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
|
Pierre-Jerome C. The peripheral nervous system: peripheral neuropathies in the diabetic foot. MYOPATHIES AND TENDINOPATHIES OF THE DIABETIC FOOT 2025:451-482. [DOI: 10.1016/b978-0-443-13328-2.00022-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
3
|
Wu Z, Hou Q, Chi H, Liu J, Mei Y, Chen T, Yang K, Zheng J, Xu J, Wei F, Wang L. Single-cell RNA sequencing reveals a distinct profile of bone immune microenvironment and decreased osteoclast differentiation in type 2 diabetic mice. Genes Dis 2024; 11:101145. [PMID: 39281831 PMCID: PMC11399629 DOI: 10.1016/j.gendis.2023.101145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 08/07/2023] [Accepted: 09/16/2023] [Indexed: 09/18/2024] Open
Abstract
The pathogenic effects of type 2 diabetes on bone tissue are gaining attention, but the cellular and molecular mechanisms underlying osteoimmunology are still unclear in diabetes-related bone diseases. We delineated the single-cell transcriptome of bone marrow cells from both wide type and type 2 diabetes mice, which provided the first detailed global profile of bone marrow cells and revealed a distinct bone immune microenvironment at the genetic level under type 2 diabetic condition. It was observed that osteoclast activity was inhibited due to a dysregulated cytokine network, which ultimately led to decreased osteoclast formation and differentiation. In type 2 diabetes mice, a specific C d 36 + cluster (cluster 18, monocytes/macrophages 2) was identified as the precursor of osteoclasts with diminished differentiation potential. AP-1 was demonstrated to be the key transcription factor in the underlying mechanism.
Collapse
Affiliation(s)
- Zimei Wu
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
- Department of Orthopedic Surgery, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Qiaodan Hou
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Heng Chi
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Jihong Liu
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
- Southern University of Science and Technology Hospital, Shenzhen, Guangdong 518055, China
| | - Yixin Mei
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Tingting Chen
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Kunkun Yang
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Jingna Zheng
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Jing Xu
- Southern University of Science and Technology Hospital, Shenzhen, Guangdong 518055, China
| | - Fuxin Wei
- Department of Orthopedic Surgery, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Lin Wang
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
- Southern University of Science and Technology Hospital, Shenzhen, Guangdong 518055, China
| |
Collapse
|
4
|
Madonna R. Endothelial heterogeneity and their relevance in cardiac development and coronary artery disease. Vascul Pharmacol 2023; 153:107242. [PMID: 37940065 DOI: 10.1016/j.vph.2023.107242] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 10/30/2023] [Accepted: 11/04/2023] [Indexed: 11/10/2023]
Abstract
Micro- and macrovascular endothelial cells (ECs) are characterized by structural and functional heterogeneity, which is also reflected in their secretory activity. The root of this heterogeneity and related regulatory mechanisms are still poorly understood. During embryogenesis, microvascular ECs participate in organogenesis prior to the development of the fetal circulation, suggesting that ECs are capable of releasing paracrine trophogens, termed angiocrine factors (AFs). These are angiocrine growth factors, adhesion molecules, and chemokines, which are intended to promote morphogenesis and repair of the adjacent parenchyma/stroma where the vessels are located. There is a tissue and organ-specificity of AFs that traces the heterogeneity of ECs. This AF heterogeneity also traces how ECs respond to pathological conditions or exposure to cardiovascular risk factors. The study of the mechanisms that regulate endothelial and paracrine heterogeneity and that contribute to endotheliopathy represents a broad and as yet understudied area of research. A better understanding of the cellular and molecular mechanisms that regulate this heterogeneity, leading to endotheliopathy is an exciting challenge. In this brief review we will discuss experimental advances in the heterogeneity of ECs and their AF, with a focus on their involvement in the pathogenesis of coronary artery disease.
Collapse
Affiliation(s)
- Rosalinda Madonna
- Cardiology Division, Department of Pathology, University of Pisa, Via Paradisa, 56124 Pisa, Italy.
| |
Collapse
|
5
|
Bonora BM, Morieri ML, Marassi M, Cappellari R, Avogaro A, Fadini GP. Improved prediction of long-term kidney outcomes in people with type 2 diabetes by levels of circulating haematopoietic stem/progenitor cells. Diabetologia 2023; 66:2346-2355. [PMID: 37712954 PMCID: PMC10627906 DOI: 10.1007/s00125-023-06002-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 07/25/2023] [Indexed: 09/16/2023]
Abstract
AIM/HYPOTHESIS We examined whether prediction of long-term kidney outcomes in individuals with type 2 diabetes can be improved by measuring circulating levels of haematopoietic stem/progenitor cells (HSPCs), which are reduced in diabetes and are associated with cardiovascular risk. METHODS We included individuals with type 2 diabetes who had a baseline determination of circulating HSPCs in 2004-2019 at the diabetes centre of the University Hospital of Padua and divided them into two groups based on their median value per ml of blood. We collected updated data on eGFR and albuminuria up to December 2022. The primary endpoint was a composite of new-onset macroalbuminuria, sustained ≥40% eGFR decline, end-stage kidney disease or death from any cause. The analyses were adjusted for known predictors of kidney disease in the population with diabetes. RESULTS We analysed 342 participants (67.8% men) with a mean age of 65.6 years. Those with low HSPC counts (n=171) were significantly older and had a greater prevalence of hypertension, heart failure and nephropathy (45.0% vs 33.9%; p=0.036), as evidenced by lower eGFR and higher albuminuria at baseline. During a median follow-up of 6.7 years, participants with high vs low HSPC counts had lower rates of the composite kidney outcome (adjusted HR 0.69 [95% CI 0.49, 0.97]), slower decline in eGFR and a similar increase in albuminuria. Adding the HSPC information to the risk score of the CKD Prognosis Consortium significantly improved discrimination of individuals with future adverse kidney outcomes. CONCLUSIONS/INTERPRETATION HSPC levels predict worsening of kidney function and improve the identification of individuals with type 2 diabetes and adverse kidney outcomes over and beyond a clinical risk score.
Collapse
Affiliation(s)
- Benedetta Maria Bonora
- Department of Medicine, University of Padova, Padua, Italy
- Veneto Institute of Molecular Medicine, 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
|
6
|
Baumbach A, Cui YX, Evans RN, Culliford L, Johnson T, Rogers CA, Reeves BC, Bucciarelli-Ducci C, Harris J, Hamilton M, Madeddu P. A cohort study of circulating progenitor cells after ST-segment elevation and non-ST segment elevation myocardial infarction in non-diabetic and diabetic patients. Front Cardiovasc Med 2022; 9:1011140. [DOI: 10.3389/fcvm.2022.1011140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 11/03/2022] [Indexed: 11/19/2022] Open
Abstract
BackgroundMyocardial infarction induces elevation of progenitor cells in the circulation, a reparative response inhibited by type-2 diabetes.ObjectivesDetermine if myocardial infarct severity and diabetes interactively influence the migratory activity of CD34+/CXCR4+ progenitor cells and if the migratory test predicts cardiac outcomes.Materials and methodsA longitudinal study was conducted on patients with or without diabetes with a STEMI or NSTEMI. CD34+/CXCR4+ cells were measured in the peripheral blood using flow cytometry, and migratory activity was tested in vitro on cells isolated from samples collected on days 0 and 4 post-infarct. Cardiac function was assessed at three months using cardiac MRI.ResultsOf 1,149 patients screened, 71 (6.3%) were eligible and consented. Fifty had STEMI (16 with diabetes) and 21 NSTEMI (8 with diabetes). The proportion of CD34+/CXCR4+ cells within blood mononuclear cells was 1.96 times higher after STEMI compared with NSTEMI (GMR = 1.96, 95% CI 0.87, 4.37) and 1.55 times higher in patients with diabetes compared to patients without diabetes (GMR = 1.55, 95% CI 0.77, 3.13). In the latter, STEMI was associated with a 2.42-times higher proportion of migrated CD34 + /CXCR4 + cells compared with NSTEMI (GMR = 2.42, 95% CI 0.66, 8.81). In patients with diabetes, the association was the opposite, with a 55% reduction in the proportion of migrated CD34+/CXCR4+ cells. No statistically significant associations were observed between the frequency in peripheral blood or in vitro migration capacity of CD34+/CXCR4+ cells and MRI outcomes.ConclusionWe document the interaction between infarct and diabetes on the migratory activity of CD34+/CXCR4+ cells. The test did not predict functional outcomes in the studied cohort.
Collapse
|
7
|
Rachubik P, Szrejder M, Rogacka D, Typiak M, Audzeyenka I, Kasztan M, Pollock DM, Angielski S, Piwkowska A. Insulin controls cytoskeleton reorganization and filtration barrier permeability via the PKGIα-Rac1-RhoA crosstalk in cultured rat podocytes. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119301. [PMID: 35642843 DOI: 10.1016/j.bbamcr.2022.119301] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 05/16/2022] [Accepted: 05/19/2022] [Indexed: 06/15/2023]
Abstract
Podocyte foot processes are an important cellular layer of the glomerular barrier that regulates glomerular permeability. Insulin via the protein kinase G type Iα (PKGIα) signaling pathway regulates the balance between contractility and relaxation (permeability) of the podocyte barrier by regulation of the actin cytoskeleton. This mechanism was shown to be disrupted in diabetes. Rho family guanosine-5'-triphosphates (GTPases) are dynamic modulators of the actin cytoskeleton and expressed in cells that form the glomerular filtration barrier. Thus, changes in Rho GTPase activity may affect glomerular permeability to albumin. The present study showed that Rho family GTPases control podocyte migration and permeability. Moreover these processes are regulated by insulin in PKGIα-dependent manner. Modulation of the PKGI-dependent activity of Rac1 and RhoA GTPases with inhibitors or small-interfering RNA impair glomerular permeability to albumin. We also demonstrated this mechanism in obese, insulin-resistant Zucker rats. We propose that PKGIα-Rac1-RhoA crosstalk is necessary in proper organization of the podocyte cytoskeleton and consequently the stabilization of glomerular architecture and regulation of filtration barrier permeability.
Collapse
Affiliation(s)
- Patrycja Rachubik
- Mossakowski Medical Research Institute, Polish Academy of Sciences, Laboratory of Molecular and Cellular Nephrology, Gdańsk, Poland
| | - Maria Szrejder
- Mossakowski Medical Research Institute, Polish Academy of Sciences, Laboratory of Molecular and Cellular Nephrology, Gdańsk, Poland
| | - Dorota Rogacka
- Mossakowski Medical Research Institute, Polish Academy of Sciences, Laboratory of Molecular and Cellular Nephrology, Gdańsk, Poland; University of Gdańsk, Faculty of Chemistry, Department of Molecular Biotechnology, Gdańsk, Poland
| | - Marlena Typiak
- Mossakowski Medical Research Institute, Polish Academy of Sciences, Laboratory of Molecular and Cellular Nephrology, Gdańsk, Poland; University of Gdańsk, Faculty of Biology, Department of General and Medical Biochemistry, Gdańsk, Poland
| | - Irena Audzeyenka
- Mossakowski Medical Research Institute, Polish Academy of Sciences, Laboratory of Molecular and Cellular Nephrology, Gdańsk, Poland; University of Gdańsk, Faculty of Chemistry, Department of Molecular Biotechnology, Gdańsk, Poland
| | - Małgorzata Kasztan
- Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - David M Pollock
- Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Stefan Angielski
- Mossakowski Medical Research Institute, Polish Academy of Sciences, Laboratory of Molecular and Cellular Nephrology, Gdańsk, Poland
| | - Agnieszka Piwkowska
- Mossakowski Medical Research Institute, Polish Academy of Sciences, Laboratory of Molecular and Cellular Nephrology, Gdańsk, Poland; University of Gdańsk, Faculty of Chemistry, Department of Molecular Biotechnology, Gdańsk, Poland.
| |
Collapse
|
8
|
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: 12] [Impact Index Per Article: 4.0] [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
|
9
|
Madonna R. Angiocrine endothelium: From physiology to atherosclerosis and cardiac repair. Vascul Pharmacol 2022; 144:106993. [DOI: 10.1016/j.vph.2022.106993] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 03/25/2022] [Accepted: 03/30/2022] [Indexed: 02/08/2023]
|
10
|
Liu K, Liu D, Cui W. Protective Effect and Mechanism of Traditional Chinese Medicine on Myocardial Ischemia Reperfusion Injury. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:6121407. [PMID: 35399643 PMCID: PMC8991389 DOI: 10.1155/2022/6121407] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 03/24/2022] [Accepted: 03/25/2022] [Indexed: 11/17/2022]
Abstract
After acute myocardial infarction, early restoration of myocardial perfusion by thrombolysis or percutaneous coronary intervention is the most effective way to reduce the size of myocardial infarction and improve clinical outcomes. However, recovery of blood flow to the ischemic myocardium may cause ischemia-reperfusion (I/R) injury, a phenomenon that instead reduces the efficacy of myocardial reperfusion. Traditional Chinese medicine (TCM) has long been used for the treatment of cardiovascular diseases and has shown remarkable efficacy. Many studies have shown that some TCMs and their active components can exert protective effects against myocardial I/R injury through different mechanisms. This review summarized the protective mechanisms and current research advances of TCMs in myocardial I/R injury.
