51
|
Morphogenesis of vascular and neuronal networks and the relationships between their remodeling processes. Brain Res Bull 2022; 186:62-69. [DOI: 10.1016/j.brainresbull.2022.05.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 05/18/2022] [Accepted: 05/29/2022] [Indexed: 11/21/2022]
|
52
|
Goswami AG, Basu S, Huda F, Pant J, Ghosh Kar A, Banerjee T, Shukla VK. An appraisal of vascular endothelial growth factor (VEGF): the dynamic molecule of wound healing and its current clinical applications. Growth Factors 2022; 40:73-88. [PMID: 35584274 DOI: 10.1080/08977194.2022.2074843] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Angiogenesis is a critical step of wound healing, and its failure leads to chronic wounds. The idea of restoring blood flow to the damaged tissues by promoting neo-angiogenesis is lucrative and has been researched extensively. Vascular endothelial growth factor (VEGF), a key dynamic molecule of angiogenesis has been investigated for its functions. In this review, we aim to appraise its biology, the comprehensive role of this dynamic molecule in the wound healing process, and how this knowledge has been translated in clinical application in various types of wounds. Although, most laboratory research on the use of VEGF is promising, its clinical applications have not met great expectations. We discuss various lacunae that might exist in making its clinical application unsuccessful for commercial use, and provide insight to the foundation for future research.
Collapse
Affiliation(s)
- Aakansha Giri Goswami
- Department of General surgery, All India Institute of Medical Sciences, Rishikesh, India
| | - Somprakas Basu
- Department of General surgery, All India Institute of Medical Sciences, Rishikesh, India
| | - Farhanul Huda
- Department of General surgery, All India Institute of Medical Sciences, Rishikesh, India
| | - Jayanti Pant
- Department of Physiology, All India Institute of Medical Sciences, Rishikesh, India
| | - Amrita Ghosh Kar
- Department of Pathology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Tuhina Banerjee
- Department of Microbiology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Vijay Kumar Shukla
- Department of General Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| |
Collapse
|
53
|
Fu Q, Duan R, Sun Y, Li Q. Hyperbaric oxygen therapy for healthy aging: From mechanisms to therapeutics. Redox Biol 2022; 53:102352. [PMID: 35649312 PMCID: PMC9156818 DOI: 10.1016/j.redox.2022.102352] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/17/2022] [Accepted: 05/23/2022] [Indexed: 12/19/2022] Open
Abstract
Hyperbaric oxygen therapy (HBOT), a technique through which 100% oxygen is provided at a pressure higher than 1 atm absolute (ATA), has become a well-established treatment modality for multiple conditions. The noninvasive nature, favorable safety profile, and common clinical application of HBOT make it a competitive candidate for several new indications, one of them being aging and age-related diseases. In fact, despite the conventional wisdom that excessive oxygen accelerates aging, appropriate HBOT protocols without exceeding the toxicity threshold have shown great promise in therapies against aging. For one thing, an extensive body of basic research has expanded our mechanistic understanding of HBOT. Interestingly, the therapeutic targets of HBOT overlap considerably with those of aging and age-related diseases. For another, pre-clinical and small-scale clinical investigations have provided validated information on the efficacy of HBOT against aging from various aspects. However, a generally applicable protocol for HBOT to be utilized in therapies against aging needs to be defined as a subsequent step. It is high time to look back and summarize the recent advances concerning biological mechanisms and therapeutic implications of HBOT in promoting healthy aging and shed light on prospective directions. Here we provide the first comprehensive overview of HBOT in the field of aging and geriatric research, which allows the scientific community to be aware of the emerging tendency and move beyond conventional wisdom to scientific findings of translational value.
