed that AEFs have similar efficacy to standard chemotherapeutic agents with a more favorable side-effects profile and improved quality of life. In particular, AEFs were shown to have limited systemic adverse effects, with the most common side effect being contact dermatitis on the scalp at the sites of transducer placement. This study prompted FDA approval of the NovoTTF-100A system in April 2011 as a standalone therapy for treatment of recurrent glioblastoma refractory to surgical and radiation treatment. In addition to discussing the available clinical evidence regarding the utilization of AEFs in glioblastoma, this article provides essential information regarding the supposed therapeutic mechanism as well as modes of potential tumor resistance to such novel therapy, delineating future perspectives regarding basic science research on the issue.
Collapse
Affiliation(s)
- Azeem A Rehman
- University of Illinois College of Medicine at Peoria, Illinois; and
| | | | | |
Collapse
|
24
|
Bekhite MM, Müller V, Tröger SH, Müller JP, Figulla HR, Sauer H, Wartenberg M. Involvement of phosphoinositide 3-kinase class IA (PI3K 110α) and NADPH oxidase 1 (NOX1) in regulation of vascular differentiation induced by vascular endothelial growth factor (VEGF) in mouse embryonic stem cells. Cell Tissue Res 2015; 364:159-74. [PMID: 26553657 DOI: 10.1007/s00441-015-2303-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 09/28/2015] [Indexed: 02/02/2023]
Abstract
The impact of reactive oxygen species and phosphoinositide 3-kinase (PI3K) in differentiating embryonic stem (ES) cells is largely unknown. Here, we show that the silencing of the PI3K catalytic subunit p110α and nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 1 (NOX1) by short hairpin RNA or pharmacological inhibition of NOX and ras-related C3 botulinum toxin substrate 1 (Rac1) abolishes superoxide production by vascular endothelial growth factor (VEGF) in mouse ES cells and in ES-cell-derived fetal liver kinase-1(+) (Flk-1(+)) vascular progenitor cells, whereas the mitochondrial complex I inhibitor rotenone does not have an effect. Silencing p110α or inhibiting Rac1 arrests vasculogenesis at initial stages in embryoid bodies, even under VEGF treatment, as indicated by platelet endothelial cell adhesion molecule-1 (PECAM-1)-positive areas and branching points. In the absence of p110α, tube-like structure formation on matrigel and cell migration of Flk-1(+) cells in scratch migration assays are totally impaired. Silencing NOX1 causes a reduction in PECAM-1-positive areas, branching points, cell migration and tube length upon VEGF treatment, despite the expression of vascular differentiation markers. Interestingly, silencing p110α but not NOX1 inhibits the activation of Rac1, Ras homologue gene family member A (RhoA) and Akt leading to the abrogation of VEGF-induced lamellipodia structure formation. Thus, our data demonstrate that the PI3K p110α-Akt/Rac1 and NOX1 signalling pathways play a pivotal role in VEGF-induced vascular differentiation and cell migration. Rac1, RhoA and Akt phosphorylation occur downstream of PI3K and upstream of NOX1 underscoring a role of PI3K p110α in the regulation of cell polarity and migration.
Collapse
Affiliation(s)
- Mohamed M Bekhite
- University Heart Center, Clinic of Internal Medicine I, Department of Cardiology, Friedrich Schiller University Jena, Erlanger Allee 101, 07743, Jena, Germany. .,Department of Zoology, Faculty of Science, Tanta University, Tanta, 31527, Egypt.
| | - Veronika Müller
- University Heart Center, Clinic of Internal Medicine I, Department of Cardiology, Friedrich Schiller University Jena, Erlanger Allee 101, 07743, Jena, Germany
| | - Sebastian H Tröger
- University Heart Center, Clinic of Internal Medicine I, Department of Cardiology, Friedrich Schiller University Jena, Erlanger Allee 101, 07743, Jena, Germany
| | - Jörg P Müller
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine, Friedrich Schiller University Jena, Jena, Germany
| | - Hans-Reiner Figulla
- University Heart Center, Clinic of Internal Medicine I, Department of Cardiology, Friedrich Schiller University Jena, Erlanger Allee 101, 07743, Jena, Germany
| | - Heinrich Sauer
- Department of Physiology, Faculty of Medicine, Justus Liebig University, Giessen, Germany
| | - Maria Wartenberg
- University Heart Center, Clinic of Internal Medicine I, Department of Cardiology, Friedrich Schiller University Jena, Erlanger Allee 101, 07743, Jena, Germany
| |
Collapse
|
25
|
Kanno M, Yazawa T, Kawabe S, Imamichi Y, Usami Y, Ju Y, Matsumura T, Mizutani T, Fujieda S, Miyamoto K. Sex-determining region Y-box 2 and GA-binding proteins regulate the transcription of liver receptor homolog-1 in early embryonic cells. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2014; 1839:406-14. [DOI: 10.1016/j.bbagrm.2014.03.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Revised: 03/26/2014] [Accepted: 03/27/2014] [Indexed: 01/08/2023]
|
26
|
Piccoli C, Agriesti F, Scrima R, Falzetti F, Di Ianni M, Capitanio N. To breathe or not to breathe: the haematopoietic stem/progenitor cells dilemma. Br J Pharmacol 2014; 169:1652-71. [PMID: 23714011 DOI: 10.1111/bph.12253] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2013] [Revised: 05/11/2013] [Accepted: 05/16/2013] [Indexed: 12/13/2022] Open
Abstract
