1
|
Shikanai A, Furukawa S, Jiang S, Fujimura S, Kutomi G, Saito M, Tanaka R. Novel breast reconstruction technique using ex vivo mononuclear (RE-01) cells and adipose-derived mesenchymal stem cells. Regen Ther 2025; 29:271-281. [PMID: 40230355 PMCID: PMC11994943 DOI: 10.1016/j.reth.2025.03.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 03/01/2025] [Accepted: 03/23/2025] [Indexed: 04/16/2025] Open
Abstract
Introduction Breast reconstruction using fat grafts presents challenges; for example, fat necrosis owing to inadequate blood flow results in reduced engraftment rates. Supplementation of adipose tissue with adipose-derived mesenchymal stem cells (ADSCs) to promote the rapid vascularization of transplanted tissue has been investigated. However, the vascularization of fat-grafted tissues using only ADSC transplantation is limited. Ex vivo cultured mononuclear cells (RE-01) are a cell population with highly vascular and tissue-regenerative properties. This study aimed to evaluate the effect of combining RE-01 cells and ADSCs on the engraftment rate of fat grafts and explore the potential of this approach as a new option for breast reconstruction surgery. We hypothesized that combining RE-01 with ADSCs might promote angiogenesis and improve the fat grafting rate, consequently reducing the number of ADSCs required. Methods ADSCs cultured from human adipose tissue discarded during liposuction were co-cultured with RE-01 cells produced from the peripheral blood of healthy volunteers. In vitro vascular regeneration and adipogenic differentiation potential were analyzed. In addition, fat grafting experiments were conducted using nude mice to verify the fat grafting efficacy of ADSCs after co-cultivation with RE-01. Results ADSCs co-cultured with RE-01 cells promoted angiogenesis and adipogenesis in vitro. This was evidenced by a significant increase in the expression of adipogenic markers FABP4 and PPARγ, as well as enhanced lipid droplet formation observed through Oil Red O staining. The in vivo results demonstrated that the fat engraftment rate was significantly improved in the mixed group of ADSCs co-cultured with RE-01 cells. The number of blood vessels and fat quality of the transplanted adipose tissue were also increased in this group, suggesting that ADSCs co-cultured with RE-01 cells were highly effective in fat transplantation. Conclusions ADSCs co-cultured with RE-01 cells may be useful for improving the engraftment rate of fat grafts. However, further studies are required to verify the mechanisms.
Collapse
Affiliation(s)
- Ayana Shikanai
- Division of Regenerative Therapy, Juntendo University Graduates School of Medicine, Tokyo, Japan
- Department of Breast Oncology, Juntendo University School of Medicine, Tokyo, Japan
| | - Satomi Furukawa
- Division of Regenerative Therapy, Juntendo University Graduates School of Medicine, Tokyo, Japan
- Center for Genomic and Regenerative Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
- ReEir. Inc., Tokyo, Japan
| | - Sen Jiang
- Division of Regenerative Therapy, Juntendo University Graduates School of Medicine, Tokyo, Japan
- Intractable Disease Research Center, Juntendo University Graduates School of Medicine, Tokyo, Japan
| | - Satoshi Fujimura
- Division of Regenerative Therapy, Juntendo University Graduates School of Medicine, Tokyo, Japan
- Intractable Disease Research Center, Juntendo University Graduates School of Medicine, Tokyo, Japan
- ReEir. Inc., Tokyo, Japan
| | - Goro Kutomi
- Department of Breast Oncology, Juntendo University School of Medicine, Tokyo, Japan
| | - Mitsue Saito
- Department of Breast Oncology, Juntendo University School of Medicine, Tokyo, Japan
| | - Rica Tanaka
- Division of Regenerative Therapy, Juntendo University Graduates School of Medicine, Tokyo, Japan
- Department of Plastic and Reconstructive Surgery, Juntendo University School of Medicine, Tokyo, Japan
- Center for Genomic and Regenerative Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Intractable Disease Research Center, Juntendo University Graduates School of Medicine, Tokyo, Japan
- ReEir. Inc., Tokyo, Japan
| |
Collapse
|
2
|
Zamproni LN, Gökçe B, Venckute Larsson J, Ceballos-Torres A, Gram M, Porcionatto MA, Herland A. Unraveling the influence of astrocytes on endothelial cell transcription: Towards understanding blood-brain barrier in vitro models' dynamics. Brain Res Bull 2025; 224:111328. [PMID: 40174788 DOI: 10.1016/j.brainresbull.2025.111328] [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: 10/16/2024] [Revised: 03/13/2025] [Accepted: 03/30/2025] [Indexed: 04/04/2025]
Abstract
In recent years, considerable advancements have been made in developing in vitro models to better understand the complex dynamics of the blood-brain barrier (BBB) and its critical role in neurological health and disease. Incorporating astrocytes into these models introduces an essential layer of complexity, allowing for a more comprehensive investigation of the cellular interactions and regulatory mechanisms that maintain BBB integrity and functionality. Despite these advances, the specific influence of astrocytes on endothelial cells in in vitro systems remains inadequately explored. This study addresses this gap by examining the transcriptional changes in primary human brain microvascular endothelial cells (HBMECs) cocultured with human astrocytes (HAs). Our findings demonstrate that astrocytes profoundly modulate endothelial pathways involved in cell cycle regulation and division while upregulating genes associated with BBB integrity, protective mechanisms, and transporter activity. Furthermore, astrocytes significantly enhanced transendothelial electrical resistance (TEER) and reduced permeability to tracer Cascade Blue dye, confirming their functional impact on BBB models. By providing a comprehensive human primary cell dataset, this research underscores the pivotal role astrocytes play in shaping endothelial cell gene expression and function in contact coculture systems. These results emphasize the necessity of incorporating astrocytes into in vitro BBB models to accurately replicate neurovascular interactions. Ultimately, this study advances our understanding of BBB physiology and highlights the importance of refining in vitro models to better reflect the complexity of the human neurovascular environment, with potential implications for studying neurological disorders and drug delivery strategies.
Collapse
Affiliation(s)
- Laura Nicoleti Zamproni
- Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil; AIMES - Center for the Advancement of Integrated Medical and Engineering Sciences at Karolinska Institutet and KTH Royal Institute of Technology, Stockholm, Sweden; Department of Neuroscience, Karolinska Institutet, Stockholm
| | - Begüm Gökçe
- AIMES - Center for the Advancement of Integrated Medical and Engineering Sciences at Karolinska Institutet and KTH Royal Institute of Technology, Stockholm, Sweden; Department of Bioengineering, Graduate School of Natural and Applied Sciences, Ege University, Izmir, Turkey
| | - Justina Venckute Larsson
- Division of Nanobiotechnology, Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, Sweden; AIMES - Center for the Advancement of Integrated Medical and Engineering Sciences at Karolinska Institutet and KTH Royal Institute of Technology, Stockholm, Sweden; Department of Neuroscience, Karolinska Institutet, Stockholm
| | - Angela Ceballos-Torres
- Division of Nanobiotechnology, Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, Sweden; AIMES - Center for the Advancement of Integrated Medical and Engineering Sciences at Karolinska Institutet and KTH Royal Institute of Technology, Stockholm, Sweden; Department of Neuroscience, Karolinska Institutet, Stockholm
| | - Magnus Gram
- Department of Clinical Sciences Lund, Pediatrics, Lund University, Lund, Sweden; Department of Neonatology, Skåne University Hospital, Lund, Sweden; Department of Biomedical Science, Faculty of Health and Society, Biofilms - Research Centre for Biointerfaces, Malmö University, Malmö, Sweden
| | | | - Anna Herland
- Division of Nanobiotechnology, Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, Sweden; AIMES - Center for the Advancement of Integrated Medical and Engineering Sciences at Karolinska Institutet and KTH Royal Institute of Technology, Stockholm, Sweden; Department of Neuroscience, Karolinska Institutet, Stockholm.
| |
Collapse
|
3
|
Alonaizan R, Purnama U, Malandraki-Miller S, Gunadasa-Rohling M, Lewis A, Smart N, Carr C. MicroRNA-210 Enhances Cell Survival and Paracrine Potential for Cardiac Cell Therapy While Targeting Mitophagy. J Funct Biomater 2025; 16:147. [PMID: 40278255 DOI: 10.3390/jfb16040147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Revised: 04/04/2025] [Accepted: 04/16/2025] [Indexed: 04/26/2025] Open
Abstract
The therapeutic potential of presumed cardiac progenitor cells (CPCs) in heart regeneration has garnered significant interest, yet clinical trials have revealed limited efficacy due to challenges in cell survival, retention, and expansion. Priming CPCs to survive the hostile hypoxic environment may be key to enhancing their regenerative capacity. We demonstrate that microRNA-210 (miR-210), known for its role in hypoxic adaptation, significantly improves CPC survival by inhibiting apoptosis through the downregulation of Casp8ap2, a ~40% reduction in caspase activity, and a ~90% decrease in DNA fragmentation. Contrary to the expected induction of Bnip3-dependent mitophagy by hypoxia, miR-210 did not upregulate Bnip3, indicating a distinct anti-apoptotic mechanism. Instead, miR-210 reduced markers of mitophagy and increased mitochondrial biogenesis and oxidative metabolism, suggesting a role in metabolic reprogramming. Furthermore, miR-210 enhanced the secretion of paracrine growth factors from CPCs, with a ~1.6-fold increase in the release of stem cell factor and of insulin growth factor 1, which promoted in vitro endothelial cell proliferation and cardiomyocyte survival. These findings elucidate the multifaceted role of miR-210 in CPC biology and its potential to enhance cell-based therapies for myocardial repair by promoting cell survival, metabolic adaptation, and paracrine signalling.
Collapse
Affiliation(s)
- Rita Alonaizan
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford OX1 3PT, UK
- King Faisal Specialist Hospital & Research Centre, Riyadh 12713, Saudi Arabia
| | - Ujang Purnama
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford OX1 3PT, UK
| | | | - Mala Gunadasa-Rohling
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford OX1 3PT, UK
| | - Andrew Lewis
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford OX1 3PT, UK
| | - Nicola Smart
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford OX1 3PT, UK
| | - Carolyn Carr
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford OX1 3PT, UK
| |
Collapse
|
4
|
Brunmaier LAE, Ozdemir T, Walker TW. Angiogenesis: Biological Mechanisms and In Vitro Models. Ann Biomed Eng 2025:10.1007/s10439-025-03721-2. [PMID: 40210793 DOI: 10.1007/s10439-025-03721-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 03/25/2025] [Indexed: 04/12/2025]
Abstract
The translation of biomedical devices and drug research is an expensive and long process with a low probability of receiving FDA approval. Developing physiologically relevant in vitro models with human cells offers a solution to not only improving the odds of FDA approval but also to expand our ability to study complex in vivo systems in a simpler fashion. Animal models remain the standard for pre-clinical testing; however, the data from animal models is an unreliable extrapolation when anticipating a human response in clinical trials, thus contributing to the low rates of translation. In this review, we focus on in vitro vascular or angiogenic models because of the incremental role that the vascular system plays in the translation of biomedical research. The first section of this review discusses the most common angiogenic cytokines that are used in vitro to initiate angiogenesis, followed by angiogenic inhibitors where both initiators and inhibitors work to maintain vascular homeostasis. Next, we evaluate previously published in vitro models, where we evaluate capturing the physical environment for biomimetic in vitro modeling. These topics provide a foundation of parameters that must be considered to improve and achieve vascular biomimicry. Finally, we summarize these topics to suggest a path forward with the goal of engineering human in vitro models that emulate the in vivo environment and provide a platform for biomedical device and drug screening that produces data to support clinical translation.
Collapse
Affiliation(s)
- Laura A E Brunmaier
- Nanoscience and Biomedical Engineering Department, South Dakota School of Mines & Technology, 501 E St. Joseph St., Rapid City, SD, 57701, USA
| | - Tugba Ozdemir
- Nanoscience and Biomedical Engineering Department, South Dakota School of Mines & Technology, 501 E St. Joseph St., Rapid City, SD, 57701, USA
| | - Travis W Walker
- Karen M. Swindler Department of Chemical and Biological Engineering, South Dakota School of Mines & Technology, 501 E St. Joseph St., Rapid City, SD, 57701, USA.
| |
Collapse
|
5
|
Urhan E, Kara CS, Oguz EF, Neselioglu S, Erel O, Altuntas HD, Bayram F. The assessment of thiol-disulfide homeostasis and ıschemia-modified albumin levels in patients with acromegaly. Pituitary 2025; 28:46. [PMID: 40186831 PMCID: PMC11972178 DOI: 10.1007/s11102-025-01519-y] [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] [Accepted: 03/23/2025] [Indexed: 04/07/2025]
Abstract
PURPOSE Data regarding the relationship between acromegaly and oxidative stress (OS) remain limited. Dynamic thiol-disulfide homeostasis (TDH) is vital for antioxidant protection, and ischemia-modified albumin (IMA) serves as a marker of OS. This study aimed to measure serum TDH parameters and IMA levels in acromegaly patients, comparing them with healthy controls. METHODS This cross-sectional study consecutively included 81 patients and 55 controls, matched for age, gender, and body mass index. Serum levels of native thiol, total thiol, and disulfide (TDH parameters) were measured using the automated spectrophotometric method developed by Erel and Neselioglu, along with serum IMA levels. RESULTS In patients, serum native and total thiol levels were significantly lower (p = 0.005 and p = 0.007), while serum IMA levels were significantly higher (p = 0.001). Disulfide levels were similar. Patients with active disease (N = 32), patients in remission (N = 49), and controls (N = 55) were compared. In post-hoc analyses; serum TDH parameters and IMA levels were similar in remission and active disease patients. Native and total thiol levels were significantly lower in patients in remission compared to controls (p = 0.01 and p = 0.04). IMA levels were significantly higher in patients in remission compared to controls (p = 0.04). Serum thiol levels positively correlated with serum insulin-like growth factor-1 levels and negatively with age and disease duration, while age independently exerted a negative impact on serum thiol levels. CONCLUSION Our findings may indicate increased OS in the acromegalic process, which may contribute to the development of acromegaly and its related complications and comorbidities.
Collapse
Affiliation(s)
- Emre Urhan
- Department of Endocrinology, Erciyes University Medical School, Kayseri, Turkey.
| | - Canan Sehit Kara
- Department of Endocrinology, Erciyes University Medical School, Kayseri, Turkey
| | - Esra Fırat Oguz
- Department of Medical Biochemistry, Ankara City Hospital, Ankara, Turkey
| | - Salim Neselioglu
- Department of Medical Biochemistry, Yildirim Beyazit University Medical School, Ankara, Turkey
| | - Ozcan Erel
- Department of Medical Biochemistry, Yildirim Beyazit University Medical School, Ankara, Turkey
| | | | - Fahri Bayram
- Department of Endocrinology, Erciyes University Medical School, Kayseri, Turkey
| |
Collapse
|
6
|
Garand M, Huang SSY, Dineen B, Glass IA, Eghtesady P. Differential Regulation of Immune-Related Genes in the Developing Heart. Pediatr Cardiol 2025; 46:442-457. [PMID: 38480572 DOI: 10.1007/s00246-024-03441-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 02/02/2024] [Indexed: 02/02/2025]
Abstract
In many congenital heart defects, it can be difficult to ascertain primary pathology from secondary consequences from altered flow through the developing heart. The molecular differences between the growing right and left ventricles (RV and LV, respectively) following the completion of septation and the impact of sex on these mechanisms have not been investigated. We analyzed RNA-seq data derived from twelve RV and LVs, one with Hypoplastic Left Heart Syndrome (HLHS), to compare the transcriptomic landscape between the ventricles during development. Differential gene expression analysis revealed a large proportion of genes unique to either the RV or LV as well as sex bias. Our GO enrichment and network analysis strategy highlighted the differential role of immune functions between the RV and LV in the developing heart. Comparatively, RNA-seq analysis of data from C57Bl6/J mice hearts collected at E14 resulted in the enrichment of similar processes related to T cells and leukocyte migration and activation. Differential gene expression analysis of an HLHS case highlighted significant downregulation of chromatin organization pathways and upregulation of genes involved in muscle organ development. This analysis also identified previously unreported upregulation of genes involved in IL-17 production pathways. In conclusion, differences exist between the gene expression profiles of RV versus LV with the expression of immune-related genes being significantly different between these two chambers. The pathogenesis of HLHS may involve alterations in the expression of chromatin and muscle gene organization as well as upregulation of the IL-17 response pathway.