Collapse
Affiliation(s)
- Kuo Liu
- Cardiology Department, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| | - Demin Liu
- Cardiology Department, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| | - Wei Cui
- Cardiology Department, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| |
Collapse
|
11
|
Hematopoietic Progenitors and the Bone Marrow Niche Shape the Inflammatory Response and Contribute to Chronic Disease. Int J Mol Sci 2022; 23:ijms23042234. [PMID: 35216355 PMCID: PMC8879433 DOI: 10.3390/ijms23042234] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/14/2022] [Accepted: 02/15/2022] [Indexed: 11/17/2022] Open
Abstract
It is now well understood that the bone marrow (BM) compartment can sense systemic inflammatory signals and adapt through increased proliferation and lineage skewing. These coordinated and dynamic alterations in responding hematopoietic stem and progenitor cells (HSPCs), as well as in cells of the bone marrow niche, are increasingly viewed as key contributors to the inflammatory response. Growth factors, cytokines, metabolites, microbial products, and other signals can cause dysregulation across the entire hematopoietic hierarchy, leading to lineage-skewing and even long-term functional adaptations in bone marrow progenitor cells. These alterations may play a central role in the chronicity of disease as well as the links between many common chronic disorders. The possible existence of a form of “memory” in bone marrow progenitor cells is thought to contribute to innate immune responses via the generation of trained immunity (also called innate immune memory). These findings highlight how hematopoietic progenitors dynamically adapt to meet the demand for innate immune cells and how this adaptive response may be beneficial or detrimental depending on the context. In this review, we will discuss the role of bone marrow progenitor cells and their microenvironment in shaping the scope and scale of the immune response in health and disease.
Collapse
|
12
|
Ortillon J, Le Bail JC, Villard E, Léger B, Poirier B, Girardot C, Beeske S, Ledein L, Blanchard V, Brieu P, Naimi S, Janiak P, Guillot E, Meloni M. High Glucose Activates YAP Signaling to Promote Vascular Inflammation. Front Physiol 2021; 12:665994. [PMID: 34149446 PMCID: PMC8213390 DOI: 10.3389/fphys.2021.665994] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 05/11/2021] [Indexed: 12/17/2022] Open
Abstract
Background and Aims The YAP/TAZ signaling is known to regulate endothelial activation and vascular inflammation in response to shear stress. Moreover, YAP/TAZ signaling plays a role in the progression of cancers and renal damage associated with diabetes. However, whether YAP/TAZ signaling is also implicated in diabetes-associated vascular complications is not known. Methods The effect of high glucose on YAP/TAZ signaling was firstly evaluated in vitro on endothelial cells cultured under static conditions or subjected to shear stress (either laminar or oscillatory flow). The impact of diabetes on YAP/TAZ signaling was additionally assessed in vivo in db/db mice. Results In vitro, we found that YAP was dephosphorylated/activated by high glucose in endothelial cells, thus leading to increased endothelial inflammation and monocyte attachment. Moreover, YAP was further activated when high glucose was combined to laminar flow conditions. YAP was also activated by oscillatory flow conditions but, in contrast, high glucose did not exert any additional effect. Interestingly, inhibition of YAP reduced endothelial inflammation and monocyte attachment. Finally, we found that YAP is also activated in the vascular wall of diabetic mice, where inflammatory markers are also increased. Conclusion With the current study we demonstrated that YAP signaling is activated by high glucose in endothelial cells in vitro and in the vasculature of diabetic mice, and we pinpointed YAP as a regulator of high glucose-mediated endothelial inflammation and monocyte attachment. YAP inhibition may represent a potential therapeutic opportunity to improve diabetes-associated vascular complications.
Collapse
Affiliation(s)
- Jeremy Ortillon
- Cardiovascular Research Unit, Sanofi R&D, Chilly-Mazarin, France
| | | | - Elise Villard
- Cardiovascular Research Unit, Sanofi R&D, Chilly-Mazarin, France
| | - Bertrand Léger
- Cardiovascular Research Unit, Sanofi R&D, Chilly-Mazarin, France
| | - Bruno Poirier
- Cardiovascular Research Unit, Sanofi R&D, Chilly-Mazarin, France
| | | | - Sandra Beeske
- Cardiovascular Research Unit, Sanofi R&D, Chilly-Mazarin, France
| | - Laetitia Ledein
- Cardiovascular Research Unit, Sanofi R&D, Chilly-Mazarin, France
| | - Véronique Blanchard
- Molecular Histopathology and Bio-Imaging Translational Sciences, Sanofi R&D, Chilly-Mazarin, France
| | - Patrice Brieu
- Molecular Histopathology and Bio-Imaging Translational Sciences, Sanofi R&D, Chilly-Mazarin, France
| | - Souâd Naimi
- Molecular Histopathology and Bio-Imaging Translational Sciences, Sanofi R&D, Chilly-Mazarin, France
| | - Philip Janiak
- Cardiovascular Research Unit, Sanofi R&D, Chilly-Mazarin, France
| | - Etienne Guillot
- Cardiovascular Research Unit, Sanofi R&D, Chilly-Mazarin, France
| | - Marco Meloni
- Cardiovascular Research Unit, Sanofi R&D, Chilly-Mazarin, France
| |
Collapse
|
13
|
Simó-Servat O, Ramos H, Bogdanov P, García-Ramírez M, Huerta J, Hernández C, Simó R. ERM Complex, a Therapeutic Target for Vascular Leakage Induced by Diabetes. Curr Med Chem 2021; 29:2189-2199. [PMID: 34042029 DOI: 10.2174/0929867328666210526114417] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/30/2021] [Accepted: 05/06/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Ezrin, radixin, and moesin (the ERM complex) interact directly with membrane proteins regulating their attachment to actin filaments. ERM protein activation modifies cytoskeleton organization and alters the endothelial barrier function, thus favoring vascular leakage. However, little is known regarding the role of ERM proteins in diabetic retinopathy (DR). OBJECTIVE This study aimed to examine whether overexpression of the ERM complex exists in db/db mice and its main regulating factors. METHOD 9 male db/db mice and 9 male db/+ aged 14 weeks were analyzed. ERM proteins were assessed by western blot and by immunohistochemistry. Vascular leakage was determined by the Evans blue method. To assess ERM regulation, HRECs were cultured in a medium containing 5.5 mM D-glucose (mimicking physiological conditions) and 25 mM D-glucose (mimicking hyperglycemia that occurs in diabetic patients). Moreover, treatment with TNF-α, IL-1β, or VEGF was added to a high glucose condition. The expression of ERM proteins was quantified by RT-PCR. Cell permeability was evaluated by measuring movements of FITC-dextran. RESULTS A significant increase of ERM in diabetic mice in comparison with non-diabetic mice was observed. A high glucose condition alone did not have any effect on ERM expression. However, TNF-α and IL-1β induced a significant increase in ERM proteins. CONCLUSIONS The increase of ERM proteins induced by diabetes could be one of the mechanisms involved in vascular leakage and could be considered as a therapeutic target. Moreover, the upregulation of the ERM complex by diabetes is induced by inflammatory mediators rather than by high glucose itself.
Collapse
Affiliation(s)
- Olga Simó-Servat
- Diabetes and Metabolism Research Unit, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
| | - Hugo Ramos
- Diabetes and Metabolism Research Unit, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
| | - Patricia Bogdanov
- Diabetes and Metabolism Research Unit, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
| | - Marta García-Ramírez
- Diabetes and Metabolism Research Unit, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
| | - Jordi Huerta
- Diabetes and Metabolism Research Unit, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
| | - Cristina Hernández
- Diabetes and Metabolism Research Unit, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
| | - Rafael Simó
- Diabetes and Metabolism Research Unit, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
| |
Collapse
|
14
|
Guan Y, Wang X. Salvianic Acid A Regulates High-Glucose-Treated Endothelial Progenitor Cell Dysfunction via the AKT/Endothelial Nitric Oxide Synthase (eNOS) Pathway. Med Sci Monit 2021; 27:e928153. [PMID: 33770068 PMCID: PMC8008975 DOI: 10.12659/msm.928153] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND The primary cause of death in patients with diabetes mellitus (DM) is diabetic macroangiopathy, a complication that related to the function and number of endothelial progenitor cells (EPCs). Salvianic acid A (SAA) is a water-soluble active ingredient of Salvia miltiorrhiza, a traditional Chinese medicine used to treat cardiovascular diseases. The purpose of this study was to explore the effects of SAA on the function of rat EPCs cultured in vitro in a high-glucose environment. MATERIAL AND METHODS Bone marrow-derived EPCs from 40 Sprague-Dawley rats were identified by fluorescence staining. Cell viability, apoptosis, tube formation, lactated dehydrogenase (LDH) release, and nitric oxide (NO) production were detected by 3-[4,5-dimethylthylthiazol-2-yl]-2,5 diphenyltetrazolium bromide assay, flow cytometry, tube formation, LDH, and 3-amino,4-aminomethyl-2',7'-difluorescein, and diacetate assays, respectively. The expression levels of proteins were examined by western blotting. RESULTS Cultured EPCs showed a cobblestone morphology and positive expression of Dil-ac-LDL and FITC-UEA-1. High glucose impaired cell viability. Different concentrations of SAA had no significant effect on EPC viability. SAA reduced the apoptosis rate and LDH release, but promoted tube formation, viability, and NO production in high-glucose-treated EPCs. The ratios of p-AKT/AKT and p-eNOS/eNOS in high-glucose-treated EPCs were elevated by SAA. Phosphoinositide 3-kinase inhibitor LY294002 blocked the rescue effects of SAA on high-glucose-treated EPCs. CONCLUSIONS SAA protected EPCs against high-glucose-induced dysfunction via the AKT/eNOS pathway.
Collapse
Affiliation(s)
- Yanhua Guan
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China (mainland).,Department of Endocrinology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China (mainland)
| | - Xu Wang
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China (mainland).,Department of Endocrinology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China (mainland)
| |
Collapse
|
15
|
Chen M, Li Y, Huang X, Gu Y, Li S, Yin P, Zhang L, Tang P. Skeleton-vasculature chain reaction: a novel insight into the mystery of homeostasis. Bone Res 2021; 9:21. [PMID: 33753717 PMCID: PMC7985324 DOI: 10.1038/s41413-021-00138-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 11/18/2020] [Accepted: 12/16/2020] [Indexed: 02/01/2023] Open
Abstract
Angiogenesis and osteogenesis are coupled. However, the cellular and molecular regulation of these processes remains to be further investigated. Both tissues have recently been recognized as endocrine organs, which has stimulated research interest in the screening and functional identification of novel paracrine factors from both tissues. This review aims to elaborate on the novelty and significance of endocrine regulatory loops between bone and the vasculature. In addition, research progress related to the bone vasculature, vessel-related skeletal diseases, pathological conditions, and angiogenesis-targeted therapeutic strategies are also summarized. With respect to future perspectives, new techniques such as single-cell sequencing, which can be used to show the cellular diversity and plasticity of both tissues, are facilitating progress in this field. Moreover, extracellular vesicle-mediated nuclear acid communication deserves further investigation. In conclusion, a deeper understanding of the cellular and molecular regulation of angiogenesis and osteogenesis coupling may offer an opportunity to identify new therapeutic targets.
Collapse
Affiliation(s)
- Ming Chen
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Yi Li
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Xiang Huang
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Ya Gu
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Shang Li
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Pengbin Yin
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China.
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China.
| | - Licheng Zhang
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China.
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China.
| | - Peifu Tang
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China.
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China.
| |
Collapse
|
16
|
Santopaolo M, Sullivan N, Thomas AC, Alvino VV, Nicholson LB, Gu Y, Spinetti G, Kallikourdis M, Blom A, Madeddu P. Activation of Bone Marrow Adaptive Immunity in Type 2 Diabetes: Rescue by Co-stimulation Modulator Abatacept. Front Immunol 2021; 12:609406. [PMID: 33746953 PMCID: PMC7969721 DOI: 10.3389/fimmu.2021.609406] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 01/27/2021] [Indexed: 01/10/2023] Open
Abstract
Background: Chronic low-grade inflammation and alterations in innate and adaptive immunity were reported in Type 2 diabetes (T2D). Here, we investigated the abundance and activation of T cells in the bone marrow (BM) of patients with T2D. We then verified the human data in a murine model and tested if the activation of T cells can be rescued by treating mice with abatacept, an immunomodulatory drug employed for the treatment of rheumatoid arthritis. Clinical evidence indicated abatacept can slow the decline in beta-cell function. Methods: A cohort of 24 patients (12 with T2D) undergoing hip replacement surgery was enrolled in the study. Flow cytometry and cytokine analyses were performed on BM leftovers from surgery. We next compared the immune profile of db/db and control wt/db mice. In an additional study, db/db mice were randomized to receive abatacept or vehicle for 4 weeks, with endpoints being immune cell profile, indices of insulin sensitivity, and heart performance. Results: Patients with T2D showed increased frequencies of BM CD4+ (2.8-fold, p = 0.001) and CD8+ T cells (1.8-fold, p = 0.01), with the upregulation of the activation marker CD69 and the homing receptor CCR7 in CD4+ (1.64-fold, p = 0.003 and 2.27-fold, p = 0.01, respectively) and CD8+ fractions (1.79-fold, p = 0.05 and 1.69-fold, p = 0.02, respectively). These differences were confirmed in a multivariable regression model. CCL19 (CCR7 receptor ligand) and CXCL10/11 (CXCR3 receptor ligands), implicated in T-cell migration and activation, were the most differentially modulated chemokines. Studies in mice confirmed the activation of adaptive immunity in T2D. Abatacept reduced the activation of T cells and the levels of proinflammatory cytokines and improved cardiac function but not insulin sensitivity. Conclusions: Results provide proof-of-concept evidence for the activation of BM adaptive immunity in T2D. In mice, treatment with abatacept dampens the activation of adaptive immunity and protects from cardiac damage.