UNLABELLED Adult haematopoietic stem/progenitor cells (HSPCs) constitute the lifespan reserve for the generation of all the cellular lineages in the blood. Although massive progress in identifying the cluster of master genes controlling self-renewal and multipotency has been achieved in the past decade, some aspects of the physiology of HSPCs still need to be clarified. In particular, there is growing interest in the metabolic profile of HSPCs in view of their emerging role as determinants of cell fate. Indeed, stem cells and progenitors have distinct metabolic profiles, and the transition from stem to progenitor cell corresponds to a critical metabolic change, from glycolysis to oxidative phosphorylation. In this review, we summarize evidence, reported in the literature and provided by our group, highlighting the peculiar ability of HSPCs to adapt their mitochondrial oxidative/bioenergetic metabolism to survive in the hypoxic microenvironment of the endoblastic niche and to exploit redox signalling in controlling the balance between quiescence versus active cycling and differentiation. Especial prominence is given to the interplay between hypoxia inducible factor-1, globins and NADPH oxidases in managing the mitochondrial dioxygen-related metabolism and biogenesis in HSPCs under different ambient conditions. A mechanistic model is proposed whereby 'mitochondrial differentiation' is a prerequisite in uncommitted stem cells, paving the way for growth/differentiation factor-dependent processes. Advancing the understanding of stem cell metabolism will, hopefully, help to (i) improve efforts to maintain, expand and manipulate HSPCs ex vivo and realize their potential therapeutic benefits in regenerative medicine; (ii) reprogramme somatic cells to generate stem cells; and (iii) eliminate, selectively, malignant stem cells. LINKED ARTICLES This article is part of a themed section on Emerging Therapeutic Aspects in Oncology. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2013.169.issue-8.
Collapse
Affiliation(s)
- C Piccoli
- Department of Medical and Experimental Medicine, University of Foggia, Foggia, Italy.
| | | | | | | | | | | |
Collapse
|
27
|
Scott E, Loya K, Mountford J, Milligan G, Baker AH. MicroRNA regulation of endothelial homeostasis and commitment-implications for vascular regeneration strategies using stem cell therapies. Free Radic Biol Med 2013; 64:52-60. [PMID: 23665307 DOI: 10.1016/j.freeradbiomed.2013.04.037] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Revised: 04/22/2013] [Accepted: 04/30/2013] [Indexed: 11/23/2022]
Abstract
Human embryonic (hESC) and induced pluripotent (hiPSC) stem cells have broad therapeutic potential in the treatment of a range of diseases, including those of the vascular system. Both hESCs and hiPSCs have the capacity for indefinite self-renewal, in addition to their ability to differentiate into any adult cell type. These cells could provide a potentially unlimited source of cells for transplantation and, therefore, provide novel treatments, e.g. in the production of endothelial cells for vascular regeneration. MicroRNAs are short, noncoding RNAs that act posttranscriptionally to control gene expression and thereby exert influence over a wide range of cellular processes, including maintenance of pluripotency and differentiation. Expression patterns of these small RNAs are tissue specific, and changes in microRNA levels have often been associated with disease states in humans, including vascular pathologies. Here, we review the roles of microRNAs in endothelial cell function and vascular disease, as well as their role in the differentiation of pluripotent stem cells to the vascular endothelial lineage. Furthermore, we discuss the therapeutic potential of stem cells and how knowledge and manipulation of microRNAs in stem cells may enhance their capacity for vascular regeneration.
Collapse
Affiliation(s)
- Elizabeth Scott
- Institute of Cardiovascular and Medical Sciences, British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow G12 8TA, UK
| | | | | | | | | |
Collapse
|
28
|
Sauer H, Ravindran F, Beldoch M, Sharifpanah F, Jedelská J, Strehlow B, Wartenberg M. α2-Macroglobulin enhances vasculogenesis/angiogenesis of mouse embryonic stem cells by stimulation of nitric oxide generation and induction of fibroblast growth factor-2 expression. Stem Cells Dev 2013; 22:1443-54. [PMID: 23379699 DOI: 10.1089/scd.2012.0640] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
α2-macroglobulin (α2M) is an acute-phase protein released upon challenges like cardiac hypertrophy and infarction. α2M signals via the low density lipoprotein receptor-related protein (LRP-1) and may induce stem cell activation. In the present study, the effects of α2M on vasculogenesis/angiogenesis and underlying signaling cascades were investigated in mouse embryonic stem (ES) cells. LRP-1 was expressed in ES cells and upregulated during differentiation. α2M dose dependently increased CD31-positive vascular structures in ES cell-derived embryoid bodies, the early cardiovascular markers isl-1, Nkx-2.5, and flk-1 as well as numbers of VE-cadherin and flk-1-positive cells, but downregulated α-smooth muscle actin. Enhancement of vasculogenesis/angiogenesis by α2M was abolished by the LRP-1 antagonist receptor-associated protein (RAP) and LRP-1 blocking antibody. Notably, α2M stimulated vascular growth in the chicken chorioallantois membrane assay, but not in a human umbilical vein endothelial cell spheroid model. α2M increased fibroblast growth factor-2 (FGF-2) protein expression, which was abolished by RAP, induced nitric oxide (NO) generation as determined by 4,5-diaminofluorescein diacetate microfluorometry, and activated nitric oxide synthase-3 (NOS-3) as well as extracellular-regulated kinase 1,2 (ERK1/2) and phosphatidyl inositol 3-kinase (PI3K). NO generation, the increase in FGF-2 expression, and the stimulation of vasculogenesis/angiogenesis by α2M were blunted by the NO synthase inhibitor L-NAME, the ERK1/2 inhibitor PD98059, and the PI3K inhibitor LY294002. Furthermore, vasculogenesis/angiogenesis by α2M was inhibited in the presence of the FGF receptor 1 antagonist SU5402. In conclusion, α2M stimulates endothelial and early cardiac, but not smooth muscle differentiation of ES cells through generation of NO, activation of ERK1/2 as well as PI3K, and induction of FGF-2 expression.