Collapse
Affiliation(s)
- Mathieu Garand
- Division of Pediatric Cardiothoracic Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Susie S Y Huang
- Division of Pediatric Cardiothoracic Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Brian Dineen
- Division of Pediatric Cardiothoracic Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Ian A Glass
- Department of Pediatrics and Medicine, University of Washington, Seattle, WA, USA
| | - Pirooz Eghtesady
- Division of Pediatric Cardiothoracic Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
7
|
Kleeberg A, Luft T, Golkowski D, Purrucker JC. Endothelial dysfunction in acute ischemic stroke: a review. J Neurol 2025; 272:143. [PMID: 39812851 PMCID: PMC11735568 DOI: 10.1007/s00415-025-12888-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 12/22/2024] [Accepted: 12/28/2024] [Indexed: 01/16/2025]
Abstract
BACKGROUND AND PURPOSE Endothelial dysfunction is considered an emerging therapeutic target to prevent complications during acute stroke and to prevent recurrent stroke. This review aims to provide an overview of the current knowledge on endothelial dysfunction, outline the diagnostic methods used to measure it and highlight the drugs currently being investigated for the treatment of endothelial dysfunction in acute ischemic stroke. METHODS The PubMed® and ClinicalTrials.gov electronic databases were searched for eligible articles/studies dealing with endothelial dysfunction and stroke. The references of the articles were screened to identify additional sources. The data were abstracted and summarized. FINDINGS AND DISCUSSION Endothelial dysfunction can be measured by serum biomarkers as well as by ultrasound or plethysmography techniques. Drugs targeting endothelial dysfunction include widely used agents such as angiotensin-converting enzyme inhibitors or isosorbide mononitrate, but also experimental therapies such as endothelial progenitor cells. CONCLUSION The role of endothelial dysfunction in acute ischemic stroke has been studied increasingly in recent years. It has been shown that there is a correlation between endothelial dysfunction and parenchymal hematoma after endovascular thrombectomy. Also, early clinical trials are conducted investigating, e.g., endothelial progenitor cells in the treatment of endothelial dysfunction in ischemic stroke. Current research focuses on the integration of novel markers of endothelial dysfunction into routine clinical practice to support decision making in the treatment of acute ischemic stroke.
Collapse
Affiliation(s)
- Antonia Kleeberg
- Department of Neurology, Heidelberg University Hospital, Heidelberg, Germany.
| | - Thomas Luft
- Department of Oncology and Hematology, University Hospital Heidelberg, Heidelberg, Germany
| | - Daniel Golkowski
- Department of Neurology, Heidelberg University Hospital, Heidelberg, Germany
| | - Jan C Purrucker
- Department of Neurology, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
8
|
Khan MZ, Zugaza JL, Torres Aleman I. The signaling landscape of insulin-like growth factor 1. J Biol Chem 2025; 301:108047. [PMID: 39638246 PMCID: PMC11748690 DOI: 10.1016/j.jbc.2024.108047] [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: 07/29/2024] [Revised: 11/22/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024] Open
Abstract
The sheer amplitude of biological actions of insulin-like growth factor I (IGF-1) affecting all types of cells in all tissues suggests a vast signaling landscape for this ubiquitous humoral signal. While the canonical signaling pathways primarily involve the Ras/MAPK and PI3K/AKT cascades, the evolutionary conservation of insulin-like peptides (ILPs) and their pathways hints at the potential for novel functions to emerge over time. Indeed, the evolutionary trajectory of ILPs opens the possibility of either novel functions for these two pathways, novel downstream routes, or both. Evidence supporting this notion includes observations of neofunctionalization in bony fishes or crustaceans, and the involvement of ILPs pathways in invertebrate eusociality or in vertebrate bone physiology, respectively. Such evolutionary processes likely contribute to the rich diversity of ILPs signaling observed today. Moreover, the interplay between conserved signaling pathways, such as those implicated in aging (predominantly involving the PI3K-AKT route), and lesser known pathways, such as those mediated by biased G-protein coupled receptors and others even less known, may underpin the context-dependent actions characteristic of ILPs signaling. While canonical IGF-1 signaling is often assumed to account for the intracellular pathways utilized by this growth factor, a comprehensive analysis of all the pathways mediated by the IGF-1 receptor (IGF-1R) remains lacking. This review aims to explore both canonical and non-canonical routes of IGF-1R action across various cell types, offering a detailed examination of the mechanisms underlying IGF-1 signaling and highlighting the significant gaps in our current understanding.
Collapse
Affiliation(s)
- Muhammad Zahid Khan
- Achucarro Basque Center for Neuroscience, Leioa, Spain; CIBERNED, Madrid, Spain
| | - Jose Luis Zugaza
- Achucarro Basque Center for Neuroscience, Leioa, Spain; Ikerbasque Science Foundation, Bilbao, Spain
| | - Ignacio Torres Aleman
- Achucarro Basque Center for Neuroscience, Leioa, Spain; CIBERNED, Madrid, Spain; Ikerbasque Science Foundation, Bilbao, Spain.
| |
Collapse
|
9
|
Ayoub M, Susin SA, Bauvois B. Tumor Cell Survival Factors and Angiogenesis in Chronic Lymphocytic Leukemia: How Hot Is the Link? Cancers (Basel) 2024; 17:72. [PMID: 39796700 PMCID: PMC11719013 DOI: 10.3390/cancers17010072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/19/2024] [Accepted: 12/24/2024] [Indexed: 01/13/2025] Open
Abstract
Chronic lymphocytic leukemia (CLL) is characterized by the accumulation of neoplastic CD5+/CD19+ B lymphocytes in the blood. These cells migrate to and proliferate in the bone marrow and lymphoid tissues. Despite the development of new therapies for CLL, drug resistance and disease relapse still occur; novel treatment approaches are therefore still needed. Inhibition of the angiogenesis involved in the progression of CLL might be a relevant therapeutic strategy. The literature data indicate that vascular endothelial growth factor, angiopoietin-2, and matrix metalloproteinase-9 are pro-angiogenic factors in CLL. A number of other CLL factors might have pro-angiogenic activity: fibroblast growth factor-2, certain chemokines (such as CXCL-12 and CXCL-2), tumor necrosis factor-α, insulin-like growth factor-1, neutrophil gelatinase-associated lipocalin, and progranulin. All these molecules contribute to the survival, proliferation, and migration of CLL cells. Here, we review the literature on these factors' respective expression profiles and roles in CLL. We also summarize the main results of preclinical and clinical trials of novel agents targeting most of these molecules in a CLL setting. Through the eradication of leukemic cells and the inhibition of angiogenesis, these therapeutic approaches might alter the course of CLL.
Collapse
Affiliation(s)
| | | | - Brigitte Bauvois
- Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, Inserm UMRS 1138, Drug Resistance in Hematological Malignancies Team, F-75006 Paris, France; (M.A.); (S.A.S.)
| |
Collapse
|
10
|
Limbu S, McCloskey KE. An Endothelial Cell Is Not Simply an Endothelial Cell. Stem Cells Dev 2024; 33:517-527. [PMID: 39030822 PMCID: PMC11564855 DOI: 10.1089/scd.2024.0088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 07/18/2024] [Indexed: 07/22/2024] Open
Abstract
Endothelial cells (ECs) are a multifaceted component of the vascular system with roles in immunity, maintaining tissue fluid balance, and vascular tone. Dysregulation or dysfunction of ECs can have far-reaching implications, leading pathologies ranging from cardiovascular diseases, such as hypertension and atherosclerosis, ischemia, chronic kidney disease, blood-brain barrier integrity, dementia, and tumor metastasis. Recent advancements in regenerative medicine have highlighted the potential of stem cell-derived ECs, particularly from induced pluripotent stem cells, to treat ischemic tissues, as well as models of vascular integrity. This review summarizes what is known in the generation of ECs with an emphasis on tissue-specific ECs and EC subphenotypes important in the development of targeted cell-based therapies for patient treatment.
Collapse
Affiliation(s)
- Shiwani Limbu
- Quantitative and System Biology Graduate Program, University of California, Merced, USA
| | - Kara E. McCloskey
- Quantitative and System Biology Graduate Program, University of California, Merced, USA
- Materials Science and Engineering Department, University of California, Merced, USA
| |
Collapse
|
11
|
Majumder S, Moriarty KL, Lee Y, Crombleholme TM. Placental Gene Therapy for Fetal Growth Restriction and Preeclampsia: Preclinical Studies and Prospects for Clinical Application. J Clin Med 2024; 13:5647. [PMID: 39337133 PMCID: PMC11432969 DOI: 10.3390/jcm13185647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/19/2024] [Accepted: 09/20/2024] [Indexed: 09/30/2024] Open
Abstract
In the last three decades, gene therapy has demonstrated significant progress. Over 700 active investigational new drug (IND) applications have been reported. Research on in utero gene therapy has advanced, but ethical and safety concerns persist. A novel approach under investigation is placental gene therapy, which holds promise for targeting diseases associated with placental dysfunction, such as fetal growth restriction (FGR) and preeclampsia. One of the underlying causes of placental insufficiency in these conditions is reduced placental growth factor-driven angiogenesis and endothelial cell dysfunction during fetal development. Studies have explored the overexpression of growth factor transgenes like IGF-1 to address FGR, yielding promising outcomes in animal models. Furthermore, intra-placental gene transfer, instead of systemic delivery of gene therapy vectors, has the potential to treat and cure these disorders. However, challenges and limitations akin to in utero gene therapy persist, including the risk of in utero infection, potential impairment of the mother's future fertility, the risk of germline integration, and possible off-target effects of gene transfer in the fetus or the mother. Consequently, additional research and deliberation within the scientific and medical communities are warranted to fully comprehend the potential benefits and risks of placental gene therapy.
Collapse
Affiliation(s)
- Sanjukta Majumder
- Molecular Fetal Therapy Laboratory, Fetal Care Center at Connecticut Children's Medical Center, Suite F254, 282 Washington Street, Hartford, CT 06106, USA
- Fetal Surgery Section, Division of Pediatric General and Thoracic Surgery, Department of Surgery, UConn Health, Farmington, CT 06030, USA
| | - Kristen Lee Moriarty
- Molecular Fetal Therapy Laboratory, Fetal Care Center at Connecticut Children's Medical Center, Suite F254, 282 Washington Street, Hartford, CT 06106, USA
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Farmington, CT 06030, USA
| | - Youngmok Lee
- Department of Pediatrics, School of Medicine, University of Connecticut, Farmington, CT 06030, USA
| | - Timothy M Crombleholme
- Molecular Fetal Therapy Laboratory, Fetal Care Center at Connecticut Children's Medical Center, Suite F254, 282 Washington Street, Hartford, CT 06106, USA
- Fetal Surgery Section, Division of Pediatric General and Thoracic Surgery, Department of Surgery, UConn Health, Farmington, CT 06030, USA
- Department of Pediatrics, School of Medicine, University of Connecticut, Farmington, CT 06030, USA
| |
Collapse
|
12
|
Wang X, Cao L, Liu S, Zhou Y, Zhou J, Zhao W, Gao S, Liu R, Shi Y, Shao C, Fang J. The critical roles of IGFs in immune modulation and inflammation. Cytokine 2024; 183:156750. [PMID: 39243567 DOI: 10.1016/j.cyto.2024.156750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/31/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024]
Abstract
Insulin-like growth factors (IGFs) are crucial for embryonic and postnatal growth and development, influencing cell survival, metabolism, myogenesis, and cancer progression. Many studies have demonstrated that IGFs also play prominent roles in the modulation of both innate and adaptive immune systems during inflammation. Strikingly, IGFs dictate the phenotype and functional properties of macrophages and T cells. Furthermore, the interplay between IGFs and inflammatory cytokines may generate tissue-protective properties during inflammation. Herein, we review the recent advances on the dialogue between immune cells and IGFs, especially zooming in on the significance of immunomodulatory properties in inflammatory conditions, cancer and autoimmune diseases. The investigation of IGFs may have broad clinical implications.
Collapse
Affiliation(s)
- Xin Wang
- The Third/Fourth Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Lijuan Cao
- The Third/Fourth Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China; Department of Experimental Medicine and Biochemical Sciences, TOR, University of Rome "Tor Vergata", Rome, Italy
| | - Shisong Liu
- The Third/Fourth Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Yipeng Zhou
- The Third/Fourth Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Jiarui Zhou
- The Third/Fourth Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Wenxuan Zhao
- The Third/Fourth Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Shengqi Gao
- The Third/Fourth Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Rui Liu
- The Third/Fourth Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China; Department of Experimental Medicine and Biochemical Sciences, TOR, University of Rome "Tor Vergata", Rome, Italy
| | - Yufang Shi
- The Third/Fourth Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China; Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Changshun Shao
- The Third/Fourth Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China.
| | - Jiankai Fang
- The Third/Fourth Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China.
| |
Collapse
|
13
|
Sahin S, Cicek E, Kocaman BB, Sulu C, Ozkaya HM, Ozkara H, Konukoglu D, Kadioglu P. The Association Between Oxidative Stress and Sperm Parameters in Patients with Acromegaly. Exp Clin Endocrinol Diabetes 2024; 132:498-506. [PMID: 38942036 DOI: 10.1055/a-2329-2998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/30/2024]
Abstract
OBJECTIVE Spermatozoa are susceptible to oxidative radicals when antioxidant defenses are inadequate. The extent to which oxidative radicals contribute to sperm damage in patients with acromegaly remains unclear. This study aimed to investigate and elucidate this relationship. METHODS The overall status of oxidants and antioxidants in both seminal plasma and serum of patients with acromegaly compared to a control group of healthy individuals was investigated. In addition, sperm parameters, including important measures such as growth hormone and insulin-like growth factor-1 concentrations. RESULTS Twenty-two patients with acromegaly with controlled disease and 14 healthy controls were included. The total oxidant status was significantly higher in the semen samples of the patients with acromegaly. A negative correlation was found between sperm total oxidant status and total sperm count and sperm concentration. Similarly, a negative correlation was found between the total sperm count and the sperm oxidative stress index. In individuals diagnosed with acromegaly, there was a statistically significant increase in sperm growth hormone levels. Conversely, the level of insulin-like growth factor 1 was significantly increased in the sperm of the control group, which consisted of healthy individuals. The correlation analysis revealed a significant relationship between venous total oxidant status and growth hormone levels in semen. CONCLUSION The elevated levels of reactive oxygen radicals in individuals with acromegaly suggest a possible link between oxidative stress and its effects on semen quality.
Collapse
Affiliation(s)
- Serdar Sahin
- Department of Endocrinology and Metabolic Diseases, Istanbul University-Cerrahpasa, Cerrahpasa School of Medicine, Istanbul, Turkey
| | - Ebru Cicek
- Department of Endocrinology and Metabolic Diseases, Istanbul University-Cerrahpasa, Cerrahpasa School of Medicine, Istanbul, Turkey
| | - Banu Betül Kocaman
- Department of Endocrinology and Metabolic Diseases, Istanbul University-Cerrahpasa, Cerrahpasa School of Medicine, Istanbul, Turkey
| | - Cem Sulu
- Department of Endocrinology and Metabolic Diseases, Istanbul University-Cerrahpasa, Cerrahpasa School of Medicine, Istanbul, Turkey
| | - Hande Mefkure Ozkaya
- Department of Endocrinology and Metabolic Diseases, Istanbul University-Cerrahpasa, Cerrahpasa School of Medicine, Istanbul, Turkey
| | - Hamdi Ozkara
- Department of Urology, Istanbul University-Cerrahpasa, Cerrahpasa School of Medicine, Istanbul, Turkey
| | - Dildar Konukoglu
- Department of Medical Biochemistry, Istanbul University-Cerrahpasa, Cerrahpasa School of Medicine, Istanbul, Turkey
| | - Pinar Kadioglu
- Department of Endocrinology and Metabolic Diseases, Istanbul University-Cerrahpasa, Cerrahpasa School of Medicine, Istanbul, Turkey
| |
Collapse
|
14
|
Wang J, Song X, Xia Z, Feng S, Zhang H, Xu C, Zhang H. Serum biomarkers for predicting microvascular complications of diabetes mellitus. Expert Rev Mol Diagn 2024; 24:703-713. [PMID: 39158206 DOI: 10.1080/14737159.2024.2391021] [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: 02/27/2024] [Accepted: 08/06/2024] [Indexed: 08/20/2024]
Abstract
INTRODUCTION Diabetic microvascular complications such as retinopathy, nephropathy, and neuropathy are primary causes of blindness, terminal renal failure, and neuropathic disorders in type 2 diabetes mellitus patients. Identifying reliable biomarkers promptly is pivotal for early detection and intervention in these severe complications. AREAS COVERED This review offers a thorough examination of the latest research concerning serum biomarkers for the prediction and assessment of diabetic microvascular complications. It encompasses biomarkers associated with glycation, oxidative stress, inflammation, endothelial dysfunction, basement membrane thickening, angiogenesis, and thrombosis. The review also highlights the potential of emerging biomarkers, such as microRNAs and long non-coding RNAs. EXPERT OPINION Serum biomarkers are emerging as valuable tools for the early assessment and therapeutic guidance of diabetic microvascular complications. The biomarkers identified not only reflect the underlying pathophysiology but also align with the extent of the disease. However, further validation across diverse populations and improvement of the practicality of these biomarkers in routine clinical practice are necessary. Pursuing these objectives is essential to advance early diagnosis, risk assessment, and individualized treatment regimens for those affected by diabetes.
Collapse
Affiliation(s)
- Jiajia Wang
- Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Department of Laboratory Medicine, Sichuan Provincial People's Hospital Chuandong Hospital & Dazhou First People's Hospital, Dazhou, China
| | - Xiaoyi Song
- School of medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Ziqiao Xia
- Laboratory medicine, Qianwei People's Hospital, Leshan, Sichuan, China
| | - Shu Feng
- Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Hangfeng Zhang
- Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Chengjie Xu
- Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Hui Zhang
- Department of Ultrasound, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
15
|
Boxhammer E, Paar V, Kopp K, Gharibeh SX, Bovenkamp-Aberger E, Rezar R, Lichtenauer M, Hoppe UC, Mirna M. Insulin-like Growth Factor-Binding Protein 2 in Severe Aortic Valve Stenosis and Pulmonary Hypertension: A Gender-Based Perspective. Int J Mol Sci 2024; 25:8220. [PMID: 39125788 PMCID: PMC11312253 DOI: 10.3390/ijms25158220] [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: 06/01/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 08/12/2024] Open
Abstract
Severe aortic valve stenosis (AS) and pulmonary hypertension (PH) are life-threatening cardiovascular conditions, necessitating early detection and intervention. Recent studies have explored the role of Insulin-like Growth Factor-Binding Protein 2 (IGF-BP2) in cardiovascular pathophysiology. Understanding its involvement may offer novel insights into disease mechanisms and therapeutic targets for these conditions. A total of 102 patients (46 female, 56 male) with severe AS undergoing a transcatheter aortic valve replacement (TAVR) in a single-center study were classified using echocardiography tests to determine systolic pulmonary artery pressure (sPAP) and the presence (sPAP ≥ 40 mmHg) or absence (sPAP < 40 mmHg) of PH. Additionally, serial laboratory determinations of IGF-BP2 before, and at 24 h, 96 h, and 3 months after intervention were conducted in all study participants. Considering the entire cohort, patients with PH had significant and continuously higher serum IGF-BP2 concentrations over time than patients without PH. After subdivision by sex, it could be demonstrated that the above-mentioned results were only verifiable in males, but not in females. In the male patients, baseline IGF-BP2 levels before the TAVR was an isolated risk factor for premature death after intervention and at 1, 3, and 5 years post-intervention. The same was valid for the combination of male and echocardiographically established PH patients. The predictive role of IGF-BP2 in severe AS and concurrent PH remains unknown. A more profound comprehension of IGF-BP2 mechanisms, particularly in males, could facilitate the earlier consideration of the TAVR as a more effective and successful treatment strategy.
Collapse
Affiliation(s)
- Elke Boxhammer
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University of Salzburg, 5020 Salzburg, Austria
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Acharya SS, Kundu CN. Havoc in harmony: Unravelling the intricacies of angiogenesis orchestrated by the tumor microenvironment. Cancer Treat Rev 2024; 127:102749. [PMID: 38714074 DOI: 10.1016/j.ctrv.2024.102749] [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: 02/20/2024] [Revised: 04/06/2024] [Accepted: 04/29/2024] [Indexed: 05/09/2024]
Abstract
Cancer cells merely exist in isolation; rather, they exist in an intricate microenvironment composed of blood vessels, signalling molecules, immune cells, stroma, fibroblasts, and the ECM. The TME provides a setting that is favourable for the successful growth and survivance of tumors. Angiogenesis is a multifaceted process that is essential for the growth, invasion, and metastasis of tumors. TME can be visualized as a "concert hall," where various cellular and non-cellular factors perform in a "symphony" to orchestrate tumor angiogenesis and create "Havoc" instead of "Harmony". In this review, we comprehensively summarized the involvement of TME in regulating tumor angiogenesis. Especially, we have focused on immune cells and their secreted factors, inflammatory cytokines and chemokines, and their role in altering the TME. We have also deciphered the crosstalk among various cell types that further aids the process of tumor angiogenesis. Additionally, we have highlighted the limitations of existing anti-angiogenic therapy and discussed various potential strategies that could be used to overcome these challenges and improve the efficacy of anti-angiogenic therapy.
Collapse
Affiliation(s)
- Sushree Subhadra Acharya
- Cancer Biology Division, School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed to be University (Institute of Eminence), Campus-11, Patia, Bhubaneswar, Odisha Pin-751024, India.
| | - Chanakya Nath Kundu
- Cancer Biology Division, School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed to be University (Institute of Eminence), Campus-11, Patia, Bhubaneswar, Odisha Pin-751024, India.
| |
Collapse
|
17
|
Lauvrud AT, Giraudo MV, Wiberg R, Wiberg M, Kingham PJ, Brohlin M. The influence of xeno-free culture conditions on the angiogenic and adipogenic differentiation properties of adipose tissue-derived stem cells. Regen Ther 2024; 26:901-910. [PMID: 39822342 PMCID: PMC11736170 DOI: 10.1016/j.reth.2024.09.013] [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: 07/18/2024] [Revised: 09/17/2024] [Accepted: 09/25/2024] [Indexed: 01/19/2025] Open
Abstract
Introduction Before performing cell therapy clinical trials, it is important to understand how cells are influenced by different growth conditions and to find optimal xeno-free medium formulations. In this study we have investigated the properties of adipose tissue-derived stem cells (ASCs) cultured under xeno-free conditions. Methods Human lipoaspirate samples were digested to yield the stromal vascular fraction cells which were then seeded in i) Minimum Essential Medium-α (MEM-α) supplemented with 10 % (v/v) fetal bovine serum (FBS), ii) MEM-α supplemented with 2 % (v/v) human platelet lysate (PLT) or iii) PRIME-XV MSC expansion XSFM xeno-free, serum free medium (XV). Flow cytometry for ASCs markers CD73, CD90 and CD105 together with the putative pericyte marker CD146 was performed. Growth rates were monitored over multiple passages and adipogenic differentiation performed at early and expanded passage culture. Growth factor gene expression was analyzed and an in vitro angiogenesis assay performed. Results Cells in FBS and PLT grew at similar rates whereas the cells cultured in XV medium proliferated significantly faster up to 60 days in culture. All cultures were >98 % positive for CD73, CD90 and CD105, whereas CD146 expression was significantly higher in XV cells. Adipogenic differentiation was most pronounced in cells which had been cultured in XV medium whilst cells grown in PLT were inferior compared with cells from the FBS cultures. IGF1 gene expression was highest in cells cultured in PLT whilst cells grown in XV medium showed 10-fold lower expression compared with FBS cells. In contrast, HGF gene expression was 90-fold greater in cells cultured in XV medium compared with those cultured in FBS. Conditioned medium from XV cultured cells showed the most angiogenic activity, inducing the greatest endothelial cell network formation and maturation. Conclusion Culture under different conditions alters the ASCs characteristics. Since cells cultured in XV medium showed the best adipogenic and angiogenic profile this might be a preferred medium formulation for preparing cells required for reconstructive surgical applications such as cell-assisted fat grafting.
Collapse
Affiliation(s)
- Anne Therese Lauvrud
- Department of Medical and Translational Biology, Umeå University, SE-901 87 Umeå, Sweden
- Department of Diagnostics and Intervention, Umeå University, SE-901 87 Umeå, Sweden
| | - Maria Vittoria Giraudo
- Department of Medical and Translational Biology, Umeå University, SE-901 87 Umeå, Sweden
| | - Rebecca Wiberg
- Department of Medical and Translational Biology, Umeå University, SE-901 87 Umeå, Sweden
- Department of Diagnostics and Intervention, Umeå University, SE-901 87 Umeå, Sweden
| | - Mikael Wiberg
- Department of Medical and Translational Biology, Umeå University, SE-901 87 Umeå, Sweden
- Department of Diagnostics and Intervention, Umeå University, SE-901 87 Umeå, Sweden
| | - Paul J. Kingham
- Department of Medical and Translational Biology, Umeå University, SE-901 87 Umeå, Sweden
| | - Maria Brohlin
- Department of Medical and Translational Biology, Umeå University, SE-901 87 Umeå, Sweden
- Department of Clinical Microbiology, Umeå University, SE-901 87 Umeå, Sweden
| |
Collapse
|
18
|
Kunnathattil M, Rahul P, Skaria T. Soluble vascular endothelial glycocalyx proteoglycans as potential therapeutic targets in inflammatory diseases. Immunol Cell Biol 2024; 102:97-116. [PMID: 37982607 DOI: 10.1111/imcb.12712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 10/30/2023] [Accepted: 10/30/2023] [Indexed: 11/21/2023]
Abstract
Reducing the activity of cytokines and leukocyte extravasation is an emerging therapeutic strategy to limit tissue-damaging inflammatory responses and restore immune homeostasis in inflammatory diseases. Proteoglycans embedded in the vascular endothelial glycocalyx, which regulate the activity of cytokines to restrict the inflammatory response in physiological conditions, are proteolytically cleaved in inflammatory diseases. Here we critically review the potential of proteolytically shed, soluble vascular endothelial glycocalyx proteoglycans to modulate pathological inflammatory responses. Soluble forms of the proteoglycans syndecan-1, syndecan-3 and biglycan exert beneficial anti-inflammatory effects by the removal of chemokines, suppression of proinflammatory cytokine expression and leukocyte migration, and induction of autophagy of proinflammatory M1 macrophages. By contrast, soluble versikine and decorin enhance proinflammatory responses by increasing inflammatory cytokine synthesis and leukocyte migration. Endogenous syndecan-2 and mimecan exert proinflammatory effects, syndecan-4 and perlecan mediate beneficial anti-inflammatory effects and glypican regulates Hh and Wnt signaling pathways involved in systemic inflammatory responses. Taken together, targeting the vascular endothelial glycocalyx-derived, soluble syndecan-1, syndecan-2, syndecan-3, syndecan-4, biglycan, versikine, mimecan, perlecan, glypican and decorin might be a potential therapeutic strategy to suppress overstimulated cytokine and leukocyte responses in inflammatory diseases.
Collapse
Affiliation(s)
- Maneesha Kunnathattil
- Department of Zoology, Government College Madappally, University of Calicut, Calicut, Kerala, India
| | - Pedapudi Rahul
- School of Biotechnology, National Institute of Technology Calicut, Calicut, Kerala, India
| | - Tom Skaria
- School of Biotechnology, National Institute of Technology Calicut, Calicut, Kerala, India
| |
Collapse
|
19
|
Li H, Yu W, Yang Y, Li S, Xu J, Gao C, Zhang W, Shi W, Jin K, Ji X, Ren C. Combination of Atractylenolide I, Atractylenolide III, and Paeoniflorin promotes angiogenesis and improves neurological recovery in a mouse model of ischemic Stroke. Chin Med 2024; 19:3. [PMID: 38178130 PMCID: PMC10768365 DOI: 10.1186/s13020-023-00872-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 12/10/2023] [Indexed: 01/06/2024] Open
Abstract
BACKGROUND Prognosis is critically important in stroke cases, with angiogenesis playing a key role in determining outcomes. This study aimed to investigate the potential protective effects of Atractylenolide I (Atr I), Atractylenolide III (Atr III), and Paeoniflorin (Pae) in promoting angiogenesis following cerebral ischemia. METHODS The bEnd.3 cell line was used to evaluate the effects of these three compounds on vascular endothelial cell proliferation, migration, and tube formation. Male C57BL/6 mice underwent transient middle cerebral artery occlusion (MCAO), followed by daily intragastric administration of the Chinese medicine compounds to assess their impact on brain protection and angiogenesis. In vivo experiments included measuring infarct size and assessing neurological function. Immunofluorescence staining and an angiogenesis antibody array were used to evaluate angiogenesis in ischemic brain tissue. Functional enrichment analysis was performed to further investigate the pathways involved in the protective effects of the compounds. Molecular docking analysis explored the potential binding affinity of the compounds to insulin-like growth factor 2 (IGF-2), and Western blotting was used to measure levels of angiogenesis-related proteins. RESULTS In vitro, the combination of Atr I, Atr III, and Pae enhanced cell proliferation, promoted migration, and stimulated tube formation. In vivo, the combined treatment significantly facilitated neurological function recovery and angiogenesis by day 14. The treatment also increased levels of angiogenesis-related proteins, including IGF-2. Pearson correlation analysis revealed a strong positive association between IGF-2 levels in ischemic brain tissue and angiogenesis, suggesting a good affinity of the compounds for the IGF-2 binding site, as supported by molecular docking analysis. CONCLUSION The administration of Atr I, Atr III, and Pae has shown significant enhancements in long-term stroke recovery in mice, likely due to the promotion of angiogenesis via increased activation of the IGF-2 pathway in ischemic brain tissue.
Collapse
Affiliation(s)
- Haiyan Li
- Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Center of Stroke, Beijing Institute of Brain Disorder, Capital Medical University, Chang Chun Road 45, Beijing, 100053, China
- School of Chinese Medicine, Beijing University of Chines Medicine, Beijing, 100029, China
| | - Wantong Yu
- Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Center of Stroke, Beijing Institute of Brain Disorder, Capital Medical University, Chang Chun Road 45, Beijing, 100053, China
| | - Yong Yang
- School of Chinese Medicine, Beijing University of Chines Medicine, Beijing, 100029, China
| | - Sijie Li
- Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Center of Stroke, Beijing Institute of Brain Disorder, Capital Medical University, Chang Chun Road 45, Beijing, 100053, China
| | - Jun Xu
- Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Center of Stroke, Beijing Institute of Brain Disorder, Capital Medical University, Chang Chun Road 45, Beijing, 100053, China
| | - Chen Gao
- Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Center of Stroke, Beijing Institute of Brain Disorder, Capital Medical University, Chang Chun Road 45, Beijing, 100053, China
| | - Wei Zhang
- Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Center of Stroke, Beijing Institute of Brain Disorder, Capital Medical University, Chang Chun Road 45, Beijing, 100053, China
| | - Wenjie Shi
- Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Center of Stroke, Beijing Institute of Brain Disorder, Capital Medical University, Chang Chun Road 45, Beijing, 100053, China
| | - Kunlin Jin
- Department of Pharmacology and Neuroscience, Texas Health Science Center, University of North, Fort Worth, TX, 76107, USA
| | - Xunming Ji
- Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Center of Stroke, Beijing Institute of Brain Disorder, Capital Medical University, Chang Chun Road 45, Beijing, 100053, China
| | - Changhong Ren
- Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Center of Stroke, Beijing Institute of Brain Disorder, Capital Medical University, Chang Chun Road 45, Beijing, 100053, China.
| |
Collapse
|
20
|
Pollenus E, Possemiers H, Knoops S, Prenen F, Vandermosten L, Thienpont C, Abdurahiman S, Demeyer S, Cools J, Matteoli G, Vanoirbeek JAJ, Vande Velde G, Van den Steen PE. Single cell RNA sequencing reveals endothelial cell killing and resolution pathways in experimental malaria-associated acute respiratory distress syndrome. PLoS Pathog 2024; 20:e1011929. [PMID: 38236930 PMCID: PMC10826972 DOI: 10.1371/journal.ppat.1011929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 01/30/2024] [Accepted: 12/29/2023] [Indexed: 01/31/2024] Open
Abstract
Plasmodium parasites cause malaria, a global health disease that is responsible for more than 200 million clinical cases and 600 000 deaths each year. Most deaths are caused by various complications, including malaria-associated acute respiratory distress syndrome (MA-ARDS). Despite the very rapid and efficient killing of parasites with antimalarial drugs, 15% of patients with complicated malaria succumb. This stresses the importance of investigating resolution mechanisms that are involved in the recovery from these complications once the parasite is killed. To study the resolution of MA-ARDS, P. berghei NK65-infected C57BL/6 mice were treated with antimalarial drugs after onset of symptoms, resulting in 80% survival. Micro-computed tomography revealed alterations of the lungs upon infection, with an increase in total and non-aerated lung volume due to edema. Whole body plethysmography confirmed a drastically altered lung ventilation, which was restored during resolution. Single-cell RNA sequencing indicated an increased inflammatory state in the lungs upon infection, which was accompanied by a drastic decrease in endothelial cells, consistent with CD8+ T cell-mediated killing. During resolution, anti-inflammatory pathways were upregulated and proliferation of endothelial cells was observed. MultiNicheNet interactome analysis identified important changes in the ligand-receptor interactions during disease resolution that warrant further exploration in order to develop new therapeutic strategies. In conclusion, our study provides insights in pro-resolving pathways that limit inflammation and promote endothelial cell proliferation in experimental MA-ARDS. This information may be useful for the design of adjunctive treatments to enhance resolution after Plasmodium parasite killing by antimalarial drugs.
