1
|
Liu Y, Li P, Yang Y. Advancements in utilizing CD34 + stem cells for repairing diabetic vascular damage. Biochem Biophys Res Commun 2025; 750:151411. [PMID: 39889623 DOI: 10.1016/j.bbrc.2025.151411] [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: 11/07/2024] [Revised: 01/16/2025] [Accepted: 01/27/2025] [Indexed: 02/03/2025]
Abstract
Diabetes-related vascular damage is a frequent complication of diabetes that results in structural and functional impairment of blood vessels. This damage significantly heightens the risk of cardiovascular events. CD34+ stem cells have shown great potential in the treatment of diabetes-related vascular damage due to their differentiation and vascular repair capabilities. This article provides a review of the research hotspots on the role and mechanisms of CD34+ stem cells in the repair of diabetes-related vascular damage, including changes in cell quantity and function during diabetes, as well as the latest research on activating, protecting, or repairing these cells to prevent or treat vascular damage. The article also summarizes the impact of diabetes on the mobilization and function of CD34+ stem cells, emphasizing how diabetes negatively affects their ability to promote angiogenesis. These deficits can result in various complications, including issues with small blood vessels, coronary heart disease, foot problems, and retinal complications. On the clinical side, the article highlights the positive effects of CD34+ stem cell therapy in improving vascular function and tissue repair in diabetic patients, while also mentioning the inconsistencies in results between diabetes models and clinical studies, which necessitate further research to optimize treatment strategies. It emphasizes the importance of enhancing the mobilization, homing, and repair capabilities of CD34+ stem cells, as well as combining them with other treatment methods, to develop more effective strategies for treating diabetes-related vascular damage.
Collapse
Affiliation(s)
- Yiting Liu
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Pengyun Li
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China.
| | - Yan Yang
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China.
| |
Collapse
|
2
|
Bobrovskikh AV, Zubairova US, Naumenko LG, Doroshkov AV. Catching the Big Fish in Big Data: A Meta-Analysis of Zebrafish Kidney scRNA-Seq Datasets Highlights Conserved Molecular Profiles of Macrophages and Neutrophils in Vertebrates. BIOLOGY 2024; 13:773. [PMID: 39452082 PMCID: PMC11505477 DOI: 10.3390/biology13100773] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/10/2024] [Accepted: 09/26/2024] [Indexed: 10/26/2024]
Abstract
The innate immune system (IIS) is an ancient and essential defense mechanism that protects animals against a wide range of pathogens and diseases. Although extensively studied in mammals, our understanding of the IIS in other taxa remains limited. The zebrafish (Danio rerio) serves as a promising model organism for investigating IIS-related processes, yet the immunogenetics of fish are not fully elucidated. To address this gap, we conducted a meta-analysis of single-cell RNA sequencing (scRNA-seq) datasets from zebrafish kidney marrow, encompassing approximately 250,000 immune cells. Our analysis confirms the presence of key genetic pathways in zebrafish innate immune cells that are similar to those identified in mammals. Zebrafish macrophages specifically express genes encoding cathepsins, major histocompatibility complex class II proteins, integral membrane proteins, and the V-ATPase complex and demonstrate the enrichment of oxidative phosphorylation ferroptosis processes. Neutrophils are characterized by the significant expression of genes encoding actins, cytoskeleton organizing proteins, the Arp2/3 complex, and glycolysis enzymes and have demonstrated their involvement in GnRH and CLR signaling pathways, adherents, and tight junctions. Both macrophages and neutrophils highly express genes of NOD-like receptors, phagosomes, and lysosome pathways and genes involved in apoptosis. Our findings reinforce the idea about the existence of a wide spectrum of immune cell phenotypes in fish since we found only a small number of cells with clear pro- or anti-inflammatory signatures.
Collapse
Affiliation(s)
- Aleksandr V. Bobrovskikh
- Department of Physics, Novosibirsk State University, 630090 Novosibirsk, Russia;
- The Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (U.S.Z.); (A.V.D.)
| | - Ulyana S. Zubairova
- The Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (U.S.Z.); (A.V.D.)
- Department of Information Technologies, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Ludmila G. Naumenko
- Department of Physics, Novosibirsk State University, 630090 Novosibirsk, Russia;
- The Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (U.S.Z.); (A.V.D.)
| | - Alexey V. Doroshkov
- The Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (U.S.Z.); (A.V.D.)
- Department of Genomics and Bioinformatics, Institute of Fundamental Biology and Biotechnology, Siberian Federal University, 660036 Krasnoyarsk, Russia
| |
Collapse
|
3
|
El Omar R, Abdellaoui N, Coulibaly ST, Fontenille L, Lanza F, Gachet C, Freund JN, Negroni M, Kissa K, Tavian M. Macrophage depletion overcomes human hematopoietic cell engraftment failure in zebrafish embryo. Cell Death Dis 2024; 15:305. [PMID: 38693109 PMCID: PMC11063059 DOI: 10.1038/s41419-024-06682-x] [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: 04/04/2023] [Revised: 04/06/2024] [Accepted: 04/15/2024] [Indexed: 05/03/2024]
Abstract
Zebrafish is widely adopted as a grafting model for studying human development and diseases. Current zebrafish xenotransplantations are performed using embryo recipients, as the adaptive immune system, responsible for host versus graft rejection, only reaches maturity at juvenile stage. However, transplanted primary human hematopoietic stem/progenitor cells (HSC) rapidly disappear even in zebrafish embryos, suggesting that another barrier to transplantation exists before the onset of adaptive immunity. Here, using a labelled macrophage zebrafish line, we demonstrated that engraftment of human HSC induces a massive recruitment of macrophages which rapidly phagocyte transplanted cells. Macrophages depletion, by chemical or pharmacological treatments, significantly improved the uptake and survival of transplanted cells, demonstrating the crucial implication of these innate immune cells for the successful engraftment of human cells in zebrafish. Beyond identifying the reasons for human hematopoietic cell engraftment failure, this work images the fate of human cells in real time over several days in macrophage-depleted zebrafish embryos.
Collapse
Affiliation(s)
- Reine El Omar
- University of Strasbourg, INSERM, EFS Grand-Est, BPPS UMR-S1255, Strasbourg, France
- Université de Lorraine, CITHEFOR, F-54505, Vandoeuvre Les Nancy, France
| | | | - Safiatou T Coulibaly
- University of Strasbourg, CNRS, Architecture et Réactivité de l'ARN, UPR 9002, Strasbourg, France
- ITI Innovec, Strasbourg, France
| | | | - François Lanza
- University of Strasbourg, INSERM, EFS Grand-Est, BPPS UMR-S1255, Strasbourg, France
| | - Christian Gachet
- University of Strasbourg, INSERM, EFS Grand-Est, BPPS UMR-S1255, Strasbourg, France
| | - Jean-Noel Freund
- ITI Innovec, Strasbourg, France
- University of Strasbourg, INSERM, IRFAC/UMR-S1113, Strasbourg, France
- INSERM, U1256 - NGERE, Université de Lorraine, 54500, Vandoeuvre-lès-Nancy, France
| | - Matteo Negroni
- University of Strasbourg, CNRS, Architecture et Réactivité de l'ARN, UPR 9002, Strasbourg, France
- ITI Innovec, Strasbourg, France
| | - Karima Kissa
- University of Montpellier, VBIC, INSERM U1047, Montpellier, France
- AZELEAD SAS, Montpellier, France
| | - Manuela Tavian
- University of Strasbourg, INSERM, EFS Grand-Est, BPPS UMR-S1255, Strasbourg, France.
- ITI Innovec, Strasbourg, France.
- University of Strasbourg, INSERM, IRFAC/UMR-S1113, Strasbourg, France.
| |
Collapse
|
4
|
Carulli E, Pompilio G, Vinci MC. Human Hematopoietic Stem/Progenitor Cells in Type One Diabetes Mellitus Treatment: Is There an Ideal Candidate? Cells 2023; 12:cells12071054. [PMID: 37048127 PMCID: PMC10093723 DOI: 10.3390/cells12071054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/21/2023] [Accepted: 03/29/2023] [Indexed: 04/03/2023] Open
Abstract
Type 1 diabetes mellitus (T1DM) is a highly prevalent autoimmune disease causing the destruction of pancreatic islet β-cells. The resulting insulin production deficiency leads to a lifelong need for insulin re-placement therapy, systemic complications, and reduced life quality and expectancy. Cell therapy has been extensively attempted to restore insulin independence (IID), and autologous nonmyeloablative hematopoietic stem cell transplantation (AHST) has appeared to give the most promising results, but with a highly variable quote of patients achieving IID across the studies. We performed a comprehensive review of the trials involving stem cells, and in particular AHST, for the treatment of T1DM. We then pooled the patients enrolled in the different trials and looked for the patient characteristics that could be associated with the achievement of IID. We found a significantly higher probability of achieving IID in older patients (OR 1.17, 95%CI 1.06–1.33, p = 0.002) and a significantly lower probability in patients with a history of ketoacidosis (OR 0.23, 95%CI 0.06–0.78, p = 0.023). This suggests that there could be a population of patients more likely to benefit from AHST, but further data would be required to depict the profile of the ideal candidate.