Collapse
Affiliation(s)
- Marianna Santopaolo
- Bristol Medical School, Translational Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Niall Sullivan
- University Hospitals Bristol NHS Trust, Bristol, United Kingdom
| | - Anita Coral Thomas
- Bristol Medical School, Translational Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Valeria Vincenza Alvino
- Bristol Medical School, Translational Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Lindsay B Nicholson
- Bristol Medical School, School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | - Yue Gu
- Bristol Medical School, Translational Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Gaia Spinetti
- Laboratory of Cardiovascular Research, Istituto di Ricovero e Cura a Carattere Scientifico MultiMedica, Milan, Italy
| | - Marinos Kallikourdis
- Department of Biomedical Sciences, Humanitas University, Milan, Italy.,Adaptive Immunity Laboratory, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Ashley Blom
- Bristol Medical School, Translational Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Paolo Madeddu
- Bristol Medical School, Translational Health Sciences, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
17
|
Gu Y, Rampin A, Alvino VV, Spinetti G, Madeddu P. Cell Therapy for Critical Limb Ischemia: Advantages, Limitations, and New Perspectives for Treatment of Patients with Critical Diabetic Vasculopathy. Curr Diab Rep 2021; 21:11. [PMID: 33651185 PMCID: PMC7925447 DOI: 10.1007/s11892-021-01378-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/13/2021] [Indexed: 12/18/2022]
Abstract
PURPOSE OF REVIEW To provide a highlight of the current state of cell therapy for the treatment of critical limb ischemia in patients with diabetes. RECENT FINDINGS The global incidence of diabetes is constantly growing with consequent challenges for healthcare systems worldwide. In the UK only, NHS costs attributed to diabetic complications, such as peripheral vascular disease, amputation, blindness, renal failure, and stroke, average £10 billion each year, with cost pressure being estimated to get worse. Although giant leaps forward have been registered in the scope of early diagnosis and optimal glycaemic control, an effective treatment for critical limb ischemia is still lacking. The present review aims to provide an update of the ongoing work in the field of regenerative medicine. Recent advancements but also limitations imposed by diabetes on the potential of the approach are addressed. In particular, the review focuses on the perturbation of non-coding RNA networks in progenitor cells and the possibility of using emerging knowledge on molecular mechanisms to design refined protocols for personalized therapy. The field of cell therapy showed rapid progress but has limitations. Significant advances are foreseen in the upcoming years thanks to a better understanding of molecular bottlenecks associated with the metabolic disorders.
Collapse
Affiliation(s)
- Y Gu
- Bristol Medical School, Translational Health Sciences, University of Bristol, Upper Maudlin Street, Bristol, BS2 8HW, UK
| | - A Rampin
- Laboratory of Cardiovascular Research, IRCCS, MultiMedica, Milan, Italy
| | - V V Alvino
- Bristol Medical School, Translational Health Sciences, University of Bristol, Upper Maudlin Street, Bristol, BS2 8HW, UK
| | - G Spinetti
- Laboratory of Cardiovascular Research, IRCCS, MultiMedica, Milan, Italy
| | - P Madeddu
- Bristol Medical School, Translational Health Sciences, University of Bristol, Upper Maudlin Street, Bristol, BS2 8HW, UK.
| |
Collapse
|
18
|
Manipulating Pericyte Function with MicroRNAs. Methods Mol Biol 2021. [PMID: 33576975 DOI: 10.1007/978-1-0716-1056-5_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/06/2023]
Abstract
MicroRNAs (miRNAs) are expressed in all cell types, including pericytes, and play essential roles in vascular development, homeostasis, and disease. Manipulation of pericytes with miRNA mimics and inhibitors represents an essential tool to study the role of pericytes in vascular development and regeneration and to better understand the therapeutic potential of miRNA manipulation in pericytes. Here we describe methods for manipulating pericyte function by using miRNA mimics and inhibitors. We also describe methods to assess pericyte function (proliferation and migration) after manipulation with miRNAs and explain how miRNA gene targets can be identified and validated in pericytes after manipulation with miRNA.
Collapse
|
19
|
Abstract
Obesity and obesity-related diseases like type 2 diabetes (T2D) are prominent global health issues; therefore, there is a need to better understand the mechanisms underlying these conditions. The onset of obesity is characterized by accumulation of proinflammatory cells, including Ly6chi monocytes (which differentiate into proinflammatory macrophages) and neutrophils, in metabolic tissues. This shift toward chronic, low-grade inflammation is an obese-state hallmark and highly linked to metabolic disorders and other obesity comorbidities. The mechanisms that induce and maintain increased inflammatory myelopoiesis are of great interest, with a recent focus on how obesity affects more primitive hematopoietic cells. The hematopoietic system is constantly replenished by proper regulation of hematopoietic stem and progenitor (HSPC) pools in the BM. While early research suggests that chronic obesity promotes expansion of myeloid-skewed HSPCs, the involvement of the hematopoietic stem cell (HSC) niche in regulating obesity-induced myelopoiesis remains undefined. In this review, we explore the role of the multicellular HSC niche in hematopoiesis and inflammation, and the potential contribution of this niche to the hematopoietic response to obesity. This review further aims to summarize the potential HSC niche involvement as a target of obesity-induced inflammation and a driver of obesity-induced myelopoiesis.
Collapse
|
20
|
Chen J, Shi W, Xu Y, Zhang H, Chen B. Hirudin prevents vascular endothelial cell apoptosis and permeability enhancement induced by the serum from rat with chronic renal failure through inhibiting RhoA/ROCK signaling pathway. Drug Dev Res 2020; 82:553-561. [PMID: 33345328 DOI: 10.1002/ddr.21773] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 11/25/2020] [Accepted: 12/02/2020] [Indexed: 11/12/2022]
Abstract
Endothelial cells injury and activation contribute to arteriovenous fistula (AVF) stenosis. Hirudin (Hiru) can inhibit the activity of thrombin, which was reported to enhance endothelial cell permeability and promote vascular inflammatory responses. RhoA/ROCK signaling pathway is also important in regulating vascular endothelial permeability. This study aimed to investigate the role of Hiru on the viability and permeability of human umbilical vein endothelial cells (HUVECs) following stimulation of serum from rat with chronic renal failure (CRF) and illustrated the effects of Hiru on RhoA/ROCK signaling. Wistar rats were randomly divided into control group and CRF group. Serum from each group was collected to stimulate HUVECs. Proliferation capability was estimated with Cell Count Kit-8 (CCK-8) assay. Transwell assay was performed to determine permeability. Cell apoptosis was examined using Tunel staining. Telomere length and telomerase activity were determined by qPCR. Moreover, the expression of RhoA, ROCK1 and ROCK2 was estimated via western blot. Results showed that the serum from CRF rat significantly inhibited cell viability while enhanced cell permeability and apoptosis. Different concentrations of Hiru prevented the above effects caused by CRF serum. Additionally, Hiru recovered the CRF serum-induced decreased telomere length and telomerase activity. Hiru also inhibited the protein expression of RhoA, ROCK1 and ROCK2, which were activated by CRF serum. Moreover, the ROCK inhibitor, Y27632, exhibited similar effects with Hiru. In conclusion, Hiru-restored HUVECs cell viability, telomere length and telomerase activity, suppressed permeability and apoptosis in the presence of CRF serum might depend on inactivating the RhoA/ROCK signaling.
Collapse
Affiliation(s)
- Jing Chen
- Department of Nephrology, Lianyungang Hospital of Traditional Chinese Medicine, Lianyungang, China
| | - Wenbin Shi
- Department of Nephrology, Lianyungang Hospital of Traditional Chinese Medicine, Lianyungang, China
| | - Yan Xu
- Department of Nephrology, Lianyungang Hospital of Traditional Chinese Medicine, Lianyungang, China
| | - Huaming Zhang
- Department of Nephrology, Lianyungang Hospital of Traditional Chinese Medicine, Lianyungang, China
| | - Bo Chen
- Department of Nephrology, Lianyungang Hospital of Traditional Chinese Medicine, Lianyungang, China
| |
Collapse
|
21
|
Regulation of JAM2 Expression in the Lungs of Streptozotocin-Induced Diabetic Mice and Human Pluripotent Stem Cell-Derived Alveolar Organoids. Biomedicines 2020; 8:biomedicines8090346. [PMID: 32932992 PMCID: PMC7555027 DOI: 10.3390/biomedicines8090346] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 08/30/2020] [Accepted: 09/09/2020] [Indexed: 12/26/2022] Open
Abstract
Hyperglycemia is a causative factor in the pathogenesis of respiratory diseases, known to induce fibrosis and inflammation in the lung. However, little attention has been paid to genes related to hyperglycemic-induced lung alterations and stem cell applications for therapeutic use. In this study, our microarray data revealed significantly increased levels of junctional adhesion molecule 2 (JAM2) in the high glucose (HG)-induced transcriptional profile in human perivascular cells (hPVCs). The elevated level of JAM2 in HG-treated hPVCs was transcriptionally and epigenetically reversible when HG treatment was removed. We further investigated the expression of JAM2 using in vivo and in vitro hyperglycemic models. Our results showed significant upregulation of JAM2 in the lungs of streptozotocin (STZ)-induced diabetic mice, which was greatly suppressed by the administration of conditioned medium obtained from human mesenchymal stem cell cultures. Furthermore, JAM2 was found to be significantly upregulated in human pluripotent stem cell-derived multicellular alveolar organoids by exposure to HG. Our results suggest that JAM2 may play an important role in STZ-induced lung alterations and could be a potential indicator for predicting the therapeutic effects of stem cells and drugs in diabetic lung complications.
Collapse
|
22
|
Qi K, Yang Y, Geng Y, Cui H, Li X, Jin C, Chen G, Tian X, Meng X. Tongxinluo attenuates oxygen-glucose-serum deprivation/restoration-induced endothelial barrier breakdown via peroxisome proliferator activated receptor-α/angiopoietin-like 4 pathway in high glucose-incubated human cardiac microvascular endothelial cells. Medicine (Baltimore) 2020; 99:e21821. [PMID: 32846824 PMCID: PMC7447398 DOI: 10.1097/md.0000000000021821] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Traditional Chinese medicine Tongxinluo (TXL) has been widely used to treat coronary artery disease in China, since it could reduce myocardial infarct size and ischemia/reperfusion injury in both non-diabetic and diabetic conditions. It has been shown that TXL could regulate peroxisome proliferator activated receptor-α (PPAR-α), a positive modulator of angiopoietin-like 4 (Angptl4), in diabetic rats. Endothelial junction substructure components, such as VE-cadherin, are involved in the protection of reperfusion injury. Thus, we hypothesized cell-intrinsic and endothelial-specific Angptl4 mediated the protection of TXL on endothelial barrier under high glucose condition against ischemia/reperfusion-injury via PPAR-α pathway. METHODS Incubated with high glucose medium, the human cardiac microvascular endothelial cells (HCMECs) were then exposed to oxygen-glucose-serum deprivation (2 hours) and restoration (2 hours) stimulation, with or without TXL, insulin, or rhAngptl4 pretreatment. RESULTS TXL, insulin, and rhAngptl4 had similar protective effects on the endothelial barrier. TXL treatment reversed the endothelial barrier breakdown in HCMECs significantly as identified by decreasing endothelial permeability, upregulating the expression of JAM-A, VE-cadherin, and integrin-α5 and increasing the membrane location of VE-cadherin and integrin-α5, and these effects of TXL were as effective as insulin and rhAngptl4. However, Angptl4 knock-down with small interfering RNA (siRNA) interference and PPAR-α inhibitor MK886 partially abrogated these beneficial effects of TXL. Western blotting also revealed that similar with insulin, TXL upregulated the expression of Angptl4 in HCMECs, which could be inhibited by Angptl4 siRNA or MK886 exposure. TXL treatment increased PPAR-α activity, which could be diminished by MK886 but not by Angptl4 siRNA. CONCLUSION These data suggest cell-intrinsic and endothelial-specific Angptl4 mediates the protection of TXL against endothelial barrier breakdown during oxygen-glucose-serum deprivation and restoration under high glucose condition partly via the PPAR-α/Angptl4 pathway.
Collapse
Affiliation(s)
- Kang Qi
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuejin Yang
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yongjian Geng
- Department of Internal Medicine, Division of Cardiology, The University of Texas Health Science Center at Houston, Houston, TX
| | - Hehe Cui
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiangdong Li
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chen Jin
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Guihao Chen
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaqiu Tian
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xianmin Meng
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
23
|
Simó-Servat O, Hernández C, Simó R. The ERM Complex: A New Player Involved in Diabetes-induced Vascular Leakage. Curr Med Chem 2020; 27:3012-3022. [PMID: 30332939 DOI: 10.2174/0929867325666181016162327] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 09/27/2018] [Accepted: 10/09/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND Microvascular complications remain an important cause of morbidity in diabetic patients, and they are associated with a significant economic burden for healthcare systems. Vascular leakage is one of the earlier hallmarks in diabetic microvascular complications. Ezrin, Radixin and Moesin (ERM) proteins have recently been involved in vascular dysfunction under the effect of molecular mediators of diabetes complications. In this review, we will present the available evidence regarding the role of these proteins in vascular leakage and their putative implication in diabetic microvascular complications. METHODS AND RESULTS A comprehensive literature search of the electronic MEDLINE database was performed between November 2017 and January 2018. As a result, 36 articles have been reviewed and discussed. DISCUSSION ERM proteins are cytoskeleton-membrane linkers, and when activated in endothelial cells are able to induce cytoskeleton reorganization in stress fibers leading to the disassembly of focal adhesions and the formation of paracellular gaps which result in an increase of vascular permeability. The activation of these proteins is induced by mediators involved in diabetic complications such as PKC activation, TNF-α, AGEs and oxidative stress. In conclusion, ERMs play an essential role in endothelium homeostasis and can be envisaged as a new therapeutic molecular target for preventing or arresting diabetes-induced vascular leakage.