Collapse
Affiliation(s)
- Emilie Pollenus
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology & Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Hendrik Possemiers
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology & Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Sofie Knoops
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology & Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Fran Prenen
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology & Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Leen Vandermosten
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology & Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Chloë Thienpont
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology & Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Saeed Abdurahiman
- Laboratory of Mucosal Immunology, Translational Research in Gastro-Intestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Sofie Demeyer
- Laboratory of Molecular Biology of Leukemia, Department of Human Genetics, VIB—KU Leuven, Leuven, Belgium
| | - Jan Cools
- Laboratory of Molecular Biology of Leukemia, Department of Human Genetics, VIB—KU Leuven, Leuven, Belgium
| | - Gianluca Matteoli
- Laboratory of Mucosal Immunology, Translational Research in Gastro-Intestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Jeroen A. J. Vanoirbeek
- Centre for Environment and Health, Department of Public Health and Primary Care, KU Leuven, Leuven, Belgium
| | - Greetje Vande Velde
- Biomedical MRI, Department of Imaging & Pathology, KU Leuven, Leuven, Belgium
| | - Philippe E. Van den Steen
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology & Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| |
Collapse
|
21
|
Liu ZL, Chen HH, Zheng LL, Sun LP, Shi L. Angiogenic signaling pathways and anti-angiogenic therapy for cancer. Signal Transduct Target Ther 2023; 8:198. [PMID: 37169756 PMCID: PMC10175505 DOI: 10.1038/s41392-023-01460-1] [Citation(s) in RCA: 342] [Impact Index Per Article: 171.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 03/20/2023] [Accepted: 04/20/2023] [Indexed: 05/13/2023] Open
Abstract
Angiogenesis, the formation of new blood vessels, is a complex and dynamic process regulated by various pro- and anti-angiogenic molecules, which plays a crucial role in tumor growth, invasion, and metastasis. With the advances in molecular and cellular biology, various biomolecules such as growth factors, chemokines, and adhesion factors involved in tumor angiogenesis has gradually been elucidated. Targeted therapeutic research based on these molecules has driven anti-angiogenic treatment to become a promising strategy in anti-tumor therapy. The most widely used anti-angiogenic agents include monoclonal antibodies and tyrosine kinase inhibitors (TKIs) targeting vascular endothelial growth factor (VEGF) pathway. However, the clinical benefit of this modality has still been limited due to several defects such as adverse events, acquired drug resistance, tumor recurrence, and lack of validated biomarkers, which impel further research on mechanisms of tumor angiogenesis, the development of multiple drugs and the combination therapy to figure out how to improve the therapeutic efficacy. Here, we broadly summarize various signaling pathways in tumor angiogenesis and discuss the development and current challenges of anti-angiogenic therapy. We also propose several new promising approaches to improve anti-angiogenic efficacy and provide a perspective for the development and research of anti-angiogenic therapy.
Collapse
Affiliation(s)
- Zhen-Ling Liu
- Department of Medicinal Chemistry, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, 210009, Nanjing, China
| | - Huan-Huan Chen
- Department of Medicinal Chemistry, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, 210009, Nanjing, China
| | - Li-Li Zheng
- Department of Medicinal Chemistry, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, 210009, Nanjing, China
| | - Li-Ping Sun
- Department of Medicinal Chemistry, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, 210009, Nanjing, China.
| | - Lei Shi
- Department of Medicinal Chemistry, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, 210009, Nanjing, China.
| |
Collapse
|
22
|
Albertine KH, Dahl MJ, Rebentisch A, Dawson E, Nabi A, Bowen S, Miers C, Wang Z, Yang H, Yu B, Null DM, Keefe D, Chung JK, Zhou Z, Barton N, Carey G, Ward R. Pilot dose-ranging of rhIGF-1/rhIGFBP-3 in a preterm lamb model of evolving bronchopulmonary dysplasia. Pediatr Res 2023; 93:1528-1538. [PMID: 36030318 PMCID: PMC9968819 DOI: 10.1038/s41390-022-02272-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 07/12/2022] [Accepted: 08/05/2022] [Indexed: 11/09/2022]
Abstract
BACKGROUND Low levels of insulin-like growth factor-1 (IGF-1) protein in preterm human infants are associated with bronchopulmonary dysplasia (BPD). We used our preterm lamb model of BPD to determine (1) dosage of recombinant human (rh) IGF-1 bound to binding protein-3 (IGFBP-3) to reach infant physiologic plasma levels; and (2) whether repletion of plasma IGF-1 improves pulmonary and cardiovascular outcomes. METHODS Group 1: normal, unventilated lambs from 128 days gestation through postnatal age 5 months defined normal plasma levels of IGF-1. Group 2: continuous infusion of rhIGF-1/rhIGFBP-3 (0.5, 1.5, or 4.5 mg/kg/day; n = 2) for 3 days in mechanically ventilated (MV) preterm lambs determined that 1.5 mg/kg/day dosage attained physiologic plasma IGF-1 concentration of ~125 ng/mL, which was infused in four more MV preterm lambs. RESULTS Group 1: plasma IGF-1 protein increased from ~75 ng/mL at 128 days gestation to ~220 ng/L at 5 months. Group 2: pilot study of the optimal dosage (1.5 mg/kg/day rhIGF-1/rhIGFBP-3) in six MV preterm lambs significantly improved some pulmonary and cardiovascular outcomes (p < 0.1) compared to six MV preterm controls. RhIGF-1/rhIGFBP-3 was not toxic to the liver, kidneys, or lungs. CONCLUSIONS Three days of continuous iv infusion of rhIGF-1/rhIGFBP-3 at 1.5 mg/kg/day improved some pulmonary and cardiovascular outcomes without toxicity. IMPACT Preterm birth is associated with rapid decreases in serum or plasma IGF-1 protein level. This decline adversely impacts the growth and development of the lung and cardiovascular system. For this pilot study, continuous infusion of optimal dosage of rhIGF-1/rhIGFBP-3 (1.5 mg/kg/day) to maintain physiologic plasma IGF-1 level of ~125 ng/mL during mechanical ventilation for 3 days statistically improved some structural and biochemical outcomes related to the alveolar formation that would favor improved gas exchange compared to vehicle-control. We conclude that 3 days of continuous iv infusion of rhIGF-1/rhIGFBP-3 improved some physiological, morphological, and biochemical outcomes, without toxicity, in mechanically ventilated preterm lambs.
Collapse
Affiliation(s)
- Kurt H Albertine
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, UT, USA.
| | - Mar Janna Dahl
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, UT, USA
| | - Andrew Rebentisch
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, UT, USA
| | - Elaine Dawson
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, UT, USA
| | - Akbar Nabi
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, UT, USA
| | - Sydney Bowen
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, UT, USA
| | - Cindy Miers
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, UT, USA
| | - Zhengming Wang
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, UT, USA
| | - Haixia Yang
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, UT, USA
| | - Baifeng Yu
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, UT, USA
| | - Donald M Null
- Division of Neonatology, Department of Pediatrics, University of California, Davis, Sacramento, CA, USA
| | - Dennis Keefe
- Takeda Pharmaceutical Company Limited, Cambridge, MA, USA
| | - J-K Chung
- Takeda Pharmaceutical Company Limited, Cambridge, MA, USA
| | - Z Zhou
- Takeda Pharmaceutical Company Limited, Cambridge, MA, USA
| | - Norman Barton
- Takeda Pharmaceutical Company Limited, Cambridge, MA, USA
| | - Galen Carey
- Takeda Pharmaceutical Company Limited, Cambridge, MA, USA
| | - Robert Ward
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
23
|
Sakata M, Kunimoto K, Kawaguchi A, Inaba Y, Kaminaka C, Yamamoto Y, Kakimoto N, Suenaga T, Tokuhara D, Jinnin M. Analysis of cytokine profiles in sera of single and multiple infantile hemangioma. J Dermatol 2023. [DOI: 10.1111/1346-8138.16781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 02/19/2023] [Accepted: 03/03/2023] [Indexed: 03/29/2023]
|
24
|
Isorhynchophylline inhibits inflammatory responses in endothelial cells and macrophages through the NF-κB/NLRP3 signaling pathway. BMC Complement Med Ther 2023; 23:80. [PMID: 36906555 PMCID: PMC10007741 DOI: 10.1186/s12906-023-03902-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 02/28/2023] [Indexed: 03/13/2023] Open
Abstract
BACKGROUND Atherosclerosis is a chronic inflammatory disease of arterial wall, which is closely related to inflammatory reaction. In this study, the anti-inflammatory effect of isorhynchophylline was studied by NF- κB / NLRP3 pathway. METHODS (1) ApoE-/- mice were fed with high-fat diet to establish atherosclerotic model, while C57 with the same genetic background was fed with common diet as control group. Body weight was recorded and blood lipids were detected. The expression of NLRP3, NF-κB, IL-18 and Caspase-1 in aorta was detected by Western-Blot and PCR, and plaque formation was detected by HE and oil red O staining. (2) Lipopolysaccharide interfered with Human Umbilical Vein Endothelial Cells (HUVECs) and RAW264.7 to form inflammatory model, and was treated with isorhynchophylline. The expression of NLRP3, NF-κB, IL-18 and Caspase-1 in aorta was detected by Western-Blot and PCR, and the ability of cell migration was detected by Transwell and scratch test. RESULTS (1) the expression of NLRP3, NF- κB, IL-18 and Caspase-1 in aorta of model group was higher than that of control group, and plaque formation was obvious. (2) the expressions of NLRP3, NF- κB, IL-18 and Caspase-1 in HUVECs and RAW264.7 model groups were higher than those in control group, while isorhynchophylline decreased their expression and enhanced cell migration ability. CONCLUSION Isorhynchophylline can reduce the inflammatory reaction induced by lipopolysaccharide and promote the ability of cell migration.
Collapse
|
25
|
Brites-Ferreira A, Taiar R, Cardoso ALBD, De Souza-Santos D, da Costa-Borges PP, Torres-Nunes L, Jaques-Albuquerque LT, Monteiro-Oliveira BB, Boyer FC, da Cunha Sá-Caputo D, Rapin A, Bernardo-Filho M. Therapeutic Approach of Whole-Body Vibration Exercise on Wound Healing in Animal Models: A Systematic Review. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:4925. [PMID: 36981833 PMCID: PMC10048796 DOI: 10.3390/ijerph20064925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/07/2023] [Accepted: 03/08/2023] [Indexed: 06/18/2023]
Abstract
Human skin wounds pose a gathering threat to the public health, carrying an immense epidemiologic and financial burden. Pharmacological and non-pharmacological (NP) treatments have been proposed to the management of wound healing. Physical exercise is a strong NP intervention considered for patients in wound healing. Particularly, a type of exercise intervention known as whole-body vibration (WBV) exercise has gained increasing interest. WBV exercise is generated due to the transmission of mechanical vibrations, produced by a vibrating platform, to the body. The aim of this review was to summarize studies in experimental animal models using WBV exercise in wound healing. Searches were performed in EMBASE, PubMed, Scopus and Web of Science including publications on 21 November 2022 using the string "whole body vibration" AND "wound healing" (animal or mice or mouse or rat or rodent). The SYRCLE tool was used to assess the risk of bias (RoB). From 48 studies, five studies met the inclusion criteria. RoB indicated that none of the studies fulfilled all methodological analyzed criteria, resulting in possible biases. The studies were homogeneous, and results suggest beneficial effects of WBV exercise in wound healing, mainly related to enhancing angiogenesis, granulation tissue formation, reducing the blood glucose level and enhancing blood microcirculation, by increasing myofiber growth and rapid re-epithelialization. In conclusion, the various biological effects of the response to the WBV exercise indicate the relevance of this intervention in wound healing in animals. Moreover, considering the translation approach, it is possible to speculate that the beneficial effects of this non-pharmacological therapy might justify clinical trials for wound healing also in humans, after criterion evaluation.
Collapse
Affiliation(s)
- Adrielli Brites-Ferreira
- Programa de Pós-Graduação em Fisiopatologia Clínica e Experimental, Faculdade de Ciências Médicas, Universidade do Estado do Rio de Janeiro, Rio de Janeiro 20551-030, Brazil
- Laboratório de Vibrações Mecânicas e Práticas Integrativas—LAVIMPI, Departamento de Biofísica e Biometria, Instituto de Biologia Roberto Alcantara Gomes and Policlínica Universitária Piquet Carneiro, Universidade do Estado do Rio de Janeiro, Rio de Janeiro 20950-003, Brazil
| | - Redha Taiar
- MATériaux et Ingénierie Mécanique (MATIM), Université de Reims, 51100 Reims, France
| | - André Luiz Bandeira Dionizio Cardoso
- Programa de Pós-Graduação em Fisiopatologia Clínica e Experimental, Faculdade de Ciências Médicas, Universidade do Estado do Rio de Janeiro, Rio de Janeiro 20551-030, Brazil
- Laboratório de Vibrações Mecânicas e Práticas Integrativas—LAVIMPI, Departamento de Biofísica e Biometria, Instituto de Biologia Roberto Alcantara Gomes and Policlínica Universitária Piquet Carneiro, Universidade do Estado do Rio de Janeiro, Rio de Janeiro 20950-003, Brazil
| | - Daysa De Souza-Santos
- Laboratório de Vibrações Mecânicas e Práticas Integrativas—LAVIMPI, Departamento de Biofísica e Biometria, Instituto de Biologia Roberto Alcantara Gomes and Policlínica Universitária Piquet Carneiro, Universidade do Estado do Rio de Janeiro, Rio de Janeiro 20950-003, Brazil
- Programa de Pós-Graduação em Saúde, Medicina Laboratorial e Tecnologia Forense, Universidade do Estado do Rio de Janeiro, Rio de Janeiro 20950-003, Brazil
| | - Patricia Prado da Costa-Borges
- Laboratório de Vibrações Mecânicas e Práticas Integrativas—LAVIMPI, Departamento de Biofísica e Biometria, Instituto de Biologia Roberto Alcantara Gomes and Policlínica Universitária Piquet Carneiro, Universidade do Estado do Rio de Janeiro, Rio de Janeiro 20950-003, Brazil
| | - Luiza Torres-Nunes
- Programa de Pós-Graduação em Fisiopatologia Clínica e Experimental, Faculdade de Ciências Médicas, Universidade do Estado do Rio de Janeiro, Rio de Janeiro 20551-030, Brazil
- Laboratório de Vibrações Mecânicas e Práticas Integrativas—LAVIMPI, Departamento de Biofísica e Biometria, Instituto de Biologia Roberto Alcantara Gomes and Policlínica Universitária Piquet Carneiro, Universidade do Estado do Rio de Janeiro, Rio de Janeiro 20950-003, Brazil
| | - Luelia Teles Jaques-Albuquerque
- Laboratório de Vibrações Mecânicas e Práticas Integrativas—LAVIMPI, Departamento de Biofísica e Biometria, Instituto de Biologia Roberto Alcantara Gomes and Policlínica Universitária Piquet Carneiro, Universidade do Estado do Rio de Janeiro, Rio de Janeiro 20950-003, Brazil
| | - Bruno Bessa Monteiro-Oliveira
- Programa de Pós-Graduação em Fisiopatologia Clínica e Experimental, Faculdade de Ciências Médicas, Universidade do Estado do Rio de Janeiro, Rio de Janeiro 20551-030, Brazil
- Laboratório de Vibrações Mecânicas e Práticas Integrativas—LAVIMPI, Departamento de Biofísica e Biometria, Instituto de Biologia Roberto Alcantara Gomes and Policlínica Universitária Piquet Carneiro, Universidade do Estado do Rio de Janeiro, Rio de Janeiro 20950-003, Brazil
| | - Francois Constant Boyer
- Centre Hospitalo-Universitaire de Reims, Département de Médecine Physique et de Réadaptation, Hôpital Sébastopol, Université de Reims Champagne-Ardenne, 51092 Reims, France
- Faculté de Médecine, VieFra, Université de Reims Champagne-Ardenne, 51097 Reims, France
| | - Danúbia da Cunha Sá-Caputo
- Laboratório de Vibrações Mecânicas e Práticas Integrativas—LAVIMPI, Departamento de Biofísica e Biometria, Instituto de Biologia Roberto Alcantara Gomes and Policlínica Universitária Piquet Carneiro, Universidade do Estado do Rio de Janeiro, Rio de Janeiro 20950-003, Brazil
| | - Amandine Rapin
- Centre Hospitalo-Universitaire de Reims, Département de Médecine Physique et de Réadaptation, Hôpital Sébastopol, Université de Reims Champagne-Ardenne, 51092 Reims, France
- Faculté de Médecine, VieFra, Université de Reims Champagne-Ardenne, 51097 Reims, France
| | - Mario Bernardo-Filho
- Laboratório de Vibrações Mecânicas e Práticas Integrativas—LAVIMPI, Departamento de Biofísica e Biometria, Instituto de Biologia Roberto Alcantara Gomes and Policlínica Universitária Piquet Carneiro, Universidade do Estado do Rio de Janeiro, Rio de Janeiro 20950-003, Brazil
| |
Collapse
|
26
|
Qu C, Liu X, Han X, Sun M, Liu H, Yang B. miR-216b-5p regulates proliferation and apoptosis of ox-LDL-stimulated VSMCs and HUVECs via IGF2. J Biochem Mol Toxicol 2023; 37:e23271. [PMID: 36510830 DOI: 10.1002/jbt.23271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 09/06/2022] [Accepted: 12/02/2022] [Indexed: 12/14/2022]
Abstract
Atherosclerosis (AS) is one of the principal causes of cardiovascular disorder. Reportedly, vascular smooth muscle cells (VSMCs) and human umbilical vein endothelial cells (HUVECs) play key roles in AS development, and microRNAs (miRNAs) regulate their functions. The function of miR-216b-5p in AS remains unknown. Human VSMCs and human HUVECs were treated with ox-LDL to establish the in vitro model of AS. MiR-216b-5p and IGF2 expressions in VSMCs and HUVECs were probed by qRT-PCR and western blot. The viability, cell cycle progression, and apoptosis of VSMCs and HUVECs were evaluated by Cell Counting Kit-8 (CCK-8), 5-ethynyl-2'-deoxyuridine, and flow cytometry assays, respectively. The binding sites between IGF2 3'UTR and miR-216b-5p were validated by dual-luciferase reporter assay. miR-216b-5p expression was declined in ox-LDL-induced VSMCs and HUVECs. In VSMCs, miR-216b-5p overexpression inhibited excessive proliferation and induced apoptosis. MiR-216b-5p could markedly restrain the viabiblity of VSMCs induced by ox-LDL and enhanced the viability of HUVECs. Additionally, IGF2 was confirmed as the direct target of miR-216b-5p and transfection of IGF2 overexpression plasmids rescued the effects of miR-216b-5p on VSMCs and HUVECs. miR-216b-5p alleviates the dysfunction of VSMCs and HUVECs caused by ox-LDL via repressing IGF2, and exerts protective functions to block the development of AS.