Collapse
Affiliation(s)
- Ermes Carulli
- Doctoral Programme in Translational Medicine, Università di Milano, 20122 Milan, Italy
- Dipartimento di Scienze Cliniche e di Comunità, Università di Milano, 20122 Milan, Italy
- National Heart and Lung Institute, Imperial College London, London SW7 2BX, UK
- Correspondence:
| | - Giulio Pompilio
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (G.P.); (M.C.V.)
- Dipartimento di Scienze Biomediche, Chirurgiche e Odontoiatriche, Università di Milano, 20122 Milan, Italy
| | - Maria Cristina Vinci
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (G.P.); (M.C.V.)
| |
Collapse
|
5
|
Fadini GP, Albiero M. Impaired haematopoietic stem / progenitor cell traffic and multi-organ damage in diabetes. Stem Cells 2022; 40:716-723. [PMID: 35552468 PMCID: PMC9406601 DOI: 10.1093/stmcls/sxac035] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 05/04/2022] [Indexed: 11/18/2022]
Abstract
During antenatal development, hematopoietic stem/progenitor cells (HSPCs) arise from a specialized endothelium and migrate from the extraembryonic mesoderm to the fetal liver before establishing hematopoiesis in the bone marrow (BM). It is still debated whether, in adulthood, HSPCs display such ontologic overlap with vascular cells and capacity for endothelial differentiation. Yet, adult HSPCs retain a prominent migratory activity and traffic in the bloodstream to secondary lymphoid organs and all peripheral tissues, before eventually returning to the BM. While patrolling parenchymatous organs, HSPCs locate close to the vasculature, where they establish local hematopoietic islands and contribute to tissue homeostasis by paracrine signals. Solid evidence shows that diabetes mellitus jeopardizes the traffic of HSPCs from BM to the circulation and peripheral tissues, a condition called “mobilopathy.” A reduction in the levels of circulating HSPCs is the most immediate and apparent consequence, which has been consistently observed in human diabetes, and is strongly associated with future risk for multi-organ damage, including micro- and macro-angiopathy. But the shortage of HSPCs in the blood is only the visible tip of the iceberg. Abnormal HSPC traffic results from a complex interplay among metabolism, innate immunity, and hematopoiesis. Notably, mobilopathy is mechanistically connected with diabetes-induced myelopoiesis. Impaired traffic of HSPCs and enhanced generation of pro-inflammatory cells synergize for tissue damage and impair the resolution of inflammation. We herein summarize the current evidence that diabetes affects HSPC traffic, which are the causes and consequences of such alteration, and how it contributes to the overall disease burden.
Collapse
Affiliation(s)
- Gian Paolo Fadini
- Department of Medicine, University of Padova, Padua, Italy.,Veneto Institute of Molecular Medicine, Padua, Italy
| | - Mattia Albiero
- Department of Medicine, University of Padova, Padua, Italy.,Veneto Institute of Molecular Medicine, Padua, Italy
| |
Collapse
|
6
|
Sforza A, Vigorelli V, Rurali E, Perrucci GL, Gambini E, Arici M, Metallo A, Rinaldi R, Fiorina P, Barbuti A, Raucci A, Sacco E, Rocchetti M, Pompilio G, Genovese S, Vinci MC. Liraglutide preserves CD34+ stem cells from dysfunction Induced by high glucose exposure. Cardiovasc Diabetol 2022; 21:51. [PMID: 35397526 PMCID: PMC8994898 DOI: 10.1186/s12933-022-01486-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 03/02/2022] [Indexed: 02/07/2023] Open
Abstract
Background Glucagon like peptide-1 receptor agonists (GLP-1RAs) have shown to reduce mortality and cardiovascular events in patients with type 2 diabetes mellitus (T2DM). Since the impairment in number and function of vasculotrophic circulating CD34+ hematopoietic stem progenitor cells (HSPCs) in T2D has been reported to increase cardiovascular (CV) risk, we hypothesized that one of the mechanisms whereby GLP-1 RAs exert CV protective effects may be related to the ability to improve CD34+ HSPC function. Methods In cord blood (CB)-derived CD34+ HSPC, the expression of GLP-1 receptor (GLP-1R) mRNA, receptor protein and intracellular signaling was evaluated by RT-qPCR and Western Blot respectively. CD34+ HSPCs were exposed to high glucose (HG) condition and GLP-1RA liraglutide (LIRA) was added before as well as after functional impairment. Proliferation, CXCR4/SDF-1α axis activity and intracellular ROS production of CD34+ HSPC were evaluated. Results CD34+ HSPCs express GLP-1R at transcriptional and protein level. LIRA treatment prevented and rescued HSPC proliferation, CXCR4/SDF-1α axis activity and metabolic imbalance from HG-induced impairment. LIRA stimulation promoted intracellular cAMP accumulation as well as ERK1/2 and AKT signaling activation. The selective GLP-1R antagonist exendin (9–39) abrogated LIRA-dependent ERK1/2 and AKT phosphorylation along with the related protective effects. Conclusion We provided the first evidence that CD34+ HSPC express GLP-1R and that LIRA can favorably impact on cell dysfunction due to HG exposure. These findings open new perspectives on the favorable CV effects of GLP-1 RAs in T2DM patients. Supplementary Information The online version contains supplementary material available at 10.1186/s12933-022-01486-9.
Collapse
|
7
|
Chen S, Li M, Sun J, Wang D, Weng C, Zeng Y, Li Y, Huo S, Huang X, Li S, Zou T, Xu H. Human Umbilical Cord Blood-Derived CD133+CD34+ Cells Protect Retinal Endothelial Cells and Ganglion Cells in X-Irradiated Rats through Angioprotective and Neurotrophic Factors. Front Cell Dev Biol 2022; 10:801302. [PMID: 35223834 PMCID: PMC8866877 DOI: 10.3389/fcell.2022.801302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 01/12/2022] [Indexed: 11/13/2022] Open
Abstract
Radiation retinopathy (RR) is a common complication following radiation therapy of globe, head, and neck malignancies, and is characterized by microangiopathy, neuroretinopathy, and the irreversible loss of visual function. To date, there is no effective treatment for RR. Stem cells have been clinically used to treat retinal degeneration. CD133+CD34+ cells from human umbilical cord blood (hUCB-CD133+CD34+ cells), a subpopulation of hematopoietic stem cells, were applied to determine their protective efficacy on irradiated rat retinas. After X-ray irradiation on the retinas, rats were intravitreally injected with hUCB-CD133+CD34+ cells. Transplantation of hUCB-CD133+CD34+ cells prevented retinal dysfunction 2 weeks post-operation and lasted at least 8 weeks. CD133+CD34+ cells were distributed along the retinal vessel and migrated to the ganglion cell layer. Moreover, grafted CD133+CD34+ cells reduced the apoptosis of endothelial and ganglion cells in irradiated rats and increased the number of survived CD31+ retinal endothelial cells and Brn3a+ ganglion cells at 2 and 4 weeks, respectively, post-operation. Co-culturing of CD133+CD34+ cells or supernatants with irradiated human retinal microvascular endothelial cells (hRECs) in vitro, confirmed that CD133+CD34+ cells ameliorated hREC apoptosis caused by irradiation. Mechanistically, we found that angioprotective mediators and neurotrophic factors were secreted by CD133+CD34+ cells, which might attenuate irradiation-induced injury of retinal endothelial cells and ganglion cells. hUCB-CD133+CD34+ cell transplantation, as a novel treatment, protects retinal endothelial and ganglion cells of X-irradiated rat retinas, possibly through angioprotective and neurotrophic factors.