Collapse
Affiliation(s)
- Olga Simó-Servat
- Diabetes and Metabolism Research Unit, Vall d'Hebron Research Institute, Barcelona, Spain.,Universitat Autònoma de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ICSIII), Madrid, Spain
| | - Cristina Hernández
- Diabetes and Metabolism Research Unit, Vall d'Hebron Research Institute, Barcelona, Spain.,Universitat Autònoma de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ICSIII), Madrid, Spain
| | - Rafael Simó
- Diabetes and Metabolism Research Unit, Vall d'Hebron Research Institute, Barcelona, Spain.,Universitat Autònoma de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ICSIII), Madrid, Spain
| |
Collapse
|
24
|
Bone marrow fat: friend or foe in people with diabetes mellitus? Clin Sci (Lond) 2020; 134:1031-1048. [PMID: 32337536 DOI: 10.1042/cs20200220] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/15/2020] [Accepted: 04/20/2020] [Indexed: 12/22/2022]
Abstract
Global trends in the prevalence of overweight and obesity put the adipocyte in the focus of huge medical interest. This review highlights a new topic in adipose tissue biology, namely the emerging pathogenic role of fat accumulation in bone marrow (BM). Specifically, we summarize current knowledge about the origin and function of BM adipose tissue (BMAT), provide evidence for the association of excess BMAT with diabetes and related cardiovascular complications, and discuss potential therapeutic approaches to correct BMAT dysfunction. There is still a significant uncertainty about the origins and function of BMAT, although several subpopulations of stromal cells have been suggested to have an adipogenic propensity. BM adipocytes are higly plastic and have a distinctive capacity to secrete adipokines that exert local and endocrine functions. BM adiposity is abundant in elderly people and has therefore been interpreted as a component of the whole-body ageing process. BM senescence and BMAT accumulation has been also reported in patients and animal models with Type 2 diabetes, being more pronounced in those with ischaemic complications. Understanding the mechanisms responsible for excess and altered function of BMAT could lead to new treatments able to preserve whole-body homeostasis.
Collapse
|
25
|
Tencerova M, Frost M, Figeac F, Nielsen TK, Ali D, Lauterlein JJL, Andersen TL, Haakonsson AK, Rauch A, Madsen JS, Ejersted C, Højlund K, Kassem M. Obesity-Associated Hypermetabolism and Accelerated Senescence of Bone Marrow Stromal Stem Cells Suggest a Potential Mechanism for Bone Fragility. Cell Rep 2020; 27:2050-2062.e6. [PMID: 31091445 DOI: 10.1016/j.celrep.2019.04.066] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 03/06/2019] [Accepted: 04/12/2019] [Indexed: 02/07/2023] Open
Abstract
Obesity is associated with increased risk for fragility fractures. However, the cellular mechanisms are unknown. Using a translational approach combining RNA sequencing and cellular analyses, we investigated bone marrow stromal stem cells (BM-MSCs) of 54 men divided into lean, overweight, and obese groups on the basis of BMI. Compared with BM-MSCs obtained from lean, obese BM-MSCs exhibited a shift of molecular phenotype toward committed adipocytic progenitors and increased expression of metabolic genes involved in glycolytic and oxidoreductase activity. Interestingly, compared with paired samples of peripheral adipose tissue-derived stromal cells (AT-MSCs), insulin signaling of obese BM-MSCs was enhanced and accompanied by increased abundance of insulin receptor positive (IR+) and leptin receptor positive (LEPR+) cells in BM-MSC cultures. Their hyper-activated metabolic state was accompanied by an accelerated senescence phenotype. Our data provide a plausible explanation for the bone fragility in obesity caused by enhanced insulin signaling leading to accelerated metabolic senescence of BM-MSCs.
Collapse
Affiliation(s)
- Michaela Tencerova
- Department of Molecular Endocrinology, KMEB, University of Southern Denmark and Odense University Hospital, 5000 Odense C, Denmark; OPEN, Odense Patient Data Explorative Network, Odense University Hospital, Odense, Denmark.
| | - Morten Frost
- Department of Molecular Endocrinology, KMEB, University of Southern Denmark and Odense University Hospital, 5000 Odense C, Denmark; Steno Diabetes Center Odense, Odense University Hospital, 5000 Odense C, Denmark
| | - Florence Figeac
- Department of Molecular Endocrinology, KMEB, University of Southern Denmark and Odense University Hospital, 5000 Odense C, Denmark
| | - Tina Kamilla Nielsen
- Department of Molecular Endocrinology, KMEB, University of Southern Denmark and Odense University Hospital, 5000 Odense C, Denmark
| | - Dalia Ali
- Department of Molecular Endocrinology, KMEB, University of Southern Denmark and Odense University Hospital, 5000 Odense C, Denmark
| | - Jens-Jacob Lindegaard Lauterlein
- Department of Molecular Endocrinology, KMEB, University of Southern Denmark and Odense University Hospital, 5000 Odense C, Denmark
| | - Thomas Levin Andersen
- Clinical Cell Biology, Department of Pathology, Odense University Hospital, 5000 Odense C, Denmark; Department of Clinical Research, University of Southern Denmark, 5000 Odense C, Denmark; Department of Molecular Medicine, University of Southern Denmark, 5000 Odense C, Denmark
| | - Anders Kristian Haakonsson
- Department of Molecular Endocrinology, KMEB, University of Southern Denmark and Odense University Hospital, 5000 Odense C, Denmark; OPEN, Odense Patient Data Explorative Network, Odense University Hospital, Odense, Denmark
| | - Alexander Rauch
- Department of Molecular Endocrinology, KMEB, University of Southern Denmark and Odense University Hospital, 5000 Odense C, Denmark
| | - Jonna Skov Madsen
- Institute of Regional Health Science, University of Southern Denmark, 5000 Odense C, Denmark; Department of Biochemistry and Immunology, Lillebaelt Hospital, 7100 Vejle, Denmark
| | - Charlotte Ejersted
- Department of Endocrinology, Odense University Hospital, 5000 Odense C, Denmark
| | - Kurt Højlund
- Steno Diabetes Center Odense, Odense University Hospital, 5000 Odense C, Denmark; Department of Clinical Research, University of Southern Denmark, 5000 Odense C, Denmark
| | - Moustapha Kassem
- Department of Molecular Endocrinology, KMEB, University of Southern Denmark and Odense University Hospital, 5000 Odense C, Denmark; Department of Cellular and Molecular Medicine, DanStem (Danish Stem Cell Center), Panum Institute, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
26
|
Vinci MC, Gambini E, Bassetti B, Genovese S, Pompilio G. When Good Guys Turn Bad: Bone Marrow's and Hematopoietic Stem Cells' Role in the Pathobiology of Diabetic Complications. Int J Mol Sci 2020; 21:ijms21113864. [PMID: 32485847 PMCID: PMC7312629 DOI: 10.3390/ijms21113864] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 05/26/2020] [Accepted: 05/27/2020] [Indexed: 12/15/2022] Open
Abstract
Diabetes strongly contributes to the development of cardiovascular disease, the leading cause of mortality and morbidity in these patients. It is widely accepted that hyperglycemia impairs hematopoietic stem/progenitor cell (HSPC) mobilization from the bone marrow (BM) by inducing stem cell niche dysfunction. Moreover, a recent study demonstrated that type 2 diabetic patients are characterized by significant depletion of circulating provascular progenitor cells and increased frequency of inflammatory cells. This unbalance, potentially responsible for the reduction of intrinsic vascular homeostatic capacity and for the establishment of a low-grade inflammatory status, suggests that bone BM-derived HSPCs are not only victims but also active perpetrators in diabetic complications. In this review, we will discuss the most recent literature on the molecular mechanisms underpinning hyperglycemia-mediated BM dysfunction and differentiation abnormality of HSPCs. Moreover, a section will be dedicated to the new glucose-lowering therapies that by specifically targeting the culprits may prevent or treat diabetic complications.
Collapse
Affiliation(s)
- Maria Cristina Vinci
- Unit of Vascular Biology and Regenerative Medicine, IRCCS Centro Cardiologico Monzino, I-20138- Milan, Italy; (E.G.); (B.B.); (G.P.)
- Correspondence: ; Tel.: +39-02-5800-2028
| | - Elisa Gambini
- Unit of Vascular Biology and Regenerative Medicine, IRCCS Centro Cardiologico Monzino, I-20138- Milan, Italy; (E.G.); (B.B.); (G.P.)
| | - Beatrice Bassetti
- Unit of Vascular Biology and Regenerative Medicine, IRCCS Centro Cardiologico Monzino, I-20138- Milan, Italy; (E.G.); (B.B.); (G.P.)
| | - Stefano Genovese
- Unit of Diabetes, Endocrine and Metabolic Diseases, IRCCS Centro Cardiologico Monzino, I-20138- Milan, Italy;
| | - Giulio Pompilio
- Unit of Vascular Biology and Regenerative Medicine, IRCCS Centro Cardiologico Monzino, I-20138- Milan, Italy; (E.G.); (B.B.); (G.P.)
| |
Collapse
|
27
|
Zhu S, Bennett S, Kuek V, Xiang C, Xu H, Rosen V, Xu J. Endothelial cells produce angiocrine factors to regulate bone and cartilage via versatile mechanisms. Am J Cancer Res 2020; 10:5957-5965. [PMID: 32483430 PMCID: PMC7255007 DOI: 10.7150/thno.45422] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 04/14/2020] [Indexed: 02/06/2023] Open
Abstract
Blood vessels are conduits distributed throughout the body, supporting tissue growth and homeostasis by the transport of cells, oxygen and nutrients. Endothelial cells (ECs) form the linings of the blood vessels, and together with pericytes, are essential for organ development and tissue homeostasis through producing paracrine signalling molecules, called angiocrine factors. In the skeletal system, ECs - derived angiocrine factors, combined with bone cells-released angiogenic factors, orchestrate intercellular crosstalk of the bone microenvironment, and the coupling of angiogenesis-to-osteogenesis. Whilst the involvement of angiogenic factors and the blood vessels of the skeleton is relatively well established, the impact of ECs -derived angiocrine factors on bone and cartilage homeostasis is gradually emerging. In this review, we survey ECs - derived angiocrine factors, which are released by endothelial cells of the local microenvironment and by distal organs, and act specifically as regulators of skeletal growth and homeostasis. These may potentially include angiocrine factors with osteogenic property, such as Hedgehog, Notch, WNT, bone morphogenetic protein (BMP), fibroblast growth factor (FGF), insulin-like growth factor (IGF), and platelet-derived growth factor (PDGF). Understanding the versatile mechanisms by which ECs-derived angiocrine factors orchestrate bone and cartilage homeostasis, and pathogenesis, is an important step towards the development of therapeutic potential for skeletal diseases.
Collapse
|
28
|
Hoyer FF, Zhang X, Coppin E, Vasamsetti SB, Modugu G, Schloss MJ, Rohde D, McAlpine CS, Iwamoto Y, Libby P, Naxerova K, Swirski FK, Dutta P, Nahrendorf M. Bone Marrow Endothelial Cells Regulate Myelopoiesis in Diabetes Mellitus. Circulation 2020; 142:244-258. [PMID: 32316750 DOI: 10.1161/circulationaha.120.046038] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND Diabetes mellitus is a prevalent public health problem that affects about one-third of the US population and leads to serious vascular complications with increased risk for coronary artery disease. How bone marrow hematopoiesis contributes to diabetes mellitus complications is incompletely understood. We investigated the role of bone marrow endothelial cells in diabetic regulation of inflammatory myeloid cell production. METHODS In 3 types of mouse diabetes mellitus, including streptozotocin, high-fat diet, and genetic induction using leptin-receptor-deficient db/db mice, we assayed leukocytes, hematopoietic stem and progenitor cells (HSPC). In addition, we investigated bone marrow endothelial cells with flow cytometry and expression profiling. RESULTS In diabetes mellitus, we observed enhanced proliferation of HSPC leading to augmented circulating myeloid cell numbers. Analysis of bone marrow niche cells revealed that endothelial cells in diabetic mice expressed less Cxcl12, a retention factor promoting HSPC quiescence. Transcriptome-wide analysis of bone marrow endothelial cells demonstrated enrichment of genes involved in epithelial growth factor receptor (Egfr) signaling in mice with diet-induced diabetes mellitus. To explore whether endothelial Egfr plays a functional role in myelopoiesis, we generated mice with endothelial-specific deletion of Egfr (Cdh5Cre Egfrfl/fl). We found enhanced HSPC proliferation and increased myeloid cell production in Cdh5Cre Egfrfl/fl mice compared with wild-type mice with diabetes mellitus. Disrupted Egfr signaling in endothelial cells decreased their expression of the HSPC retention factor angiopoietin-1. We tested the functional relevance of these findings for wound healing and atherosclerosis, both implicated in complications of diabetes mellitus. Inflammatory myeloid cells accumulated more in skin wounds of diabetic Cdh5Cre Egfrfl/fl mice, significantly delaying wound closure. Atherosclerosis was accelerated in Cdh5Cre Egfrfl/fl mice, leading to larger and more inflamed atherosclerotic lesions in the aorta. CONCLUSIONS In diabetes mellitus, bone marrow endothelial cells participate in the dysregulation of bone marrow hematopoiesis. Diabetes mellitus reduces endothelial production of Cxcl12, a quiescence-promoting niche factor that reduces stem cell proliferation. We describe a previously unknown counterregulatory pathway, in which protective endothelial Egfr signaling curbs HSPC proliferation and myeloid cell production.