Collapse
|
54
|
Watson AMD, Chen YC, Peter K. Vascular Aging and Vascular Disease Have Much in Common! Arterioscler Thromb Vasc Biol 2022; 42:1077-1080. [PMID: 35735019 DOI: 10.1161/atvbaha.122.317892] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Anna M D Watson
- Atherothrombosis and Vascular Biology Program, Baker Heart and Diabetes Institute, Central Clinical School, Monash University, Melbourne, VIC, Australia. Department of Cardiometabolic Health, University of Melbourne, VIC, Australia
| | - Yung-Chih Chen
- Atherothrombosis and Vascular Biology Program, Baker Heart and Diabetes Institute, Central Clinical School, Monash University, Melbourne, VIC, Australia. Department of Cardiometabolic Health, University of Melbourne, VIC, Australia
| | - Karlheinz Peter
- Atherothrombosis and Vascular Biology Program, Baker Heart and Diabetes Institute, Central Clinical School, Monash University, Melbourne, VIC, Australia. Department of Cardiometabolic Health, University of Melbourne, VIC, Australia
| |
Collapse
|
55
|
Richter RP, Payne GA, Ambalavanan N, Gaggar A, Richter JR. The endothelial glycocalyx in critical illness: A pediatric perspective. Matrix Biol Plus 2022; 14:100106. [PMID: 35392182 PMCID: PMC8981764 DOI: 10.1016/j.mbplus.2022.100106] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 02/28/2022] [Accepted: 03/01/2022] [Indexed: 12/18/2022] Open
Abstract
The vascular endothelium is the interface between circulating blood and end organs and thus has a critical role in preserving organ function. The endothelium is lined by a glycan-rich glycocalyx that uniquely contributes to endothelial function through its regulation of leukocyte and platelet interactions with the vessel wall, vascular permeability, coagulation, and vasoreactivity. Degradation of the endothelial glycocalyx can thus promote vascular dysfunction, inflammation propagation, and organ injury. The endothelial glycocalyx and its role in vascular pathophysiology has gained increasing attention over the last decade. While studies characterizing vascular glycocalyx injury and its downstream consequences in a host of adult human diseases and in animal models has burgeoned, studies evaluating glycocalyx damage in pediatric diseases are relatively few. As children have unique physiology that differs from adults, significant knowledge gaps remain in our understanding of the causes and effects of endothelial glycocalyx disintegrity in pediatric critical illness. In this narrative literature overview, we offer a unique perspective on the role of the endothelial glycocalyx in pediatric critical illness, drawing from adult and preclinical data in addition to pediatric clinical experience to elucidate how marked derangement of the endothelial surface layer may contribute to aberrant vascular biology in children. By calling attention to this nascent field, we hope to increase research efforts to address important knowledge gaps in pediatric vascular biology that may inform the development of novel therapeutic strategies.
Collapse
Key Words
- ACE2, angiotensin-converting enzyme 2
- CD, cell differentiation marker
- COVID-19, coronavirus disease 2019
- CPB, cardiopulmonary bypass
- CT, component therapy
- Children
- Critical illness
- DENV NS1, dengue virus nonstructural protein 1
- DM, diabetes mellitus
- ECLS, extracorporeal life support
- ECMO, extracorporeal membrane oxygenation
- EG, endothelial glycocalyx
- Endothelial glycocalyx
- FFP, fresh frozen plasma
- GAG, glycosaminoglycan
- GPC, glypican
- HPSE, heparanase
- HSV, herpes simplex virus
- IV, intravenous
- MIS-C, multisystem inflammatory syndrome in children
- MMP, matrix metalloproteinase
- Pragmatic, Randomized Optimal Platelet and Plasma Ratios
- RHAMM, receptor for hyaluronan-mediated motility
- S protein, spike protein
- SAFE, Saline versus Albumin Fluid Evaluation
- SARS-CoV-2, severe acute respiratory syndrome coronavirus 2
- SDC, syndecan
- SDF, sidestream darkfield
- SIRT1, sirtuin 1
- TBI, traumatic brain injury
- TBSA, total body surface area
- TMPRSS2, transmembrane protease serine 2
- Th2, type 2 helper T cell
- VSMC, vascular smooth muscle cell
- Vascular biology
- WB+CT, whole blood and component therapy
- eNOS, endothelial nitric oxide synthase
Collapse
Affiliation(s)
- Robert P. Richter
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
- Program in Protease and Matrix Biology, University of Alabama at Birmingham, Birmingham, AL, USA
- Center for Injury Science, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Gregory A. Payne
- Program in Protease and Matrix Biology, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Namasivayam Ambalavanan
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
- Translational Research in Normal and Disordered Development Program, University of Alabama, Birmingham, AL, USA
| | - Amit Gaggar
- Program in Protease and Matrix Biology, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jillian R. Richter
- Center for Injury Science, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
56
|
Weigel R, Schilling L, Krauss JK. The pathophysiology of chronic subdural hematoma revisited: emphasis on aging processes as key factor. GeroScience 2022; 44:1353-1371. [DOI: 10.1007/s11357-022-00570-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 04/07/2022] [Indexed: 12/24/2022] Open
|
57
|
Abstract
There have been published regenerative endodontic protocols for treating immature teeth in young patients, but there are no clinical considerations for the adult teeth. The goal of the present review is to propose a specific clinical protocol for both mature and immature adult teeth with necrotic pulps. Research was performed from January to April of 2021. From the 539 studies identified through the initial search, 23 studies were qualified for the final analysis (3 randomized controlled trials and 20 case reports). The results in mature adult teeth indicate a success rate of 96.35 and 100% in bone healing through the randomized controlled trials and case reports, respectively; 100% in absence of clinical symptoms, and 58 and 62.5% in positive response to sensibility tests. The success rate in the case reports in teeth with open apex reported a 61.5% of root development, 100% of bone healing, 96.15% of absence of clinical symptoms, and 43.7% of positive response to sensibility tests. The current evidence is scarce but emerging, so REPs may be a promising alternative for treating adult necrotic teeth. The clinical protocol proposed is based on the evidence available and age considerations, and should be updated in the future.