Collapse
Affiliation(s)
- Chuan Qu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China.,Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan, Hubei, China
| | - Xin Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China.,Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan, Hubei, China
| | - Xueyu Han
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China.,Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan, Hubei, China
| | - Meng Sun
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China.,Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan, Hubei, China
| | - Haixia Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China.,Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan, Hubei, China
| | - Bo Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China.,Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
27
|
Sukhanov S, Higashi Y, Yoshida T, Danchuk S, Alfortish M, Goodchild T, Scarborough A, Sharp T, Jenkins JS, Garcia D, Ivey J, Tharp DL, Schumacher J, Rozenbaum Z, Kolls JK, Bowles D, Lefer D, Delafontaine P. Insulin-like growth factor 1 reduces coronary atherosclerosis in pigs with familial hypercholesterolemia. JCI Insight 2023; 8:e165713. [PMID: 36602878 PMCID: PMC9990768 DOI: 10.1172/jci.insight.165713] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 01/03/2023] [Indexed: 01/06/2023] Open
Abstract
Although murine models of coronary atherosclerotic disease have been used extensively to determine mechanisms, limited new therapeutic options have emerged. Pigs with familial hypercholesterolemia (FH pigs) develop complex coronary atheromas that are almost identical to human lesions. We reported previously that insulin-like growth factor 1 (IGF-1) reduced aortic atherosclerosis and promoted features of stable plaque in a murine model. We administered human recombinant IGF-1 or saline (control) in atherosclerotic FH pigs for 6 months. IGF-1 decreased relative coronary atheroma in vivo (intravascular ultrasound) and reduced lesion cross-sectional area (postmortem histology). IGF-1 increased plaque's fibrous cap thickness, and reduced necrotic core, macrophage content, and cell apoptosis, consistent with promotion of a stable plaque phenotype. IGF-1 reduced circulating triglycerides, markers of systemic oxidative stress, and CXCL12 chemokine levels. We used spatial transcriptomics (ST) to identify global transcriptome changes in advanced plaque compartments and to obtain mechanistic insights into IGF-1 effects. ST analysis showed that IGF-1 suppressed FOS/FOSB factors and gene expression of MMP9 and CXCL14 in plaque macrophages, suggesting possible involvement of these molecules in IGF-1's effect on atherosclerosis. Thus, IGF-1 reduced coronary plaque burden and promoted features of stable plaque in a pig model, providing support for consideration of clinical trials.
Collapse
Affiliation(s)
- Sergiy Sukhanov
- Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Yusuke Higashi
- Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Tadashi Yoshida
- Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Svitlana Danchuk
- Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Mitzi Alfortish
- Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Traci Goodchild
- Cardiovascular Center of Excellence, School of Medicine, Louisiana State University, New Orleans, Louisiana, USA
| | - Amy Scarborough
- Cardiovascular Center of Excellence, School of Medicine, Louisiana State University, New Orleans, Louisiana, USA
| | - Thomas Sharp
- Cardiovascular Center of Excellence, School of Medicine, Louisiana State University, New Orleans, Louisiana, USA
| | | | | | - Jan Ivey
- Ochsner Medical Center, New Orleans, Louisiana, USA
| | - Darla L. Tharp
- Department of Biomedical Sciences, University of Missouri-Columbia, Missouri, USA
| | - Jeffrey Schumacher
- Cardiovascular Center of Excellence, School of Medicine, Louisiana State University, New Orleans, Louisiana, USA
| | - Zach Rozenbaum
- Tulane University School of Medicine, New Orleans, Louisiana, USA
- Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Jay K. Kolls
- Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Douglas Bowles
- Department of Biomedical Sciences, University of Missouri-Columbia, Missouri, USA
| | - David Lefer
- Cardiovascular Center of Excellence, School of Medicine, Louisiana State University, New Orleans, Louisiana, USA
| | | |
Collapse
|
28
|
Systemic Cytokines in Retinopathy of Prematurity. J Pers Med 2023; 13:jpm13020291. [PMID: 36836525 PMCID: PMC9966226 DOI: 10.3390/jpm13020291] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 01/30/2023] [Accepted: 01/30/2023] [Indexed: 02/09/2023] Open
Abstract
Retinopathy of prematurity (ROP), a vasoproliferative vitreoretinal disorder, is the leading cause of childhood blindness worldwide. Although angiogenic pathways have been the main focus, cytokine-mediated inflammation is also involved in ROP etiology. Herein, we illustrate the characteristics and actions of all cytokines involved in ROP pathogenesis. The two-phase (vaso-obliteration followed by vasoproliferation) theory outlines the evaluation of cytokines in a time-dependent manner. Levels of cytokines may even differ between the blood and the vitreous. Data from animal models of oxygen-induced retinopathy are also valuable. Although conventional cryotherapy and laser photocoagulation are well established and anti-vascular endothelial growth factor agents are available, less destructive novel therapeutics that can precisely target the signaling pathways are required. Linking the cytokines involved in ROP to other maternal and neonatal diseases and conditions provides insights into the management of ROP. Suppressing disordered retinal angiogenesis via the modulation of hypoxia-inducible factor, supplementation of insulin-like growth factor (IGF)-1/IGF-binding protein 3 complex, erythropoietin, and its derivatives, polyunsaturated fatty acids, and inhibition of secretogranin III have attracted the attention of researchers. Recently, gut microbiota modulation, non-coding RNAs, and gene therapies have shown promise in regulating ROP. These emerging therapeutics can be used to treat preterm infants with ROP.
Collapse
|
29
|
Chen H, Aneman I, Nikolic V, Karadzov Orlic N, Mikovic Z, Stefanovic M, Cakic Z, Jovanovic H, Town SEL, Padula MP, McClements L. Maternal plasma proteome profiling of biomarkers and pathogenic mechanisms of early-onset and late-onset preeclampsia. Sci Rep 2022; 12:19099. [PMID: 36351970 PMCID: PMC9646706 DOI: 10.1038/s41598-022-20658-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 09/16/2022] [Indexed: 11/10/2022] Open
Abstract
Preeclampsia is still the leading cause of morbidity and mortality in pregnancy without a cure. There are two phenotypes of preeclampsia, early-onset (EOPE) and late-onset (LOPE) with poorly defined pathogenic differences. This study aimed to facilitate better understanding of the mechanisms of pathophysiology of EOPE and LOPE, and identify specific biomarkers or therapeutic targets. In this study, we conducted an untargeted, label-free quantitative proteomic analyses of plasma samples from pregnant women with EOPE (n = 17) and LOPE (n = 11), and age, BMI-matched normotensive controls (n = 18). Targeted proteomics approach was also employed to validate a subset of proteins (n = 17). In total, there were 26 and 20 differentially abundant proteins between EOPE or LOPE, and normotensive controls, respectively. A series of angiogenic and inflammatory proteins, including insulin-like growth factor-binding protein 4 (IGFBP4; EOPE: FDR = 0.0030 and LOPE: FDR = 0.00396) and inter-alpha-trypsin inhibitor heavy chain H2-4 (ITIH2-4), were significantly altered in abundance in both phenotypes. Through validation we confirmed that ITIH2 was perturbed only in LOPE (p = 0.005) whereas ITIH3 and ITIH4 were perturbed in both phenotypes (p < 0.05). Overall, lipid metabolism/transport proteins associated with atherosclerosis were highly abundant in LOPE, however, ECM proteins had a more pronounced role in EOPE. The complement cascade and binding and uptake of ligands by scavenger receptors, pathways, were associated with both EOPE and LOPE.
Collapse
Affiliation(s)
- Hao Chen
- grid.117476.20000 0004 1936 7611School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW Australia
| | - Ingrid Aneman
- grid.117476.20000 0004 1936 7611School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW Australia
| | - Valentina Nikolic
- grid.11374.300000 0001 0942 1176Department of Pharmacology and Toxicology, Faculty of Medicine, University of Nis, Nis, Serbia
| | - Natasa Karadzov Orlic
- Department of Gynaecology and Obstetrics, Narodni Front, Belgrade, Serbia ,grid.7149.b0000 0001 2166 9385Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Zeljko Mikovic
- Department of Gynaecology and Obstetrics, Narodni Front, Belgrade, Serbia ,grid.7149.b0000 0001 2166 9385Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Milan Stefanovic
- grid.11374.300000 0001 0942 1176Department of Gynaecology and Obstetrics, Faculty of Medicine, University of Nis, Nis, Serbia ,grid.418653.d0000 0004 0517 2741Gynaecology and Obstetrics Clinic, Clinical Centre Nis, Nis, Serbia
| | - Zoran Cakic
- Department of Gynaecology and Obstetrics, General Hospital of Leskovac, Leskovac, Serbia
| | - Hristina Jovanovic
- grid.11374.300000 0001 0942 1176Department of Pharmacology and Toxicology, Faculty of Medicine, University of Nis, Nis, Serbia
| | - Stephanie E. L. Town
- grid.117476.20000 0004 1936 7611School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW Australia
| | - Matthew P. Padula
- grid.117476.20000 0004 1936 7611School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW Australia
| | - Lana McClements
- grid.117476.20000 0004 1936 7611School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW Australia ,grid.117476.20000 0004 1936 7611Institute for Biomedical Materials and Devices, Faculty of Science, University of Technology Sydney, Ultimo, NSW Australia
| |
Collapse
|
30
|
Nadine S, Fernandes IJ, Correia CR, Mano JF. Close-to-native bone repair via tissue-engineered endochondral ossification approaches. iScience 2022; 25:105370. [PMID: 36339269 PMCID: PMC9626746 DOI: 10.1016/j.isci.2022.105370] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
In order to solve the clinical challenges related to bone grafting, several tissue engineering (TE) strategies have been proposed to repair critical-sized defects. Generally, the classical TE approaches are designed to promote bone repair via intramembranous ossification. Although promising, strategies that direct the osteogenic differentiation of mesenchymal stem/stromal cells are usually characterized by a lack of functional vascular supply, often resulting in necrotic cores. A less explored alternative is engineering bone constructs through a cartilage-mediated approach, resembling the embryological process of endochondral ossification. The remodeling of an intermediary hypertrophic cartilaginous template triggers vascular invasion and bone tissue deposition. Thus, employing this knowledge can be a promising direction for the next generation of bone TE constructs. This review highlights the most recent biomimetic strategies for applying endochondral ossification in bone TE while discussing the plethora of cell types, culture conditions, and biomaterials essential to promote a successful bone regeneration process.
Collapse
|
31
|
Moskalik A, Ratajska A, Majchrzak B, Jankowska-Steifer E, Bartkowiak K, Bartkowiak M, Niderla-Bielińska J. miR-31-5p-Modified RAW 264.7 Macrophages Affect Profibrotic Phenotype of Lymphatic Endothelial Cells In Vitro. Int J Mol Sci 2022; 23:13193. [PMID: 36361979 PMCID: PMC9657882 DOI: 10.3390/ijms232113193] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/23/2022] [Accepted: 10/27/2022] [Indexed: 07/30/2023] Open
Abstract
Cardiac lymphatic vessel (LyV) remodeling as a contributor to heart failure has not been extensively evaluated in metabolic syndrome (MetS). Our studies have shown structural changes in cardiac LyV in MetS that contribute to the development of edema and lead to myocardial fibrosis. Tissue macrophages may affect LyV via secretion of various substances, including noncoding RNAs. The aim of the study was to evaluate the influence of macrophages modified by miR-31-5p, a molecule that regulates fibrosis and lymphangiogenesis, on lymphatic endothelial cells (LECs) in vitro. The experiments were carried out on the RAW 264.7 macrophage cell line and primary dermal lymphatic endothelial cells. RAW 264.7 macrophages were transfected with miR-31-5p and supernatant from this culture was used for LEC stimulation. mRNA expression levels for genes associated with lymphangiogenesis and fibrosis were measured with qRT-PCR. Selected results were confirmed with ELISA or Western blotting. miR-31-5p-modified RAW 264.7 macrophages secreted increased amounts of VEGF-C and TGF-β and a decreased amount of IGF-1. The supernatant from miR-31-5p-modified RAW 264.7 downregulated the mRNA expression for genes regulating endothelial-to-mesenchymal transition (EndoMT) and fibrosis in LECs. Our results suggest that macrophages under the influence of miR-31-5p show the potential to inhibit LEC-dependent fibrosis. However, more studies are needed to confirm this effect in vivo.
Collapse
Affiliation(s)
- Aneta Moskalik
- Postgraduate School of Molecular Medicine, Collegium Anatomicum, Medical University of Warsaw, 02-004 Warsaw, Poland
| | - Anna Ratajska
- Department of Pathology, Collegium Anatomicum, Medical University of Warsaw, 02-004 Warsaw, Poland
| | - Barbara Majchrzak
- Department of Pathology, Collegium Anatomicum, Medical University of Warsaw, 02-004 Warsaw, Poland
| | - Ewa Jankowska-Steifer
- Department of Histology and Embryology, Collegium Anatomicum, Medical University of Warsaw, 02-004 Warsaw, Poland
| | - Krzysztof Bartkowiak
- Department of Histology and Embryology, Collegium Anatomicum, Medical University of Warsaw, 02-004 Warsaw, Poland
| | - Mateusz Bartkowiak
- Department of History of Medicine, Medical University of Warsaw, 00-581 Warsaw, Poland
| | - Justyna Niderla-Bielińska
- Department of Histology and Embryology, Collegium Anatomicum, Medical University of Warsaw, 02-004 Warsaw, Poland
| |
Collapse
|
32
|
Systemic Biomarkers and Unique Pathways in Different Phenotypes of Heart Failure with Preserved Ejection Fraction. Biomolecules 2022; 12:biom12101419. [PMID: 36291628 PMCID: PMC9599828 DOI: 10.3390/biom12101419] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 09/28/2022] [Accepted: 09/29/2022] [Indexed: 11/16/2022] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) accounts for around 50% of all heart failure cases. It is a heterogeneous condition with poorly understood pathogenesis. Here, we aimed to identify unique pathogenic mechanisms in acute and chronic HFpEF and hypertrophic cardiomyopathy (HCM). We performed unbiased, comprehensive proteomic analyses of plasma samples from gender- and BMI-matched patients with acute HFpEF (n = 8), chronic HFpEF (n = 9) and HCM (n = 14) using liquid chromatography–mass spectrometry. Distinct molecular signatures were observed in different HFpEF forms. Clusters of biomarkers differentially abundant between HFpEF forms were predominantly associated with microvascular inflammation. New candidate protein markers were also identified, including leucine-rich alpha-2-glycoprotein 1 (LRG1), serum amyloid A1 (SAA1) and inter-alpha-trypsin inhibitor heavy chain 3 (ITIH3). Our study is the first to apply systematic, quantitative proteomic screening of plasma samples from patients with different subtypes of HFpEF and identify candidate biomarkers for improved management of acute and chronic HFpEF and HCM.