Collapse
Affiliation(s)
- Friedrich Felix Hoyer
- Center for Systems Biology and Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, Boston (F.F.H., M.J.S., D.R., C.S.A., Y.I., K.N., F.K.S., M.N.)
| | - Xinyi Zhang
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, PA (X.Z., E.C., S.B.V., G.M., P.D.).,The Third Xiangya Hospital, Central South University, Changsha, Hunan, China (X.Z.)
| | - Emilie Coppin
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, PA (X.Z., E.C., S.B.V., G.M., P.D.)
| | - Sathish Babu Vasamsetti
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, PA (X.Z., E.C., S.B.V., G.M., P.D.)
| | - Ganesh Modugu
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, PA (X.Z., E.C., S.B.V., G.M., P.D.)
| | - Maximilian J Schloss
- Center for Systems Biology and Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, Boston (F.F.H., M.J.S., D.R., C.S.A., Y.I., K.N., F.K.S., M.N.)
| | - David Rohde
- Center for Systems Biology and Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, Boston (F.F.H., M.J.S., D.R., C.S.A., Y.I., K.N., F.K.S., M.N.)
| | - Cameron S McAlpine
- Center for Systems Biology and Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, Boston (F.F.H., M.J.S., D.R., C.S.A., Y.I., K.N., F.K.S., M.N.)
| | - Yoshiko Iwamoto
- Center for Systems Biology and Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, Boston (F.F.H., M.J.S., D.R., C.S.A., Y.I., K.N., F.K.S., M.N.)
| | - Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (P.L.)
| | - Kamila Naxerova
- Center for Systems Biology and Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, Boston (F.F.H., M.J.S., D.R., C.S.A., Y.I., K.N., F.K.S., M.N.)
| | - Filip K Swirski
- Center for Systems Biology and Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, Boston (F.F.H., M.J.S., D.R., C.S.A., Y.I., K.N., F.K.S., M.N.)
| | - Partha Dutta
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, PA (X.Z., E.C., S.B.V., G.M., P.D.)
| | - Matthias Nahrendorf
- Center for Systems Biology and Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, Boston (F.F.H., M.J.S., D.R., C.S.A., Y.I., K.N., F.K.S., M.N.).,Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston (M.N.).,Department of Internal Medicine I, University Hospital Würzburg, Germany (M.N.)
| |
Collapse
|
29
|
Santopaolo M, Sambataro M, Spinetti G, Madeddu P. Bone marrow as a target and accomplice of vascular complications in diabetes. Diabetes Metab Res Rev 2020; 36 Suppl 1:e3240. [PMID: 31840418 DOI: 10.1002/dmrr.3240] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 11/19/2019] [Indexed: 12/16/2022]
Abstract
Peripheral vascular complications are common in diabetic patients. While pathogenic mechanisms have received much consideration, only recently regenerative processes captured attention. There is now a consensus that the bone marrow is a source of reparative cells and that this healing mechanism is lost in people with diabetes, especially in those suffering from ischemic complications. This failure was thought to occur due to a negative impact of diabetes on the mobilization of stem/progenitor cells with angiogenic properties from the bone marrow to the circulation. Moreover, those patients showing severely reduced bone marrow cell mobilization also bared a very high risk for adverse cardiovascular events. More recently, the structural integrity of the bone marrow was recognized to be altered because of the rarefaction of local microvasculature and innervation, thus mirroring anatomical features that typically occur in peripheral tissues. Ensuing hypoxia, nutrient starvation, and creation of an acidic and oxidative environment concur in causing the depletion of stem/progenitor cells in the endosteal niche and in forcing stromal cells to activate an adipogenesis program. Moreover, stem/progenitor cells acquire a pathogenic phenotype and, once mobilized, can pass harmful signalling molecules to vascular cells of peripheral tissues thereby contributing to ischemic complications. These new pieces of evidence indicate that the bone marrow should deserve more attention in the current care of critical limb ischemia and diabetic foot. Owing to powerful reserve capacities, the bone marrow integrity could be preserved and even rescued using rehabilitation programs and pharmacological treatments with consequent benefit for local and whole-organism homeostasis.
Collapse
Affiliation(s)
- Marianna Santopaolo
- Bristol Heart Institute, Translational Health Sciences, University of Bristol, Bristol Royal Infirmary, Bristol, UK
| | - Maria Sambataro
- Endocrine, Metabolism, and Nutrition Disease Unit, Santa Maria di Ca' Foncello Hospital, Treviso, Italy
| | - Gaia Spinetti
- Laboratory of Cardiovascular Research, IRCCS MultiMedica, Milan, Italy
| | - Paolo Madeddu
- Bristol Heart Institute, Translational Health Sciences, University of Bristol, Bristol Royal Infirmary, Bristol, UK
| |
Collapse
|
30
|
Madeddu P, Avolio E, Alvino VV, Santopaolo M, Spinetti G. Personalized Cardiovascular Regenerative Medicine: Targeting the Extreme Stages of Life. Front Cardiovasc Med 2019; 6:177. [PMID: 31828079 PMCID: PMC6890607 DOI: 10.3389/fcvm.2019.00177] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 11/14/2019] [Indexed: 12/28/2022] Open
Abstract
Cardiovascular regenerative medicine is an exciting new approach that promises to change the current care of million people world-wide. Major emphasis was given to the quality and quantities of regenerative products, but recent evidence points to the importance of a better specification of the target population that may take advantage of these advanced medical treatments. Patient stratification is an important step in drug development. Tailoring treatment to the patient's specificity allowed significant improvement in cancer therapy, but personalized regenerative medicine is still at the initial stage in the cardiovascular field. For example, new-borns with a congenital heart condition and elderly people require dedicated therapeutic approaches, which adapt to their lifetime needs. In these people, personalized treatments may overcome the benefits delivered by standard protocols. In this review, we provide insights into these extreme stages of life as potential targets for cardiovascular reconstitution.
Collapse
Affiliation(s)
- Paolo Madeddu
- Translational Health Sciences, Bristol Heart Institute, Bristol Royal Infirmary, University of Bristol, Bristol, United Kingdom
| | - Elisa Avolio
- Translational Health Sciences, Bristol Heart Institute, Bristol Royal Infirmary, University of Bristol, Bristol, United Kingdom
| | - Valeria Vincenza Alvino
- Translational Health Sciences, Bristol Heart Institute, Bristol Royal Infirmary, University of Bristol, Bristol, United Kingdom
| | - Marianna Santopaolo
- Translational Health Sciences, Bristol Heart Institute, Bristol Royal Infirmary, University of Bristol, Bristol, United Kingdom
| | - Gaia Spinetti
- Laboratory of Cardiovascular Research, IRCCS MultiMedica, Milan, Italy
| |
Collapse
|
31
|
Andrzejewska A, Catar R, Schoon J, Qazi TH, Sass FA, Jacobi D, Blankenstein A, Reinke S, Krüger D, Streitz M, Schlickeiser S, Richter S, Souidi N, Beez C, Kamhieh-Milz J, Krüger U, Zemojtel T, Jürchott K, Strunk D, Reinke P, Duda G, Moll G, Geissler S. Multi-Parameter Analysis of Biobanked Human Bone Marrow Stromal Cells Shows Little Influence for Donor Age and Mild Comorbidities on Phenotypic and Functional Properties. Front Immunol 2019; 10:2474. [PMID: 31781089 PMCID: PMC6857652 DOI: 10.3389/fimmu.2019.02474] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 10/03/2019] [Indexed: 12/28/2022] Open
Abstract
Heterogeneous populations of human bone marrow-derived stromal cells (BMSC) are among the most frequently tested cellular therapeutics for treating degenerative and immune disorders, which occur predominantly in the aging population. Currently, it is unclear whether advanced donor age and commonly associated comorbidities affect the properties of ex vivo-expanded BMSCs. Thus, we stratified cells from adult and elderly donors from our biobank (n = 10 and n = 13, mean age 38 and 72 years, respectively) and compared their phenotypic and functional performance, using multiple assays typically employed as minimal criteria for defining multipotent mesenchymal stromal cells (MSCs). We found that BMSCs from both cohorts meet the standard criteria for MSC, exhibiting similar morphology, growth kinetics, gene expression profiles, and pro-angiogenic and immunosuppressive potential and the capacity to differentiate toward adipogenic, chondrogenic, and osteogenic lineages. We found no substantial differences between cells from the adult and elderly cohorts. As positive controls, we studied the impact of in vitro aging and inflammatory cytokine stimulation. Both conditions clearly affected the cellular properties, independent of donor age. We conclude that in vitro aging rather than in vivo donor aging influences BMSC characteristics.
Collapse
Affiliation(s)
- Anastazja Andrzejewska
- BIH Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health (BIH), Berlin, Germany.,Berlin-Brandenburg School for Regenerative Therapies, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, BIH, Berlin, Germany.,Julius Wolff Institute, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, BIH, Berlin, Germany
| | - Rusan Catar
- BIH Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health (BIH), Berlin, Germany.,Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, BIH, Berlin, Germany
| | - Janosch Schoon
- BIH Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health (BIH), Berlin, Germany.,Berlin-Brandenburg School for Regenerative Therapies, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, BIH, Berlin, Germany.,Julius Wolff Institute, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, BIH, Berlin, Germany
| | - Taimoor Hasan Qazi
- BIH Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health (BIH), Berlin, Germany.,Berlin-Brandenburg School for Regenerative Therapies, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, BIH, Berlin, Germany.,Julius Wolff Institute, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, BIH, Berlin, Germany
| | - Frauke Andrea Sass
- BIH Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health (BIH), Berlin, Germany.,Berlin-Brandenburg School for Regenerative Therapies, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, BIH, Berlin, Germany.,Julius Wolff Institute, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, BIH, Berlin, Germany
| | - Dorit Jacobi
- BIH Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health (BIH), Berlin, Germany.,Julius Wolff Institute, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, BIH, Berlin, Germany
| | - Antje Blankenstein
- BIH Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health (BIH), Berlin, Germany.,Julius Wolff Institute, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, BIH, Berlin, Germany
| | - Simon Reinke
- BIH Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health (BIH), Berlin, Germany.,Julius Wolff Institute, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, BIH, Berlin, Germany
| | - David Krüger
- Julius Wolff Institute, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, BIH, Berlin, Germany
| | - Mathias Streitz
- BIH Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health (BIH), Berlin, Germany.,Institute of Medical Immunology, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, BIH, Berlin, Germany
| | - Stephan Schlickeiser
- BIH Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health (BIH), Berlin, Germany.,Institute of Medical Immunology, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, BIH, Berlin, Germany
| | - Sarina Richter
- BIH Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health (BIH), Berlin, Germany.,Institute of Medical Immunology, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, BIH, Berlin, Germany
| | - Naima Souidi
- BIH Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health (BIH), Berlin, Germany.,Berlin-Brandenburg School for Regenerative Therapies, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, BIH, Berlin, Germany.,Institute of Medical Immunology, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, BIH, Berlin, Germany
| | - Christien Beez
- BIH Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health (BIH), Berlin, Germany.,Berlin-Brandenburg School for Regenerative Therapies, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, BIH, Berlin, Germany.,Institute of Medical Immunology, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, BIH, Berlin, Germany
| | - Julian Kamhieh-Milz
- Department of Transfusion Medicine, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, BIH, Berlin, Germany
| | - Ulrike Krüger
- BIH Core Unit Genomics Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Tomasz Zemojtel
- BIH Core Unit Genomics Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Karsten Jürchott
- BIH Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health (BIH), Berlin, Germany
| | - Dirk Strunk
- Berlin Center for Advanced Therapies, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, BIH, Berlin, Germany
| | - Petra Reinke
- BIH Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health (BIH), Berlin, Germany.,Institute of Medical Immunology, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, BIH, Berlin, Germany.,Spinal Cord Injury and Tissue Regeneration Center, Experimental and Clinical Cell Therapy Institute, Paracelsus Medical University, Salzburg, Austria
| | - Georg Duda
- BIH Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health (BIH), Berlin, Germany.,Berlin-Brandenburg School for Regenerative Therapies, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, BIH, Berlin, Germany.,Julius Wolff Institute, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, BIH, Berlin, Germany
| | - Guido Moll
- BIH Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health (BIH), Berlin, Germany.,Berlin-Brandenburg School for Regenerative Therapies, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, BIH, Berlin, Germany.,Julius Wolff Institute, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, BIH, Berlin, Germany.,Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, BIH, Berlin, Germany
| | - Sven Geissler
- BIH Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health (BIH), Berlin, Germany.,Berlin-Brandenburg School for Regenerative Therapies, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, BIH, Berlin, Germany.,Julius Wolff Institute, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, BIH, Berlin, Germany
| |
Collapse
|
32
|
Mangialardi G, Ferland-McCollough D, Maselli D, Santopaolo M, Cordaro A, Spinetti G, Sambataro M, Sullivan N, Blom A, Madeddu P. Bone marrow pericyte dysfunction in individuals with type 2 diabetes. Diabetologia 2019; 62:1275-1290. [PMID: 31001672 PMCID: PMC6560025 DOI: 10.1007/s00125-019-4865-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 03/04/2019] [Indexed: 12/17/2022]
Abstract
AIMS/HYPOTHESIS Previous studies have shown that diabetes mellitus destabilises the integrity of the microvasculature in different organs by damaging the interaction between pericytes and endothelial cells. In bone marrow, pericytes exert trophic functions on endothelial cells and haematopoietic cells through paracrine mechanisms. However, whether bone marrow pericytes are a target of diabetes-induced damage remains unknown. Here, we investigated whether type 2 diabetes can affect the abundance and function of bone marrow pericytes. METHODS We conducted an observational clinical study comparing the abundance and molecular/functional characteristics of CD146+ pericytes isolated from the bone marrow of 25 individuals without diabetes and 14 individuals with uncomplicated type 2 diabetes, referring to our Musculoskeletal Research Unit for hip reconstructive surgery. RESULTS Immunohistochemistry revealed that diabetes causes capillary rarefaction and compression of arteriole size in bone marrow, without changing CD146+ pericyte counts. These data were confirmed by flow cytometry on freshly isolated bone marrow cells. We then performed an extensive functional and molecular characterisation of immunosorted CD146+ pericytes. Type 2 diabetes caused a reduction in pericyte proliferation, viability, migration and capacity to support in vitro angiogenesis, while inducing apoptosis. AKT is a key regulator of the above functions and its phosphorylation state is reportedly reduced in the bone marrow endothelium of individuals with diabetes. Surprisingly, we could not find a difference in AKT phosphorylation (at either Ser473 or Thr308) in bone marrow pericytes from individuals with and without diabetes. Nonetheless, the angiocrine signalling reportedly associated with AKT was found to be significantly downregulated, with lower levels of fibroblast growth factor-2 (FGF2) and C-X-C motif chemokine ligand 12 (CXCL12), and activation of the angiogenesis inhibitor angiopoietin 2 (ANGPT2). Transfection with the adenoviral vector carrying the coding sequence for constitutively active myristoylated AKT rescued functional defects and angiocrine signalling in bone marrow pericytes from diabetic individuals. Furthermore, an ANGPT2 blocking antibody restored the capacity of pericytes to promote endothelial networking. CONCLUSIONS/INTERPRETATION This is the first demonstration of pericyte dysfunction in bone marrow of people with type 2 diabetes. An altered angiocrine signalling from pericytes may participate in bone marrow microvascular remodelling in individuals with diabetes.