Collapse
|
58
|
Kiss T, Nyúl-Tóth Á, Gulej R, Tarantini S, Csipo T, Mukli P, Ungvari A, Balasubramanian P, Yabluchanskiy A, Benyo Z, Conley SM, Wren JD, Garman L, Huffman DM, Csiszar A, Ungvari Z. Old blood from heterochronic parabionts accelerates vascular aging in young mice: transcriptomic signature of pathologic smooth muscle remodeling. GeroScience 2022; 44:953-981. [PMID: 35124764 PMCID: PMC9135944 DOI: 10.1007/s11357-022-00519-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 01/16/2022] [Indexed: 02/07/2023] Open
Abstract
Vascular aging has a central role in the pathogenesis of cardiovascular diseases contributing to increased mortality of older adults. There is increasing evidence that, in addition to the documented role of cell-autonomous mechanisms of aging, cell-nonautonomous mechanisms also play a critical role in the regulation of vascular aging processes. Our recent transcriptomic studies (Kiss T. et al. Geroscience. 2020;42(2):727-748) demonstrated that circulating anti-geronic factors from young blood promote vascular rejuvenation in aged mice. The present study was designed to expand upon the results of this study by testing the hypothesis that circulating pro-geronic factors also contribute to the genesis of vascular aging phenotypes. To test this hypothesis, through heterochronic parabiosis, we determined the extent to which shifts in the vascular transcriptome (RNA-seq) are modulated by the old systemic environment. We reanalyzed existing RNA-seq data, comparing the transcriptome in the aorta arch samples isolated from isochronic parabiont aged (20-month-old) C57BL/6 mice [A-(A); parabiosis for 8 weeks] and young isochronic parabiont (6-month-old) mice [Y-(Y)] and also assessing transcriptomic changes in the aortic arch in young (6-month-old) parabiont mice [Y-(A); heterochronic parabiosis for 8 weeks] induced by the presence of old blood derived from aged (20-month-old) parabionts. We identified 528 concordant genes whose expression levels differed in the aged phenotype and were shifted towards the aged phenotype by the presence of old blood in young Y-(A) animals. Among them, the expression of 221 concordant genes was unaffected by the presence of young blood in A-(Y) mice. GO enrichment analysis suggests that old blood-regulated genes may contribute to pathologic vascular remodeling. IPA Upstream Regulator analysis (performed to identify upstream transcriptional regulators that may contribute to the observed transcriptomic changes) suggests that the mechanism of action of pro-geronic factors present in old blood may include inhibition of pathways mediated by SRF (serum response factor), insulin-like growth factor-1 (IGF-1) and VEGF-A. In conclusion, relatively short-term exposure to old blood can accelerate vascular aging processes. Our findings provide additional evidence supporting the significant plasticity of vascular aging and the existence of circulating pro-geronic factors mediating pathological remodeling of the vascular smooth muscle cells and the extracellular matrix.
Collapse
Affiliation(s)
- Tamas Kiss
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
- International Training Program in Geroscience, First Department of Pediatrics, Semmelweis University, Budapest, Hungary
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Ádám Nyúl-Tóth
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
- International Training Program in Geroscience, Institute of Biophysics, Biological Research Centre, Eötvös Loránd Research Network (ELKH), Szeged, Hungary
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Rafal Gulej
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
| | - Stefano Tarantini
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104 USA
| | - Tamas Csipo
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Peter Mukli
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Anna Ungvari
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
| | - Priya Balasubramanian
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
| | - Zoltan Benyo
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Shannon M. Conley
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
| | - Jonathan D. Wren
- Oklahoma Medical Research Foundation, Genes & Human Disease Research Program, Oklahoma City, OK USA
| | - Lori Garman
- Oklahoma Medical Research Foundation, Genes & Human Disease Research Program, Oklahoma City, OK USA
| | - Derek M. Huffman
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461 USA
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY USA
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY USA
| | - Anna Csiszar
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Translational Medicine, Semmelweis University, Budapest, Hungary
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104 USA
- International Training Program in Geroscience, Theoretical Medicine Doctoral School, University of Szeged, Szeged, Hungary
| | - Zoltan Ungvari
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104 USA
| |
Collapse
|
59
|
Brinkmann V, Romeo M, Larigot L, Hemmers A, Tschage L, Kleinjohann J, Schiavi A, