1
|
Zhou G, Lu D. Proteomics screening uncovers HMGA1 as a promising negative regulator for γ-globin expression in response to decreased β-globin levels. J Proteomics 2023; 286:104957. [PMID: 37423548 DOI: 10.1016/j.jprot.2023.104957] [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: 05/05/2023] [Revised: 06/28/2023] [Accepted: 06/30/2023] [Indexed: 07/11/2023]
Abstract
Reactivation of fetal hemoglobin (HbF) is a critical goal for the treatment of patients with hemoglobinopathies. β-globin disorders can trigger stress erythropoiesis in red blood cells (RBCs). Cell-intrinsic erythroid stress signals promote erythroid precursors to express high levels of fetal hemoglobin, which is also known as γ-globin. However, the molecular mechanism underlying γ-globin production during cell-intrinsic erythroid stress remains to be elucidated. Here, we utilized CRISPR-Cas9 to model a stressed state caused by reduced levels of adult β-globin in HUDEP2 human erythroid progenitor cells. We found that a decrease in β-globin expression correlates with the upregulation of γ-globin expression. We also identified transcription factor high-mobility group A1 (HMGA1; formerly HMG-I/Y) as a potential γ-globin regulator that responds to reduced β-globin levels. Upon erythroid stress, there is a downregulation of HMGA1, which normally binds -626 to -610 base pairs upstream from the STAT3 promoter, to downregulate STAT3 expression. STAT3 is a known γ-globin repressor, so the downregulation of HMGA1 ultimately upregulates γ-globin expression. SIGNIFICANCE: This study demonstrated HMGA1 as a potential regulator in the poorly understood phenomenon of stress-induced globin compensation, and after further validation these results might inform new strategies to treat patients with sickle cell disease and β-thalassemia.
Collapse
Affiliation(s)
- Guoqiang Zhou
- Greater Bay Area Institute of Precision Medicine (Guangzhou), School of Life Sciences, Fudan University, Guangzhou, 511458, China
| | - Daru Lu
- Greater Bay Area Institute of Precision Medicine (Guangzhou), School of Life Sciences, Fudan University, Guangzhou, 511458, China; NHC Key Laboratory of Birth Defects and Reproductive Health, Chongqing Key Laboratory of Birth Defects and Reproductive Health, Chongqing Population and Family Planning, Science and Technology Research Institute, Chongqing, China.
| |
Collapse
|
2
|
Catapano R, Sessa R, Trombetti S, Cesaro E, Russo F, Izzo P, Makis A, Grosso M. Identification and Functional Analysis of Known and New Mutations in the Transcription Factor KLF1 Linked with β-Thalassemia-like Phenotypes. BIOLOGY 2023; 12:biology12040510. [PMID: 37106711 PMCID: PMC10135830 DOI: 10.3390/biology12040510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/16/2023] [Accepted: 03/24/2023] [Indexed: 03/31/2023]
Abstract
The erythroid transcriptional factor Krüppel-like factor 1 (KLF1) is a master regulator of erythropoiesis. Mutations that cause KLF1 haploinsufficiency have been linked to increased fetal hemoglobin (HbF) and hemoglobin A2 (HbA2) levels with ameliorative effects on the severity of β-thalassemia. With the aim of determining if KLF1 gene variations might play a role in the modulation of β-thalassemia, in this study we screened 17 subjects showing a β-thalassemia-like phenotype with a slight or marked increase in HbA2 and HbF levels. Overall, seven KLF1 gene variants were identified, of which two were novel. Functional studies were performed in K562 cells to clarify the pathogenic significance of these mutations. Our study confirmed the ameliorative effect on the thalassemia phenotype for some of these variants but also raised the notion that certain mutations may have deteriorating effects by increasing KLF1 expression levels or enhancing its transcriptional activity. Our results indicate that functional studies are required to evaluate the possible effects of KLF1 mutations, particularly in the case of the co-existence of two or more mutations that could differently contribute to KLF1 expression or transcriptional activity and consequently to the thalassemia phenotype.
Collapse
Affiliation(s)
- Rosa Catapano
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, 80131 Naples, Italy
| | - Raffaele Sessa
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, 80131 Naples, Italy
| | - Silvia Trombetti
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, 80131 Naples, Italy
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, 80137 Naples, Italy
| | - Elena Cesaro
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, 80131 Naples, Italy
| | - Filippo Russo
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, 80131 Naples, Italy
| | - Paola Izzo
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, 80131 Naples, Italy
| | - Alexandros Makis
- Department of Pediatrics, University Hospital of Ioannina, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Michela Grosso
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, 80131 Naples, Italy
- Correspondence:
| |
Collapse
|
3
|
Huang H, Liang L, Sun D, Li J, Wang W, Zha L, Yang J, Pan K, Fan X, He C, Tang X, Zhang P. Rab37 Promotes Endothelial Differentiation and Accelerates ADSC-Mediated Diabetic Wound Healing through Regulating Secretion of Hsp90α and TIMP1. Stem Cell Rev Rep 2023; 19:1019-1033. [PMID: 36627432 DOI: 10.1007/s12015-022-10491-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/07/2022] [Indexed: 01/12/2023]
Abstract
Accumulating evidence indicates that adipose tissue-derived mesenchymal stem cells (ADSCs) are an effective treatment for diabetic refractory wounds. However, the application of ADSCs to diabetic wounds is still limited, indicating that we still lack sufficient knowledge regarding regulators/mediators of ADSCs during wound healing. Rab37, a member of RabGTPase, may function as regulator of vesicle trafficking, which is a crucial event for the secretion of cytokines by ADSCs. Our previous study indicated that Rab37 promotes the adiopogenic differentiation of ADSCs. In this study, we explored the role of Rab37 in ADSC-mediated diabetic wound healing. An in vivo study in db/db diabetic mice showed that Rab37-expressing ADSCs shortened the wound closure time, improved re-epithelialization and collagen deposition, and promoted angiogenesis during wound healing. An in vitro study showed that Rab37 promoted the proliferation, migration and endothelial differentiation of ADSCs. LC-MS/MS analysis identified Hsp90α and TIMP1 as up-regulated cytokines in conditioned media of Rab37-ADSCs. The up-regulation of Rab37 enhanced the secretion of Hsp90α and TIMP1 during endothelial differentiation and under high-glucose exposure. Interestingly, Rab37 promoted the expression of TIMP1, but not Hsp90α, during endothelial differentiation. PLA showed that Rab37 can directly bind to Hsp90α orTIMP1 in ADSCs. Moreover, Hsp90α and TIMP1 knockdown compromised the promoting effects of Rab37 on the proliferation, migration and endothelial differentiation of ADSCs. In conclusion, Rab37 promotes the proliferation, migration and endothelial differentiation of ADSCs and accelerates ADSC-mediated diabetic wound healing through regulating the secretion of Hsp90α and TIMP1.
Collapse
Affiliation(s)
- Haili Huang
- Department of Plastic Surgery, Affiliated Hospital of Guangdong Medical University, No. 57 Renmin Avenue South, Xiashan District, Zhanjiang City, 524001, Guangdong Province, China
| | - Ling Liang
- Department of Plastic Surgery, Affiliated Hospital of Guangdong Medical University, No. 57 Renmin Avenue South, Xiashan District, Zhanjiang City, 524001, Guangdong Province, China
| | - Dan Sun
- Department of Plastic Surgery, Affiliated Hospital of Guangdong Medical University, No. 57 Renmin Avenue South, Xiashan District, Zhanjiang City, 524001, Guangdong Province, China
| | - Jin Li
- Department of Plastic Surgery, Affiliated Hospital of Guangdong Medical University, No. 57 Renmin Avenue South, Xiashan District, Zhanjiang City, 524001, Guangdong Province, China
| | - Wentao Wang
- Department of Plastic Surgery, Affiliated Hospital of Guangdong Medical University, No. 57 Renmin Avenue South, Xiashan District, Zhanjiang City, 524001, Guangdong Province, China
| | - Lixia Zha
- Department of Plastic Surgery, Affiliated Hospital of Guangdong Medical University, No. 57 Renmin Avenue South, Xiashan District, Zhanjiang City, 524001, Guangdong Province, China
| | - Jiaqi Yang
- Department of Plastic Surgery, Affiliated Hospital of Guangdong Medical University, No. 57 Renmin Avenue South, Xiashan District, Zhanjiang City, 524001, Guangdong Province, China
| | - Kunyan Pan
- Department of Plastic Surgery, Affiliated Hospital of Guangdong Medical University, No. 57 Renmin Avenue South, Xiashan District, Zhanjiang City, 524001, Guangdong Province, China
| | - Xianmou Fan
- Department of Plastic Surgery, Affiliated Hospital of Guangdong Medical University, No. 57 Renmin Avenue South, Xiashan District, Zhanjiang City, 524001, Guangdong Province, China
| | - Chengzhang He
- Department of Plastic Surgery, Affiliated Hospital of Guangdong Medical University, No. 57 Renmin Avenue South, Xiashan District, Zhanjiang City, 524001, Guangdong Province, China
| | - Xudong Tang
- Institute of Biochemistry and Molecular Biology, Collaborative Innovation Center for Antitumor Active Substance Research and development, Guangdong Medical University, Zhanjiang, China
| | - Peihua Zhang
- Department of Plastic Surgery, Affiliated Hospital of Guangdong Medical University, No. 57 Renmin Avenue South, Xiashan District, Zhanjiang City, 524001, Guangdong Province, China.
| |
Collapse
|
4
|
Expression of Tight Junction Proteins Is Altered in Bladder Cancer. ACTA ACUST UNITED AC 2020; 2020:6341256. [PMID: 33282635 PMCID: PMC7685791 DOI: 10.1155/2020/6341256] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 11/03/2020] [Indexed: 11/17/2022]
Abstract
Bladder cancer (BC) is one of the tumors which occur most frequently in urological system, but less is known about the expression of tight junction proteins and its clinical significance in BC. In this study, expression of claudin-4, zonula occludens-1 (ZO-1) and zonula occludens-1 nucleic acid-binding protein (ZONAB), in BC tissues, adjacent nontumor tissue (ANTT), and BC cell lines was examined by Western blotting, semiquantitative RT-PCR, and immunohistochemistry, and then, the clinical significance of these proteins was investigated. The mRNA and protein expression of ZONAB were significantly upregulated, while those of ZO-1 was significantly downregulated in some BC cell lines and tissues in comparison with nontumor urothelial cell lines and ANTT. High expression rate of ZO-1 and ZONAB had negative correlation in BC tissues and was also correlated with muscle-invasive lesions in BC tissues. In conclusion, the expression of tight junction proteins is significantly altered in BC and ZO-1, and ZONAB interaction might be involved in BC development.
Collapse
|
5
|
Riccio P, Sessa R, de Nicola S, Petruzziello F, Trombetti S, Menna G, Pepe G, Maddalena P, Izzo P, Grosso M. GATA-1 isoforms differently contribute to the production and compartmentation of reactive oxygen species in the myeloid leukemia cell line K562. J Cell Physiol 2019; 234:20829-20846. [PMID: 31049966 PMCID: PMC6767011 DOI: 10.1002/jcp.28688] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 03/12/2019] [Accepted: 03/19/2019] [Indexed: 12/11/2022]
Abstract
Maintenance of a balanced expression of the two isoforms of the transcription factor GATA‐1, the full‐length protein (GATA‐1FL) and a shorter isoform (GATA‐1
S), contributes to control hematopoiesis, whereas their dysregulation can alter the differentiation/proliferation potential of hematopoietic precursors thereby eventually leading to a variety of hematopoietic disorders. Although it is well established that these isoforms play opposite roles in these remarkable processes, most of the molecular pathways involved remain unknown. Here, we demonstrate that GATA‐1FL and GATA‐1S are able to differently influence intracellular redox states and reactive oxygen species (ROS) compartmentation in the erythroleukemic K562 cell line, thus shedding novel mechanistic insights into the processes of cell proliferation and apoptosis resistance in myeloid precursors. Furthermore, given the role played by ROS signaling as a strategy to escape apoptosis and evade cell‐mediated immunity in myeloid cells, this study highlights a mechanism through which aberrant expression of GATA‐1 isoforms could play a role in the leukemogenic process.
Collapse
Affiliation(s)
- Patrizia Riccio
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Raffaele Sessa
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Sergio de Nicola
- CNR-SPIN, National Research Council, Institute for Superconductors, Innovative Materials and Devices, Naples, Italy
| | - Fara Petruzziello
- Pediatric Hematology Unit, Santobono-Pausilipon Hospital, Naples, Italy
| | - Silvia Trombetti
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Giuseppe Menna
- Pediatric Hematology Unit, Santobono-Pausilipon Hospital, Naples, Italy
| | - Giampiero Pepe
- CNR-SPIN, National Research Council, Institute for Superconductors, Innovative Materials and Devices, Naples, Italy.,Department of Physics, University of Naples Federico II, Naples, Italy
| | | | - Paola Izzo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Michela Grosso
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| |
Collapse
|
6
|
Liu RT, Wang GR, Liu C, Qiu J, Yan LK, Li XJ, Wang XQ. RNAi-mediated downregulation of DNA binding protein A inhibits tumorigenesis in colorectal cancer. Int J Mol Med 2016; 38:703-12. [PMID: 27430286 PMCID: PMC4990294 DOI: 10.3892/ijmm.2016.2662] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2015] [Accepted: 06/22/2016] [Indexed: 12/14/2022] Open
Abstract
DNA binding protein A (dbpA) belongs to the Y-box binding protein family and has been reported to play an important role in carcinogenesis. Our previous study demonstrated that the knockdown of dbpA in gastric cancer cells inhibited cell proliferation by modulating the cell cycle. However, the role of dbpA in human colorectal cancer (CRC) remains unclear. In this study, immunohistochemical (IHC) staining and clinicopathological parameter analysis were employed to detect dbpA expression in 44 paired CRC samples and 7 CRC cell lines. Lentivirus-mediated short hairpin RNA (shRNA) was used to silence dbpA, and the effects of dbpA knockdown on cell proliferation were determined by MTT assay, colony formation assay and flow cytometry. Furthermore, a xenograft model was established to observe tumor growth in vivo. Functional analysis indicated that dbpA was overexpressed in the CRC tissues and cell lines, and a high dbpA expression was associated with the depth of invasion (p<0.001), the degree of differentiation (p<0.001), lymphatic metastasis (p<0.001) and vessel invasion (p<0.001). The suppression of dbpA expression resulted in decreased cell proliferation in vitro and tumor growth in vivo, and it induced cell cycle arrest and promoted the apoptosis of the CRC cells. As a whole, our findings illustrate the crucial role of dbpA in colorectal tumorigenesis. Thus, dbpA may be used as a novel and potent therapeutic target in CRC.
Collapse
Affiliation(s)
- Rui-Ting Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, College of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Guo-Rong Wang
- Department of General Surgery, The Third Affiliated Hospital, College of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710068, P.R. China
| | - Chang Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, College of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Jian Qiu
- Department of General Surgery, The Third Affiliated Hospital, College of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710068, P.R. China
| | - Li-Kun Yan
- Department of General Surgery, The Third Affiliated Hospital, College of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710068, P.R. China
| | - Xiao-Jun Li
- Department of General Surgery, The Third Affiliated Hospital, College of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710068, P.R. China
| | - Xiao-Qiang Wang
- Department of General Surgery, The Third Affiliated Hospital, College of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710068, P.R. China
| |
Collapse
|
7
|
Zhu C, Sauter E, Schreiter A, van Roeyen CRC, Ostendorf T, Floege J, Gembardt F, Hugo CP, Isermann B, Lindquist JA, Mertens PR. Cold Shock Proteins Mediate GN with Mesangioproliferation. J Am Soc Nephrol 2016; 27:3678-3689. [PMID: 27151923 DOI: 10.1681/asn.2015121367] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 03/09/2016] [Indexed: 11/03/2022] Open
Abstract
DNA binding protein A (DbpA) is a member of the human cold shock domain-containing protein superfamily, with known functions in cell proliferation, differentiation, and stress responses. DbpA mediates tight junction-associated activities in tubular epithelial cells, but the function of DbpA in mesangial cells is unknown. Here, we found DbpA protein expression restricted to vascular smooth muscle cells in healthy human kidney tissue but profound induction of DbpA protein expression within the glomerular mesangial compartment in mesangioproliferative nephritis. In vitro, depletion or overexpression of DbpA using lentiviral constructs led to inhibition or promotion, respectively, of mesangial cell proliferation. Because platelet-derived growth factor B (PDGF-B) signaling has a pivotal role in mesangial cell proliferation, we examined the regulatory effect of PDGF-B on DbpA. In vitro studies of human and rat mesangial cells confirmed a stimulatory effect of PDGF-B on DbpA transcript numbers and protein levels. Additional in vivo investigations showed DbpA upregulation in experimental rat anti-Thy1.1 nephritis and murine mesangioproliferative nephritis models. To interfere with PDGF-B signaling, we injected nephritic rats with PDGF-B neutralizing aptamers or the MEK/ERK inhibitor U0126. Both interventions markedly decreased DbpA protein expression. Conversely, continuous PDGF-B infusion in healthy rats induced DbpA expression predominantly within the mesangial compartment. Taken together, these results indicate that DbpA is a novel target of PDGF-B signaling and a key mediator of mesangial cell proliferation.
Collapse
Affiliation(s)
- Cheng Zhu
- Department of Nephrology, Hypertension, Diabetes and Endocrinology and
| | - Eva Sauter
- Department of Nephrology, Hypertension, Diabetes and Endocrinology and
| | - Anja Schreiter
- Department of Nephrology, Hypertension, Diabetes and Endocrinology and
| | - Claudia R C van Roeyen
- Department of Nephrology and Immunology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany; and
| | - Tammo Ostendorf
- Department of Nephrology and Immunology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany; and
| | - Jürgen Floege
- Department of Nephrology and Immunology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany; and
| | - Florian Gembardt
- Division of Nephrology, Department of Internal Medicine III, Dresden University of Technology, Dresden, Germany
| | - Christian P Hugo
- Division of Nephrology, Department of Internal Medicine III, Dresden University of Technology, Dresden, Germany
| | - Berend Isermann
- Institute of Clinical Chemistry and Pathobiochemistry, Medical Faculty, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | | | - Peter R Mertens
- Department of Nephrology, Hypertension, Diabetes and Endocrinology and
| |
Collapse
|
8
|
Forristal CE, Christensen DR, Chinnery FE, Petruzzelli R, Parry KL, Sanchez-Elsner T, Houghton FD. Environmental oxygen tension regulates the energy metabolism and self-renewal of human embryonic stem cells. PLoS One 2013; 8:e62507. [PMID: 23671606 PMCID: PMC3645991 DOI: 10.1371/journal.pone.0062507] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Accepted: 03/25/2013] [Indexed: 01/07/2023] Open
Abstract
Energy metabolism is intrinsic to cell viability but surprisingly has been little studied in human embryonic stem cells (hESCs). The current study aims to investigate the effect of environmental O2 tension on carbohydrate utilisation of hESCs. Highly pluripotent hESCs cultured at 5% O2 consumed significantly more glucose, less pyruvate and produced more lactate compared to those maintained at 20% O2. Moreover, hESCs cultured at atmospheric O2 levels expressed significantly less OCT4, SOX2 and NANOG than those maintained at 5% O2. To determine whether this difference in metabolism was a reflection of the pluripotent state, hESCs were cultured at 5% O2 in the absence of FGF2 for 16 hours leading to a significant reduction in the expression of SOX2. In addition, these cells consumed less glucose and produced significantly less lactate compared to those cultured in the presence of FGF2. hESCs maintained at 5% O2 were found to consume significantly less O2 than those cultured in the absence of FGF2, or at 20% O2. GLUT1 expression correlated with glucose consumption and using siRNA and chromatin immunoprecipitation was found to be directly regulated by hypoxia inducible factor (HIF)-2α at 5% O2. In conclusion, highly pluripotent cells associated with hypoxic culture consume low levels of O2, high levels of glucose and produce large amounts of lactate, while at atmospheric conditions glucose consumption and lactate production are reduced and there is an increase in oxidative metabolism. These data suggest that environmental O2 regulates energy metabolism and is intrinsic to the self-renewal of hESCs.
Collapse
Affiliation(s)
- Catherine E. Forristal
- Centre for Human Development, Stem Cells and Regeneration, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - David R. Christensen
- Centre for Human Development, Stem Cells and Regeneration, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Fay E. Chinnery
- Centre for Human Development, Stem Cells and Regeneration, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Raffaella Petruzzelli
- Centre for Human Development, Stem Cells and Regeneration, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Kate L. Parry
- Centre for Human Development, Stem Cells and Regeneration, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Tilman Sanchez-Elsner
- Centre for Human Development, Stem Cells and Regeneration, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Franchesca D. Houghton
- Centre for Human Development, Stem Cells and Regeneration, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- * E-mail:
| |
Collapse
|