Collapse
Affiliation(s)
- Siyu Chen
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Key Lab of Visual Damage and Regeneration and Restoration of Chongqing, Chongqing, China
| | - Minghui Li
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Key Lab of Visual Damage and Regeneration and Restoration of Chongqing, Chongqing, China
| | - Jianguo Sun
- Cancer Institute, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Dan Wang
- Department of Obstetrics and Gynecology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Chuanhuang Weng
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Key Lab of Visual Damage and Regeneration and Restoration of Chongqing, Chongqing, China
| | - Yuxiao Zeng
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Key Lab of Visual Damage and Regeneration and Restoration of Chongqing, Chongqing, China
| | - Yijian Li
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Key Lab of Visual Damage and Regeneration and Restoration of Chongqing, Chongqing, China
| | - Shujia Huo
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Key Lab of Visual Damage and Regeneration and Restoration of Chongqing, Chongqing, China
| | - Xiaona Huang
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Key Lab of Visual Damage and Regeneration and Restoration of Chongqing, Chongqing, China
| | - Shiying Li
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Key Lab of Visual Damage and Regeneration and Restoration of Chongqing, Chongqing, China
| | - Ting Zou
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Key Lab of Visual Damage and Regeneration and Restoration of Chongqing, Chongqing, China
- *Correspondence: Ting Zou, ; Haiwei Xu,
| | - Haiwei Xu
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Key Lab of Visual Damage and Regeneration and Restoration of Chongqing, Chongqing, China
- *Correspondence: Ting Zou, ; Haiwei Xu,
| |
Collapse
|
8
|
Pouresmaeil V, Ebre S, Yazdi MS. Assessment of the Relationship Between CD34 Antigen and Vitamin D Level and Insulin Resistance in Patients With Type 2 Diabetes. Clin Diabetes 2022; 40:425-433. [PMID: 36385969 PMCID: PMC9606555 DOI: 10.2337/cd21-0091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Diabetes causes disorders in the performance of endothelial progenitor cells, and obesity and vitamin D deficiency are associated with endothelial dysfunction and cardiovascular disease. This case-control study investigated the relationship between serum CD34 antigen and vitamin D levels and insulin resistance in type 2 diabetes. The results showed that CD34 has a significant inverse relationship with BMI, A1C, fasting blood glucose, insulin resistance, and insulin levels and has a significant direct relationship with vitamin D levels. Both CD34 and vitamin D were found to be significantly associated with type 2 diabetes. The association between reduced CD34 and vitamin D levels with type 2 diabetes and increased insulin resistance suggests that these parameters may be helpful in assessing diabetes and predicting its complications.
Collapse
Affiliation(s)
- Vahid Pouresmaeil
- Department of Biochemistry, Faculty of Medicine, Mashhad Medical Sciences, Islamic Azad University, Mashhad, Iran
- Innovative Medical Research Center, Faculty of Medicine, Mashhad Medical Sciences, Islamic Azad University, Mashhad, Iran
| | - Shaker Ebre
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Mohammad Sarafraz Yazdi
- Department of Internal Medicine, Faculty of Medicine, Mashhad Medical Sciences, Islamic Azad University, Mashhad, Iran
| |
Collapse
|
9
|
Bonora BM, Albiero M, Morieri ML, Cappellari R, Amendolagine FI, Mazzucato M, Zambon A, Iori E, Avogaro A, Fadini GP. Fenofibrate increases circulating haematopoietic stem cells in people with diabetic retinopathy: a randomised, placebo-controlled trial. Diabetologia 2021; 64:2334-2344. [PMID: 34368894 DOI: 10.1007/s00125-021-05532-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 05/18/2021] [Indexed: 11/27/2022]
Abstract
AIM/HYPOTHESIS In two large RCTs, fenofibrate reduced the progression of diabetic retinopathy. We investigated whether fenofibrate increases circulating haematopoietic stem/progenitor cells (HSPCs), which have vascular properties and have been shown to protect from retinopathy. METHODS We conducted a 12 week parallel-group RCT comparing fenofibrate vs placebo. Patients with diabetic retinopathy and without other conditions that would affect HSPCs were enrolled at a tertiary diabetes outpatient clinic and randomised to receive fenofibrate or placebo based on a computer-generated sequence. Patients and study staff assessing the outcomes were blinded to group assignment. The primary endpoint was the change in the levels of circulating HSPCs, defined by expression of the stem cell markers CD34 and/or CD133. Secondary endpoints were the changes in endothelial progenitor cells, lipids, soluble mediators and gene expression. We used historical data on the association between HSPCs and retinopathy outcomes to estimate the effect of fenofibrate on retinopathy progression. RESULTS Forty-two participants with diabetic retinopathy were randomised and 41 completed treatment and were analysed (20 in the placebo group and 21 in the fenofibrate group). Mean age was 57.4 years, diabetes duration was 18.2 years and baseline HbA1c was 60 mmol/mol (7.6%). When compared with placebo, fenofibrate significantly increased levels of HSPCs expressing CD34 and/or CD133. CD34+ HSPCs non-significantly declined in the placebo group (mean ± SD -44.2 ± 31.6 cells/106) and significantly increased in the fenofibrate group (53.8 ± 31.1 cells/106). The placebo-subtracted increase in CD34+ HSPCs from baseline was 30% (99.3 ± 43.3 cells/106; p = 0.027) which, projected onto the relationship between HSPC levels and retinopathy outcomes, yielded an OR of retinopathy progression of 0.67 for fenofibrate vs placebo. Endothelial differentiation of CD34+ cells, estimated by the %KDR (kinase insert domain receptor) expression, was significantly reduced by fenofibrate. Fenofibrate decreased serum triacylglycerols, but the change in triacylglycerols was unrelated to the change in HSPCs. No effect was observed for endothelial progenitor cells, cytokines/chemokines (stromal-cell derived factor-1, vascular endothelial growth factor, monocyte chemoattractant protein-1) and gene expression in peripheral blood mononuclear cells. CONCLUSIONS/INTERPRETATION Fenofibrate increased HSPC levels in participants with diabetic retinopathy and this mechanism may explain why fenofibrate reduced retinopathy progression in previous studies. TRIAL REGISTRATION ClinicalTrials.gov NCT01927315.
Collapse
Affiliation(s)
- Benedetta Maria Bonora
- Department of Medicine, University of Padova, Padua, Italy
- Veneto Institute of Molecular Medicine, Padua, Italy
| | - Mattia Albiero
- Department of Medicine, University of Padova, Padua, Italy
- Veneto Institute of Molecular Medicine, Padua, Italy
| | | | | | | | | | - Alberto Zambon
- Department of Medicine, University of Padova, Padua, Italy
| | | | - Angelo Avogaro
- Department of Medicine, University of Padova, Padua, Italy
| | - Gian Paolo Fadini
- Department of Medicine, University of Padova, Padua, Italy.
- Veneto Institute of Molecular Medicine, Padua, Italy.
| |
Collapse
|
10
|
Tripaldi R, Lanuti P, Simeone PG, Liani R, Bologna G, Ciotti S, Simeone P, Di Castelnuovo A, Marchisio M, Cipollone F, Santilli F. Endogenous PCSK9 may influence circulating CD45 neg/CD34 bright and CD45 neg/CD34 bright/CD146 neg cells in patients with type 2 diabetes mellitus. Sci Rep 2021; 11:9659. [PMID: 33958634 PMCID: PMC8102605 DOI: 10.1038/s41598-021-88941-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 04/05/2021] [Indexed: 12/22/2022] Open
Abstract
Protease proprotein convertase subtilisin/kexin type 9 (PCSK9) is a regulator of LDL cholesterol clearance and has been associated with cardiovascular risk. PCSK9 inhibitors increase in vivo circulating endothelial progenitor cells (EPCs), a subtype of immature cells involved in ongoing endothelial repair. We hypothesized that the effect of PCSK9 on vascular homeostasis may be mediated by EPCs in patients with or without type 2 diabetes mellitus (T2DM). Eighty-two patients (45 with, 37 without T2DM) at high cardiovascular risk were enrolled in this observational study. Statin treatment was associated with higher circulating levels of PCSK9 in patients with and without T2DM (p < 0.001 and p = 0.036) and with reduced CD45neg/CD34bright (total EPC compartment) (p = 0.016) and CD45neg/CD34bright/CD146neg (early EPC) (p = 0.040) only among patients with T2DM. In the whole group of patients, statin treatment was the only independent predictor of low number of CD45neg/CD34bright (β = - 0.230; p = 0.038, adjusted R2 = 0.041). Among T2DM patients, PCSK9 circulating levels were inversely related and predicted both the number of CD45neg/CD34bright (β = - 0.438; p = 0.003, adjusted R2 = 0.173), and CD45neg/CD34bright/CD146neg (β = - 0.458; p = 0.002, adjusted R2 = 0.191) independently of age, gender, BMI and statin treatment. In high-risk T2DM patients, high endogenous levels of PCSK9 may have a detrimental effect on EPCs by reducing the endothelial repair and worsening the progression of atherothrombosis.
