1
|
Ramakrishnan VM, Thaker H, Ocampo GL, Adam RM, Estrada CR. Pediatric bladder tissue engineering: Where have we been and where do we go next? J Pediatr Urol 2025:S1477-5131(25)00001-4. [PMID: 39827049 DOI: 10.1016/j.jpurol.2025.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 12/11/2024] [Accepted: 01/01/2025] [Indexed: 01/22/2025]
Abstract
OBJECTIVES This review aims to (a) provide a concise overview of early clinical trials in bladder tissue engineering and the associated challenges, (b) evaluate significant advancements over the past 15 years in addressing key limitations in angiogenesis, scaffolding, cell sourcing, and immunomodulation, and (c) explore the individual and synergistic contributions of each domain toward the development of a viable engineered solution. MATERIALS AND METHODS Relevant papers for this narrative review were selected through a PubMed search for "bladder tissue engineering" studies published between 01/01/2009 and 12/31/2024, as well as earlier clinical trials that predate this period. RESULTS Along with reviewing four major clinical trials, this review highlights nearly 20 distinct studies that showcase progress in the critical domains of angiogenesis, scaffolding, cell sourcing, and immunomodulation. CONCLUSIONS Are we close to developing an off-the-shelf bladder substitute? Not yet. Current efforts are focused on addressing two major knowledge gaps: (a) the lack of testing in clinically relevant disease models and (b) the need for a more comprehensive understanding of engineered tissue physiology.
Collapse
Affiliation(s)
- Venkat M Ramakrishnan
- Department of Urology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Hatim Thaker
- Department of Urology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Gabriel-Luis Ocampo
- Department of Urology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Rosalyn M Adam
- Department of Urology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Carlos R Estrada
- Department of Urology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
2
|
Zakharova IS, Shevchenko AI, Arssan MA, Sleptcov AA, Nazarenko MS, Zarubin AA, Zheltysheva NV, Shevchenko VA, Tmoyan NA, Saaya SB, Ezhov MV, Kukharchuk VV, Parfyonova YV, Zakian SM. iPSC-Derived Endothelial Cells Reveal LDLR Dysfunction and Dysregulated Gene Expression Profiles in Familial Hypercholesterolemia. Int J Mol Sci 2024; 25:689. [PMID: 38255763 PMCID: PMC10815294 DOI: 10.3390/ijms25020689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/28/2023] [Accepted: 01/03/2024] [Indexed: 01/24/2024] Open
Abstract
Defects in the low-density lipoprotein receptor (LDLR) are associated with familial hypercholesterolemia (FH), manifested by atherosclerosis and cardiovascular disease. LDLR deficiency in hepatocytes leads to elevated blood cholesterol levels, which damage vascular cells, especially endothelial cells, through oxidative stress and inflammation. However, the distinctions between endothelial cells from individuals with normal and defective LDLR are not yet fully understood. In this study, we obtained and examined endothelial derivatives of induced pluripotent stem cells (iPSCs) generated previously from conditionally healthy donors and compound heterozygous FH patients carrying pathogenic LDLR alleles. In normal iPSC-derived endothelial cells (iPSC-ECs), we detected the LDLR protein predominantly in its mature form, whereas iPSC-ECs from FH patients have reduced levels of mature LDLR and show abolished low-density lipoprotein uptake. RNA-seq of mutant LDLR iPSC-ECs revealed a unique transcriptome profile with downregulated genes related to monocarboxylic acid transport, exocytosis, and cell adhesion, whereas upregulated signaling pathways were involved in cell secretion and leukocyte activation. Overall, these findings suggest that LDLR defects increase the susceptibility of endothelial cells to inflammation and oxidative stress. In combination with elevated extrinsic cholesterol levels, this may result in accelerated endothelial dysfunction, contributing to early progression of atherosclerosis and other cardiovascular pathologies associated with FH.
Collapse
Affiliation(s)
- Irina S. Zakharova
- Federal Research Centre Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (I.S.Z.); (A.I.S.); (M.A.A.); (N.V.Z.); (V.A.S.)
| | - Alexander I. Shevchenko
- Federal Research Centre Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (I.S.Z.); (A.I.S.); (M.A.A.); (N.V.Z.); (V.A.S.)
| | - Mhd Amin Arssan
- Federal Research Centre Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (I.S.Z.); (A.I.S.); (M.A.A.); (N.V.Z.); (V.A.S.)
| | - Aleksei A. Sleptcov
- Research Institute of Medical Genetics, Tomsk National Research Medical Centre, Russian Academy of Science, 634050 Tomsk, Russia; (A.A.S.); (M.S.N.); (A.A.Z.)
| | - Maria S. Nazarenko
- Research Institute of Medical Genetics, Tomsk National Research Medical Centre, Russian Academy of Science, 634050 Tomsk, Russia; (A.A.S.); (M.S.N.); (A.A.Z.)
| | - Aleksei A. Zarubin
- Research Institute of Medical Genetics, Tomsk National Research Medical Centre, Russian Academy of Science, 634050 Tomsk, Russia; (A.A.S.); (M.S.N.); (A.A.Z.)
| | - Nina V. Zheltysheva
- Federal Research Centre Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (I.S.Z.); (A.I.S.); (M.A.A.); (N.V.Z.); (V.A.S.)
| | - Vlada A. Shevchenko
- Federal Research Centre Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (I.S.Z.); (A.I.S.); (M.A.A.); (N.V.Z.); (V.A.S.)
| | - Narek A. Tmoyan
- Federal State Budgetary Institution, National Medical Research Centre of Cardiology Named after Academician E.I. Chazov, Ministry of Health of Russian Federation, 121552 Moscow, Russia; (N.A.T.); (M.V.E.); (V.V.K.); (Y.V.P.)