Collapse
Affiliation(s)
- Giuseppe Mangialardi
- Bristol Heart Institute, University of Bristol, Bristol Royal Infirmary, Level 7, Upper Maudlin Street, Bristol, BS2 8HW, UK
| | - David Ferland-McCollough
- Bristol Heart Institute, University of Bristol, Bristol Royal Infirmary, Level 7, Upper Maudlin Street, Bristol, BS2 8HW, UK
| | - Davide Maselli
- Bristol Heart Institute, University of Bristol, Bristol Royal Infirmary, Level 7, Upper Maudlin Street, Bristol, BS2 8HW, UK
- IRCCS Multimedica, Milan, Italy
- Department of Biochemistry, University of Sassari, Sassari, Italy
| | - Marianna Santopaolo
- Bristol Heart Institute, University of Bristol, Bristol Royal Infirmary, Level 7, Upper Maudlin Street, Bristol, BS2 8HW, UK
| | - Andrea Cordaro
- Bristol Heart Institute, University of Bristol, Bristol Royal Infirmary, Level 7, Upper Maudlin Street, Bristol, BS2 8HW, UK
| | | | - Maria Sambataro
- Department of Specialized Medicines, Endocrine, Metabolic and Nutrition Diseases Unit, Santa Maria of Ca' Foncello Hospital, Treviso, Italy
| | - Niall Sullivan
- Muscloskeletal Research Unit, School of Clinical Sciences, University of Bristol, Bristol, UK
| | - Ashley Blom
- Muscloskeletal Research Unit, School of Clinical Sciences, University of Bristol, Bristol, UK
| | - Paolo Madeddu
- Bristol Heart Institute, University of Bristol, Bristol Royal Infirmary, Level 7, Upper Maudlin Street, Bristol, BS2 8HW, UK.
| |
Collapse
|
33
|
Manning P, Munasinghe PE, Bellae Papannarao J, Gray AR, Sutherland W, Katare R. Acute Weight Loss Restores Dysregulated Circulating MicroRNAs in Individuals Who Are Obese. J Clin Endocrinol Metab 2019; 104:1239-1248. [PMID: 30383229 DOI: 10.1210/jc.2018-00684] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 10/26/2018] [Indexed: 12/17/2022]
Abstract
CONTEXT Obesity is a global epidemic and an independent risk factor for several diseases. miRNAs are gaining interest as early molecular regulators of various pathological processes. OBJECTIVE To examine the miRNA signatures in women who are obese and determine the response of miRNAs to acute weight loss. METHODS Plasma samples were collected from women who are obese (n = 80) before and after acute weight loss (mean, 7.2%). Plasma samples from age-matched lean volunteers (n = 80) were used as controls. Total RNA was extracted from the plasma samples and subjected to NanoString analysis of 822 miRNAs. The expression level of candidate miRNAs was validated in all participants using quantitative real-time PCR analysis. RESULTS NanoString analysis identified substantial dysregulation of 21 miRNAs in women who are obese that were associated with impaired glucose tolerance, senescence, cardiac hypertrophy, angiogenesis, inflammation, and cell death. Acute weight loss reversed the expression pattern of 18 of these miRNAs toward those seen in the lean control group. Furthermore, real-time PCR validation of all the samples for 13 miRNAs with at least twofold upregulation or downregulation confirmed substantial dysregulation of all the chosen miRNAs in women who are obese at baseline. After acute weight loss, the levels of seven miRNAs in women who are obese and who are lean were comparable, with no statistically significant evidence for differences between the two groups. CONCLUSIONS Our study has provided evidence that the circulating miRNAs associated with various disorders are dysregulated in women who are obese. We also found that seven of these miRNAs showed levels comparable to those in lean controls after acute weight loss in women who are obese.
Collapse
Affiliation(s)
- Patrick Manning
- Department of Medicine, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Pujika Emani Munasinghe
- Department of Physiology, HeartOtago, School of Biomedical Science, University of Otago, Dunedin, New Zealand
| | - Jayanthi Bellae Papannarao
- Department of Physiology, HeartOtago, School of Biomedical Science, University of Otago, Dunedin, New Zealand
| | - Andrew R Gray
- Biostatistics Unit, Dunedin School of Medicine, Health Sciences, University of Otago, Dunedin, New Zealand
| | - Wayne Sutherland
- Department of Medicine, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Rajesh Katare
- Department of Physiology, HeartOtago, School of Biomedical Science, University of Otago, Dunedin, New Zealand
| |
Collapse
|
34
|
Brakenhielm E, Richard V. Therapeutic vascular growth in the heart. VASCULAR BIOLOGY 2019; 1:H9-H15. [PMID: 32923948 PMCID: PMC7439849 DOI: 10.1530/vb-19-0006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 03/28/2019] [Indexed: 12/03/2022]
Abstract
Despite tremendous efforts in preclinical research over the last decades, the clinical translation of therapeutic angiogenesis to grow stable and functional blood vessels in patients with ischemic diseases continues to prove challenging. In this mini review, we briefly present the current main approaches applied to improve pro-angiogenic therapies. Specific examples from research on therapeutic cardiac angiogenesis and arteriogenesis will be discussed, and finally some suggestions for future therapeutic developments will be presented.
Collapse
Affiliation(s)
- Ebba Brakenhielm
- Normandy University, UniRouen, Inserm (Institut National de la Santé et de la Recherche Médicale) UMR1096 (EnVI Laboratory), FHU REMOD-VHF, Rouen, France
| | - Vincent Richard
- Normandy University, UniRouen, Inserm (Institut National de la Santé et de la Recherche Médicale) UMR1096 (EnVI Laboratory), FHU REMOD-VHF, Rouen, France
| |
Collapse
|
35
|
Picke AK, Campbell G, Napoli N, Hofbauer LC, Rauner M. Update on the impact of type 2 diabetes mellitus on bone metabolism and material properties. Endocr Connect 2019; 8:R55-R70. [PMID: 30772871 PMCID: PMC6391903 DOI: 10.1530/ec-18-0456] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 01/24/2019] [Indexed: 11/23/2022]
Abstract
The prevalence of type 2 diabetes mellitus (T2DM) is increasing worldwide, especially as a result of our aging society, high caloric intake and sedentary lifestyle. Besides the well-known complications of T2DM on the cardiovascular system, the eyes, kidneys and nerves, bone strength is also impaired in diabetic patients. Patients with T2DM have a 40-70% increased risk for fractures, despite having a normal to increased bone mineral density, suggesting that other factors besides bone quantity must account for increased bone fragility. This review summarizes the current knowledge on the complex effects of T2DM on bone including effects on bone cells, bone material properties and other endocrine systems that subsequently affect bone, discusses the effects of T2DM medications on bone and concludes with a model identifying factors that may contribute to poor bone quality and increased bone fragility in T2DM.
Collapse
Affiliation(s)
- Ann-Kristin Picke
- Institute of Comparative Molecular Endocrinology, Ulm University, Ulm, Germany
| | - Graeme Campbell
- Institute of Biomechanics, TUHH Hamburg University of Technology, Hamburg, Germany
| | - Nicola Napoli
- Diabetes and Bone Network, Department Endocrinology and Diabetes, University Campus Bio-Medico of Rome, Rome, Italy
- Division of Bone and Mineral Diseases, Washington University in St Louis, St Louis, Missouri, USA
| | - Lorenz C Hofbauer
- Department of Medicine III & Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Martina Rauner
- Department of Medicine III & Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
- Correspondence should be addressed to M Rauner:
| |
Collapse
|
36
|
Zhou J, Zhang Z, Qian G. Neuropathy and inflammation in diabetic bone marrow. Diabetes Metab Res Rev 2019; 35:e3083. [PMID: 30289199 DOI: 10.1002/dmrr.3083] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Revised: 09/05/2018] [Accepted: 10/02/2018] [Indexed: 12/14/2022]
Abstract
Diabetes impairs the bone marrow (BM) architecture and function as well as the mobilization of immature cells into the bloodstream and number of potential regenerative cells. Circadian regulation of bone immature cell migration is regulated by β-adrenergic receptors, which are expressed on haematopoietic stem cells, mesenchymal stem cells, and osteoblasts in the BM. Diabetes is associated with a substantially lower number of sympathetic nerve terminal endings in the BM; thus, diabetic neuropathy plays a critical role in BM dysfunction. Treatment with mesenchymal stem cells, BM mononuclear cells, haematopoietic stem cells, and stromal cells ameliorates the dysfunction of diabetic neuropathy, which occurs, in part, through secreted neurotrophic factors, growth factors, adipokines, and polarizing macrophage M2 cells and inhibiting inflammation. Inflammation may be a therapeutic target for BM stem cells to improve diabetic neuropathy. Given that angiogenic and neurotrophic effects are two major barriers to effective diabetic neuropathy therapy, targeting BM stem cells may provide a novel approach to develop these types of treatments.
Collapse
Affiliation(s)
- Jiyin Zhou
- National Drug Clinical Trial Institution, The Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Zuo Zhang
- National Drug Clinical Trial Institution, The Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Guisheng Qian
- Institute of Respiratory Diseases, The Second Affiliated Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
37
|
Endothelial Progenitor Cells Biology in Diabetes Mellitus and Peripheral Arterial Disease and their Therapeutic Potential. Stem Cell Rev Rep 2018; 15:157-165. [DOI: 10.1007/s12015-018-9863-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
38
|
S AlSalhi M, Devanesan S, E AlZahrani K, AlShebly M, Al-Qahtani F, Farhat K, Masilamani V. Impact of Diabetes Mellitus on Human Erythrocytes: Atomic Force Microscopy and Spectral Investigations. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2018; 15:ijerph15112368. [PMID: 30373127 PMCID: PMC6266196 DOI: 10.3390/ijerph15112368] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 10/21/2018] [Accepted: 10/24/2018] [Indexed: 12/24/2022]
Abstract
Diabetes mellitus (DM) is a common metabolic disease indicated by high sugar levels in the blood over a prolonged period. When left untreated, it can lead to long-term complications, such as cardiovascular disease, stroke, and diabetic retinopathy or foot ulcers. Approximately 415 million people (about 8.3% of the world’s population) had diabetes worldwide in 2015, with 90% of the cases classified as Type 2 DM, which is caused by insulin resistance that arises mostly from being overweight and from a lack of exercise. DM affects every part of the body, including the erythrocytes. The aim of the present report is to gain insight into the damage done to the erythrocytes of patients classified with pre-diabetes and diabetes (plenty are found in the Kingdom of Saudi Arabia, a country where young people encompass a large segment of the population). The study presents results on the morphological analysis of erythrocytes by atomic force microscopy (AFM) and molecular investigations by fluorescence spectroscopy (FS). Our results indicate significant differences (in the morphology, size, and hemolytic end products) between the erythrocytes of diabetic patients (HbA1C, glycated hemoglobin, levels of 8–10%) and normal controls. It is well-known that DM and smoking are two major contributory factors for cardiovascular diseases (CVDs), and our observations presented in this study suggest that diabetes plays a relatively less damaging role than smoking for CVD.
Collapse
Affiliation(s)
- Mohamad S AlSalhi
- Department of Physics and Astronomy, College of Science, King Saud University, Riyadh 11451, Saudi Arabia.
- Research Chair in Laser Diagnosis of Cancers, Department of Physics and Astronomy, College of Science, King Saud University, Riyadh 11451, Saudi Arabia.
| | - Sandhanasamy Devanesan
- Department of Physics and Astronomy, College of Science, King Saud University, Riyadh 11451, Saudi Arabia.
- Research Chair in Laser Diagnosis of Cancers, Department of Physics and Astronomy, College of Science, King Saud University, Riyadh 11451, Saudi Arabia.
| | - Khalid E AlZahrani
- Department of Physics and Astronomy, College of Science, King Saud University, Riyadh 11451, Saudi Arabia.
- King Abdullah Institute for Nanotechnology, King Saud University, Riyadh 1451, Saudi Arabia.
| | - Mashael AlShebly
- Department of Obstetrics and Gynecology, College of Medicine, King Khalid University Hospital, King Saud University, Riyadh 11451, Saudi Arabia.
| | - Fatima Al-Qahtani
- Hematology Unit, Department of Pathology, College of Medicine, King Saud University and King Saud University Medical City, Riyadh 11451, Saudi Arabia.
| | - Karim Farhat
- Cancer Research Chair, College of Medicine, King Saud University, Riyadh 11451, Saudi Arabia.
| | - Vadivel Masilamani
- Department of Physics and Astronomy, College of Science, King Saud University, Riyadh 11451, Saudi Arabia.
- Research Chair in Laser Diagnosis of Cancers, Department of Physics and Astronomy, College of Science, King Saud University, Riyadh 11451, Saudi Arabia.
| |
Collapse
|
39
|
Qi K, Li X, Geng Y, Cui H, Jin C, Wang P, Li Y, Yang Y. Tongxinluo attenuates reperfusion injury in diabetic hearts by angiopoietin-like 4-mediated protection of endothelial barrier integrity via PPAR-α pathway. PLoS One 2018; 13:e0198403. [PMID: 29912977 PMCID: PMC6005559 DOI: 10.1371/journal.pone.0198403] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Accepted: 05/20/2018] [Indexed: 12/02/2022] Open
Abstract
Objective Endothelial barrier function in the onset and Tongxinluo (TXL) protection of myocardial ischemia/reperfusion (I/R) injury, and TXL can induce the secretion of Angiopoietin-like 4 (Angptl4) in human cardiac microvascular endothelial cells during hypoxia/reoxygenation. We intend to demonstrate whether TXL can attenuate myocardial I/R injury in diabetes, characterized with microvascular endothelial barrier disruption, by induction of Angptl4-mediated protection of endothelial barrier integrity. Methods and results I/R injury was created by coronary ligation in ZDF diabetic and non-diabetic control rats. The animals were anesthetized and randomized to sham operation or I/R injury with or without the exposure to insulin, rhAngptl4, TXL, Angptl4 siRNA, and the PPAR-α inhibitor MK886. Tongxinluo, insulin and rhAngptl4 have the similar protective effect on diabetic hearts against I/R injury. In I/R-injured diabetic hearts, TXL treatment remarkably reduced the infarct size, and protected endothelial barrier integrity demonstrated by decreased endothelial cells apoptosis, microvascular permeability, and myocardial hemorrhage, fortified tight junction, and upregulated expression of JAM-A, integrin-α5, and VE-cadherin, and these effects of TXL were as effective as insulin and rhAngptl4. However, Angptl4 knock-down with siRNA interference and inhibition of PPAR-α with MK886 partially diminished these beneficial effects of TXL and rhAngptl4. TXL induced the expression of Angptl4 in I/R-injured diabetic hearts, and was canceled by Angptl4 siRNA and MK886. TXL treatment increased myocardial PPAR-α activity, and was abolished by MK886 but not by Angptl4 siRNA. Conclusions TXL protects diabetic hearts against I/R injury by activating Angptl4-mediated restoration of endothelial barrier integrity via the PPAR-α pathway.
Collapse
Affiliation(s)
- Kang Qi
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiangdong Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yongjian Geng
- Division of Cardiovascular Medicine, University of Texas Health Science Center at Houston, Houston, TX, United States of America
| | - Hehe Cui
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chen Jin
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Peihe Wang
- Peking Key Laboratory for Pre-clinical Evaluation of Cardiovascular Implant Material, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Animal Experimental Center, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yue Li
- Peking Key Laboratory for Pre-clinical Evaluation of Cardiovascular Implant Material, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Animal Experimental Center, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuejin Yang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- * E-mail:
| |
Collapse
|
40
|
Wang B, Yu J, Wang T, Shen Y, Lin D, Xu X, Wang Y. Identification of megakaryocytes as a target of advanced glycation end products in diabetic complications in bone marrow. Acta Diabetol 2018; 55:419-427. [PMID: 29417230 DOI: 10.1007/s00592-018-1109-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 01/17/2018] [Indexed: 10/18/2022]
Abstract
AIMS To define the possible effect of diabetic conditions on megakaryocytes, the long-know precursors of platelets and lately characterized modulator of hematopoietic stem quiescence-activation transition. METHODS Megakaryoblastic MEG-01 cell culture and TPO/SCF/IL-3-induced differentiation of human umbilical blood mononuclear cells toward megakaryocytes were used to test effects of glycated bovine serum albumin (BSA-AGEs). The ob/ob mice and streptozotocin-treated mice were used as models of hyperglycemia. MTT was used to measure cell proliferation, FACS for surface marker and cell cycle, and RT-qPCR for the expression of interested genes. Megakaryocytes at different stages in marrow smear were checked under microscope. RESULTS When added in MEG-01 cultures at 200 μg/ml, BSA-AGEs increased proliferation of cells and enhanced mRNA expression of RAGE, VEGFα and PF4 in the cells. None of cell cycle distribution, PMA-induced platelet-like particles production, expression of GATA1/NF-E2/PU-1/IL-6/OPG/PDGF in MEG-01 cells nor TPO/SCF/IL-3 induced umbilical cord blood cells differentiation into megakaryocyte was affected by BSA-AGEs. In the ob/ob diabetic mice, MKs percentages in marrow cells and platelets in peripheral blood were significantly increased compared with control mice. In streptozotocin-induced diabetic mice, however, MKs percentage in marrow cells was decreased though peripheral platelet counts were not altered. Gene expression assay showed that the change in MKs in these two diabetic conditions might be explained by the alteration of GATA1 and NF-E2 expression, respectively. CONCLUSIONS Diabetic condition in animals might exert its influence on hematopoiesis via megakaryocytes-the newly identified modulator of hematopoietic stem cells in bone marrow.
Collapse
Affiliation(s)
- Benfang Wang
- MOH Key Lab of Thrombosis and Hemostasis, Collaborative Innovation Center of Hematology-Thrombosis and Hemostasis Group, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Soochow University, 708 Renmin Road, Suzhou, 215007, China
| | - Jianjiang Yu
- Department of Clinical Laboratory, The Affiliated Jiangyin Hospital of Southeast University, Jiangyin, 214400, China
| | - Ting Wang
- MOH Key Lab of Thrombosis and Hemostasis, Collaborative Innovation Center of Hematology-Thrombosis and Hemostasis Group, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Soochow University, 708 Renmin Road, Suzhou, 215007, China
| | - Ying Shen
- MOH Key Lab of Thrombosis and Hemostasis, Collaborative Innovation Center of Hematology-Thrombosis and Hemostasis Group, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Soochow University, 708 Renmin Road, Suzhou, 215007, China
| | - Dandan Lin
- MOH Key Lab of Thrombosis and Hemostasis, Collaborative Innovation Center of Hematology-Thrombosis and Hemostasis Group, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Soochow University, 708 Renmin Road, Suzhou, 215007, China
| | - Xin Xu
- Department of Hematology, The Affiliated Jiangyin Hospital of Southeast University, Jiangyin, 214400, China
| | - Yiqiang Wang
- MOH Key Lab of Thrombosis and Hemostasis, Collaborative Innovation Center of Hematology-Thrombosis and Hemostasis Group, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Soochow University, 708 Renmin Road, Suzhou, 215007, China.
| |
Collapse
|
41
|
Accelerated cellular senescence as underlying mechanism for functionally impaired bone marrow-derived progenitor cells in ischemic heart disease. Atherosclerosis 2017; 260:138-146. [DOI: 10.1016/j.atherosclerosis.2017.03.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 02/20/2017] [Accepted: 03/17/2017] [Indexed: 02/05/2023]
|
42
|
Nollet E, Hoymans VY, Rodrigus IR, De Bock D, Dom M, Vanassche B, Van Hoof VOM, Cools N, Van Ackeren K, Wouters K, Vermeulen K, Vrints CJ, Van Craenenbroeck EM. Bone Marrow-Derived Progenitor Cells Are Functionally Impaired in Ischemic Heart Disease. J Cardiovasc Transl Res 2016; 9:266-78. [PMID: 27456951 PMCID: PMC5031720 DOI: 10.1007/s12265-016-9707-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 07/07/2016] [Indexed: 12/22/2022]
Abstract
To determine whether the presence of ischemic heart disease (IHD) per se, or rather the co-presence of heart failure (HF), is the primum movens for less effective stem cell products in autologous stem cell therapy, we assessed numbers and function of bone marrow (BM)-derived progenitor cells in patients with coronary artery disease (n = 17), HF due to ischemic cardiomyopathy (n = 8), non-ischemic HF (n = 7), and control subjects (n = 11). Myeloid and erythroid differentiation capacity of BM-derived mononuclear cells was impaired in patients with underlying IHD but not with non-ischemic HF. Migration capacity decreased with increasing IHD severity. Hence, IHD, with or without associated cardiomyopathy, is an important determinant of progenitor cell function. No depletion of hematopoietic and endothelial progenitor cells (EPC) within the BM was observed, while circulating EPC numbers were increased in the presence of IHD, suggesting active recruitment. The observed myelosuppression was not driven by inflammation and thus other mechanisms are at play.
Collapse
Affiliation(s)
- Evelien Nollet
- Laboratory of Cellular and Molecular Cardiology, Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium.
- Cardiovascular Diseases, Department of Translational Pathophysiological Research, University of Antwerp, Antwerp, Belgium.
| | - Vicky Y Hoymans
- Laboratory of Cellular and Molecular Cardiology, Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium
- Cardiovascular Diseases, Department of Translational Pathophysiological Research, University of Antwerp, Antwerp, Belgium
| | - Inez R Rodrigus
- Department of Cardiac Surgery, Antwerp University Hospital, Antwerp, Belgium
| | - Dina De Bock
- Department of Cardiac Surgery, Antwerp University Hospital, Antwerp, Belgium
| | - Marc Dom
- Department of Oral and Maxillofacial Surgery, General Hospital Sint-Maarten, Duffel, Belgium
| | - Bruno Vanassche
- Department of Oral and Maxillofacial Surgery, General Hospital Monica, Antwerp, Belgium
| | - Viviane O M Van Hoof
- Department of Clinical Chemistry, Antwerp University Hospital, Antwerp, Belgium
- Biochemistry, Department of Translational Pathophysiological Research, University of Antwerp, Antwerp, Belgium
| | - Nathalie Cools
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Katrijn Van Ackeren
- Laboratory of Cellular and Molecular Cardiology, Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium
- Cardiovascular Diseases, Department of Translational Pathophysiological Research, University of Antwerp, Antwerp, Belgium
| | - Kristien Wouters
- Department of Scientific Coordination and Biostatistics, Antwerp University Hospital, Antwerp, Belgium
| | - Katrien Vermeulen
- Laboratory of Hematology, Antwerp University Hospital, Antwerp, Belgium
| | - Christiaan J Vrints
- Laboratory of Cellular and Molecular Cardiology, Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium
- Cardiovascular Diseases, Department of Translational Pathophysiological Research, University of Antwerp, Antwerp, Belgium
- Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium
| | - Emeline M Van Craenenbroeck
- Laboratory of Cellular and Molecular Cardiology, Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium
- Cardiovascular Diseases, Department of Translational Pathophysiological Research, University of Antwerp, Antwerp, Belgium
- Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium
| |
Collapse
|
43
|
Reni C, Mangialardi G, Meloni M, Madeddu P. Diabetes Stimulates Osteoclastogenesis by Acidosis-Induced Activation of Transient Receptor Potential Cation Channels. Sci Rep 2016; 6:30639. [PMID: 27468810 PMCID: PMC4965751 DOI: 10.1038/srep30639] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 07/07/2016] [Indexed: 01/10/2023] Open
Abstract
Patients with type 1 diabetes have lower bone mineral density and higher risk of fractures. The role of osteoblasts in diabetes-related osteoporosis is well acknowledged whereas the role of osteoclasts (OCLs) is still unclear. We hypothesize that OCLs participate in pathological bone remodeling. We conducted studies in animals (streptozotocin-induced type 1 diabetic mice) and cellular models to investigate canonical and non-canonical mechanisms underlying excessive OCL activation. Diabetic mice show an increased number of active OCLs. In vitro studies demonstrate the involvement of acidosis in OCL activation and the implication of transient receptor potential cation channel subfamily V member 1 (TRPV1). In vivo studies confirm the establishment of local acidosis in the diabetic bone marrow (BM) as well as the ineffectiveness of insulin in correcting the pH variation and osteoclast activation. Conversely, treatment with TRPV1 receptor antagonists re-establishes a physiological OCL availability. These data suggest that diabetes causes local acidosis in the BM that in turn increases osteoclast activation through the modulation of TRPV1. The use of clinically available TRPV1 antagonists may provide a new means to combat bone problems associated with diabetes.