Collapse
Affiliation(s)
- Romina Tripaldi
- Department of Medicine and Aging Sciences, Center for Advanced Studies and Technology (CAST), Via Luigi Polacchi, Chieti, Italy
| | - Paola Lanuti
- Department of Medicine and Aging Sciences, Center for Advanced Studies and Technology (CAST), Via Luigi Polacchi, Chieti, Italy
| | - Paola Giustina Simeone
- Department of Medicine and Aging Sciences, Center for Advanced Studies and Technology (CAST), Via Luigi Polacchi, Chieti, Italy
| | - Rossella Liani
- Department of Medicine and Aging Sciences, Center for Advanced Studies and Technology (CAST), Via Luigi Polacchi, Chieti, Italy
| | - Giuseppina Bologna
- Department of Medicine and Aging Sciences, Center for Advanced Studies and Technology (CAST), Via Luigi Polacchi, Chieti, Italy
| | - Sonia Ciotti
- Department of Medicine and Aging Sciences, Center for Advanced Studies and Technology (CAST), Via Luigi Polacchi, Chieti, Italy
| | - Pasquale Simeone
- Department of Medicine and Aging Sciences, Center for Advanced Studies and Technology (CAST), Via Luigi Polacchi, Chieti, Italy
| | | | - Marco Marchisio
- Department of Medicine and Aging Sciences, Center for Advanced Studies and Technology (CAST), Via Luigi Polacchi, Chieti, Italy
| | - Francesco Cipollone
- Department of Medicine and Aging Sciences, Center for Advanced Studies and Technology (CAST), Via Luigi Polacchi, Chieti, Italy
| | - Francesca Santilli
- Department of Medicine and Aging Sciences, Center for Advanced Studies and Technology (CAST), Via Luigi Polacchi, Chieti, Italy.
| |
Collapse
|
11
|
Somasagara RR, Huang X, Xu C, Haider J, Serody JS, Armistead PM, Leung T. Targeted therapy of human leukemia xenografts in immunodeficient zebrafish. Sci Rep 2021; 11:5715. [PMID: 33707624 PMCID: PMC7952715 DOI: 10.1038/s41598-021-85141-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 02/25/2021] [Indexed: 01/05/2023] Open
Abstract
Personalized medicine holds tremendous promise for improving safety and efficacy of drug therapies by optimizing treatment regimens. Rapidly developed patient-derived xenografts (pdx) could be a helpful tool for analyzing the effect of drugs against an individual's tumor by growing the tumor in an immunodeficient animal. Severe combined immunodeficiency (SCID) mice enable efficient in vivo expansion of vital tumor cells and generation of personalized xenografts. However, they are not amenable to large-scale rapid screening, which is critical in identifying new compounds from large compound libraries. The development of a zebrafish model suitable for pdx could facilitate large-scale screening of drugs targeted against specific malignancies. Here, we describe a novel strategy for establishing a zebrafish model for drug testing in leukemia xenografts. We used chronic myelogenous leukemia and acute myeloid leukemia for xenotransplantation into SCID zebrafish to evaluate drug screening protocols. We showed the in vivo efficacy of the ABL inhibitor imatinib, MEK inhibitor U0126, cytarabine, azacitidine and arsenic trioxide. We performed corresponding in vitro studies, demonstrating that combination of MEK- and FLT3-inhibitors exhibit an enhanced effect in vitro. We further evaluated the feasibility of zebrafish for transplantation of primary human hematopoietic cells that can survive at 15 day-post-fertilization. Our results provide critical insights to guide development of high-throughput platforms for evaluating leukemia.
Collapse
Affiliation(s)
- Ranganatha R Somasagara
- The Julius L. Chambers Biomedical/Biotechnology Research Institute, North Carolina Central University, North Carolina Research Campus, Kannapolis, NC, 28081, USA
| | - Xiaoyan Huang
- The Julius L. Chambers Biomedical/Biotechnology Research Institute, North Carolina Central University, North Carolina Research Campus, Kannapolis, NC, 28081, USA
| | - Chunyu Xu
- The Julius L. Chambers Biomedical/Biotechnology Research Institute, North Carolina Central University, North Carolina Research Campus, Kannapolis, NC, 28081, USA
| | - Jamil Haider
- The Julius L. Chambers Biomedical/Biotechnology Research Institute, North Carolina Central University, North Carolina Research Campus, Kannapolis, NC, 28081, USA
| | - Jonathan S Serody
- Division of Hematology/Oncology, Department of Medicine, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Paul M Armistead
- Division of Hematology/Oncology, Department of Medicine, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - TinChung Leung
- The Julius L. Chambers Biomedical/Biotechnology Research Institute, North Carolina Central University, North Carolina Research Campus, Kannapolis, NC, 28081, USA. .,Department of Biological & Biomedical Sciences, North Carolina Central University, Durham, NC, 27707, USA.
| |
Collapse
|
12
|
Albiero M, Bonora BM, Fadini GP. Diabetes pharmacotherapy and circulating stem/progenitor cells. State of the art and evidence gaps. Curr Opin Pharmacol 2020; 55:151-156. [PMID: 33271409 DOI: 10.1016/j.coph.2020.10.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 10/22/2020] [Accepted: 10/23/2020] [Indexed: 12/13/2022]
Abstract
Diabetes is burdened with the development of several end-organ complications leading to excess mortality. Though the causes of such organ damage are far from being clarified, diabetes has been redefined as a disease of impaired damage control, wherein ongoing damage is not adequately compensated by activation of repair processes. Bone marrow-derived hematopoietic stem/progenitor cells (HSPCs) and their descendants endothelial progenitor cells (EPCs) have been extensively studied as major players in tissue homeostasis as well as biomarkers of diabetic complication risk. Thus, strategies to raise the levels of circulating HSPCs/EPCs have attracted interest for their potential to modify the future risk of complications. We herein discuss state-of-the-art of the effects exerted by diabetes pharmacotherapy on such cell populations. Further, we highlight which outstanding questions remain to be addressed for a more comprehensive understanding of this topic.
Collapse
Affiliation(s)
- Mattia Albiero
- Department of Medicine, University of Padova, 35128 Padova, Italy; Veneto Institute of Molecular Medicine, 35128 Padova, Italy
| | - Benedetta Maria Bonora
- Department of Medicine, University of Padova, 35128 Padova, Italy; Veneto Institute of Molecular Medicine, 35128 Padova, Italy
| | - Gian Paolo Fadini
- Department of Medicine, University of Padova, 35128 Padova, Italy; Veneto Institute of Molecular Medicine, 35128 Padova, Italy.
| |
Collapse
|
13
|
Díaz del Moral S, Barrena S, Muñoz-Chápuli R, Carmona R. Embryonic circulating endothelial progenitor cells. Angiogenesis 2020; 23:531-541. [DOI: 10.1007/s10456-020-09732-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 06/22/2020] [Indexed: 12/26/2022]
|
14
|
Fadini GP, Albiero M, Bonora BM, Avogaro A. Angiogenic Abnormalities in Diabetes Mellitus: Mechanistic and Clinical Aspects. J Clin Endocrinol Metab 2019; 104:5431-5444. [PMID: 31211371 DOI: 10.1210/jc.2019-00980] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 06/12/2019] [Indexed: 12/25/2022]
Abstract
CONTEXT Diabetes causes severe pathological changes to the microvasculature in many organs and tissues and is at the same time associated with an increased risk of coronary and peripheral macrovascular events. We herein review alterations in angiogenesis observed in human and experimental diabetes and how they contribute to diabetes onset and development of vascular complications. EVIDENCE ACQUISITION The English language medical literature was searched for articles reporting on angiogenesis/vasculogenesis abnormalities in diabetes and their clinical manifestations, mechanistic aspects, and possible therapeutic implications. EVIDENCE SYNTHESIS Angiogenesis is a complex process, driven by a multiplicity of molecular mechanisms and involved in several physiological and pathological conditions. Incompetent angiogenesis is pervasive in diabetic vascular complications, with both excessive and defective angiogenesis observed in various tissues. A striking different angiogenic response typically occurs in the retina vs the myocardium and peripheral circulation, but some commonalities in abnormal angiogenesis can explain the well-known association between microangiopathy and macroangiopathy. Impaired angiogenesis can also affect endocrine islet and adipose tissue function, providing a link to diabetes onset. Exposure to high glucose itself directly affects angiogenic/vasculogenic processes, and the mechanisms include defective responses to hypoxia and proangiogenic factors, impaired nitric oxide bioavailability, shortage of proangiogenic cells, and loss of pericytes. CONCLUSIONS Dissecting the molecular drivers of tissue-specific alterations of angiogenesis/vasculogenesis is an important challenge to devise new therapeutic approaches. Angiogenesis-modulating therapies should be carefully evaluated in view of their potential off-target effects. At present, glycemic control remains the most reasonable therapeutic strategy to normalize angiogenesis in diabetes.