Collapse
Affiliation(s)
- Heinrich Sauer
- Department of Physiology, Justus Liebig University Giessen, Giessen, Germany.
| | | | | | | | | | | | | |
Collapse
|
29
|
Rath D, Barcikowski S, de Graaf S, Garrels W, Grossfeld R, Klein S, Knabe W, Knorr C, Kues W, Meyer H, Michl J, Moench-Tegeder G, Rehbock C, Taylor U, Washausen S. Sex selection of sperm in farm animals: status report and developmental prospects. Reproduction 2013; 145:R15-30. [DOI: 10.1530/rep-12-0151] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Pre-selection of spermatozoa based on the relative DNA difference between X- and Y-chromosome bearing populations by flow cytometry is an established method that has been introduced into commercial cattle production. Although several important improvements have increased the sort efficiency, the fertilising ability of sexed spermatozoa based on offspring per insemination is still behind farmers' expectations. The main stress factors, especially on mitochondria, that reduce the lifespan of spermatozoa are described, and new technical as well as biological solutions to maintain the natural sperm integrity and to increase the sorting efficiency are discussed. Among these methods are the identification of Y-chromosome bearing spermatozoa by bi-functionalised gold nanoparticles and triplex hybridisationin vivoas well as new laser-controlled deflection system that replaces the deflection of spermatozoa in the electrostatic field. Additionally, as well as a new nonsurgical transfer system of spermatozoa into the oviduct of cows has been developed and allows a significant reduction of spermatozoa per transfer. Altogether, the improvements made in the recent years will allow a broader use of sex-sorted spermatozoa even in those species that require more cells than cows and sheep.
Collapse
|
30
|
Urao N, Ushio-Fukai M. Redox regulation of stem/progenitor cells and bone marrow niche. Free Radic Biol Med 2013; 54:26-39. [PMID: 23085514 PMCID: PMC3637653 DOI: 10.1016/j.freeradbiomed.2012.10.532] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Revised: 10/02/2012] [Accepted: 10/05/2012] [Indexed: 01/19/2023]
Abstract
Bone marrow (BM)-derived stem and progenitor cell functions including self-renewal, differentiation, survival, migration, proliferation, and mobilization are regulated by unique cell-intrinsic and -extrinsic signals provided by their microenvironment, also termed the "niche." Reactive oxygen species (ROS), especially hydrogen peroxide (H(2)O(2)), play important roles in regulating stem and progenitor cell functions in various physiologic and pathologic responses. The low level of H(2)O(2) in quiescent hematopoietic stem cells (HSCs) contributes to maintaining their "stemness," whereas a higher level of H(2)O(2) within HSCs or their niche promotes differentiation, proliferation, migration, and survival of HSCs or stem/progenitor cells. Major sources of ROS are NADPH oxidase and mitochondria. In response to ischemic injury, ROS derived from NADPH oxidase are increased in the BM microenvironment, which is required for hypoxia and hypoxia-inducible factor-1α expression and expansion throughout the BM. This, in turn, promotes progenitor cell expansion and mobilization from BM, leading to reparative neovascularization and tissue repair. In pathophysiological states such as aging, atherosclerosis, heart failure, hypertension, and diabetes, excess amounts of ROS create an inflammatory and oxidative microenvironment, which induces cell damage and apoptosis of stem and progenitor cells. Understanding the molecular mechanisms of how ROS regulate the functions of stem and progenitor cells and their niche in physiological and pathological conditions will lead to the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Norifumi Urao
- Department of Pharmacology, Center for Lung and Vascular Biology, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, IL 60612, USA
| | | |
Collapse
|
31
|
Balint R, Cassidy NJ, Cartmell SH. Electrical stimulation: a novel tool for tissue engineering. TISSUE ENGINEERING PART B-REVIEWS 2012; 19:48-57. [PMID: 22873689 DOI: 10.1089/ten.teb.2012.0183] [Citation(s) in RCA: 242] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
New advances in tissue engineering are being made through the application of different types of electrical stimuli to influence cell proliferation and differentiation. Developments made in the last decade have allowed us to improve the structure and functionality of tissue-engineered products through the use of growth factors, hormones, drugs, physical stimuli, bioreactor use, and two-dimensional (2-D) and three-dimensional (3-D) artificial extracellular matrices (with various material properties and topography). Another potential type of stimulus is electricity, which is important in the physiology and development of the majority of all human tissues. Despite its great potential, its role in tissue regeneration and its ability to influence cell migration, orientation, proliferation, and differentiation has rarely been considered in tissue engineering. This review highlights the importance of endogenous electrical stimulation, gathering the current knowledge on its natural occurrence and role in vivo, discussing the novel methods of delivering this stimulus and examining its cellular and tissue level effects, while evaluating how the technique could benefit the tissue engineering discipline in the future.