Collapse
Affiliation(s)
- Carlotta Reni
- Division of Experimental Cardiovascular Medicine, Bristol Heart Institute, University of Bristol, UK
| | - Giuseppe Mangialardi
- Division of Experimental Cardiovascular Medicine, Bristol Heart Institute, University of Bristol, UK
| | - Marco Meloni
- Vascular Pathology and Regeneration, Bristol Heart Institute, University of Bristol, UK.,University/British Heart Foundation Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, UK
| | - Paolo Madeddu
- Division of Experimental Cardiovascular Medicine, Bristol Heart Institute, University of Bristol, UK
| |
Collapse
|
44
|
Fadini GP, Ciciliot S, Albiero M. Concise Review: Perspectives and Clinical Implications of Bone Marrow and Circulating Stem Cell Defects in Diabetes. Stem Cells 2016; 35:106-116. [PMID: 27401837 DOI: 10.1002/stem.2445] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 05/27/2016] [Accepted: 05/28/2016] [Indexed: 12/12/2022]
Abstract
Diabetes mellitus is a complex systemic disease characterized by severe morbidity and excess mortality. The burden of its multiorgan complications relies on an imbalance between hyperglycemic cell damage and defective endogenous reparative mechanisms. Inflammation and abnormalities in several hematopoietic components are typically found in diabetes. The discovery that diabetes reduces circulating stem/progenitor cells and impairs their function has opened an entire new field of study where diabetology comes into contact with hematology and regenerative medicine. It is being progressively recognized that such rare circulating cell populations mirror finely regulated processes involved in hematopoiesis, immunosurveillance, and peripheral tissue homeostasis. From a clinical perspective, pauperization of circulating stem cells predicts adverse outcomes and death. Furthermore, studies in murine models and humans have identified the bone marrow (BM) as a previously neglected site of diabetic end-organ damage, characterized by microangiopathy, neuropathy, fat deposition, and inflammation. As a result, diabetes impairs the mobilization of BM stem/progenitor cells, a defect known as mobilopathy or myelokathexis, with negative consequences for physiologic hematopoiesis, immune regulation, and tissue regeneration. A better understanding of the molecular and cellular processes that govern the BM stem cell niche, cell mobilization, and kinetics in peripheral tissues may uncover new therapeutic strategies for patients with diabetes. This concise review summarizes the current knowledge on the interplay between the BM, circulating stem cells, and diabetes, and sets the stages for future developments in the field. Stem Cells 2017;35:106-116.
Collapse
Affiliation(s)
- Gian Paolo Fadini
- Department of Medicine, University of Padova, and Venetian Institute of Molecular Medicine, Padova, 35128, Italy
| | - Stefano Ciciliot
- Department of Medicine, University of Padova, and Venetian Institute of Molecular Medicine, Padova, 35128, Italy
| | - Mattia Albiero
- Department of Medicine, University of Padova, and Venetian Institute of Molecular Medicine, Padova, 35128, Italy
| |
Collapse
|
45
|
Abstract
Diabetes is one of the main economic burdens in health care, which threatens to worsen dramatically if prevalence forecasts are correct. What makes diabetes harmful is the multi-organ distribution of its microvascular and macrovascular complications. Regenerative medicine with cellular therapy could be the dam against life-threatening or life-altering complications. Bone marrow-derived stem cells are putative candidates to achieve this goal. Unfortunately, the bone marrow itself is affected by diabetes, as it can develop a microangiopathy and neuropathy similar to other body tissues. Neuropathy leads to impaired stem cell mobilization from marrow, the so-called mobilopathy. Here, we review the role of bone marrow-derived stem cells in diabetes: how they are affected by compromised bone marrow integrity, how they contribute to other diabetic complications, and how they can be used as a treatment for these. Eventually, we suggest new tactics to optimize stem cell therapy.
Collapse
Affiliation(s)
- Giuseppe Mangialardi
- Bristol Heart Institute, University of Bristol, Level 7, Bristol Royal Infirmary, Upper Maudlin Street, Bristol, BS28HW UK
| | - Paolo Madeddu
- Bristol Heart Institute, University of Bristol, Level 7, Bristol Royal Infirmary, Upper Maudlin Street, Bristol, BS28HW UK
| |
Collapse
|
46
|
Ma T, Xue YX. MiRNA-200b Regulates RMP7-Induced Increases in Blood-Tumor Barrier Permeability by Targeting RhoA and ROCKII. Front Mol Neurosci 2016; 9:9. [PMID: 26903801 PMCID: PMC4742559 DOI: 10.3389/fnmol.2016.00009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 01/18/2016] [Indexed: 12/20/2022] Open
Abstract
The primary goals of this study were to investigate the potential roles of miR-200b in regulating RMP7-induced increases in blood-tumor barrier (BTB) permeability and some of the possible molecular mechanisms associated with this effect. Microarray analysis revealed 34 significantly deregulated miRNAs including miR-200b in the BTB as induced by RMP7 and 8 significantly up-regulated miRNAs in the BTB by RMP7. RMP7 induced tight junction (TJ) opening of the BTB, thereby increasing BTB permeability. Associated with this effect of RMP7 was a decrease in miR-200b expression within the human cerebral microvascular endothelial cells line hCMEC/D3 (ECs) of the BTB. Overexpression of miR-200b inhibited endothelial leakage and restored normal transendothelial electric resistance values. A simultaneous shift in occludin and claudin-5 distributions from insoluble to soluble fractions were observed to be significantly reduced. In addition, overexpression of miR-200b inhibited the relocation of occludin and claudin-5 from cellular borders into the cytoplasm as well as the production of stress fiber formation in GECs (ECs with U87 glioma cells co-culturing) of the BTB. MiR-200b silencing produced opposite results as that obtained from that of the miR-200b overexpression group. Overexpression of miR-200b was also associated with a down-regulation in RhoA and ROCKII expression, concomitant with a decrease in BTB permeability. Again, results which were opposite to that obtained with the miR-200b silencing group. We further found that miR-200b regulated BTB permeability by directly targeting RhoA and ROCKII. Collectively, these results suggest that miR-200b's contribution to the RMP7-induced increase in BTB permeability was associated with stress fiber formation and TJ disassembly as achieved by directly targeting RhoA and ROCKII.
Collapse
Affiliation(s)
- Teng Ma
- Department of Neurobiology, College of Basic Medicine, China Medical UniversityShenyang, China; Institute of Pathology and Pathophysiology, China Medical UniversityShenyang, China
| | - Yi-Xue Xue
- Department of Neurobiology, College of Basic Medicine, China Medical UniversityShenyang, China; Institute of Pathology and Pathophysiology, China Medical UniversityShenyang, China
| |
Collapse
|
47
|
Caruso M, Zhang X, Ma D, Yang Z, Qi Y, Yi Z. Novel Endogenous, Insulin-Stimulated Akt2 Protein Interaction Partners in L6 Myoblasts. PLoS One 2015; 10:e0140255. [PMID: 26465754 PMCID: PMC4605787 DOI: 10.1371/journal.pone.0140255] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 09/23/2015] [Indexed: 12/28/2022] Open
Abstract
Insulin resistance and Type 2 diabetes are marked by an aberrant response in the insulin signaling network. The phosphoinositide-dependent serine/threonine kinase, Akt2, plays a key role in insulin signaling and glucose uptake, most notably within skeletal muscle. Protein-protein interaction regulates the functional consequence of Akt2 and in turn, Akt2's role in glucose uptake. However, only few insulin-responsive Akt2 interaction partners have been identified in skeletal muscle cells. In the present work, rat L6 myoblasts, a widely used insulin sensitive skeletal muscle cell line, were used to examine endogenous, insulin-stimulated Akt2 protein interaction partners. Akt2 co-immunoprecipitation was coupled with 1D-SDS-PAGE and fractions were analyzed by HPLC-ESI-MS/MS to reveal Akt2 protein-protein interactions. The pull-down assay displayed specificity for the Akt2 isoform; Akt1 and Akt3 unique peptides were not detected. A total of 49 were detected with a significantly increased (47) or decreased (2) association with Akt2 following insulin administration (n = 4; p<0.05). Multiple pathways were identified for the novel Akt2 interaction partners, such as the EIF2 and ubiquitination pathways. These data suggest that multiple new endogenous proteins may associate with Akt2 under basal as well as insulin-stimulated conditions, providing further insight into the insulin signaling network. Data are available via ProteomeXchange with identifier PXD002557.
Collapse
Affiliation(s)
- Michael Caruso
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy/Health Sciences, Wayne State University, Detroit, MI, United States of America
| | - Xiangmin Zhang
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy/Health Sciences, Wayne State University, Detroit, MI, United States of America
| | - Danjun Ma
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy/Health Sciences, Wayne State University, Detroit, MI, United States of America
| | - Zhao Yang
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy/Health Sciences, Wayne State University, Detroit, MI, United States of America
| | - Yue Qi
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy/Health Sciences, Wayne State University, Detroit, MI, United States of America
| | - Zhengping Yi
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy/Health Sciences, Wayne State University, Detroit, MI, United States of America
| |
Collapse
|
48
|
Albiero M, Poncina N, Ciciliot S, Cappellari R, Menegazzo L, Ferraro F, Bolego C, Cignarella A, Avogaro A, Fadini GP. Bone Marrow Macrophages Contribute to Diabetic Stem Cell Mobilopathy by Producing Oncostatin M. Diabetes 2015; 64:2957-68. [PMID: 25804939 DOI: 10.2337/db14-1473] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2014] [Accepted: 03/16/2015] [Indexed: 11/13/2022]
Abstract
Diabetes affects bone marrow (BM) structure and impairs mobilization of stem cells (SCs) into peripheral blood (PB). This amplifies multiorgan complications because BMSCs promote vascular repair. Because diabetes skews macrophage phenotypes and BM macrophages (BMMΦ) prevent SC mobilization, we hypothesized that excess BMMΦ contribute to diabetic SC mobilopathy. We show that patients with diabetes have increased M1 macrophages, whereas diabetic mice have increased CD169(+) BMMΦ with SC-retaining activity. Depletion of BMMΦ restored SC mobilization in diabetic mice. We found that CD169 labels M1 macrophages and that conditioned medium (CM) from M1 macrophages, but not from M0 and M2 macrophages, induced chemokine (C-X-C motif) ligand 12 (CXCL12) expression by mesenchymal stem/stromal cells. In silico data mining and in vitro validation identified oncostatin M (OSM) as the soluble mediator contained in M1 CM that induces CXCL12 expression via a mitogen-activated protein kinase kinase-p38-signal transducer and activator of a transcription 3-dependent pathway. In diabetic mice, OSM neutralization prevented CXCL12 induction and improved granulocyte-colony stimulating factor and ischemia-induced mobilization, SC homing to ischemic muscles, and vascular recovery. In patients with diabetes, BM plasma OSM levels were higher and correlated with the BM-to-PB SC ratio. In conclusion, BMMΦ prevent SC mobilization by OSM secretion, and OSM antagonism is a strategy to restore BM function in diabetes, which can translate into protection mediated by BMSCs.
Collapse
Affiliation(s)
- Mattia Albiero
- Department of Medicine, University of Padova, Padova, Italy Venetian Institute of Molecular Medicine, Padova, Italy
| | - Nicol Poncina
- Department of Medicine, University of Padova, Padova, Italy Venetian Institute of Molecular Medicine, Padova, Italy
| | - Stefano Ciciliot
- Department of Medicine, University of Padova, Padova, Italy Venetian Institute of Molecular Medicine, Padova, Italy
| | | | - Lisa Menegazzo
- Department of Medicine, University of Padova, Padova, Italy Venetian Institute of Molecular Medicine, Padova, Italy
| | - Francesca Ferraro
- Pennsylvania Hospital, University of Pennsylvania Health System, Philadelphia, PA Fox Chase Cancer Center, Philadelphia, PA
| | - Chiara Bolego
- Department of Pharmaceutical Sciences, University of Padova, Italy
| | | | - Angelo Avogaro
- Department of Medicine, University of Padova, Padova, Italy Venetian Institute of Molecular Medicine, Padova, Italy
| | - Gian Paolo Fadini
- Department of Medicine, University of Padova, Padova, Italy Venetian Institute of Molecular Medicine, Padova, Italy
| |
Collapse
|
49
|
Cheng CI, Chen PH, Lin YC, Kao YH. High glucose activates Raw264.7 macrophages through RhoA kinase-mediated signaling pathway. Cell Signal 2015; 27:283-92. [DOI: 10.1016/j.cellsig.2014.11.012] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 11/08/2014] [Indexed: 11/24/2022]
|
50
|
Iacobazzi D, Mangialardi G, Gubernator M, Hofner M, Wielscher M, Vierlinger K, Reni C, Oikawa A, Spinetti G, Vono R, Sangalli E, Montagnani M, Madeddu P. Increased antioxidant defense mechanism in human adventitia-derived progenitor cells is associated with therapeutic benefit in ischemia. Antioxid Redox Signal 2014; 21:1591-604. [PMID: 24512058 PMCID: PMC4174427 DOI: 10.1089/ars.2013.5404] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
AIMS Vascular wall-resident progenitor cells hold great promise for cardiovascular regenerative therapy. This study evaluates the impact of oxidative stress on the viability and functionality of adventitia-derived progenitor cells (APCs) from vein remnants of coronary artery bypass graft (CABG) surgery. We also investigated the antioxidant enzymes implicated in the resistance of APCs to oxidative stress-induced damage and the effect of interfering with one of them, the extracellular superoxide dismutase (EC-SOD/SOD3), on APC therapeutic action in a model of peripheral ischemia. RESULTS After exposure to hydrogen peroxide, APCs undergo apoptosis to a smaller extent than endothelial cells (ECs). This was attributed to up-regulation of antioxidant enzymes, especially SODs and catalase. Pharmacological inhibition of SODs increases reactive oxygen species (ROS) levels in APCs and impairs their survival. Likewise, APC differentiation results in SOD down-regulation and ROS-induced apoptosis. Oxidative stress increases APC migratory activity, while being inhibitory for ECs. In addition, oxidative stress does not impair APC capacity to promote angiogenesis in vitro. In a mouse limb ischemia model, an injection of naïve APCs, but not SOD3-silenced APCs, helps perfusion recovery and neovascularization, thus underlining the importance of this soluble isoform in protection from ischemia. INNOVATION This study newly demonstrates that APCs are endowed with enhanced detoxifier and antioxidant systems and that SOD3 plays an important role in their therapeutic activity in ischemia. CONCLUSIONS APCs from vein remnants of CABG patients express antioxidant defense mechanisms, which enable them to resist stress. These properties highlight the potential of APCs in cardiovascular regenerative medicine.
Collapse
Affiliation(s)
- Dominga Iacobazzi
- 1 Expermental Cardiovascular Medicine, School of Clinical Sciences, University of Bristol , Bristol, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|