Collapse
Affiliation(s)
- Gian Paolo Fadini
- Department of Medicine, University of Padova, Padova, Italy
- Venetian Institute of Molecular Medicine, Padova, Italy
| | - Mattia Albiero
- Department of Medicine, University of Padova, Padova, Italy
- Venetian Institute of Molecular Medicine, Padova, Italy
| | - Benedetta Maria Bonora
- Department of Medicine, University of Padova, Padova, Italy
- Venetian Institute of Molecular Medicine, Padova, Italy
| | - Angelo Avogaro
- Department of Medicine, University of Padova, Padova, Italy
| |
Collapse
|
15
|
Facchin F, Alviano F, Canaider S, Bianconi E, Rossi M, Bonsi L, Casadei R, Biava PM, Ventura C. Early Developmental Zebrafish Embryo Extract to Modulate Senescence in Multisource Human Mesenchymal Stem Cells. Int J Mol Sci 2019; 20:2646. [PMID: 31146388 PMCID: PMC6600478 DOI: 10.3390/ijms20112646] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 05/24/2019] [Accepted: 05/25/2019] [Indexed: 12/14/2022] Open
Abstract
Stem cells undergo senescence both in vivo, contributing to the progressive decline in self-healing mechanisms, and in vitro during prolonged expansion. Here, we show that an early developmental zebrafish embryo extract (ZF1) could act as a modulator of senescence in human mesenchymal stem cells (hMSCs) isolated from both adult tissues, including adipose tissue (hASCs), bone marrow (hBM-MSCs), dental pulp (hDP-MSCs), and a perinatal tissue such as the Wharton's Jelly (hWJ-MSCs). In all the investigated hMSCs, ZF1 decreased senescence-associated β-galactosidase (SA β-gal) activity and enhanced the transcription of TERT, encoding the catalytic telomerase core. In addition, it was associated, only in hASCs, with a transcriptional induction of BMI1, a pleiotropic repressor of senescence. In hBM-MSCs, hDP-MSCs, and hWJ-MSCs, TERT over-expression was concomitant with a down-regulation of two repressors of TERT, TP53 (p53), and CDKN1A (p21). Furthermore, ZF1 increased the natural ability of hASCs to perform adipogenesis. These results indicate the chance of using ZF1 to modulate stem cell senescence in a source-related manner, to be potentially used as a tool to affect stem cell senescence in vitro. In addition, its anti-senescence action could also set the basis for future in vivo approaches promoting tissue rejuvenation bypassing stem cell transplantation.
Collapse
Affiliation(s)
- Federica Facchin
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy.
- National Laboratory of Molecular Biology and Stem Cell Bioengineering of the National Institute of Biostructures and Biosystems (NIBB)-Eldor Lab, at the Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy.
| | - Francesco Alviano
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy.
| | - Silvia Canaider
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy.
- National Laboratory of Molecular Biology and Stem Cell Bioengineering of the National Institute of Biostructures and Biosystems (NIBB)-Eldor Lab, at the Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy.
| | - Eva Bianconi
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy.
- National Laboratory of Molecular Biology and Stem Cell Bioengineering of the National Institute of Biostructures and Biosystems (NIBB)-Eldor Lab, at the Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy.
| | - Martina Rossi
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy.
| | - Laura Bonsi
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy.
| | - Raffaella Casadei
- Department for Life Quality Studies (QuVi), University of Bologna, Corso D'Augusto 237, 47921 Rimini, Italy.
| | - Pier Mario Biava
- Scientific Institute of Research and Care Multimedica, Via Milanese 300, 20099 Sesto San Giovanni (Milano), Italy.
| | - Carlo Ventura
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy.
- National Laboratory of Molecular Biology and Stem Cell Bioengineering of the National Institute of Biostructures and Biosystems (NIBB)-Eldor Lab, at the Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy.
| |
Collapse
|
16
|
Bonora BM, Cappellari R, Albiero M, Avogaro A, Fadini GP. Effects of SGLT2 Inhibitors on Circulating Stem and Progenitor Cells in Patients With Type 2 Diabetes. J Clin Endocrinol Metab 2018; 103:3773-3782. [PMID: 30113651 DOI: 10.1210/jc.2018-00824] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 07/27/2018] [Indexed: 02/08/2023]
Abstract
CONTEXT Reduction in the levels of circulating stem cells (CSCs) and endothelial progenitor cells (EPCs) predicts development or progression of microangiopathy and macroangiopathy in patients with type 2 diabetes (T2D). OBJECTIVE We tested whether treatment with sodium glucose cotransporter-2 (SGLT2) inhibitors affected the levels of CSCs and EPCs. DESIGN A randomized trial of dapagliflozin vs placebo with open-label extension, and an open-label observational study of empagliflozin treatment. SETTING Tertiary referral diabetes outpatient clinic. PATIENTS Patients with T2D aged 18 to 75 years. INTERVENTION Dapagliflozin at 10 mg vs placebo (n = 31); empagliflozin at 10 mg (n = 15). MAIN OUTCOME MEASURES We measured CSCs (CD34+) and EPCs (CD34+KDR+) by flow cytometry at baseline, at 12 weeks, and after the extension period. RESULTS After 12 weeks, CSCs declined nonsignificantly in the dapagliflozin group, remained stable in the placebo group, and the change from baseline was not significantly different between the two groups. EPCs declined nonsignificantly in the dapagliflozin group, increased nonsignificantly in the placebo group, and the change from baseline was significantly different between the two groups. After an open-label extension period of about 1.5 years, CSCs remained stable over time, whereas EPCs significantly increased in patients who received dapagliflozin. In all patients, irrespectively of treatment, EPCs increased significantly from baseline to the end of observation, concomitantly with improvement in HbA1c. In a cohort of 15 patients who received open-label empagliflozin for 12 weeks, CSCs declined nonsignificantly, whereas EPCs remained stable. CONCLUSION SGLT2 inhibitors do not significantly increase CSCs or EPCs. Thus, cardiovascular protection by SGLT2 inhibitors may not directly involve stem/progenitor cells.
Collapse
Affiliation(s)
| | - Roberta Cappellari
- Department of Medicine, University of Padova, Padova, Italy
- Venetian Institute of Molecular Medicine, Padova, Italy
| | - Mattia Albiero
- Department of Medicine, University of Padova, Padova, Italy
- Venetian Institute of Molecular Medicine, Padova, Italy
| | - Angelo Avogaro
- Department of Medicine, University of Padova, Padova, Italy
| | - Gian Paolo Fadini
- Department of Medicine, University of Padova, Padova, Italy
- Venetian Institute of Molecular Medicine, Padova, Italy
| |
Collapse
|
17
|
Facchin F, Bianconi E, Canaider S, Basoli V, Biava PM, Ventura C. Tissue Regeneration without Stem Cell Transplantation: Self-Healing Potential from Ancestral Chemistry and Physical Energies. Stem Cells Int 2018; 2018:7412035. [PMID: 30057626 PMCID: PMC6051063 DOI: 10.1155/2018/7412035] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Accepted: 06/20/2018] [Indexed: 12/17/2022] Open
Abstract
The human body constantly regenerates after damage due to the self-renewing and differentiating properties of its resident stem cells. To recover the damaged tissues and regenerate functional organs, scientific research in the field of regenerative medicine is firmly trying to understand the molecular mechanisms through which the regenerative potential of stem cells may be unfolded into a clinical application. The finding that some organisms are capable of regenerative processes and the study of conserved evolutionary patterns in tissue regeneration may lead to the identification of natural molecules of ancestral species capable to extend their regenerative potential to human tissues. Such a possibility has also been strongly suggested as a result of the use of physical energies, such as electromagnetic fields and mechanical vibrations in human adult stem cells. Results from scientific studies on stem cell modulation confirm the possibility to afford a chemical manipulation of stem cell fate in vitro and pave the way to the use of natural molecules, as well as electromagnetic fields and mechanical vibrations to target human stem cells in their niche inside the body, enhancing human natural ability for self-healing.
Collapse
Affiliation(s)
- Federica Facchin
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
- National Laboratory of Molecular Biology and Stem Cell Bioengineering of the National Institute of Biostructures and Biosystems (NIBB) - Eldor Lab, Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy
| | - Eva Bianconi
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
- National Laboratory of Molecular Biology and Stem Cell Bioengineering of the National Institute of Biostructures and Biosystems (NIBB) - Eldor Lab, Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy
| | - Silvia Canaider
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
- National Laboratory of Molecular Biology and Stem Cell Bioengineering of the National Institute of Biostructures and Biosystems (NIBB) - Eldor Lab, Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy
| | - Valentina Basoli
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy
| | - Pier Mario Biava
- Scientific Institute of Research and Care Multimedica, Via Milanese 300, 20099 Sesto San Giovanni, Italy
| | - Carlo Ventura
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
- National Laboratory of Molecular Biology and Stem Cell Bioengineering of the National Institute of Biostructures and Biosystems (NIBB) - Eldor Lab, Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy
| |
Collapse
|
18
|
Fadini GP, Boscari F, Cappellari R, Galasso S, Rigato M, Bonora BM, D'Anna M, Bruttomesso D, Avogaro A. Effects of Hypoglycemia on Circulating Stem and Progenitor Cells in Diabetic Patients. J Clin Endocrinol Metab 2018; 103:1048-1055. [PMID: 29300991 DOI: 10.1210/jc.2017-02187] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 12/21/2017] [Indexed: 01/07/2023]
Abstract
CONTEXT Iatrogenic hypoglycemia is the most common acute diabetic complication, and it significantly increases morbidity. In people with diabetes, reduction in the levels of circulating stem and progenitor cells predicts adverse outcomes. OBJECTIVE To evaluate whether hypoglycemia in diabetes affects circulating stem cells and endothelial progenitor cells (EPCs). DESIGN We performed an experimental hypoglycemia study (Study 1) and a case-control study (Study 2). SETTING Tertiary referral inpatient clinic. PATIENTS AND OTHER PARTICIPANTS Type 1 diabetic patients (Study 1, n = 19); diabetic patients hospitalized for severe iatrogenic hypoglycemia, matched inpatient and outpatient controls (Study 2, n = 22/group). INTERVENTIONS Type 1 diabetic patients underwent two in-hospital sessions of glucose monitoring during a breakfast meal with or without induction of hypoglycemia in random order. In Study 2, patients hospitalized for hypoglycemia and matched controls were compared. MAIN OUTCOME MEASURE Circulating stem cells and EPCs were measured by flow cytometry based on the expression of CD34 and kinase insert domain receptor (KDR). RESULTS In Study 1, the physiologic decline of CD34+KDR+ EPCs from 8 am to 2 pm was abolished by insulin-induced hypoglycemia in type 1 diabetic patients. In Study 2, diabetic patients hospitalized for severe iatrogenic hypoglycemia had significantly lower levels of CD34+ stem cells and CD34+KDR+ EPCs compared with diabetic inpatients or outpatient controls. CONCLUSIONS In diabetic patients, a single mild hypoglycemic episode can compromise the physiologic EPC fluctuation, whereas severe hypoglycemia is associated with a marked reduction in stem cells and EPCs. These data provide a possible link between hypoglycemia and adverse outcomes of diabetes.