Collapse
Affiliation(s)
- Richard Balint
- Materials Science Centre, University of Manchester, Manchester, United Kingdom
| | | | | |
Collapse
|
32
|
Sheikh AQ, Taghian T, Hemingway B, Cho H, Kogan AB, Narmoneva DA. Regulation of endothelial MAPK/ERK signalling and capillary morphogenesis by low-amplitude electric field. J R Soc Interface 2012; 10:20120548. [PMID: 22993248 DOI: 10.1098/rsif.2012.0548] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Low-amplitude electric field (EF) is an important component of wound-healing response and can promote vascular tissue repair; however, the mechanisms of action on endothelium remain unclear. We hypothesized that physiological amplitude EF regulates angiogenic response of microvascular endothelial cells via activation of the mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) pathway. A custom set-up allowed non-thermal application of EF of high (7.5 GHz) and low (60 Hz) frequency. Cell responses following up to 24 h of EF exposure, including proliferation and apoptosis, capillary morphogenesis, vascular endothelial growth factor (VEGF) expression and MAPK pathways activation were quantified. A db/db mouse model of diabetic wound healing was used for in vivo validation. High-frequency EF enhanced capillary morphogenesis, VEGF release, MEK-cRaf complex formation, MEK and ERK phosphorylation, whereas no MAPK/JNK and MAPK/p38 pathways activation was observed. The endothelial response to EF did not require VEGF binding to VEGFR2 receptor. EF-induced MEK phosphorylation was reversed in the presence of MEK and Ca(2+) inhibitors, reduced by endothelial nitric oxide synthase inhibition, and did not depend on PI3K pathway activation. The results provide evidence for a novel intracellular mechanism for EF regulation of endothelial angiogenic response via frequency-sensitive MAPK/ERK pathway activation, with important implications for EF-based therapies for vascular tissue regeneration.
Collapse
Affiliation(s)
- Abdul Q Sheikh
- Biomedical Engineering, SEEBME, University of Cincinnati, 2901 Woodside Drive, Cincinnati, OH 45221-0012, USA
| | | | | | | | | | | |
Collapse
|
33
|
Chen H, Liu R, Liu J, Tang J. Growth inhibition of malignant melanoma by intermediate frequency alternating electric fields, and the underlying mechanisms. J Int Med Res 2012; 40:85-94. [PMID: 22429348 DOI: 10.1177/147323001204000109] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE This study investigated the antitumour effects of intermediate frequency alternating electric fields (IF-AEF) in a murine melanoma cell line (B16F10) and a mouse tumour model. METHODS IF-AEF was applied at 100 kHz. Proliferation of B16F10 cells in vitro was assessed by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide assay. IF-AEF was applied in vivo to mice bearing B16F10 tumours. Terminal deoxy nucleotidyl transferase-mediated dUTP nick end labelling (TUNEL) assay for apoptosis, and immunohistochemical detection of CD34 and vascular endothelial growth factor (VEGF), were performed. RESULTS IF-AEF inhibited the proliferation of B16F10 cells in an electrical intensity and time-dependent manner. Treatment with IF-AEF for 7 days significantly inhibited the growth of tumours compared with untreated controls. IF-AEF induced apoptosis in vivo and reduced CD34-positive cell numbers; CD34 is a special marker of microvessel density. CONCLUSION IF-AEF reduced microvessel density related to tumour growth and may serve as a therapeutic strategy for cancer treatment.
Collapse
Affiliation(s)
- H Chen
- Department of Oncology, Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| | | | | | | |
Collapse
|
34
|
Boga A, Binokay S, Emre M, Sertdemir Y. The embryonic development of Xenopus laevis under a low frequency electric field. In Vitro Cell Dev Biol Anim 2012; 48:385-91. [DOI: 10.1007/s11626-012-9519-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Accepted: 05/18/2012] [Indexed: 10/28/2022]
|
35
|
Korraah A, Odenthal M, Kopp M, Vigneswaran N, Sacks PG, Dienes HP, Stützer H, Niedermeier W. Induction of apoptosis and up-regulation of cellular proliferation in oral leukoplakia cell lines inside electric field. Oral Surg Oral Med Oral Pathol Oral Radiol 2012; 113:644-54. [PMID: 22668623 DOI: 10.1016/j.oooo.2011.11.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Revised: 11/15/2011] [Accepted: 11/17/2011] [Indexed: 11/26/2022]
Abstract
OBJECTIVE In dentistry, metallic alloys are used for dentures, restorative materials, and orthodontic devices. Electric voltages up to 950 mV may occur between different dental alloys in the oral cavity. This study aimed to investigate physiologic reactions of oral leukoplakia cells in vitro to electric fields. STUDY DESIGN A human leukoplakia cell line (MSK-LEUK1), cultivated in keratinocyte growth medium (KGM-2) supplemented with growth factors in 5% CO(2) humidified air at 37°C, was exposed to electric field strength of 1-20 V/m for 24 hours in a custom-made pulse chamber. The cells were then analyzed for proliferation with the use of BrdU assay and for apoptosis with the use of TUNEL assay. Findings were assessed with the use of fluorescent microscopy. Ultrastructural changes were studied by transmission electron microscopy. RESULTS Electric field strength of 1-10 V/m led to up-regulation of cell proliferation rate from 10.64% to 44.06% (P = .0001). The apoptotic index increased significantly (P = .0001) from 20.03% at 1 V/m to 46.56% at 10 V/m. Individual cell keratinization was seen in leukoplakia cells treated with 16 V/m. CONCLUSIONS Oral galvanism induces subcellular changes in oral precancer cells in vitro that closely simulate some of the morphologic features of oral squamous cell carcinoma cells in vivo.
Collapse
Affiliation(s)
- Ahmed Korraah
- Department of Prosthetic Dentistry, Dental School, University of Cologne, Germany
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Bir SC, Kolluru GK, Fang K, Kevil CG. Redox balance dynamically regulates vascular growth and remodeling. Semin Cell Dev Biol 2012; 23:745-57. [PMID: 22634069 DOI: 10.1016/j.semcdb.2012.05.003] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Revised: 05/11/2012] [Accepted: 05/16/2012] [Indexed: 02/07/2023]
Abstract
Vascular growth and remodeling responses entail several complex biochemical, molecular, and cellular responses centered primarily on endothelial cell activation and function. Recent studies reveal that changes in endothelial cell redox status critically influence numerous cellular events that are important for vascular growth under different conditions. It has been known for some time that oxidative stress actively participates in many aspects of angiogenesis and vascular remodeling. Initial studies in this field were largely exploratory with minimal insight into specific molecular mechanisms and how these responses could be regulated. However, it is now clear that intracellular redox mechanisms involving hypoxia, NADPH oxidases (NOX), xanthine oxidase (XO), nitric oxide and its synthases, and intracellular antioxidant defense pathways collectively orchestrate a redox balance system whereby reactive oxygen and nitrogen species integrate cues controlling vascular growth and remodeling. In this review, we discuss key redox regulation pathways that are centrally important for vascular growth in tissue health and disease. Important unresolved questions and issues are also addressed that requires future investigation.
Collapse
Affiliation(s)
- Shyamal C Bir
- Department of Pathology, LSU Health Sciences Center-Shreveport, 1501 Kings Hwy.,Shreveport, LA 71130, United States
| | | | | | | |
Collapse
|
37
|
Takayama Y, Saito A, Moriguchi H, Kotani K, Suzuki T, Mabuchi K, Jimbo Y. Simultaneous induction of calcium transients in embryoid bodies using microfabricated electrode substrates. J Biosci Bioeng 2011; 112:624-9. [PMID: 21903466 DOI: 10.1016/j.jbiosc.2011.08.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2011] [Revised: 08/05/2011] [Accepted: 08/09/2011] [Indexed: 12/20/2022]
Abstract
Precise control of differentiation processes of pluripotent stem cells is a key component for the further development of regenerative medicine. For this purpose, combining a cell-aggregate-size treatment for regulating intercellular signal transmissions and an electrical stimulation technique for inducing cellular responses is a promising approach. In the present study, we developed microfabricated electrode substrates that allow simultaneous stimulation of embryoid bodies (EBs) of P19 cells. Mouse embryonal carcinoma P19 cells can be induced to differentiate into three germ layers and serve as a promising stem cell model. Microcavity-array patterns were fabricated onto indium-tin-oxide (ITO) substrates using a standard photo-lithography technique, and uniform-sized EBs of P19 cells were inserted into each microcavity. Electrical stimulation was applied to the EBs through substrate electrodes and stimulus-induced intracellular calcium transients were monitored. We confirmed that the developed electrode device could simultaneously stimulate smaller (200μm diameter) and larger (500μm diameter) EBs inserted in the microcavities and induce specific spatio-temporal patterns of intracellular calcium transients in the EBs with fine reproducibility. We concluded that the developed microcavity array with embedded electrodes could simultaneously and effectively stimulate uniform-sized EBs inserted in it. Therefore, it is a promising experimental tool for precisely controlling cell differentiation processes.
Collapse
Affiliation(s)
- Yuzo Takayama
- Graduate School of Information Science and Technology, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.
| | | | | | | | | | | | | |
Collapse
|
38
|
Bekhite MM, Finkensieper A, Binas S, Müller J, Wetzker R, Figulla HR, Sauer H, Wartenberg M. VEGF-mediated PI3K class IA and PKC signaling in cardiomyogenesis and vasculogenesis of mouse embryonic stem cells. J Cell Sci 2011; 124:1819-30. [PMID: 21540297 DOI: 10.1242/jcs.077594] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
VEGF-, phosphoinositide 3-kinase (PI3K)- and protein kinase C (PKC)-regulated signaling in cardiac and vascular differentiation was investigated in mouse ES cells and in ES cell-derived Flk-1⁺ cardiovascular progenitor cells. Inhibition of PI3K by wortmannin and LY294002, disruption of PI3K catalytic subunits p110α and p110δ using short hairpin RNA (shRNA), or inhibition of p110α with compound 15e and of p110δ with IC-87114 impaired cardiac and vascular differentiation. By contrast, TGX-221, an inhibitor of p110β, and shRNA knockdown of p110β were without significant effects. Antagonists of the PKC family, i.e. bisindolylmaleimide-1 (BIM-1), GÖ 6976 (targeting PKCα/βII) and rottlerin (targeting PKCδ) abolished vasculogenesis, but not cardiomyogenesis. Inhibition of Akt blunted cardiac as well as vascular differentiation. VEGF induced phosphorylation of PKCα/βII and PKCδ but not PKCζ. This was abolished by PI3K inhibitors and the VEGFR-2 antagonist SU5614. Furthermore, phosphorylation of Akt and phosphoinositide-dependent kinase-1 (PDK1) was blunted upon inhibition of PI3K, but not upon inhibition of PKC by BIM-1, suggesting that activation of Akt and PDK1 by VEGF required PI3K but not PKC. In summary, we demonstrate that PI3K catalytic subunits p110α and p110δ are central to cardiovasculogenesis of ES cells. Akt downstream of PI3K is involved in both cardiomyogenesis and vasculogenesis, whereas PKC is involved only in vasculogenesis.
Collapse
Affiliation(s)
- Mohamed M Bekhite
- Department of Internal Medicine I, Cardiology Division, Friedrich Schiller University, 07743 Jena, Germany
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Kane NM, Xiao Q, Baker AH, Luo Z, Xu Q, Emanueli C. Pluripotent stem cell differentiation into vascular cells: a novel technology with promises for vascular re(generation). Pharmacol Ther 2011; 129:29-49. [PMID: 20965210 DOI: 10.1016/j.pharmthera.2010.10.004] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2010] [Accepted: 10/05/2010] [Indexed: 12/15/2022]
Abstract
Several types of stem and progenitor cells are currently under investigation for their potential to accomplish vascular regeneration. This review focuses on embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs). We will discuss the technologies allowing for their derivation, culture expansion and maintenance in a pluripotent status. Moreover, both ESCs and iPSCs can be differentiated in endothelial cells (ECs) and mural cell, including vascular smooth muscle cells (VSMCs). Here, we will describe the involvements of growth factors (vascular endothelial growth factors-VEGFs-, platet-derived growth factors-PDGFs-), Wnt and Notch signal pathways, reactive oxygen species (ROS), histone deacetylases (HDACs), and microRNAs (miRNAs) in vascular cell differentiation from pluripotent stem cells. We will additionally describe the therapeutic potential of stem cells for vascular medicine.
Collapse
Affiliation(s)
- Nicole M Kane
- British Heart Foundation Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland, UK
| | | | | | | | | | | |
Collapse
|
40
|
Milosevic N, Bekhite MM, Sharifpanah F, Ruhe C, Wartenberg M, Sauer H. Redox stimulation of cardiomyogenesis versus inhibition of vasculogenesis upon treatment of mouse embryonic stem cells with thalidomide. Antioxid Redox Signal 2010; 13:1813-27. [PMID: 20722506 DOI: 10.1089/ars.2010.3139] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Thalidomide [α-(N-phthalimido)-glutarimide] exerts antiangiogenic properties and causes cardiac malformations in embryos. Herein the effects of thalidomide on cardiovascular differentiation were investigated in mouse embryonic stem (ES) cell-derived embryoid bodies. Thalidomide inhibited the formation of capillary-like blood vessels and decreased tumor-induced angiogenesis in confrontation cultures of embryoid bodies and multicellular prostate tumor spheroids, but stimulated cardiomyogenesis of ES cells. The number of CD31- and CD144-positive endothelial cells was not impaired, suggesting that thalidomide acted on vascular tube formation and cell migration rather than endothelial differentiation. Thalidomide increased reactive oxygen species generation, which was abolished by the NADPH oxidase inhibitor VAS2870 and the complex I respiratory chain inhibitor rotenone. Conversely, thalidomide decreased nitric oxide (NO) generation and endothelial NO synthase activity. VAS2870 abrogated thalidomide stimulation of cardiomyogenesis, whereas inhibition of vasculogenesis persisted. In NOX-1 and NOX-4 shRNA gene-inactivated ES cells, cardiomyogenesis was severely impaired and thalidomide failed to stimulate cardiac cell commitment. The NO donor S-nitrosopenicillamine reversed the antiangiogenic effect of thalidomide and increased capillary structure formation, whereas scavenging NO by 2-(4-carboxyphenyl)-4,4,5,5-tetramethylimidazoline-1-oxyl-3-oxide and inhibition of endothelial NO synthase by N(G)-nitro-l-arginine methyl ester decreased cardiovascular differentiation. Our data demonstrate that thalidomide causes an imbalance of reactive oxygen species/NO generation, thus stimulating cardiomyogenesis and impairing vascular sprout formation.
Collapse
Affiliation(s)
- Nada Milosevic
- Department of Physiology, Justus Liebig University Giessen , Giessen, Germany
| | | | | | | | | | | |
Collapse
|
41
|
Bekhite MM, Finkensieper A, Abou-Zaid FA, El-Shourbagy IK, Omar KM, Figulla HR, Sauer H, Wartenberg M. Static electromagnetic fields induce vasculogenesis and chondro-osteogenesis of mouse embryonic stem cells by reactive oxygen species-mediated up-regulation of vascular endothelial growth factor. Stem Cells Dev 2010; 19:731-43. [PMID: 19788349 DOI: 10.1089/scd.2008.0266] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Electromagnetic fields (EMFs) are used to treat bone diseases. Herein, the effects of static EMFs on chondroosteogenesis and vasculogenesis of embryonic stem (ES) cells and bone mineralization of mouse fetuses were investigated. Treatment of differentiating ES cells with static EMFs (0.4-2 mT) stimulated vasculogenesis and chondro-osteogenesis and increased reactive oxygen species (ROS), which was abolished by the free radical scavengers trolox, 1,10-phenanthroline (phen), and the NAD(P)H oxidase inhibitor diphenylen iodonium (DPI). In contrast, EMFs of 10 mT field strength exerted inhibitory effects on vasculogenesis and chondro-osteogenesis despite robust ROS generation. EMFs of 1 mT and 10 mT increased and decreased vascular endothelial growth factor (VEGF) expression, respectively, which was abolished by DPI and radical scavengers. EMFs activated extracellular-regulated kinase 1/2 (ERK1/2), p38, and c-jun N-terminal kinase (JNK), which was sensitive to DPI treatment. The increase in VEGF by EMFs was inhibited by the ERK1/2 inhibitor U0126 but not by SB203580 and SP600125, which are p38 and JNK inhibitors, respectively, suggesting VEGF regulation by ERK1/2. Chondroosteogenesis and vasculogenesis of ES cells was blunted by trolox, DPI, and the VEGF receptor-2 (flk-1) antagonist SU5614. In mouse fetuses 1 mT EMFs increased and 10 mT EMFs decreased bone mineralization, which was abolished in the presence of trolox. Hence, EMFs induced chondro-osteogenesis and vasculogenesis in ES cells and bone mineralization of mouse fetuses by a ROS-dependent up-regulation of VEGF expression.
Collapse
Affiliation(s)
- Mohamed M Bekhite
- Department of Internal Medicine I, Cardiology Division, Friedrich Schiller University Jena, Germany
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Hammerick KE, James AW, Huang Z, Prinz FB, Longaker MT. Pulsed direct current electric fields enhance osteogenesis in adipose-derived stromal cells. Tissue Eng Part A 2010; 16:917-31. [PMID: 19824802 DOI: 10.1089/ten.tea.2009.0267] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Adipose-derived stromal cells (ASCs) constitute a promising source of cells for regenerative medicine applications. Previous studies of osteogenic potential in ASCs have focused on chemicals, growth factors, and mechanical stimuli. Citing the demonstrated role electric fields play in enhancing healing in bone fractures and defects, we investigated the ability of pulsed direct current electric fields to drive osteogenic differentiation in mouse ASCs. Employing 50 Hz direct current electric fields in concert with and without osteogenic factors, we demonstrated increased early osteoblast-specific markers. We were also able to establish that commonly reported artifacts of electric field stimulation are not the primary mediators of the observed effects. The electric fields caused marked changes in the cytoskeleton. We used atomic force microscopy-based force spectroscopy to record an increase in the cytoskeletal tension after treatment with electric fields. We abolished the increased cytoskeletal stresses with the rho-associated protein kinase inhibitor, Y27632, and did not see any decrease in osteogenic gene expression, suggesting that the pro-osteogenic effects of the electric fields are not transduced via cytoskeletal tension. Electric fields may show promise as candidate enhancers of osteogenesis of ASCs and may be incorporated into cell-based strategies for skeletal regeneration.
Collapse
Affiliation(s)
- Kyle E Hammerick
- Department of Mechanical Engineering, School of Engineering, Stanford University, Stanford, California 94305, USA
| | | | | | | | | |
Collapse
|
43
|
Matos MA, Cicerone MT. Alternating current electric field effects on neural stem cell viability and differentiation. Biotechnol Prog 2010; 26:664-70. [DOI: 10.1002/btpr.389] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
44
|
Finkensieper A, Kieser S, Bekhite MM, Richter M, Mueller JP, Graebner R, Figulla HR, Sauer H, Wartenberg M. The 5-lipoxygenase pathway regulates vasculogenesis in differentiating mouse embryonic stem cells. Cardiovasc Res 2009; 86:37-44. [DOI: 10.1093/cvr/cvp385] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
|
45
|
Baptista AF, Goes BT, Menezes D, Gomes FCA, Zugaib J, Stipursky J, Gomes JRS, Oliveira JÃT, Vannier-Santos MA, Martinez AMB. PEMF fails to enhance nerve regeneration after sciatic nerve crush lesion. J Peripher Nerv Syst 2009; 14:285-93. [DOI: 10.1111/j.1529-8027.2009.00240.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
46
|
Ushio-Fukai M, Urao N. Novel role of NADPH oxidase in angiogenesis and stem/progenitor cell function. Antioxid Redox Signal 2009; 11:2517-33. [PMID: 19309262 PMCID: PMC2821135 DOI: 10.1089/ars.2009.2582] [Citation(s) in RCA: 108] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Neovascularization is involved in normal development and wound repair as well as ischemic heart disease and peripheral artery disease. Both angiogenesis and vasculogenesis [de novo new vessel formation through mobilization of stem/progenitor cells from bone marrow (BM) and their homing to the ischemic sites] contribute to the formation of new blood vessels after tissue ischemia. Angiogenesis is dependent on cell proliferation, migration, and capillary tube formation in endothelial cells (ECs). Stem/progenitor cells have been used for cell-based therapy to promote revascularization after peripheral or myocardial ischemia. Excess amounts of reactive oxygen species (ROS) are involved in senescence and apoptosis of ECs and stem/progenitor cells, causing defective neovascularization. ROS at low levels function as signaling molecules to mediate cell proliferation, migration, differentiation, and gene expression. NADPH oxidase is one of the major sources of ROS in ECs and stem/progenitor cells, and is activated by various growth factors, cytokines, hypoxia, and ischemia. ROS derived from NADPH oxidase play an important role in redox signaling linked to angiogenesis ECs, as well as stem/progenitor cell mobilization, homing, and differentiation, thereby promoting neovascularization. Understanding these mechanisms may provide insight into NADPH oxidase and its mediators as potential therapeutic targets for ischemic heart and limb disease.
Collapse
Affiliation(s)
- Masuko Ushio-Fukai
- Department of Pharmacology, Center for Lung and Vascular Biology, University of Illinois at Chicago, Chicago, Illinois 60612, USA.
| | | |
Collapse
|
47
|
Herrmann JL, Markel TA, Abarbanell AM, Weil BR, Wang M, Wang Y, Tan J, Meldrum DR. Proinflammatory stem cell signaling in cardiac ischemia. Antioxid Redox Signal 2009; 11:1883-96. [PMID: 19187005 PMCID: PMC2872207 DOI: 10.1089/ars.2009.2434] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Cardiovascular disease remains a leading cause of mortality in developed nations, despite continued advancement in modern therapy. Progenitor and stem cell-based therapy is a novel treatment for cardiovascular disease, and modest benefits in cardiac recovery have been achieved in small clinical trials. This therapeutic modality remains challenged by limitations of low donor-cell survival rates, transient recovery of cardiac function, and the technical difficulty of applying directed cell therapy. Understanding the signaling mechanisms involved in the stem cell response to ischemia has revealed opportunities to modify directly aspects of these pathways to improve their cardioprotective abilities. This review highlights general considerations of stem cell therapy for cardiac disease, reviews the major proinflammatory signaling pathways of mesenchymal stem cells, and reviews ex vivo modifications of stem cells based on these pathways.
Collapse
Affiliation(s)
- Jeremy L Herrmann
- Clarian Cardiovascular Surgery, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Intracellular adenosine triphosphate delivery enhanced skin wound healing in rabbits. Ann Plast Surg 2009; 62:180-6. [PMID: 19158531 DOI: 10.1097/sap.0b013e31817fe47e] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Small unilamellar lipid vesicles were used to encapsulate adenosine triphosphate (ATP-vesicles) for intracellular energy delivery. This technique was tested in full-thickness skin wounds in 16 adult rabbits. One ear was rendered ischemic by using a minimally invasive surgery. The other ear served as a normal control. Four circular full-thickness wounds were created on the ventral side of each ear. ATP-vesicles or saline was used and the wounds were covered with Tegaderm (3M, St. Paul, MN). Dressing was changed and digital photos were taken daily until all the wounds were healed. The mean healing times of ATP-vesicles-treated wounds were significantly shorter than that of saline-treated wounds on ischemic and nonischemic ears. Histologic study indicated better-developed granular tissue and reepithelialization in the ATP-vesicles-treated wounds. The wounds treated by ATP-vesicles exhibited extremely fast granular tissue growth. More CD31 positive cells were seen in the ATP-vesicles-treated wounds. This preliminary study shows that direct intracellular delivery of ATP can accelerate the healing process of skin wounds on ischemic and nonischemic rabbit ears. The extremely fast granular tissue growth was something never seen or reported in the past.
Collapse
|
49
|
Chan EC, Jiang F, Peshavariya HM, Dusting GJ. Regulation of cell proliferation by NADPH oxidase-mediated signaling: Potential roles in tissue repair, regenerative medicine and tissue engineering. Pharmacol Ther 2009; 122:97-108. [DOI: 10.1016/j.pharmthera.2009.02.005] [Citation(s) in RCA: 125] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2009] [Accepted: 02/05/2009] [Indexed: 12/30/2022]
|
50
|
Sen CK, Roy S. Redox signals in wound healing. BIOCHIMICA ET BIOPHYSICA ACTA 2008; 1780:1348-61. [PMID: 18249195 PMCID: PMC2574682 DOI: 10.1016/j.bbagen.2008.01.006] [Citation(s) in RCA: 222] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/10/2007] [Revised: 12/31/2007] [Accepted: 01/14/2008] [Indexed: 12/19/2022]
Abstract
Physical trauma represents one of the most primitive challenges that threatened survival. Healing a problem wound requires a multi-faceted comprehensive approach. First and foremost, the wound environment will have to be made receptive to therapies. Second, the appropriate therapeutic regimen needs to be identified and provided while managing systemic limitations that could secondarily limit the healing response. Unfortunately, most current solutions seem to aim at designing therapeutic regimen with little or no consideration of the specific details of the wound environment and systemic limitations. One factor that is centrally important in making the wound environment receptive is correction of wound hypoxia. Recent work have identified that oxygen is not only required to disinfect wounds and fuel healing but that oxygen-dependent redox-sensitive signaling processes represent an integral component of the healing cascade. Over a decade ago, it was proposed that in biological systems oxidants are not necessarily always the triggers for oxidative damage and that oxidants such as H2O2 could actually serve as signaling messengers and drive several aspects of cellular signaling. Today, that concept is much more developed and mature. Evidence supporting the role of oxidants such as H2O2 as signaling messenger is compelling. A complete understanding of the continuum between the classical and emergent roles of oxygen requires a thorough consideration of current concepts in redox biology. The objective of this review is to describe our current understanding of how redox-sensitive processes may drive dermal tissue repair.
Collapse
Affiliation(s)
- Chandan K Sen
- Comprehensive Wound Center, Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, Ohio 43210, USA.
| | | |
Collapse
|