Collapse
|
33
|
Weaver SRC, Rendeiro C, Lucas RAI, Cable NT, Nightingale TE, McGettrick HM, Lucas SJE. Non-pharmacological interventions for vascular health and the role of the endothelium. Eur J Appl Physiol 2022; 122:2493-2514. [PMID: 36149520 PMCID: PMC9613570 DOI: 10.1007/s00421-022-05041-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 09/05/2022] [Indexed: 12/11/2022]
Abstract
The most common non-pharmacological intervention for both peripheral and cerebral vascular health is regular physical activity (e.g., exercise training), which improves function across a range of exercise intensities and modalities. Numerous non-exercising approaches have also been suggested to improved vascular function, including repeated ischemic preconditioning (IPC); heat therapy such as hot water bathing and sauna; and pneumatic compression. Chronic adaptive responses have been observed across a number of these approaches, yet the precise mechanisms that underlie these effects in humans are not fully understood. Acute increases in blood flow and circulating signalling factors that induce responses in endothelial function are likely to be key moderators driving these adaptations. While the impact on circulating factors and environmental mechanisms for adaptation may vary between approaches, in essence, they all centre around acutely elevating blood flow throughout the circulation and stimulating improved endothelium-dependent vascular function and ultimately vascular health. Here, we review our current understanding of the mechanisms driving endothelial adaptation to repeated exposure to elevated blood flow, and the interplay between this response and changes in circulating factors. In addition, we will consider the limitations in our current knowledge base and how these may be best addressed through the selection of more physiologically relevant experimental models and research. Ultimately, improving our understanding of the unique impact that non-pharmacological interventions have on the vasculature will allow us to develop superior strategies to tackle declining vascular function across the lifespan, prevent avoidable vascular-related disease, and alleviate dependency on drug-based interventions.
Collapse
Affiliation(s)
- Samuel R C Weaver
- School of Sport, Exercise and Rehabilitation Sciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham, UK.
- Centre for Human Brain Health, University of Birmingham, Birmingham, UK.
| | - Catarina Rendeiro
- School of Sport, Exercise and Rehabilitation Sciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham, UK
- Centre for Human Brain Health, University of Birmingham, Birmingham, UK
| | - Rebekah A I Lucas
- School of Sport, Exercise and Rehabilitation Sciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham, UK
| | - N Timothy Cable
- Institute of Sport, Manchester Metropolitan University, Manchester, UK
| | - Tom E Nightingale
- School of Sport, Exercise and Rehabilitation Sciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham, UK
| | - Helen M McGettrick
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Samuel J E Lucas
- School of Sport, Exercise and Rehabilitation Sciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham, UK
- Centre for Human Brain Health, University of Birmingham, Birmingham, UK
| |
Collapse
|
34
|
Huang Y, Song C, He J, Li M. Research progress in endothelial cell injury and repair. Front Pharmacol 2022; 13:997272. [PMID: 36176426 PMCID: PMC9513221 DOI: 10.3389/fphar.2022.997272] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 08/24/2022] [Indexed: 11/30/2022] Open
Abstract
Endothelial cells, which are important metabolic and endocrine cells, play an important role in regulating vascular function. The occurrence and development of various cardiovascular and cerebrovascular diseases are associated with endothelial dysfunction. However, the underlying mechanism of vascular endothelial injury is not fully understood. It has been reported that the mechanism of endothelial injury mainly involves inflammation and oxidative stress. Moreover, endothelial progenitor cells are regarded as important contributors in repairing damaged endothelium. Multiple interventions (including chemical drugs and traditional Chinese medicines) exert endothelial protection by decreasing the release of inducing factors, suppressing inflammation and oxidative stress, and preventing endothelial cell senescence.
Collapse
Affiliation(s)
- Yongpan Huang
- Medicine School, Changsha Social Work College, Changsha, Hunan, China
| | - Chong Song
- Medicine School, Changsha Social Work College, Changsha, Hunan, China
| | - Jianbin He
- Department of Respiratory and Critical Care Medicine, The First People’s Hospital of Huaihua, Affiliated to University of South China, Huaihua, Hunan, China
- *Correspondence: Jianbin He, ; Min Li,
| | - Min Li
- Medicine School, Changsha Social Work College, Changsha, Hunan, China
- *Correspondence: Jianbin He, ; Min Li,
| |
Collapse
|
35
|
Seo SH, Hwang S, Hwang S, Han S, Park H, Lee Y, Rho SB, Kwon Y. Hypoxia‐induced ELF3 promotes tumor angiogenesis through IGF1/IGF1R. EMBO Rep 2022; 23:e52977. [PMID: 35695065 PMCID: PMC9346469 DOI: 10.15252/embr.202152977] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/09/2022] [Accepted: 05/19/2022] [Indexed: 11/09/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is one of the most lethal gynecological cancers despite a relatively low incidence. Angiogenesis, one of the hallmarks of cancer, is essential for the pathogenesis of EOC, which is related to the induction of angiogenic factors. We found that ELF3 was highly expressed in EOCs under hypoxia and functioned as a transcription factor for IGF1. The ELF3‐mediated increase in the secretion of IGF1 and VEGF promoted endothelial cell proliferation, migration, and EOC angiogenesis. Although this situation was much exaggerated under hypoxia, ELF3 silencing under hypoxia significantly attenuated angiogenic activity in endothelial cells by reducing the expression and secretion of IGF1 and VEGF. ELF3 silencing attenuated angiogenesis and tumorigenesis in ex vivo and xenograft mouse models. Consequently, ELF3 plays an important role in the induction of angiogenesis and tumorigenesis in EOC as a transcription factor of IGF1. A detailed understanding of the biological mechanism of ELF3 may both improve current antiangiogenic therapies and have anticancer effects for EOC.
Collapse
Affiliation(s)
- Seung Hee Seo
- College of Pharmacy Graduate School of Pharmaceutical Sciences Ewha Womans University Seoul Korea
| | - Soo‐Yeon Hwang
- College of Pharmacy Graduate School of Pharmaceutical Sciences Ewha Womans University Seoul Korea
| | - Seohui Hwang
- College of Pharmacy Graduate School of Pharmaceutical Sciences Ewha Womans University Seoul Korea
| | - Sunjung Han
- College of Pharmacy Graduate School of Pharmaceutical Sciences Ewha Womans University Seoul Korea
| | - Hyojin Park
- College of Pharmacy Graduate School of Pharmaceutical Sciences Ewha Womans University Seoul Korea
| | - Yun‐Sil Lee
- College of Pharmacy Graduate School of Pharmaceutical Sciences Ewha Womans University Seoul Korea
| | - Seung Bae Rho
- Research Institute National Cancer Center Goyang‐si Gyeonggi‐do Korea
| | - Youngjoo Kwon
- College of Pharmacy Graduate School of Pharmaceutical Sciences Ewha Womans University Seoul Korea
| |
Collapse
|
36
|
Xu W, Wu C, Zhu X, Wu J, Zhang Z, Wei Z, Cao Y, Zhou P, Wang J. UC-MSCs promote frozen-thawed ovaries angiogenesis via activation of the Wnt/β-catenin pathway in vitro ovarian culture system. Stem Cell Res Ther 2022; 13:296. [PMID: 35841074 PMCID: PMC9284710 DOI: 10.1186/s13287-022-02989-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 04/07/2022] [Indexed: 12/01/2022] Open
Abstract
Background Ovarian tissue cryopreservation and transplantation are novel therapeutic approaches for fertility preservation. However, follicle loss caused by ischemic and hypoxic damage is one of the issues after frozen-thawed ovarian tissue transplantation. Promoting angiogenesis in grafts is the key to restore cryopreserved ovarian function. Mesenchymal stem cells (MSCs) have been reported to facilitate angiogenesis in the cryopreserved ovarian tissue transplantation. However, the risk of embolization, immunogenic effect and tumorigenesis hinders the clinical application of MSCs to human organ transplantation. In this study, we established an in vitro ovarian culture system to restore frozen-thawed ovarian function before transplantation with the application of umbilical cord mesenchymal stem cells (UC-MSCs), and explored the effects of UC-MSCs on frozen-thawed ovaries in vitro ovarian culture system and the mechanisms of UC-MSCs on the angiogenesis of frozen-thawed ovaries. Methods A simple in vitro three dimensional (3D) ovarian culture system using Matrigel was established to support to an ideal niche, and ovary was alone cultured in the 24-well plate as a control. We also evaluated the effects of UC-MSCs treatment on ovarian function with or without Matrigel support. All thawed ovaries were randomly divided into control group (Matrigel−/UC-MSCs−), Matrigel group (Matrigel+/UC-MSCs−), UC-MSCs group (Matrigel−/UC-MSCs+) and UC-MSCs + Matrigel group (Matrigel+/UC-MSCs+). HE staining was used to detect the histological structure of follicles and TUNEL staining was used to detect cell apoptosis. The number of microvessels was counted to evaluate neovascularization. The mRNA expression of VEGFA, IGF1 and ANGPT2 were detected by RT-PCR. Western blotting was used to measure the expression of GSK-3β, β-catenin and p-β-catenin. Results In the absence of UC-MSCs, 3D culture system supported by Matrigel showed significantly improved follicular development and microvascular number. Additionally, UC-MSCs were also found to effectively improve follicular development and microvascular number regardless of the culture condition used. However, alleviated follicular apoptosis, increased mRNA expression of angiogenesis-related gene and activated Wnt/β-catenin pathway occurred only in the UC-MSCs + Matrigel group. Besides, with the application of IWP-2 in UC-MSCs + Matrigel group, Wnt//β-catenin pathway could be blocked by IWP-2 serving as one of Wnt/β-catenin pathway inhibitors. Conclusions This in vitro study showed the beneficial effects of UC-MSCs on thawed ovaries and explored a potential mechanism inducing angiogenesis. In particular, 3D ovarian culture system supported by Matrigel further improved UC-MSCs treatment. The in vitro culture system using Matrigel and UC-MSCs may provide a potential treatment strategy for improving the success rate of thawed ovaries transplantation. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-02989-8.
Collapse
Affiliation(s)
- Wenjuan Xu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, 230022, Anhui, China.,Anhui Province Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China.,NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Caiyun Wu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, 230022, Anhui, China.,Anhui Province Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China.,NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Xiaoqian Zhu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, 230022, Anhui, China.,Anhui Province Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China.,NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Jingjing Wu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, 230022, Anhui, China.,Anhui Province Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China.,NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Zhiguo Zhang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, 230022, Anhui, China.,Anhui Province Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China.,NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Zhaolian Wei
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, 230022, Anhui, China.,Anhui Province Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China.,NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Yunxia Cao
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, 230022, Anhui, China.,Anhui Province Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China.,NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Ping Zhou
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, 230022, Anhui, China. .,Anhui Province Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China. .,NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China.
| | - Jianye Wang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, 230022, Anhui, China. .,Anhui Province Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China. .,NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China.
| |
Collapse
|
37
|
Grismaldo A, Sobrevia L, Morales L. Role of platelet-derived growth factor c on endothelial dysfunction in cardiovascular diseases. Biochim Biophys Acta Gen Subj 2022; 1866:130188. [PMID: 35691459 DOI: 10.1016/j.bbagen.2022.130188] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 06/02/2022] [Accepted: 06/05/2022] [Indexed: 01/01/2023]
Abstract
Loss of endothelial function is a common feature to all cardiovascular diseases (CVDs). One of the risk factors associated with the development of CVDs is the hyperglycaemia that occurs in patients with metabolic disorders such as Type 1 and Type 2 diabetes mellitus. Hyperglycaemia causes endothelial dysfunction through increased production of reactive oxygen species (ROS) from different cellular sources leading to oxidative stress. Vascular endothelial growth factor (VEGF) is essential in the stimulation and maintenance of endothelial functional aspects and, although it can mitigate the impact of ROS, VEGF-mediated signalling is partially inhibited in diabetes mellitus. The search for therapeutic strategies that preserve, protect and improve the functions of the endothelium is of great relevance in the investigation of CVDs associated with hyperglycaemia. Platelet-derived growth factor C (PDGF-C) is a peptide with angiogenic properties, independent of VEGF, that stimulates angiogenesis and revascularization of ischemic tissue. In a diabetic mouse model, PDGF-C stimulates mature endothelial cell migration, angiogenesis, endothelial progenitor cell mobilization, and increased neovascularization, and protects blood vessels in a retinal degeneration model activating anti-apoptosis and proliferation signalling pathways in endothelial cells. This review summarizes the information on the damage that high d-glucose causes on endothelial function and the beneficial effects that PDGF-CC could exert in this condition.
Collapse
Affiliation(s)
- Adriana Grismaldo
- Experimental and Computational Biochemistry Group, Faculty of Sciences, Nutrition and Biochemistry Department, Pontificia Universidad Javeriana, Bogotá, DC, Colombia; Cellular and Molecular Physiology Laboratory, Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile.
| | - Luis Sobrevia
- Cellular and Molecular Physiology Laboratory, Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; Department of Physiology, Faculty of Pharmacy, Universidad de Sevilla, Seville E-41012, Spain; Medical School (Faculty of Medicine), Sao Paulo State University (UNESP), Brazil; University of Queensland Centre for Clinical Research (UQCCR), Faculty of Medicine and Biomedical Sciences, University of Queensland, Herston, QLD 4029, Australia; Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, 9713GZ Groningen, the Netherlands; Tecnologico de Monterrey, Eutra, The Institute for Obesity Research (IOR), School of Medicine and Health Sciences, Monterrey, Nuevo León. Mexico..
| | - Ludis Morales
- Experimental and Computational Biochemistry Group, Faculty of Sciences, Nutrition and Biochemistry Department, Pontificia Universidad Javeriana, Bogotá, DC, Colombia.
| |
Collapse
|
38
|
Wang Y, Du J, Liu Y, Yang S, Wang Q. microRNA-301a-3p is a potential biomarker in venous ulcers vein and gets involved in endothelial cell dysfunction. Bioengineered 2022; 13:14138-14158. [PMID: 35734851 PMCID: PMC9342147 DOI: 10.1080/21655979.2022.2083821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 05/23/2022] [Accepted: 05/24/2022] [Indexed: 11/04/2022] Open
Abstract
Venous ulcer is a common contributor to chronic venous insufficiency (CVI) of lower limbs, which seriously affects the life quality of patients. In this study, we researched the expression characteristics of microRNA-301a-3p (miR-301a-3p) in patients with CVI and investigated the impact of miR-301a-3p on the dysfunction of human umbilical vein endothelial cells (HUVECs). The plasma level of miR-301a-3p in normal controls, patients with varicose great saphenous vein, and patients with the venous ulcer of lower limbs were measured. We adopted Interleukin-1β (IL-1β), H2O2, and oxygen and glucose deprivation (OGD) to induce endothelial cell injury in vitro. In this way, we evaluated the influence of miR-301a-3p on HUVEC viability, apoptosis, inflammatory response, and oxidative stress. Our data showed that miR-301a-3p was substantially overexpressed in patients with lower limb venous ulcers. The viability of HUVECs decreased, and miR-301a-3p was up-regulated after IL-1β, H2O2, and OGD treatment. miR-301a-3p inhibition greatly ameliorated the dysfunction and cell damage of HUVECs, promoted IGF1/PI3K/Akt/PPARγ, and down-regulated NF-κB/MMPs. The phosphatidylinositol 3-kinase (PI3K) inhibitor (LY294002) or the peroxisome proliferator-activated receptor-γ (PPARγ) inhibitor (GW9661) reversed the anti-inflammatory, antioxidant, and anti-apoptotic effects mediated by miR-301a-3p down-regulation. The nuclear factor-κB (NF-κB) inhibitor lessened cell injury mediated by miR-301a-3p overexpression. In terms of the mechanism, miR-301a-3p targeted the 3'UTR of Insulin-like growth factor-1 (IGF1) and repressed the profile of IGF1. Thus, miR-301a-3p mediates venous endothelial cell damage by targeting IGF1 and regulating the IGF1/PI3K/Akt/PPARγ/NF-κB/MMPs pathway.