| | - Shoraan B. Saaya
- E.N. Meshalkin National Medical Research Centre, Ministry of Health Care of the Russian Federation, 630055 Novosibirsk, Russia;
| | - Marat V. Ezhov
- Federal State Budgetary Institution, National Medical Research Centre of Cardiology Named after Academician E.I. Chazov, Ministry of Health of Russian Federation, 121552 Moscow, Russia; (N.A.T.); (M.V.E.); (V.V.K.); (Y.V.P.)
| | - Valery V. Kukharchuk
- Federal State Budgetary Institution, National Medical Research Centre of Cardiology Named after Academician E.I. Chazov, Ministry of Health of Russian Federation, 121552 Moscow, Russia; (N.A.T.); (M.V.E.); (V.V.K.); (Y.V.P.)
| | - Yelena V. Parfyonova
- Federal State Budgetary Institution, National Medical Research Centre of Cardiology Named after Academician E.I. Chazov, Ministry of Health of Russian Federation, 121552 Moscow, Russia; (N.A.T.); (M.V.E.); (V.V.K.); (Y.V.P.)
| | - Suren M. Zakian
- Federal Research Centre Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (I.S.Z.); (A.I.S.); (M.A.A.); (N.V.Z.); (V.A.S.)
| |
Collapse
|
3
|
Taheri F, Taghizadeh E, Baniamerian F, Rostami D, Rozeian A, Mohammad Gheibi Hayat S, Jamialahmadi T, Reiner Ž, Sahebkar A. Cellular and Molecular Aspects of Managing Familial Hypercholesterolemia: Recent and Emerging Therapeutic Approaches. Endocr Metab Immune Disord Drug Targets 2022; 22:1018-1028. [PMID: 35532248 DOI: 10.2174/1871530322666220509040844] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 03/08/2022] [Accepted: 03/18/2022] [Indexed: 11/22/2022]
Abstract
Familial hypercholesterolemia (FH) as a high-frequency genetic disorder is diagnosed based on family and/or patient's history of coronary heart disease (CHD) or some other atherosclerotic disease, LDL-C levels and/or clinical signs such as tendonous xantomata, arcus cornealis before age 45 years as well as functional mutation in the LDLR, apoB or PCSK9 gene. Its clinical features are detectable since early childhood. Early diagnosis and timely treatment increase life expectancy in most patients with FH. Current FH therapies decrease the level of low-density lipoprotein up to ≥50% from baseline with diet, pharmacotherapeutic treatment, lipid apheresis, and liver transplantation. The cornerstone of medical therapy is the use of more potent statins in higher doses, to which often ezetimibe has to be added, but some FH patients do not achieve the target LDL-C with this therapy Therefore, besides these and the most recent but already established therapeutic approaches including PCSK9 inhibitors, inclisiran, and bempedoic acid, new therapies are on the horizon such as gene therapy, CRISPR/Cas9 strategy etc. This paper focuses on cellular and molecular potential strategies for the treatment of FH.
Collapse
Affiliation(s)
- Forough Taheri
- Sharekord Branch, Islamic Azad University, Sharekord, Iran
| | - Eskandar Taghizadeh
- Department of Medical Genetics, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Fatemeh Baniamerian
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Daryoush Rostami
- Department of Anesthesia, school of Paramedical Sciences, Zabol University of Medical Sciences, Zabol, Iran
| | - Ahmad Rozeian
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Seyed Mohammad Gheibi Hayat
- Department of Medical Genetics, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Tannaz Jamialahmadi
- Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Željko Reiner
- Department of Internal Medicine, University Hospital Center Zagreb, School of Medicine University of Zagreb, Croatia
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,School of Medicine, The University of Western Australia, Perth, Australia.,Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
4
|
Bailly A, Milhavet O, Lemaitre JM. RNA-Based Strategies for Cell Reprogramming toward Pluripotency. Pharmaceutics 2022; 14:317. [PMID: 35214051 PMCID: PMC8876983 DOI: 10.3390/pharmaceutics14020317] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 01/16/2022] [Accepted: 01/25/2022] [Indexed: 02/04/2023] Open
Abstract
Cell therapy approaches to treat a wide range of pathologies have greatly benefited from cell reprogramming techniques that allow the conversion of a somatic cell into a pluripotent cell. Many technological developments have been made since the initial major discovery of this biological process. Recently reprogramming methods based on the use of RNA have emerged and seem very promising. Thus, in this review we will focus on presenting the interest of such methods for cell reprogramming but also how these RNA-based strategies can be extended to eventually lead to medical applications to improve healthspan and longevity.
Collapse
Affiliation(s)
- Anaëlle Bailly
- IRMB, University Montpellier, INSERM, 34295 Montpellier, France
- INGRAALYS, SA, IRMB, Incubator Cyborg, 34295 Montpellier, France
| | - Ollivier Milhavet
- IRMB, University Montpellier, INSERM, CNRS, 34295 Montpellier, France
- SAFE-iPSC Facility, CHU Montpellier, 34295 Montpellier, France
| | - Jean-Marc Lemaitre
- IRMB, University Montpellier, INSERM, 34295 Montpellier, France
- SAFE-iPSC Facility, CHU Montpellier, 34295 Montpellier, France
| |
Collapse
|
5
|
Wang AYL. Application of Modified mRNA in Somatic Reprogramming to Pluripotency and Directed Conversion of Cell Fate. Int J Mol Sci 2021; 22:8148. [PMID: 34360910 PMCID: PMC8348611 DOI: 10.3390/ijms22158148] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/25/2021] [Accepted: 07/27/2021] [Indexed: 02/07/2023] Open
Abstract
Modified mRNA (modRNA)-based somatic reprogramming is an effective and safe approach that overcomes the genomic mutation risk caused by viral integrative methods. It has improved the disadvantages of conventional mRNA and has better stability and immunogenicity. The modRNA molecules encoding multiple pluripotent factors have been applied successfully in reprogramming somatic cells such as fibroblasts, mesenchymal stem cells, and amniotic fluid stem cells to generate pluripotent stem cells (iPSCs). Moreover, it also can be directly used in the terminal differentiation of stem cells and fibroblasts into functional therapeutic cells, which exhibit great promise in disease modeling, drug screening, cell transplantation therapy, and regenerative medicine. In this review, we summarized the reprogramming applications of modified mRNA in iPSC generation and therapeutic applications of functionally differentiated cells.
Collapse
Affiliation(s)
- Aline Yen Ling Wang
- Center for Vascularized Composite Allotransplantation, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| |
Collapse
|
6
|
Omer L, Hudson EA, Hudgins LC, Boyd NL. Cohort Generation and Characterization of Patient-Specific Familial Hypercholesterolemia Induced Pluripotent Stem Cells. Stem Cells Dev 2021; 30:632-640. [PMID: 34029164 DOI: 10.1089/scd.2021.0004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Homozygous familial hypercholesterolemia (hoFH) is a rare disorder caused primarily by pathological mutations in the low-density lipoprotein receptor (LDLR), which disrupts LDL-cholesterol (LDL-C) metabolism homeostasis. hoFH patients are at extremely high risk for cardiovascular disease and are resistant to standard therapies. LDLR knockout animals and in vitro cell models overexpressing different mutations have proved useful, but may not fully recapitulate human LDLR mutation biology. We and others have generated induced pluripotent stem cells (iPSC) from hoFH patient's fibroblasts and T cells and demonstrated their ability to recapitulate hoFH biology. In this study, we present the generation and characterization of a cohort of seven hoFH-iPSC lines derived from peripheral blood mononuclear cells (PBMC) collected from four homozygous and three compound heterozygous patients. The hoFH-iPSC cohort demonstrated a wide range of LDLR expression and LDL-C internalization in response to rosuvastatin that correlated with the predicted pathogenicity of the mutation. We were able to confirm that hoFH-iPSC cohort were pluripotent by differentiation toward all three germ layers and specifically to hepatocyte-like cells (HLC), the cell with primary LDL-C metabolic regulatory control, by expression of hepatocyte markers. hoFH patient PBMC-derived iPSC recapitulate the LDLR dysfunction of their specific mutation. They were capable of differentiating to HLC and could be useful for early developmental studies, pharmacology/toxicology, and potentially autologous cell therapy.
Collapse
Affiliation(s)
- Linda Omer
- Department of Biochemistry and Molecular Genetics and University of Louisville School of Medicine, Louisville, Kentucky, USA.,Cardiovascular Innovation Institute, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Elizabeth A Hudson
- Department of Biochemistry and Molecular Genetics and University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Lisa C Hudgins
- Rogosin Institute, Weill Cornell Medical College, New York, New York, USA
| | - Nolan L Boyd
- Department of Biochemistry and Molecular Genetics and University of Louisville School of Medicine, Louisville, Kentucky, USA.,Department of Physiology, University of Louisville School of Medicine, Louisville, Kentucky, USA
| |
Collapse
|
7
|
Larsen LE, Smith MA, Abbey D, Korn A, Reeskamp LF, Hand NJ, Holleboom AG. Hepatocyte-like cells derived from induced pluripotent stem cells: A versatile tool to understand lipid disorders. Atherosclerosis 2020; 303:8-14. [PMID: 32460140 DOI: 10.1016/j.atherosclerosis.2020.03.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 02/19/2020] [Accepted: 03/18/2020] [Indexed: 12/12/2022]
Abstract
Dyslipidemias are strongly linked to the development of atherosclerotic cardiovascular disease. Most dyslipidemias find their origin in the liver. In recent years, the differentiation of induced pluripotent stem cells (iPSCs) into hepatocyte-like cells has provided a versatile platform for the functional study of various dyslipidemias, both rare genetic dyslipidemia as well as common lipid disorders associated with insulin resistance or non-alcoholic fatty liver disease. In addition, iPSC-derived hepatocytes can serve as a cell model for developing novel lipid lowering therapies and have the potential of regenerative medicine. This review provides an overview of these developments.
Collapse
Affiliation(s)
- Lars E Larsen
- Department of Experimental Vascular Medicine, Academic Medical Center, Amsterdam, the Netherlands
| | - Mikhaila A Smith
- Departments of Genetics and Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, USA
| | - Deepti Abbey
- Departments of Genetics and Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, USA
| | - Amber Korn
- Department of Experimental Vascular Medicine, Academic Medical Center, Amsterdam, the Netherlands; Department of Vascular Medicine, Academic Medical Center, Amsterdam, the Netherlands
| | - Laurens F Reeskamp
- Department of Experimental Vascular Medicine, Academic Medical Center, Amsterdam, the Netherlands; Department of Vascular Medicine, Academic Medical Center, Amsterdam, the Netherlands
| | - Nicholas J Hand
- Departments of Genetics and Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, USA.
| | - Adriaan G Holleboom
- Department of Experimental Vascular Medicine, Academic Medical Center, Amsterdam, the Netherlands; Department of Vascular Medicine, Academic Medical Center, Amsterdam, the Netherlands.
| |
Collapse
|
8
|
Wang AYL, Loh CYY. Episomal Induced Pluripotent Stem Cells: Functional and Potential Therapeutic Applications. Cell Transplant 2019; 28:112S-131S. [PMID: 31722555 PMCID: PMC7016470 DOI: 10.1177/0963689719886534] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 06/11/2019] [Accepted: 10/07/2019] [Indexed: 12/19/2022] Open
Abstract
The term episomal induced pluripotent stem cells (EiPSCs) refers to somatic cells that are reprogrammed into induced pluripotent stem cells (iPSCs) using non-integrative episomal vector methods. This reprogramming process has a better safety profile compared with integrative methods using viruses. There is a current trend toward using episomal plasmid reprogramming to generate iPSCs because of the improved safety profile. Clinical reports of potential human cell sources that have been successfully reprogrammed into EiPSCs are increasing, but no review or summary has been published. The functional applications of EiPSCs and their potential uses in various conditions have been described, and these may be applicable to clinical scenarios. This review summarizes the current direction of EiPSC research and the properties of these cells with the aim of explaining their potential role in clinical applications and functional restoration.
Collapse
Affiliation(s)
- Aline Yen Ling Wang
- Center for Vascularized Composite Allotransplantation, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- *Both the authors contributed equally to this article
| | - Charles Yuen Yung Loh
- St Andrew’s Center for Burns and Plastic Surgery, Chelmsford, United Kingdom
- *Both the authors contributed equally to this article
| |
Collapse
|
9
|
Caron J, Pène V, Tolosa L, Villaret M, Luce E, Fourrier A, Heslan JM, Saheb S, Bruckert E, Gómez-Lechón MJ, Nguyen TH, Rosenberg AR, Weber A, Dubart-Kupperschmitt A. Low-density lipoprotein receptor-deficient hepatocytes differentiated from induced pluripotent stem cells allow familial hypercholesterolemia modeling, CRISPR/Cas-mediated genetic correction, and productive hepatitis C virus infection. Stem Cell Res Ther 2019; 10:221. [PMID: 31358055 PMCID: PMC6664765 DOI: 10.1186/s13287-019-1342-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 07/03/2019] [Accepted: 07/14/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Familial hypercholesterolemia type IIA (FH) is due to mutations in the low-density lipoprotein receptor (LDLR) resulting in elevated levels of low-density lipoprotein cholesterol (LDL-c) in plasma and in premature cardiovascular diseases. As hepatocytes are the only cells capable of metabolizing cholesterol, they are therefore the target cells for cell/gene therapy approaches in the treatment of lipid metabolism disorders. Furthermore, the LDLR has been reported to be involved in hepatitis C virus (HCV) entry into hepatocytes; however, its role in the virus infection cycle is still disputed. METHODS We generated induced pluripotent stem cells (iPSCs) from a homozygous LDLR-null FH-patient (FH-iPSCs). We constructed a correction cassette bearing LDLR cDNA under the control of human hepatic apolipoprotein A2 promoter that targets the adeno-associated virus integration site AAVS1. We differentiated both FH-iPSCs and corrected FH-iPSCs (corr-FH-iPSCs) into hepatocytes to study statin-mediated regulation of genes involved in cholesterol metabolism. Upon HCV particle inoculation, viral replication and production were quantified in these cells. RESULTS We showed that FH-iPSCs displayed the disease phenotype. Using homologous recombination mediated by the CRISPR/Cas9 system, FH-iPSCs were genetically corrected by the targeted integration of a correction cassette at the AAVS1 locus. Both FH-iPSCs and corr-FH-iPSCs were then differentiated into functional polarized hepatocytes using a stepwise differentiation approach (FH-iHeps and corr-FH-iHeps). The correct insertion and expression of the correction cassette resulted in restoration of LDLR expression and function (LDL-c uptake) in corr-FH-iHeps. We next demonstrated that pravastatin treatment increased the expression of genes involved in cholesterol metabolism in both cell models. Moreover, LDLR expression and function were also enhanced in corr-FH-iHeps after pravastatin treatment. Finally, we demonstrated that both FH-iHeps and corr-FH-iHeps were as permissive to viral infection as primary human hepatocytes but that virus production in FH-iHeps was significantly decreased compared to corr-FH-iHeps, suggesting a role of the LDLR in HCV morphogenesis. CONCLUSIONS Our work provides the first LDLR-null FH cell model and its corrected counterpart to study the regulation of cholesterol metabolism and host determinants of HCV life cycle, and a platform to screen drugs for treating dyslipidemia and HCV infection.
Collapse
Affiliation(s)
- Jérôme Caron
- INSERM UMR_S1193, Hôpital Paul Brousse, Villejuif, France; UMR-S1193, Université Paris-Saclay, Hôpital Paul Brousse, Villejuif, France; DHU Hepatinov, Hôpital Paul Brousse, Villejuif, France
| | | | - Laia Tolosa
- Unidad de Hepatología Experimental, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | | | - Eléanor Luce
- INSERM UMR_S1193, Hôpital Paul Brousse, Villejuif, France; UMR-S1193, Université Paris-Saclay, Hôpital Paul Brousse, Villejuif, France; DHU Hepatinov, Hôpital Paul Brousse, Villejuif, France
| | - Angélique Fourrier
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France
| | - Jean-Marie Heslan
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France
| | - Samir Saheb
- Service d'Endocrinologie Métabolisme, Hôpital Pitié-Salpêtrière, Paris, France
| | - Eric Bruckert
- Service d'Endocrinologie Métabolisme, Hôpital Pitié-Salpêtrière, Paris, France
| | - María José Gómez-Lechón
- Unidad de Hepatología Experimental, Instituto de Investigación Sanitaria La Fe, Valencia, Spain.,CIBERehd, FIS, Barcelona, Spain
| | - Tuan Huy Nguyen
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France
| | - Arielle R Rosenberg
- Université Paris Descartes, EA4474, Paris, France.,AP-HP, Hôpital Cochin, Service de Virologie, Paris, France
| | - Anne Weber
- INSERM UMR_S1193, Hôpital Paul Brousse, Villejuif, France; UMR-S1193, Université Paris-Saclay, Hôpital Paul Brousse, Villejuif, France; DHU Hepatinov, Hôpital Paul Brousse, Villejuif, France
| | - Anne Dubart-Kupperschmitt
- INSERM UMR_S1193, Hôpital Paul Brousse, Villejuif, France; UMR-S1193, Université Paris-Saclay, Hôpital Paul Brousse, Villejuif, France; DHU Hepatinov, Hôpital Paul Brousse, Villejuif, France.
| |
Collapse
|
10
|
Hajighasemi S, Mahdavi Gorabi A, Bianconi V, Pirro M, Banach M, Ahmadi Tafti H, Reiner Ž, Sahebkar A. A review of gene- and cell-based therapies for familial hypercholesterolemia. Pharmacol Res 2019; 143:119-132. [PMID: 30910740 DOI: 10.1016/j.phrs.2019.03.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 03/10/2019] [Accepted: 03/20/2019] [Indexed: 12/20/2022]
|
11
|
Rodriguez-Calvo R, Masana L. Review of the scientific evolution of gene therapy for the treatment of homozygous familial hypercholesterolaemia: past, present and future perspectives. J Med Genet 2019; 56:711-717. [DOI: 10.1136/jmedgenet-2018-105713] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 02/12/2019] [Accepted: 02/16/2019] [Indexed: 11/03/2022]
Abstract
Familial hypercholesterolaemia (FH) is a devastating genetic disease that leads to extremely high cholesterol levels and severe cardiovascular disease, mainly caused by mutations in any of the main genes involved in low-density lipoprotein cholesterol (LDL-C) uptake. Among these genes, mutations in the LDL receptor (LDLR) are responsible for 80%–90% of the FH cases. The severe homozygous variety (HoFH) is not successfully treated with standard cholesterol-lowering therapies, and more aggressive strategies must be considered to mitigate the effects of this disease, such as weekly/biweekly LDL apheresis. However, development of new therapeutic approaches is needed to cure HoFH. Because HoFH is mainly due to mutations in theLDLR, this disease has been proposed as an ideal candidate for gene therapy. Several preclinical studies have proposed that the transference of functional copies of theLDLRgene reduces circulating LDL-C levels in several models of HoFH, which has led to the first clinical trials in humans. Additionally, the recent development of clustered regularly interspaced short palindromic repeat/CRISPR-associated 9 technology for genome editing has opened the door to therapies aimed at directly correcting the specific mutation in the endogenousLDLRgene. In this article, we review the genetic basis of the FH disease, paying special attention to the severe HoFH as well as the challenges in its diagnosis and clinical management. Additionally, we discuss the current therapies for this disease and the new emerging advances in gene therapy to target a definitive cure for this disease.
Collapse
|
12
|
Ramakrishnan VM, Boyd NL. The Adipose Stromal Vascular Fraction as a Complex Cellular Source for Tissue Engineering Applications. TISSUE ENGINEERING. PART B, REVIEWS 2018; 24:289-299. [PMID: 28316259 PMCID: PMC6080106 DOI: 10.1089/ten.teb.2017.0061] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 03/17/2017] [Indexed: 12/27/2022]
Abstract
A major challenge in tissue engineering is the generation of sufficient volumes of viable tissue for organ transplant. The development of a stable, mature vasculature is required to sustain the metabolic and functional activities of engineered tissues. Adipose stromal vascular fraction (SVF) cells are an easily accessible, heterogeneous cell system comprised of endothelial cells, macrophages, pericytes, and various stem cell populations. Collectively, SVF has been shown to spontaneously form vessel-like networks in vitro and robust, patent, and functional vasculatures in vivo. Capitalizing on this ability, we and others have demonstrated adipose SVF's utility in generating and augmenting engineered liver, cardiac, and vascular tissues, to name a few. This review highlights the scientific origins of SVF, the use of SVF as a clinically relevant vascular source, various SVF constituents and their roles, and practical considerations associated with isolating SVF for various tissue engineering applications.
Collapse
Affiliation(s)
- Venkat M. Ramakrishnan
- Cardiovascular Innovation Institute, Department of Physiology, University of Louisville School of Medicine, Louisville, Kentucky
| | - Nolan L. Boyd
- Cardiovascular Innovation Institute, Department of Physiology, University of Louisville School of Medicine, Louisville, Kentucky
| |
Collapse
|
13
|
Systems Analysis of the Liver Transcriptome in Adult Male Zebrafish Exposed to the Plasticizer (2-Ethylhexyl) Phthalate (DEHP). Sci Rep 2018; 8:2118. [PMID: 29391432 PMCID: PMC5794889 DOI: 10.1038/s41598-018-20266-8] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 01/15/2018] [Indexed: 02/08/2023] Open
Abstract
The organic compound diethylhexyl phthalate (DEHP) represents a high production volume chemical found in cosmetics, personal care products, laundry detergents, and household items. DEHP, along with other phthalates causes endocrine disruption in males. Exposure to endocrine disrupting chemicals has been linked to the development of several adverse health outcomes with apical end points including Non-Alcoholic Fatty Liver Disease (NAFLD). This study examined the adult male zebrafish (Danio rerio) transcriptome after exposure to environmental levels of DEHP and 17α-ethinylestradiol (EE2) using both DNA microarray and RNA-sequencing technologies. Our results show that exposure to DEHP is associated with differentially expressed (DE) transcripts associated with the disruption of metabolic processes in the liver, including perturbation of five biological pathways: ‘FOXA2 and FOXA3 transcription factor networks’, ‘Metabolic pathways’, ‘metabolism of amino acids and derivatives’, ‘metabolism of lipids and lipoproteins’, and ‘fatty acid, triacylglycerol, and ketone body metabolism’. DE transcripts unique to DEHP exposure, not observed with EE2 (i.e. non-estrogenic effects) exhibited a signature related to the regulation of transcription and translation, and ruffle assembly and organization. Collectively our results indicate that exposure to low DEHP levels modulates the expression of liver genes related to fatty acid metabolism and the development of NAFLD.
Collapse
|
14
|
Kumar A, Kumar V, Rattan V, Jha V, Pal A, Bhattacharyya S. Molecular spectrum of secretome regulates the relative hepatogenic potential of mesenchymal stem cells from bone marrow and dental tissue. Sci Rep 2017; 7:15015. [PMID: 29118330 PMCID: PMC5678086 DOI: 10.1038/s41598-017-14358-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 10/10/2017] [Indexed: 01/10/2023] Open
Abstract
Liver regeneration is a spontaneous process that occurs after liver injury, but acute liver failure is a complex and fatal disease which is difficult to treat. Cell-based therapies are promising alternative therapeutic approach for liver failure and different cell sources have been tested in this regard. We investigated the comparative hepatogenic potential of human bone marrow stem cells (BMSC) with stem cells derived from human dental pulp (DPSC), apical papilla (SCAP) and follicle (DFSC) during this study. Hepatogenic potential of stem cells was assessed by functional assays at both genetic and protein level. We observed higher expression of most of the hepatic markers post differentiation in DPSCs compared to other cell types. LC-MS/MS analysis of stem cell secretome revealed the presence of different proteins related to hepatogenic lineage like growth arrest specific protein 6, oncostatin M, hepatocyte growth factor receptor etc. Interactome and Reactome pathway analysis revealed the interaction of DPSC/SCAP secretome proteins and these proteins were found to be associated with various pathways involved in lipid transport and metabolism. To the best of our knowledge, this is the first study regarding detailed investigation of hepatogenic potential of BMSCs v/s DMSCs (DPSC, SCAP & DFSC) along-with secretome characterization.
Collapse
Affiliation(s)
- Ajay Kumar
- Department of Biophysics, PGIMER, Chandigarh, India
| | - Vinod Kumar
- Department of Nephrology, PGIMER, Chandigarh, India
| | - Vidya Rattan
- Unit of Oral and Maxillofacial surgery, Oral health science centre, PGIMER, Chandigarh, India
| | - Vivekananda Jha
- Department of Nephrology, PGIMER, Chandigarh, India.,University of Oxford, Oxford, UK
| | - Arnab Pal
- Department of Biochemistry, PGIMER, Chandigarh, India
| | | |
Collapse
|
15
|
Omer L, Hudson EA, Zheng S, Hoying JB, Shan Y, Boyd NL. CRISPR Correction of a Homozygous Low-Density Lipoprotein Receptor Mutation in Familial Hypercholesterolemia Induced Pluripotent Stem Cells. Hepatol Commun 2017; 1:886-898. [PMID: 29130076 PMCID: PMC5677509 DOI: 10.1002/hep4.1110] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 09/12/2017] [Indexed: 01/26/2023] Open
Abstract
UNLABELLED Familial hypercholesterolemia (FH) is a hereditary disease primarily due to mutations in the low-density lipoprotein receptor (LDLR) that lead to elevated cholesterol and premature development of cardiovascular disease. Homozygous FH patients (HoFH) with two dysfunctional LDLR alleles are not as successfully treated with standard hypercholesterol therapies, and more aggressive therapeutic approaches to control cholesterol levels must be considered. Liver transplant can resolve HoFH, and hepatocyte transplantation has shown promising results in animals and humans. However, demand for donated livers and high-quality hepatocytes overwhelm the supply. Human pluripotent stem cells can differentiate to hepatocyte-like cells (HLCs) with the potential for experimental and clinical use. To be of future clinical use as autologous cells, LDLR genetic mutations in derived FH-HLCs need to be corrected. Genome editing technology clustered-regularly-interspaced-short-palindromic-repeats/CRISPR-associated 9 (CRISPR/Cas9) can repair pathologic genetic mutations in human induced pluripotent stem cells. CONCLUSION We used CRISPR/Cas9 genome editing to permanently correct a 3-base pair homozygous deletion in LDLR exon 4 of patient-derived HoFH induced pluripotent stem cells. The genetic correction restored LDLR-mediated endocytosis in FH-HLCs and demonstrates the proof-of-principle that CRISPR-mediated genetic modification can be successfully used to normalize HoFH cholesterol metabolism deficiency at the cellular level.
Collapse
Affiliation(s)
- Linda Omer
- Cardiovascular Innovation InstituteUniversity of Louisville School of MedicineLouisvilleKY
- Department of Biochemistry and Molecular GeneticsUniversity of Louisville School of MedicineLouisvilleKY
| | - Elizabeth A. Hudson
- Department of Biochemistry and Molecular GeneticsUniversity of Louisville School of MedicineLouisvilleKY
| | - Shirong Zheng
- Cardiovascular Innovation InstituteUniversity of Louisville School of MedicineLouisvilleKY
| | - James B. Hoying
- Cardiovascular Innovation InstituteUniversity of Louisville School of MedicineLouisvilleKY
- Department of PhysiologyUniversity of Louisville School of MedicineLouisvilleKY
| | - Yuan Shan
- Departments of Anatomic Pathology and Neurosurgery, Scott & White Medical Center, College of MedicineTexas A&M UniversityTempleTX
| | - Nolan L. Boyd
- Cardiovascular Innovation InstituteUniversity of Louisville School of MedicineLouisvilleKY
- Department of PhysiologyUniversity of Louisville School of MedicineLouisvilleKY
| |
Collapse
|
16
|
Lipid Metabolism and Emerging Targets for Lipid-Lowering Therapy. Can J Cardiol 2017; 33:872-882. [DOI: 10.1016/j.cjca.2016.12.019] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 12/12/2016] [Accepted: 12/26/2016] [Indexed: 12/25/2022] Open
|
17
|
Generation of Human Liver Chimeric Mice with Hepatocytes from Familial Hypercholesterolemia Induced Pluripotent Stem Cells. Stem Cell Reports 2017; 8:605-618. [PMID: 28262545 PMCID: PMC5355732 DOI: 10.1016/j.stemcr.2017.01.027] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 01/25/2017] [Accepted: 01/26/2017] [Indexed: 12/11/2022] Open
Abstract
Familial hypercholesterolemia (FH) causes elevation of low-density lipoprotein cholesterol (LDL-C) in blood and carries an increased risk of early-onset cardiovascular disease. A caveat for exploration of new therapies for FH is the lack of adequate experimental models. We have created a comprehensive FH stem cell model with differentiated hepatocytes (iHeps) from human induced pluripotent stem cells (iPSCs), including genetically engineered iPSCs, for testing therapies for FH. We used FH iHeps to assess the effect of simvastatin and proprotein convertase subtilisin/kexin type 9 (PCSK9) antibodies on LDL-C uptake and cholesterol lowering in vitro. In addition, we engrafted FH iHeps into the liver of Ldlr-/-/Rag2-/-/Il2rg-/- mice, and assessed the effect of these same medications on LDL-C clearance and endothelium-dependent vasodilation in vivo. Our iHep models recapitulate clinical observations of higher potency of PCSK9 antibodies compared with statins for reversing the consequences of FH, demonstrating the utility for preclinical testing of new therapies for FH patients.
Collapse
|
18
|
Santos RD, Gidding SS, Hegele RA, Cuchel MA, Barter PJ, Watts GF, Baum SJ, Catapano AL, Chapman MJ, Defesche JC, Folco E, Freiberger T, Genest J, Hovingh GK, Harada-Shiba M, Humphries SE, Jackson AS, Mata P, Moriarty PM, Raal FJ, Al-Rasadi K, Ray KK, Reiner Z, Sijbrands EJG, Yamashita S. Defining severe familial hypercholesterolaemia and the implications for clinical management: a consensus statement from the International Atherosclerosis Society Severe Familial Hypercholesterolemia Panel. Lancet Diabetes Endocrinol 2016; 4:850-61. [PMID: 27246162 DOI: 10.1016/s2213-8587(16)30041-9] [Citation(s) in RCA: 305] [Impact Index Per Article: 33.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 03/24/2016] [Accepted: 04/06/2016] [Indexed: 12/26/2022]
Abstract
Familial hypercholesterolaemia is common in individuals who had a myocardial infarction at a young age. As many as one in 200 people could have heterozygous familial hypercholesterolaemia, and up to one in 300 000 individuals could be homozygous. The phenotypes of heterozygous and homozygous familial hypercholesterolaemia overlap considerably; the response to treatment is also heterogeneous. In this Review, we aim to define a phenotype for severe familial hypercholesterolaemia and identify people at highest risk for cardiovascular disease, based on the concentration of LDL cholesterol in blood and individuals' responsiveness to conventional lipid-lowering treatment. We assess the importance of molecular characterisation and define the role of other cardiovascular risk factors and advanced subclinical coronary atherosclerosis in risk stratification. Individuals with severe familial hypercholesterolaemia might benefit in particular from early and more aggressive cholesterol-lowering treatment (eg, with PCSK9 inhibitors). In addition to better tailored therapy, more precise characterisation of individuals with severe familial hypercholesterolaemia could improve resource use.
Collapse
Affiliation(s)
- Raul D Santos
- Lipid Clinic Heart Institute (InCor), University of São Paulo Medical School Hospital, and Preventive Medicine Centre and Cardiology Program, Hospital Israelita Albert Einstein, São Paulo, Brazil.
| | - Samuel S Gidding
- Nemours Cardiac Center, A I DuPont Hospital for Children, Wilmington, DE, USA
| | - Robert A Hegele
- Department of Medicine and Robarts Research Institute, Schulich School of Medicine, Western University, London, ON, Canada
| | - Marina A Cuchel
- Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Philip J Barter
- School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Gerald F Watts
- Lipid Disorders Clinic, Royal Perth Hospital, The University of Western Australia, Perth, WA, Australia
| | - Seth J Baum
- Preventive Cardiology, Christine E Lynn Women's Health & Wellness Institute, Boca Raton Regional Hospital, Boca Raton, FL, USA
| | - Alberico L Catapano
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy; IRCCS Multimedica, Milan, Italy
| | | | - Joep C Defesche
- University of Amsterdam, Academic Medical Center (AMC), Amsterdam, Netherlands
| | | | - Tomas Freiberger
- Molecular Genetics Lab, Centre for Cardiovascular Surgery and Transplantation, and Ceitec, Masaryk University, Brno, Czech Republic
| | - Jacques Genest
- McGill University Health Center, Royal Victoria Hospital, Montreal, QC, Canada
| | - G Kees Hovingh
- University of Amsterdam, Academic Medical Center (AMC), Amsterdam, Netherlands
| | - Mariko Harada-Shiba
- National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Steve E Humphries
- Centre for Cardiovascular Genetics, Institute of Cardiovascular Science, University College of London, London, UK
| | - Ann S Jackson
- International Atherosclerosis Society, Houston, TX, USA
| | - Pedro Mata
- Fundación Hipercolesterolemia Familiar, Madrid, Spain
| | - Patrick M Moriarty
- Atherosclerosis and Lipoprotein-Apheresis Center, University of Kansas Medical Center, Kansas City, KS, USA
| | - Frederick J Raal
- Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | | | - Kausik K Ray
- School of Public Health, Imperial College London, London, UK
| | - Zelijko Reiner
- European Association for Cardiovascular Prevention and Rehabilitations, Zagreb, Croatia
| | | | | | | |
Collapse
|
19
|
Ajufo E, Cuchel M. Recent Developments in Gene Therapy for Homozygous Familial Hypercholesterolemia. Curr Atheroscler Rep 2016; 18:22. [PMID: 26980316 DOI: 10.1007/s11883-016-0579-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Homozygous familial hypercholesterolemia (HoFH) is a life-threatening Mendelian disorder with a mean life expectancy of 33 years despite maximally tolerated standard lipid-lowering therapies. This disease is an ideal candidate for gene therapy, and in the last few years, a number of exciting developments have brought this approach closer to the clinic than ever before. In this review, we discuss in detail the most advanced of these developments, a recombinant adeno-associated virus (AAV) vector carrying a low-density lipoprotein receptor (LDLR) transgene which has recently entered phase 1/2a testing. We also review ongoing development of approaches to enhance transgene expression, improve the efficiency of hepatocyte transduction, and minimize the AAV capsid-specific adaptive immune response. We include a summary of key gene therapy approaches for HoFH in pre-clinical development, including RNA silencing of the gene encoding HMG-CoA reductase (HMGCR) and induced pluripotent stem cell transplant therapy.
Collapse
Affiliation(s)
- Ezim Ajufo
- Department of Medicine, Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Marina Cuchel
- Department of Medicine, Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
20
|
Ramakrishnan VM, Tien KT, McKinley TR, Bocard BR, McCurry TM, Williams SK, Hoying JB, Boyd NL. Wnt5a Regulates the Assembly of Human Adipose Derived Stromal Vascular Fraction-Derived Microvasculatures. PLoS One 2016; 11:e0151402. [PMID: 26963616 PMCID: PMC4786226 DOI: 10.1371/journal.pone.0151402] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 02/27/2016] [Indexed: 01/05/2023] Open
Abstract
Human adipose-derived stromal vascular fraction (hSVF) cells are an easily accessible, heterogeneous cell system that can spontaneously self-assemble into functional microvasculatures in vivo. However, the mechanisms underlying vascular self-assembly and maturation are poorly understood, therefore we utilized an in vitro model to identify potential in vivo regulatory mechanisms. We utilized passage one (P1) hSVF because of the rapid UEA1+ endothelium (EC) loss at even P2 culture. We exposed hSVF cells to a battery of angiogenesis inhibitors and found that the pan-Wnt inhibitor IWP2 produced the most significant hSVF-EC networking decrease (~25%). To determine which Wnt isoform(s) and receptor(s) may be involved, hSVF was screened by PCR for isoforms associated with angiogenesis, with only WNT5A and its receptor, FZD4, being expressed for all time points observed. Immunocytochemistry confirmed Wnt5a protein expression by hSVF. To see if Wnt5a alone could restore IWP2-induced EC network inhibition, recombinant human Wnt5a (0-150 ng/ml) was added to IWP2-treated cultures. The addition of rhWnt5a significantly increased EC network area and significantly decreased the ratio of total EC network length to EC network area compared to untreated controls. To determine if Wnt5a mediates in vivo microvascular self-assembly, 3D hSVF constructs containing an IgG isotype control, anti-Wnt5a neutralizing antibody or rhWnt5a were implanted subcutaneously for 2w in immune compromised mice. Compared to IgG controls, anti-Wnt5a treatment significantly reduced vessel length density by ~41%, while rhWnt5a significantly increased vessel length density by ~62%. However, anti-Wnt5a or rhWnt5a did not significantly affect the density of segments and nodes, both of which measure vascular complexity. Taken together, this data demonstrates that endogenous Wnt5a produced by hSVF plays a regulatory role in microvascular self-assembly in vivo. These findings also suggest that manipulating Wnt signaling could enhance control of hSVF vascularization in tissue engineering applications.
Collapse
Affiliation(s)
- Venkat M. Ramakrishnan
- Division of Cardiovascular Therapeutics, Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky, United States of America
- Department of Physiology, University of Louisville, Louisville, Kentucky, United States of America
| | - Kevin T. Tien
- Division of Cardiovascular Therapeutics, Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky, United States of America
| | - Thomas R. McKinley
- Division of Cardiovascular Therapeutics, Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky, United States of America
| | - Braden R. Bocard
- Division of Cardiovascular Therapeutics, Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky, United States of America
- Georgetown College, Georgetown, Kentucky, United States of America
| | - Terry M. McCurry
- Division of Plastic Surgery, Department of Surgery, University of Louisville, Louisville, Kentucky, United States of America
| | - Stuart K. Williams
- Division of Cardiovascular Therapeutics, Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky, United States of America
- Department of Physiology, University of Louisville, Louisville, Kentucky, United States of America
| | - James B. Hoying
- Division of Cardiovascular Therapeutics, Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky, United States of America
- Department of Physiology, University of Louisville, Louisville, Kentucky, United States of America
| | - Nolan L. Boyd
- Division of Cardiovascular Therapeutics, Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky, United States of America
- Department of Physiology, University of Louisville, Louisville, Kentucky, United States of America
| |
Collapse
|