Collapse
Affiliation(s)
| | | | | | - Silvia Galasso
- Department of Medicine, University of Padova, Padova, Italy
| | - Mauro Rigato
- Department of Medicine, University of Padova, Padova, Italy
| | | | | | | | - Angelo Avogaro
- Department of Medicine, University of Padova, Padova, Italy
| |
Collapse
|
19
|
The utility of stem cells in pediatric urinary bladder regeneration. Pediatr Res 2018; 83:258-266. [PMID: 28915233 DOI: 10.1038/pr.2017.229] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 09/07/2017] [Indexed: 02/06/2023]
Abstract
Pediatric patients with a neurogenic urinary bladder, caused by developmental abnormalities including spina bifida, exhibit chronic urological problems. Surgical management in the form of enterocystoplasty is used to enlarge the bladder, but is associated with significant clinical complications. Thus, alternative methods to enterocystoplasty have been explored through the incorporation of stem cells with tissue engineering strategies. Within the context of this review, we will examine the use of bone marrow stem cells and induced pluripotent stem cells (iPSCs), as they relate to bladder regeneration at the anatomic and molecular levels. The use of bone marrow stem cells has demonstrated significant advances in bladder tissue regeneration as multiple aspects of bladder tissue have been recapitulated including the urothelium, bladder smooth muscle, vasculature, and peripheral nerves. iPSCs, on the other hand, have been well characterized and used in multiple tissue-regenerative settings, yet iPSC research is still in its infancy with regards to bladder tissue regeneration with recent studies describing the differentiation of iPSCs to the bladder urothelium. Finally, we examine the role of the Sonic Hedgehog signaling cascade that mediates the proliferative response during regeneration between bladder smooth muscle and urothelium. Taken together, this review provides a current, comprehensive perspective on bladder regeneration.
Collapse
|
20
|
Fadini GP, Tura A, Pacini G, Avogaro A, Vigili de Kreutzenberg S. Reduced circulating stem cells associate with excess fasting and post-load NEFA exposure in healthy adults with normal glucose tolerance. Atherosclerosis 2017; 261:117-123. [DOI: 10.1016/j.atherosclerosis.2017.03.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 02/20/2017] [Accepted: 03/01/2017] [Indexed: 12/27/2022]
|
21
|
Fadini GP, Rigato M, Cappellari R, Bonora BM, Avogaro A. Long-term Prediction of Cardiovascular Outcomes by Circulating CD34+ and CD34+CD133+ Stem Cells in Patients With Type 2 Diabetes. Diabetes Care 2017; 40:125-131. [PMID: 27815289 DOI: 10.2337/dc16-1755] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2016] [Accepted: 10/19/2016] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Cardiovascular risk varies substantially in the population with diabetes, and biomarkers can improve risk stratification. Circulating stem cells predict future cardiovascular events and death, but data for the population with diabetes are scant. In this study we evaluated the ability of circulating stem cell levels to predict future cardiovascular outcomes and improve risk discrimination in patients with type 2 diabetes. RESEARCH DESIGN AND METHODS A cohort of 187 patients with type 2 diabetes was monitored for a median of 6.1 years. The primary outcome was time to a first cardiovascular event, defined as 3-point major adverse cardiovascular event (cardiovascular death, nonfatal myocardial infarction, or nonfatal stroke) plus hospitalization for cardiovascular causes. At baseline, we measured six stem/progenitor cell phenotypes in peripheral blood based on expression of CD34, CD133, and KDR. RESULTS The primary outcome occurred in 48 patients (4.5/100 patient-years). Patients with incident cardiovascular events had significantly lower CD34+ and CD34+CD133+ cells than those without. Higher rates of cardiovascular events occurred in patients with below median levels of CD34+ and CD34+CD133+. In Cox proportional hazards regression analyses, a reduced CD34+ (hazard ratio 2.21 [95% CI 1.14-4.29]) and CD34+CD133+ (2.98 [1.46-6.08]) cell count independently predicted future events. Addition of the CD34+ cell count to the reference model or the UK Prospective Diabetes Study risk engine improved C statistics, continuous net reclassification improvement, and/or integrated discrimination index. CONCLUSIONS In patients with type 2 diabetes, a reduced baseline level of circulating CD34+ stem cells predicts adverse cardiovascular outcomes up to 6 years later and improves risk stratification.
Collapse
Affiliation(s)
| | - Mauro Rigato
- Department of Medicine, University of Padova, Padova, Italy
| | | | | | - Angelo Avogaro
- Department of Medicine, University of Padova, Padova, Italy
| |
Collapse
|
22
|
Nadlacki B, Suuronen EJ. Biomaterial strategies to improve the efficacy of bone marrow cell therapy for myocardial infarction. Expert Opin Biol Ther 2016; 16:1501-1516. [DOI: 10.1080/14712598.2016.1235149] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
23
|
Hlushchuk R, Brönnimann D, Correa Shokiche C, Schaad L, Triet R, Jazwinska A, Tschanz SA, Djonov V. Zebrafish Caudal Fin Angiogenesis Assay-Advanced Quantitative Assessment Including 3-Way Correlative Microscopy. PLoS One 2016; 11:e0149281. [PMID: 26950851 PMCID: PMC4780710 DOI: 10.1371/journal.pone.0149281] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 01/08/2016] [Indexed: 11/24/2022] Open
Abstract
Background Researchers evaluating angiomodulating compounds as a part of scientific projects or pre-clinical studies are often confronted with limitations of applied animal models. The rough and insufficient early-stage compound assessment without reliable quantification of the vascular response counts, at least partially, to the low transition rate to clinics. Objective To establish an advanced, rapid and cost-effective angiogenesis assay for the precise and sensitive assessment of angiomodulating compounds using zebrafish caudal fin regeneration. It should provide information regarding the angiogenic mechanisms involved and should include qualitative and quantitative data of drug effects in a non-biased and time-efficient way. Approach & Results Basic vascular parameters (total regenerated area, vascular projection area, contour length, vessel area density) were extracted from in vivo fluorescence microscopy images using a stereological approach. Skeletonization of the vasculature by our custom-made software Skelios provided additional parameters including “graph energy” and “distance to farthest node”. The latter gave important insights into the complexity, connectivity and maturation status of the regenerating vascular network. The employment of a reference point (vascular parameters prior amputation) is unique for the model and crucial for a proper assessment. Additionally, the assay provides exceptional possibilities for correlative microscopy by combining in vivo-imaging and morphological investigation of the area of interest. The 3-way correlative microscopy links the dynamic changes in vivo with their structural substrate at the subcellular level. Conclusions The improved zebrafish fin regeneration model with advanced quantitative analysis and optional 3-way correlative morphology is a promising in vivo angiogenesis assay, well-suitable for basic research and preclinical investigations.