Collapse
Affiliation(s)
- Ying Wang
- Department of Vascular Surgery, The Second Affiliated Hospital of Qiqihar Medical College, Qiqihar, Heilongjiang, China
| | - Jingchen Du
- Department of Vascular Surgery, Heilongjiang Provincial Hospital, Harbin, Heilongjiang, China
| | - Yu Liu
- Department of Vascular Surgery, The First Hospital of Qiqihar, Qiqihar, Heilongjiang, China
| | - Shuhui Yang
- Department of Vascular Surgery, Heilongjiang Provincial Hospital, Harbin, Heilongjiang, China
| | - Qingshan Wang
- Department of Vascular Surgery, Heilongjiang Provincial Hospital, Harbin, Heilongjiang, China
- Department of Vascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
39
|
Zhang C, Wu S, Chen E, Yu L, Wang J, Wu M. ALX1-transcribed LncRNA AC132217.4 promotes osteogenesis and bone healing via IGF-AKT signaling in mesenchymal stem cells. Cell Mol Life Sci 2022; 79:328. [PMID: 35639207 PMCID: PMC11073114 DOI: 10.1007/s00018-022-04338-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 04/23/2022] [Accepted: 04/29/2022] [Indexed: 11/03/2022]
Abstract
The osteogenic potential of bone marrow mesenchymal stem cells (BMSCs) is critical for bone formation and regeneration. A high non-/delayed-union rate of fracture healing still occurs in specific populations, implying an urgent need to discover novel targets for promoting osteogenesis and bone regeneration. Long non-coding (lnc)RNAs are emerging regulators of multiple physiological processes, including osteogenesis. Based on differential expression analysis of RNA sequencing data, we found that lncRNA AC132217.4, a 3'UTR-overlapping lncRNA of insulin growth factor 2 (IGF2), was highly induced during osteogenic differentiation of BMSCs. Afterward, both gain-of-function and loss-of-function experiments proved that AC132217.4 promotes osteoblast development from BMSCs. As for its molecular mechanism, we found that AC132217.4 binds with IGF2 mRNA to regulate its expression and downstream AKT activation to control osteoblast maturation and function. Furthermore, we identified two splicing factors, splicing component 35 KDa (SC35) and heterogeneous nuclear ribonucleoprotein A1 (HNRNPA1), which regulate the biogenesis of AC132217.4 at the post-transcriptional level. We also identified a transcription factor, ALX1, which regulates AC132217.7 expression at the transcriptional level to promote osteogenesis. Importantly, in-vivo over-expression of AC132217.4 essentially promotes the bone healing process in a murine tibial drill-hole model. Our study demonstrates that lncRNA AC132217.4 is a novel anabolic regulator of BMSC osteogenesis and could be a plausible therapeutic target for improving bone regeneration.
Collapse
Affiliation(s)
- Cui Zhang
- Department of Cell and Developmental Biology, College of Life Sciences, Zhejiang University, Hangzhou, China
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shali Wu
- Department of Cell and Developmental Biology, College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Erman Chen
- Department of Orthopedics, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Luyang Yu
- Department of Cell and Developmental Biology, College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Jinfu Wang
- Department of Cell and Developmental Biology, College of Life Sciences, Zhejiang University, Hangzhou, China.
| | - Mengrui Wu
- Department of Cell and Developmental Biology, College of Life Sciences, Zhejiang University, Hangzhou, China.
| |
Collapse
|
40
|
AT2R activation increases in vitro angiogenesis in pregnant human uterine artery endothelial cells. PLoS One 2022; 17:e0267826. [PMID: 35486619 PMCID: PMC9053770 DOI: 10.1371/journal.pone.0267826] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 04/15/2022] [Indexed: 12/04/2022] Open
Abstract
Angiogenesis is vital during pregnancy for remodeling and enhancing vasodilation of maternal uterine arteries, and increasing uterine blood flow. Abnormal angiogenesis is associated with decreased uteroplacental blood flow and development of pregnancy disorders such as gestational hypertension, preeclampsia, fetal growth restriction, preterm delivery, stillbirth, and miscarriage. The mechanisms that contribute to normal angiogenesis remain obscure. Our previous studies demonstrated that expression of the angiotensin type 2 receptor (AT2R) is increased while the angiotensin type 1 receptor (AT1R) is unchanged in the endothelium of uterine arteries, and that AT2R-mediated pregnancy adaptation facilitates enhanced vasodilation and uterine arterial blood flow. However, the role of AT2R in regulating angiogenesis during pregnancy has never been studied. This study examines whether or not AT2R activation induces angiogenesis and, if so, what mechanisms are involved. To this end, we used primary human uterine artery endothelial cells (hUAECs) isolated from pregnant and nonpregnant women undergoing hysterectomy. The present study shows that Compound 21, a selective AT2R agonist, induced proliferation of pregnant-hUAECs, but not nonpregnant-hUAECs, in a concentration-dependent manner, and that this C21-induced mitogenic effect was blocked by PD123319, a selective AT2R antagonist. The mitogenic effects induced by C21 were inhibited by blocking JNK—but not ERK, PI3K, and p38—signaling pathways. In addition, C21 concentration dependently increased cell migration and capillary-like tube formation in pregnant-hUAECs. The membrane-based antibody array showed that C21 increased expression of multiple angiogenic proteins, including EGF, bFGF, leptin, PLGF, IGF-1, and angiopoietins. Our qPCR analysis demonstrates that C21-induced increase in expression of these angiogenic proteins correlates with a proportional increase in mRNA expression, indicating that AT2R activates angiogenic proteins at the transcriptional level. In summary, the present study shows that AT2R activation induces angiogenesis of hUAECs in a pregnancy-specific manner through JNK-mediated pathways with associated transcriptional upregulation of multiple proangiogenic proteins.
Collapse
|
41
|
Betzmann D, Döring M, Blumenstock G, Erdmann F, Grabow D, Lang P, Binder G. Impact of serum insulin-like growth factor-1 on HSCT outcome in pediatric cancer patients. Transplant Cell Ther 2022; 28:355.e1-355.e9. [PMID: 35405367 DOI: 10.1016/j.jtct.2022.03.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 12/20/2021] [Accepted: 03/30/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Hematopoietic stem cell transplantation (HSCT) is associated with severe medical complications and variable outcome depending on the recipient's disease stage and health condition. Biomarkers predicting outcome may have therapeutic relevance in pediatric cancer care. Insulin like growth factor 1 (IGF 1) is a mitogenic and anabolic peptide hormone that is expressed in almost all tissues. This hormone is the major growth factor in childhood. As IGF 1 is decreased in conditions that cause catabolic metabolism, it may reflect the degree of physical robustness of the patient and serve as predictive biomarker for transplant outcome. OBJECTIVES AND STUDY DESIGN We evaluated the impact of pre-transplant serum-IGF 1 on both survival and adverse events in 587 pediatric cancer patients, who underwent autologous or allogeneic HSCT between 1987 and 2014 at the University Children's Hospital Tübingen, Germany. Survival probabilities of the entire cohort and of defined subgroups according to pre-transplant serum-IGF-1 were estimated using the Kaplan-Meier method. RESULTS Mean pre-transplant IGF 1 (n = 498) was low: -1.67 SDS (SD, 1.54). Completeness of follow-up three and ten years post HSCT was 96 % and 83 %, respectively. The ten-year overall survival was 44.8 % (95 % confidence interval [CI], 40.6-48.9). With decreasing IGF-1 SDS, there was a significant increase of transplant-related mortality (p = 0.027), sinusoidal obstruction syndrome (quartiles 4 to 1: 3; 1; 12; 12%; p < 0.001) and thrombotic microangiopathy (quartiles 4 to 1: 0: 0: 2; 5%; p = 0.004). IGF 1 decile 1 showed a significantly poorer outcome (p=0.042) with lower median (12 versus 68 months) and ten-year overall survival (37 % versus 52 %) when compared to decile 2-10. CONCLUSIONS This retrospective study suggests pre-transplant serum-IGF 1 as a predictor of survival and selected vascular adverse events that may have diagnostic and therapeutic relevance in pediatric cancer care.
Collapse
Affiliation(s)
- Deborah Betzmann
- University Children's Hospital Tübingen, Hoppe-Seyler-Str.1, 72076 Tübingen, Germany
| | - Michaela Döring
- University Children's Hospital Tübingen, Hoppe-Seyler-Str.1, 72076 Tübingen, Germany
| | - Gunnar Blumenstock
- Department of Clinical Epidemiology and Applied Biometry, University of Tübingen, Silcherstraße 5, 72076 Tübingen, Germany
| | - Friederike Erdmann
- Division of Childhood Cancer Epidemiology, German Childhood Cancer Registry, Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI) University Medical Center of the Johannes Gutenberg University Mainz, Mainz
| | - Desiree Grabow
- Division of Childhood Cancer Epidemiology, German Childhood Cancer Registry, Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI) University Medical Center of the Johannes Gutenberg University Mainz, Mainz
| | - Peter Lang
- University Children's Hospital Tübingen, Hoppe-Seyler-Str.1, 72076 Tübingen, Germany
| | - Gerhard Binder
- University Children's Hospital Tübingen, Hoppe-Seyler-Str.1, 72076 Tübingen, Germany.
| |
Collapse
|
42
|
Yang Z, Liang X, Yang L. Circular RNA circ_0001445 alleviates the ox-LDL-induced endothelial injury in human primary aortic endothelial cells through regulating ABCG1 via acting as a sponge of miR-208b-5p. Gan To Kagaku Ryoho 2022; 70:779-792. [PMID: 35391605 DOI: 10.1007/s11748-022-01799-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 03/01/2022] [Indexed: 02/08/2023]
Abstract
BACKGROUND Coronary artery disease (CAD) originates from the blockage of the inner walls of the coronary arteries due to a plaque buildup. Circular RNA (circRNA) circ_0001445 has been reported to be downregulated in patients with a higher coronary atherosclerotic burden. This study is designed to explore the role and mechanism of circ_0001445 on the oxidized low-density lipoprotein (ox-LDL)-induced endothelial cell damage. METHODS Circ_0001445, microRNA-208b-5p (miR-208b-5p), and ATP-binding cassette sub-family G member 1 (ABCG1) levels were detected by real-time quantitative polymerase chain reaction (RT-qPCR). Inflammatory cytokines levels, cell viability, proliferation, migration were detected by Enzyme-linked immunosorbent assay (ELISA) kits, Cell Counting Kit-8 (CCK-8), 5-ethynyl-2'-deoxyuridine (EdU), and transwell assays, respectively. Protein levels were determined by western blot assay. The binding between miR-208b-5p and circ_0001445 or ABCG1 was predicted by circBank or TargetScan, and then verified by a dual-luciferase reporter, RNA Immunoprecipitation (RIP), and RNA pull-down assays. RESULTS Circ_0001445 and ABCG1 were decreased, and miR-208b-5p was increased in CAD patients and ox-LDL-treated HAECs. Also, circ_0001445 overexpression could weaken ox-LDL-triggered HAEC injury by boosting proliferation, migration, and repressing inflammation and extracellular matrix (ECM). Mechanically, circ_0001445 directly targeted miR-208b-5p. Furthermore, miR-208b-5p mediated the modulation of circ_0001445 in ox-LDL-induced HAEC injury. ABCG1 acted as a direct target of miR-208b-5p, and the downregulation of miR-208b-5p relieved ox-LDL-induced HAEC damage by interacting with ABCG1. Additionally, circ_0001445 regulated ABCG1 expression by sponging miR-208b-5p. CONCLUSION Circ_0001445 could abate ox-LDL-mediated HAEC damage by the miR-208b-5p/ABCG1 axis, providing a novel insight into the pathogenesis and treatment of CAD.
Collapse
Affiliation(s)
- Zhihua Yang
- Department of Cardiovascular Medicine, 920 Hospital of Joint Logistics Support Force, No. 212, Daguan Road, Xishan District, Kunming, Yunnan Province, China
| | - Xing Liang
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, 288 Tianwen Avenue, Nan'an District, Chongqing, China
| | - Lixia Yang
- Department of Cardiovascular Medicine, 920 Hospital of Joint Logistics Support Force, No. 212, Daguan Road, Xishan District, Kunming, Yunnan Province, China.
| |
Collapse
|
43
|
Nies MK, Yang J, Griffiths M, Damico R, Zhu J, Vaydia D, Fu Z, Brandal S, Austin ED, Ivy DD, Hassoun PM, Van Eyk JE, Everett AD. Proteomics discovery of pulmonary hypertension biomarkers: Insulin-like growth factor binding proteins are associated with disease severity. Pulm Circ 2022; 12:e12039. [PMID: 35514776 PMCID: PMC9063962 DOI: 10.1002/pul2.12039] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 12/09/2021] [Accepted: 12/21/2021] [Indexed: 11/16/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive disease characterized by sustained elevations of pulmonary artery pressure. To date, we lack circulating, diagnostic, and prognostic markers that correlate to clinical and functional parameters. In this study, we performed mass spectrometry-based proteomics analysis to identify circulating biomarkers of PAH. Plasma samples from patients with idiopathic pulmonary arterial hypertension (IPAH, N = 9) and matched normal controls (N = 9) were digested with trypsin and analyzed using data-dependent acquisition on an Orbitrap mass spectrometer. A total of 826 (false discovery rate [FDR] 0.047) and 461 (FDR 0.087) proteins were identified across all plasma samples obtained from IPAH and control subjects, respectively. Of these, 153 proteins showed >2 folds change (p < 0.05) between groups. Circulating levels of carbonic anhydrase 2 (CA2), plasma kallikrein (KLKB1), and the insulin-like growth factor binding proteins (IGFBP1-7) were quantified by immunoassay in an independent verification cohort (N = 36 PAH and N = 35 controls). CA2 and KLKB1 were significantly different in PAH versus control but were not associated with any functional or hemodynamic measurements. Whereas, IGFBP1 and 2 were associated with higher pulmonary vascular resistance, IGFBP2, 4, and 7 with decreased 6-min walk distance (6MWD), and IGFBP1, 2, 4, and 7 with worse survival. This plasma proteomic discovery analysis suggests the IGF axis may serve as important new biomarkers for PAH and play an important role in PAH pathogenesis.
Collapse
Affiliation(s)
- Melanie K. Nies
- Department of Pediatrics, Division of CardiologyJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Jun Yang
- Department of Pediatrics, Division of CardiologyJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Megan Griffiths
- Department of Pediatrics, Division of CardiologyJohns Hopkins UniversityBaltimoreMarylandUSA
- Department of Pediatrics, Division of Pediatric CardiologyColumbia UniversityNew YorkNew YorkUSA
| | - Rachel Damico
- Department of Medicine, Division of Pulmonary and Critical Care MedicineJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Jie Zhu
- Department of Pediatrics, Division of CardiologyJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Dhananjay Vaydia
- Department of Medicine, Division of Pulmonary and Critical Care MedicineJohns Hopkins UniversityBaltimoreMarylandUSA
- Department of Epidemiology, School of Public HealthJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Zongming Fu
- Department of Pediatrics, Division of HematologyJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Stephanie Brandal
- Department of Pediatrics, Division of CardiologyJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Eric D. Austin
- Department of Pediatrics, Division of Allergy, Immunology, and Pulmonary MedicineVanderbilt UniversityNashvilleTennesseeUSA
| | - Dunbar D. Ivy
- Department of Pediatric CardiologyChildren's Hospital ColoradoAuroraColoradoUSA
| | - Paul M. Hassoun
- Department of Medicine, Division of Pulmonary and Critical Care MedicineJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Jennifer E. Van Eyk
- Department of Internal Medicine, Division of CardiologyJohns Hopkins UniversityBaltimoreMarylandUSA
- Advanced Clinical Biosystems Research InstituteCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | - Allen D. Everett
- Department of Pediatrics, Division of CardiologyJohns Hopkins UniversityBaltimoreMarylandUSA
| |
Collapse
|
44
|
Uimari O, Subramaniam KS, Vollenhoven B, Tapmeier TT. Uterine Fibroids (Leiomyomata) and Heavy Menstrual Bleeding. FRONTIERS IN REPRODUCTIVE HEALTH 2022; 4:818243. [PMID: 36303616 PMCID: PMC9580818 DOI: 10.3389/frph.2022.818243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 02/09/2022] [Indexed: 11/13/2022] Open
Abstract
Uterine Fibroids, or leiomyomata, affect millions of women world-wide, with a high incidence of 75% within women of reproductive age. In ~30% of patients, uterine fibroids cause menorrhagia, or heavy menstrual bleeding, and more than half of the patients experience symptoms such as heavy menstrual bleeding, pelvic pain, or infertility. Treatment is symptomatic with limited options including hysterectomy as the most radical solution. The genetic foundations of uterine fibroid growth have been traced to somatic driver mutations (MED12, HMGA2, FH -/-, and COL4A5-A6). These also lead to downstream expression of angiogenic factors including IGF-1 and IGF-2, as opposed to the VEGF-driven mechanism found in the angiogenesis of hypoxic tumors. The resulting vasculature supplying the fibroid with nutrients and oxygen is highly irregular. Of particular interest is the formation of a pseudocapsule around intramural fibroids, a unique structure within tumor angiogenesis. These aberrations in vascular architecture and network could explain the heavy menstrual bleeding observed. However, other theories have been proposed such as venous trunks, or venous lakes caused by the blocking of normal blood flow by uterine fibroids, or the increased local action of vasoactive growth factors. Here, we review and discuss the evidence for the various hypotheses proposed.
Collapse
Affiliation(s)
- Outi Uimari
- Department of Obstetrics and Gynecology, Oulu University, Oulu, Finland
- Research Unit for Pediatrics, Pediatric Neurology, Pediatric Surgery, Child Psychiatry, Dermatology, Clinical Genetics, Obstetrics and Gynecology, Otorhinolaryngology and Ophthalmology (PEDEGO) Research Unit and Medical Research Center Oulu, Oulu University Hospital, Oulu, Finland
| | - Kavita S. Subramaniam
- St John's Institute of Dermatology, King's College London, Guy's Hospital, London, United Kingdom
- Endometriosis CaRe Centre, Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Beverley Vollenhoven
- Women's and Newborn Program, Monash Health, Clayton, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| | - Thomas T. Tapmeier
- Endometriosis CaRe Centre, Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| |
Collapse
|
45
|
Rosano S, Parab S, Noghero A, Corà D, Bussolino F. Long Non-Coding RNA LINC02802 Regulates In Vitro Sprouting Angiogenesis by Sponging microRNA-486-5p. Int J Mol Sci 2022; 23:ijms23031653. [PMID: 35163581 PMCID: PMC8836176 DOI: 10.3390/ijms23031653] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 01/28/2022] [Accepted: 01/28/2022] [Indexed: 12/10/2022] Open
Abstract
In the last several years, accumulating evidence indicates that noncoding RNAs, especially long-noncoding RNAs (lncRNAs) and microRNAs, play essential roles in regulating angiogenesis. However, the contribution of lncRNA-mediated competing-endogenous RNA (ceRNA) activity in the control of capillary sprouting from the pre-existing ones has not been described so far. Here, by exploiting the transcriptomic profile of VEGF-A-activated endothelial cells in a consolidate three-dimensional culture system, we identified a list of lncRNAs whose expression was modified during the sprouting process. By crossing the lncRNAs with a higher expression level and the highest fold change value between unstimulated and VEGF-A-stimulated endothelial cells, we identified the unknown LINC02802 as the best candidate to take part in sprouting regulation. LINC02802 was upregulated after VEGF-A stimulation and its knockdown resulted in a significant reduction in sprouting activity. Mechanistically, we demonstrated that LINC02802 acts as a ceRNA in the post-transcriptional regulation of Mastermind-like-3 (MAML3) gene expression through a competitive binding with miR-486-5p. Taken together, these results suggest that LINC02802 plays a critical role in preventing the miR-486-5p anti-angiogenic effect and that this inhibitory effect results from the reduction in MAML3 expression.
Collapse
Affiliation(s)
- Stefania Rosano
- Department of Oncology, University of Torino, 10124 Orbassano, Italy; (S.P.); (F.B.)
- Candiolo Cancer Institute, IRCCS-FPO, 10060 Candiolo, Italy
- Correspondence:
| | - Sushant Parab
- Department of Oncology, University of Torino, 10124 Orbassano, Italy; (S.P.); (F.B.)
- Candiolo Cancer Institute, IRCCS-FPO, 10060 Candiolo, Italy
| | - Alessio Noghero
- Lovelace Biomedical Research Institute, Albuquerque, NM 87108, USA;
| | - Davide Corà
- Department of Translational Medicine, Piemonte Orientale University, 28100 Novara, Italy;
- Center for Translational Research on Autoimmune and Allergic Diseases—CAAD, 28100 Novara, Italy
| | - Federico Bussolino
- Department of Oncology, University of Torino, 10124 Orbassano, Italy; (S.P.); (F.B.)
- Candiolo Cancer Institute, IRCCS-FPO, 10060 Candiolo, Italy
| |
Collapse
|
46
|
Sandovici I, Georgopoulou A, Pérez-García V, Hufnagel A, López-Tello J, Lam BYH, Schiefer SN, Gaudreau C, Santos F, Hoelle K, Yeo GSH, Burling K, Reiterer M, Fowden AL, Burton GJ, Branco CM, Sferruzzi-Perri AN, Constância M. The imprinted Igf2-Igf2r axis is critical for matching placental microvasculature expansion to fetal growth. Dev Cell 2022; 57:63-79.e8. [PMID: 34963058 PMCID: PMC8751640 DOI: 10.1016/j.devcel.2021.12.005] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 09/30/2021] [Accepted: 12/03/2021] [Indexed: 11/21/2022]
Abstract
In all eutherian mammals, growth of the fetus is dependent upon a functional placenta, but whether and how the latter adapts to putative fetal signals is currently unknown. Here, we demonstrate, through fetal, endothelial, hematopoietic, and trophoblast-specific genetic manipulations in the mouse, that endothelial and fetus-derived IGF2 is required for the continuous expansion of the feto-placental microvasculature in late pregnancy. The angiocrine effects of IGF2 on placental microvasculature expansion are mediated, in part, through IGF2R and angiopoietin-Tie2/TEK signaling. Additionally, IGF2 exerts IGF2R-ERK1/2-dependent pro-proliferative and angiogenic effects on primary feto-placental endothelial cells ex vivo. Endothelial and fetus-derived IGF2 also plays an important role in trophoblast morphogenesis, acting through Gcm1 and Synb. Thus, our study reveals a direct role for the imprinted Igf2-Igf2r axis on matching placental development to fetal growth and establishes the principle that hormone-like signals from the fetus play important roles in controlling placental microvasculature and trophoblast morphogenesis.
Collapse
Affiliation(s)
- Ionel Sandovici
- Department of Obstetrics and Gynaecology and National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge, Cambridge CB2 0SW, UK; Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, Cambridge CB2 0QQ, UK; Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK.
| | - Aikaterini Georgopoulou
- Department of Obstetrics and Gynaecology and National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge, Cambridge CB2 0SW, UK; Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| | - Vicente Pérez-García
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK; Epigenetics Programme, Babraham Institute, Cambridge CB22 3AT, UK; Centro de Investigación Príncipe Felipe, Eduardo Primo Yúfera, 46012 Valencia, Spain
| | - Antonia Hufnagel
- Department of Obstetrics and Gynaecology and National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge, Cambridge CB2 0SW, UK
| | - Jorge López-Tello
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| | - Brian Y H Lam
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Samira N Schiefer
- Department of Obstetrics and Gynaecology and National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge, Cambridge CB2 0SW, UK
| | - Chelsea Gaudreau
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Fátima Santos
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK; Epigenetics Programme, Babraham Institute, Cambridge CB22 3AT, UK
| | - Katharina Hoelle
- Department of Obstetrics and Gynaecology and National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge, Cambridge CB2 0SW, UK
| | - Giles S H Yeo
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Keith Burling
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Moritz Reiterer
- Physiological Laboratory, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK; Center for Cancer Research and Cell Biology, Queen's University Belfast, Belfast BT9 7AE, UK
| | - Abigail L Fowden
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| | - Graham J Burton
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| | - Cristina M Branco
- Physiological Laboratory, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK; Center for Cancer Research and Cell Biology, Queen's University Belfast, Belfast BT9 7AE, UK
| | - Amanda N Sferruzzi-Perri
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| | - Miguel Constância
- Department of Obstetrics and Gynaecology and National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge, Cambridge CB2 0SW, UK; Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, Cambridge CB2 0QQ, UK; Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK.
| |
Collapse
|
47
|
Aggarwal C, Saini K, Reddy ES, Singla M, Nayak K, Chawla YM, Maheshwari D, Singh P, Sharma P, Bhatnagar P, Kumar S, Gottimukkala K, Panda H, Gunisetty S, Davis CW, Kissick HT, Kabra SK, Lodha R, Medigeshi GR, Ahmed R, Murali-Krishna K, Chandele A. Immunophenotyping and Transcriptional Profiling of Human Plasmablasts in Dengue. J Virol 2021; 95:e0061021. [PMID: 34523972 PMCID: PMC8577383 DOI: 10.1128/jvi.00610-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 09/11/2021] [Indexed: 12/07/2022] Open
Abstract
Plasmablasts represent a specialized class of antibody-secreting effector B cells that transiently appear in blood circulation following infection or vaccination. The expansion of these cells generally tends to be massive in patients with systemic infections such as dengue or Ebola that cause hemorrhagic fever. To gain a detailed understanding of human plasmablast responses beyond antibody expression, here, we performed immunophenotyping and RNA sequencing (RNA-seq) analysis of the plasmablasts from dengue febrile children in India. We found that plasmablasts expressed several adhesion molecules and chemokines or chemokine receptors that are involved in endothelial interactions or homing to inflamed tissues, including skin, mucosa, and intestine, and upregulated the expression of several cytokine genes that are involved in leukocyte extravasation and angiogenesis. These plasmablasts also upregulated the expression of receptors for several B-cell prosurvival cytokines that are known to be induced robustly in systemic viral infections such as dengue, some of which generally tend to be relatively higher in patients manifesting hemorrhage and/or shock than in patients with mild febrile infection. These findings improve our understanding of human plasmablast responses during the acute febrile phase of systemic dengue infection. IMPORTANCE Dengue is globally spreading, with over 100 million clinical cases annually, with symptoms ranging from mild self-limiting febrile illness to more severe and sometimes life-threatening dengue hemorrhagic fever or shock, especially among children. The pathophysiology of dengue is complex and remains poorly understood despite many advances indicating a key role for antibody-dependent enhancement of infection. While serum antibodies have been extensively studied, the characteristics of the early cellular factories responsible for antibody production, i.e., plasmablasts, are only beginning to emerge. This study provides a comprehensive understanding of the transcriptional profiles of human plasmablasts from dengue patients.
Collapse
Affiliation(s)
- Charu Aggarwal
- ICGEB-Emory Vaccine Center, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Keshav Saini
- ICGEB-Emory Vaccine Center, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Elluri Seetharami Reddy
- ICGEB-Emory Vaccine Center, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
- Kusuma School of Biological Sciences, Indian Institute of Technology, New Delhi, India
| | - Mohit Singla
- Department of Pediatrics, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| | - Kaustuv Nayak
- ICGEB-Emory Vaccine Center, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Yadya M. Chawla
- ICGEB-Emory Vaccine Center, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Deepti Maheshwari
- ICGEB-Emory Vaccine Center, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Prabhat Singh
- ICGEB-Emory Vaccine Center, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Pragati Sharma
- ICGEB-Emory Vaccine Center, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
- Department of Biotechnology, School of Chemical and Life Sciences, New Delhi, India
| | - Priya Bhatnagar
- ICGEB-Emory Vaccine Center, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
- TERI School of Advanced Studies, New Delhi, India
| | - Sanjeev Kumar
- ICGEB-Emory Vaccine Center, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Kamalvishnu Gottimukkala
- ICGEB-Emory Vaccine Center, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Harekrushna Panda
- ICGEB-Emory Vaccine Center, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Sivaram Gunisetty
- Department of Pediatrics, Division of Infectious Disease, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Carl W. Davis
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Haydn Thomas Kissick
- Department of Microbiology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Sushil Kumar Kabra
- Department of Pediatrics, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| | - Rakesh Lodha
- Department of Pediatrics, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| | | | - Rafi Ahmed
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia, USA
- Department of Microbiology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Kaja Murali-Krishna
- ICGEB-Emory Vaccine Center, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
- Department of Pediatrics, Division of Infectious Disease, Emory University School of Medicine, Atlanta, Georgia, USA
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Anmol Chandele
- ICGEB-Emory Vaccine Center, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| |
Collapse
|
48
|
Ma J, Zhang Z, Wang Y, Shen H. Investigation of miR-126-3p loaded on adipose stem cell-derived exosomes for wound healing of full-thickness skin defects. Exp Dermatol 2021; 31:362-374. [PMID: 34694648 DOI: 10.1111/exd.14480] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 09/01/2021] [Accepted: 10/16/2021] [Indexed: 11/29/2022]
Abstract
OBJECTIVE To investigate the function of miR-126-3p loaded on adipose stem cell (ADSC)-derived exosomes (ADSC-Exos) in wound healing of full-thickness skin defects. METHODS ADSCs transfected with miR-126-3p mimic, miR-126-3p inhibitor or pcDNA3.1-PIK3R2, or PKH26-marked ADSC-Exos were cultured with fibroblasts or human umbilical vein endothelial cells (HUVECs). The proliferation and migration rates of fibroblasts and angiogenesis of HUVECs were measured. Rats with full-thickness skin defects were injected with ADSC-Exos or exosomes extracted from ADSCs transfected with miR-126-3p inhibitor and the wound healing rates were measured. The wound bed, collagen deposition and angiogenesis in injured rats were assessed. RESULTS ADSC-Exos could be ingested by fibroblasts and HUVECs. Co-incubation with ADSCs or ADSC-Exos promoted the proliferation and migration of fibroblasts and angiogenesis of HUVECs, which was further enhanced by miR-126-3p overexpression. Inhibition of ADSC-Exos or miR-126-3p or PIK3R2 overexpression suppressed the proliferation and migration of fibroblasts and angiogenesis of HUVECs. ADSC-derived exosomal miR-126-3p increased wound healing rate, collagen deposition and newly formed vessels in injured rats. CONCLUSION ADSC-derived exosomal miR-126-3p promotes wound healing of full-thickness skin defects by targeting PIK3R2.
Collapse
Affiliation(s)
- Jie Ma
- Department of Plastic Surgery, Shanghai General Hospital, Shanghai, 201620, P.R. China
| | - Zhaofeng Zhang
- Department of Plastic Surgery, Shanghai General Hospital, Shanghai, 201620, P.R. China
| | - Yinmin Wang
- Department of Plastic Surgery, Shanghai General Hospital, Shanghai, 201620, P.R. China
| | - Hua Shen
- Department of Plastic Surgery, Shanghai General Hospital, Shanghai, 201620, P.R. China
| |
Collapse
|
49
|
Ishii T, Warabi E, Mann GE. Mechanisms underlying unidirectional laminar shear stress-mediated Nrf2 activation in endothelial cells: Amplification of low shear stress signaling by primary cilia. Redox Biol 2021; 46:102103. [PMID: 34425388 PMCID: PMC8379703 DOI: 10.1016/j.redox.2021.102103] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/07/2021] [Accepted: 08/12/2021] [Indexed: 12/14/2022] Open
Abstract
Endothelial cells are sensitive to mechanical stress and respond differently to oscillatory flow versus unidirectional flow. This review highlights the mechanisms by which a wide range of unidirectional laminar shear stress induces activation of the redox sensitive antioxidant transcription factor nuclear factor-E2-related factor 2 (Nrf2) in cultured endothelial cells. We propose that fibroblast growth factor-2 (FGF-2), brain-derived neurotrophic factor (BDNF) and 15-Deoxy-Δ12,14-prostaglandin J2 (15d-PGJ2) are potential Nrf2 activators induced by laminar shear stress. Shear stress-dependent secretion of FGF-2 and its receptor-mediated signaling is tightly controlled, requiring neutrophil elastase released by shear stress, αvβ3 integrin and the cell surface glycocalyx. We speculate that primary cilia respond to low laminar shear stress (<10 dyn/cm2), resulting in secretion of insulin-like growth factor 1 (IGF-1), which facilitates αvβ3 integrin-dependent FGF-2 secretion. Shear stress induces generation of heparan-binding epidermal growth factor-like growth factor (HB-EGF), which contributes to FGF-2 secretion and gene expression. Furthermore, HB-EGF signaling modulates FGF-2-mediated NADPH oxidase 1 activation that favors casein kinase 2 (CK2)-mediated phosphorylation/activation of Nrf2 associated with caveolin 1 in caveolae. Higher shear stress (>15 dyn/cm2) induces vesicular exocytosis of BDNF from endothelial cells, and we propose that BDNF via the p75NTR receptor could induce CK2-mediated Nrf2 activation. Unidirectional laminar shear stress upregulates gene expression of FGF-2 and BDNF and generation of 15d-PGJ2, which cooperate in sustaining Nrf2 activation to protect endothelial cells against oxidative damage.
Collapse
Affiliation(s)
- Tetsuro Ishii
- School of Medicine, University of Tsukuba, Tsukuba, Ibaraki, 305-8577, Japan.
| | - Eiji Warabi
- School of Medicine, University of Tsukuba, Tsukuba, Ibaraki, 305-8577, Japan.
| | - Giovanni E Mann
- King's British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine & Sciences, King's College London, 150 Stamford Street, London, SE1 9NH, UK.
| |
Collapse
|
50
|
Cerebrovascular alterations in NAFLD: Is it increasing our risk of Alzheimer's disease? Anal Biochem 2021; 636:114387. [PMID: 34537182 DOI: 10.1016/j.ab.2021.114387] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/27/2021] [Accepted: 09/15/2021] [Indexed: 02/07/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a multisystem disease, which has been classified as an emerging epidemic not only confined to liver-related morbidity and mortality. It is also becoming apparent that NAFLD is associated with moderate cerebral dysfunction and cognitive decline. A possible link between NAFLD and Alzheimer's disease (AD) has only recently been proposed due to the multiple shared genes and pathological mechanisms contributing to the development of these conditions. Although AD is a progressive neurodegenerative disease, the exact pathophysiological mechanism remains ambiguous and similarly to NAFLD, currently available pharmacological therapies have mostly failed in clinical trials. In addition to the usual suspects (inflammation, oxidative stress, blood-brain barrier alterations and ageing) that could contribute to the NAFLD-induced development and progression of AD, changes in the vasculature, cerebral perfusion and waste clearance could be the missing link between these two diseases. Here, we review the most recent literature linking NAFLD and AD, focusing on cerebrovascular alterations and the brain's clearance system as risk factors involved in the development and progression of AD, with the aim of promoting further research using neuroimaging techniques and new mechanism-based therapeutic interventions.
Collapse
|