Collapse
Affiliation(s)
| | | | | | - Laura Schaad
- Institute of Anatomy, University of Bern, Bern, Switzerland
| | - Ramona Triet
- Institute of Anatomy, University of Bern, Bern, Switzerland
| | - Anna Jazwinska
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | | | - Valentin Djonov
- Institute of Anatomy, University of Bern, Bern, Switzerland
- * E-mail:
| |
Collapse
|
24
|
Ishibashi T, Yokota T, Tanaka H, Ichii M, Sudo T, Satoh Y, Doi Y, Ueda T, Tanimura A, Hamanaka Y, Ezoe S, Shibayama H, Oritani K, Kanakura Y. ESAM is a novel human hematopoietic stem cell marker associated with a subset of human leukemias. Exp Hematol 2016; 44:269-81.e1. [PMID: 26774386 DOI: 10.1016/j.exphem.2015.12.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 12/24/2015] [Accepted: 12/28/2015] [Indexed: 12/26/2022]
Abstract
Reliable markers are essential to increase our understanding of the biological features of human hematopoietic stem cells and to facilitate the application of hematopoietic stem cells in the field of transplantation and regenerative medicine. We previously identified endothelial cell-selective adhesion molecule (ESAM) as a novel functional marker of hematopoietic stem cells in mice. Here, we found that ESAM can also be used to purify human hematopoietic stem cells from all the currently available sources (adult bone marrow, mobilized peripheral blood, and cord blood). Multipotent colony-forming units and long-term hematopoietic-reconstituting cells in immunodeficient mice were found exclusively in the ESAM(High) fraction of CD34(+)CD38(-) cells. The CD34(+)CD38(-) fraction of cord blood and collagenase-treated bone marrow contained cells exhibiting extremely high expression of ESAM; these cells are likely to be related to the endothelial lineage. Leukemia cell lines of erythroid and megakaryocyte origin, but not those of myeloid or lymphoid descent, were ESAM positive. However, high ESAM expression was observed in some primary acute myeloid leukemia cells. Furthermore, KG-1a myeloid leukemia cells switched from ESAM negative to ESAM positive with repeated leukemia reconstitution in vivo. Thus, ESAM is a useful marker for studying both human hematopoietic stem cells and leukemia cells.
Collapse
Affiliation(s)
- Tomohiko Ishibashi
- Department of Hematology and Oncology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Takafumi Yokota
- Department of Hematology and Oncology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.
| | - Hirokazu Tanaka
- Department of Hematology and Rheumatology, Kinki University Faculty of Medicine, Osakasayama, Osaka, Japan
| | - Michiko Ichii
- Department of Hematology and Oncology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Takao Sudo
- Department of Hematology and Oncology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yusuke Satoh
- Department of Hematology and Oncology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan; Department of Lifestyle Studies, Kobe Shoin Women's University, Kobe, Japan
| | - Yukiko Doi
- Department of Hematology and Oncology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Tomoaki Ueda
- Department of Hematology and Oncology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Akira Tanimura
- Department of Hematology and Oncology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yuri Hamanaka
- Department of Hematology and Oncology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Sachiko Ezoe
- Department of Hematology and Oncology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Hirohiko Shibayama
- Department of Hematology and Oncology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Kenji Oritani
- Department of Hematology and Oncology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yuzuru Kanakura
- Department of Hematology and Oncology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| |
Collapse
|
25
|
Martini G, Biscaro F, Boscaro E, Calabrese F, Lunardi F, Facco M, Agostini C, Zulian F, Fadini GP. Reduced levels of circulating progenitor cells in juvenile idiopathic arthritis are counteracted by anti TNF-α therapy. BMC Musculoskelet Disord 2015; 16:103. [PMID: 25925313 PMCID: PMC4418050 DOI: 10.1186/s12891-015-0555-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 04/16/2015] [Indexed: 02/07/2023] Open
Abstract
Background Endothelial progenitor cells (EPC) promote angiogenesis and vascular repair. Though reduced EPC levels have been shown in rheumatoid arthritis, no study has so far evaluated EPCs in children with juvenile idiopathic arthritis (JIA). We aimed to study circulating EPCs in children with JIA, their relation to disease activity, and effects of anti TNF-α treatment. Methods Circulating EPCs were quantified by flow cytometry based on CD34, CD133 and KDR expression in peripheral blood of 22 patients with oligoarticular JIA and 29 age-matched controls. EPCs were re-assessed in children with methotrexate-resistant oligo-extended JIA before and up to 12 month after initiation of anti-TNF-alpha therapy. Plasma concentrations of inflammatory and EPC-regulating factors were measured using a multiplex array. Confocal immunofluorescence was used to demonstrate EPCs in synovial tissues. Results Children with active JIA showed a significant reduction of relative and absolute counts of circulating progenitor cells and EPCs compared to age-matched healthy controls. CD34+ cell levels were modestly and inversely correlated to disease activity. A strong inverse correlation was found between serum TNF-α and EPC levels. In 8 patients treated with anti TNF-α agents, the number of EPCs rose to values similar to healthy controls. CD34+KDR+ EPCs were found in the synovial tissue of JIA children, but not in control. Conclusions Children with JIA have reduced levels of the vasculoprotective and proangiogenic EPCs. While EPCs may contribute to synovial tissue remodelling, EPC pauperization may indicate an excess cardiovascular risk if projected later in life.
Collapse
Affiliation(s)
- Giorgia Martini
- Paediatric Rheumatology Unit, Department of Paediatrics, 35128, Padova, Italy.
| | - Francesca Biscaro
- Paediatric Rheumatology Unit, Department of Paediatrics, 35128, Padova, Italy.
| | - Elisa Boscaro
- Department of Medicine, University of Padova, Via Giustiniani, 2, 35128, Padova, Italy.
| | - Fiorella Calabrese
- Department of Cardiovascular and Thoracic Sciences, University of Padova, Via Giustiniani, 2, 35128, Padova, Italy.
| | - Francesca Lunardi
- Department of Cardiovascular and Thoracic Sciences, University of Padova, Via Giustiniani, 2, 35128, Padova, Italy.
| | - Monica Facco
- Department of Medicine, University of Padova, Via Giustiniani, 2, 35128, Padova, Italy.
| | - Carlo Agostini
- Department of Medicine, University of Padova, Via Giustiniani, 2, 35128, Padova, Italy.
| | - Francesco Zulian
- Paediatric Rheumatology Unit, Department of Paediatrics, 35128, Padova, Italy.
| | - Gian Paolo Fadini
- Department of Medicine, University of Padova, Via Giustiniani, 2, 35128, Padova, Italy.
| |
Collapse
|
26
|
Albiero M, Poncina N, Tjwa M, Ciciliot S, Menegazzo L, Ceolotto G, Vigili de Kreutzenberg S, Moura R, Giorgio M, Pelicci P, Avogaro A, Fadini GP. Diabetes causes bone marrow autonomic neuropathy and impairs stem cell mobilization via dysregulated p66Shc and Sirt1. Diabetes 2014; 63:1353-65. [PMID: 24270983 DOI: 10.2337/db13-0894] [Citation(s) in RCA: 121] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Diabetes compromises the bone marrow (BM) microenvironment and reduces the number of circulating CD34(+) cells. Diabetic autonomic neuropathy (DAN) may impact the BM, because the sympathetic nervous system is prominently involved in BM stem cell trafficking. We hypothesize that neuropathy of the BM affects stem cell mobilization and vascular recovery after ischemia in patients with diabetes. We report that, in patients, cardiovascular DAN was associated with fewer circulating CD34(+) cells. Experimental diabetes (streptozotocin-induced and ob/ob mice) or chemical sympathectomy in mice resulted in BM autonomic neuropathy, impaired Lin(-)cKit(+)Sca1(+) (LKS) cell and endothelial progenitor cell (EPC; CD34(+)Flk1(+)) mobilization, and vascular recovery after ischemia. DAN increased the expression of the 66-kDa protein from the src homology and collagen homology domain (p66Shc) and reduced the expression of sirtuin 1 (Sirt1) in mice and humans. p66Shc knockout (KO) in diabetic mice prevented DAN in the BM, and rescued defective LKS cell and EPC mobilization. Hematopoietic Sirt1 KO mimicked the diabetic mobilization defect, whereas hematopoietic Sirt1 overexpression in diabetes rescued defective mobilization and vascular repair. Through p66Shc and Sirt1, diabetes and sympathectomy elevated the expression of various adhesion molecules, including CD62L. CD62L KO partially rescued the defective stem/progenitor cell mobilization. In conclusion, autonomic neuropathy in the BM impairs stem cell mobilization in diabetes with dysregulation of the life-span regulators p66Shc and Sirt1.
Collapse
|
27
|
Liu CL, Kwok HF, Cheng L, Ko CH, Wong CW, Ho TWF, Leung PC, Fung KP, Lau CBS. Molecular mechanisms of angiogenesis effect of active sub-fraction from root of Rehmannia glutinosa by zebrafish sprout angiogenesis-guided fractionation. JOURNAL OF ETHNOPHARMACOLOGY 2014; 151:565-75. [PMID: 24247081 DOI: 10.1016/j.jep.2013.11.019] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Revised: 08/17/2013] [Accepted: 11/10/2013] [Indexed: 05/04/2023]
Abstract
ETHNOPHARMACOLOGICAL IMPORTANCE The root of Rehmannia glutinosa (Rehmanniae Radix (RR)) is clinically used as a wound-healing agent in traditional Chinese medicine. Angiogenesis acts crucially in the pathogenesis of chronic wound healing. The present study investigated the angiogenesis effect and its underlying mechanism of RR through zebrafish sprout angiogenesis guided-fractionation. MATERIALS AND METHODS The in vivo angiogenesis effect was studied by analyzing the number of ectopic sprouts formed upon sub-intestinal vessel of transgenic TG(fli1:EGFP)(y1)/+(AB) zebrafish embryos by fluorescence microscopy. Quantitative real-time PCR gene expression of the zebrafish embryos was further performed using a panel of 30 angiogenesis-associated genes designed for zebrafish sprout angiogenesis. Classical in vitro angiogenesis assays using human microvascular endothelial cells (HMEC-1) was accompanied. RESULTS We demonstrated that among all RR sub-fractions tested, C1-1 treated-zebrafish embryos possessed the most potent angiogenesis activities (from 190 to 780 ng/ml, p<0.001) in sprout formation in the zebrafish model. Quantitative gene expression of the treated embryos demonstrated significant up-regulation in MMP-9 (p<0.05), ANGPT1 (p<0.05), EGFR (p<0.05), EPHB4 (p<0.01), and significant down-regulation in Ephrin B2 (p<0.05), Flt-1 (p<0.05) and Ets-1 (p<0.05). C1-1 treatment could also significantly (p<0.001-0.05) stimulate HMEC-1 cell migration in scratch assay. Significant increase (p<0.05) in mean tubule length was observed in the C1-1-treated HMEC-1 cells in the tubule formation assay. CONCLUSIONS Our zebrafish sprout angiogenesis model-guided fractionation revealed that C1-1 possessed the most potent angiogenesis effect in RR. The design of the panel with 30 tailor-made angiogenesis-associated genes exhibited in zebrafish gene expression analysis showed that C1-1 could trigger differential expression of various angiogenesis-associated genes, such as VEGFR3 and MMP9, which played key role in angiogenesis. The pro-angiogenic activity of C1-1 was further confirmed in the translated study in motogenic and tubule-inducing effect using HMEC-1.
Collapse
Affiliation(s)
- Cheuk-Lun Liu
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, Hong Kong; State Key Laboratory of Phytochemistry and Plant Resources in West China, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, Hong Kong; School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, Hong Kong
| | - Hin-Fai Kwok
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, Hong Kong; State Key Laboratory of Phytochemistry and Plant Resources in West China, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, Hong Kong
| | - Ling Cheng
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, Hong Kong; State Key Laboratory of Phytochemistry and Plant Resources in West China, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, Hong Kong
| | - Chun-Hay Ko
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, Hong Kong; State Key Laboratory of Phytochemistry and Plant Resources in West China, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, Hong Kong
| | - Chun-Wai Wong
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, Hong Kong; State Key Laboratory of Phytochemistry and Plant Resources in West China, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, Hong Kong
| | - Tina Wai Fong Ho
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, Hong Kong; State Key Laboratory of Phytochemistry and Plant Resources in West China, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, Hong Kong
| | - Ping-Chung Leung
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, Hong Kong; State Key Laboratory of Phytochemistry and Plant Resources in West China, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, Hong Kong
| | - Kwok-Pui Fung
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, Hong Kong; State Key Laboratory of Phytochemistry and Plant Resources in West China, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, Hong Kong; School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, Hong Kong
| | - Clara Bik-San Lau
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, Hong Kong; State Key Laboratory of Phytochemistry and Plant Resources in West China, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, Hong Kong.
| |
Collapse
|
28
|
Orlova VV, Drabsch Y, Freund C, Petrus-Reurer S, van den Hil FE, Muenthaisong S, Dijke PT, Mummery CL. Functionality of Endothelial Cells and Pericytes From Human Pluripotent Stem Cells Demonstrated in Cultured Vascular Plexus and Zebrafish Xenografts. Arterioscler Thromb Vasc Biol 2014; 34:177-86. [DOI: 10.1161/atvbaha.113.302598] [Citation(s) in RCA: 154] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Valeria V. Orlova
- From the Department of Anatomy and Embryology (V.V.O., C.F., S.P.-R., F.E.v.d.H., S.M., C.L.M.) and Department of Molecular Cell Biology (V.V.O., Y.D., P.t.D.), Cancer Genomics Centre and Centre for Biomedical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Yvette Drabsch
- From the Department of Anatomy and Embryology (V.V.O., C.F., S.P.-R., F.E.v.d.H., S.M., C.L.M.) and Department of Molecular Cell Biology (V.V.O., Y.D., P.t.D.), Cancer Genomics Centre and Centre for Biomedical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Christian Freund
- From the Department of Anatomy and Embryology (V.V.O., C.F., S.P.-R., F.E.v.d.H., S.M., C.L.M.) and Department of Molecular Cell Biology (V.V.O., Y.D., P.t.D.), Cancer Genomics Centre and Centre for Biomedical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Sandra Petrus-Reurer
- From the Department of Anatomy and Embryology (V.V.O., C.F., S.P.-R., F.E.v.d.H., S.M., C.L.M.) and Department of Molecular Cell Biology (V.V.O., Y.D., P.t.D.), Cancer Genomics Centre and Centre for Biomedical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Francijna E. van den Hil
- From the Department of Anatomy and Embryology (V.V.O., C.F., S.P.-R., F.E.v.d.H., S.M., C.L.M.) and Department of Molecular Cell Biology (V.V.O., Y.D., P.t.D.), Cancer Genomics Centre and Centre for Biomedical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Suchitra Muenthaisong
- From the Department of Anatomy and Embryology (V.V.O., C.F., S.P.-R., F.E.v.d.H., S.M., C.L.M.) and Department of Molecular Cell Biology (V.V.O., Y.D., P.t.D.), Cancer Genomics Centre and Centre for Biomedical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Peter ten Dijke
- From the Department of Anatomy and Embryology (V.V.O., C.F., S.P.-R., F.E.v.d.H., S.M., C.L.M.) and Department of Molecular Cell Biology (V.V.O., Y.D., P.t.D.), Cancer Genomics Centre and Centre for Biomedical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Christine L. Mummery
- From the Department of Anatomy and Embryology (V.V.O., C.F., S.P.-R., F.E.v.d.H., S.M., C.L.M.) and Department of Molecular Cell Biology (V.V.O., Y.D., P.t.D.), Cancer Genomics Centre and Centre for Biomedical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
29
|
An in vivo method to quantify lymphangiogenesis in zebrafish. PLoS One 2012; 7:e45240. [PMID: 23028871 PMCID: PMC3441694 DOI: 10.1371/journal.pone.0045240] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Accepted: 08/17/2012] [Indexed: 11/19/2022] Open
Abstract
Background Lymphangiogenesis is a highly regulated process involved in the pathogenesis of disease. Current in vivo models to assess lymphangiogenesis are largely unphysiologic. The zebrafish is a powerful model system for studying development, due to its rapid growth and transparency during early stages of life. Identification of a network of trunk lymphatic capillaries in zebrafish provides an opportunity to quantify lymphatic growth in vivo. Methods and Results Late-phase microangiography was used to detect trunk lymphatic capillaries in zebrafish 2- and 3-days post-fertilization. Using this approach, real-time changes in lymphatic capillary development were measured in response to modulators of lymphangiogenesis. Recombinant human vascular endothelial growth factor (VEGF)-C added directly to the zebrafish aqueous environment as well as human endothelial and mouse melanoma cell transplantation resulted in increased lymphatic capillary growth, while morpholino-based knockdown of vegfc and chemical inhibitors of lymphangiogenesis added to the aqueous environment resulted in decreased lymphatic capillary growth. Conclusion Lymphatic capillaries in embryonic and larval zebrafish can be quantified using late-phase microangiography. Human activators and small molecule inhibitors of lymphangiogenesis, as well as transplanted human endothelial and mouse melanoma cells, alter lymphatic capillary development in zebrafish. The ability to rapidly quantify changes in lymphatic growth under physiologic conditions will allow for broad screening of lymphangiogenesis modulators, as well as help define cellular roles and elucidate pathways of lymphatic development.
Collapse
|
30
|
Fadini GP, Losordo D, Dimmeler S. Critical reevaluation of endothelial progenitor cell phenotypes for therapeutic and diagnostic use. Circ Res 2012; 110:624-37. [PMID: 22343557 DOI: 10.1161/circresaha.111.243386] [Citation(s) in RCA: 515] [Impact Index Per Article: 39.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Diverse subsets of endothelial progenitor cells (EPCs) are used for the treatment of ischemic diseases in clinical trials, and circulating EPCs levels are considered as biomarkers for coronary and peripheral artery disease. However, despite significant steps forward in defining their potential for both therapeutic and diagnostic purposes, further progress has been mired by unresolved questions around the definition and the mechanism of action of EPCs. Diverse culturing methods and detection of various combinations of different surface antigens were used to enrich and identify EPCs. These attempts were particularly challenged by the close relationship and overlapping markers of the endothelial and hematopoietic lineages. This article will critically review the most commonly used protocols to define EPCs by culture assays or by fluorescence-activated cell sorter in the context of their therapeutic or diagnostic use. We also delineate new research avenues to move forward our knowledge on EPC biology.
Collapse
Affiliation(s)
- Gian Paolo Fadini
- Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, University of Frankfurt, Theodor-Stern-Kai 7, Frankfurt, Germany
| | | | | |
Collapse
|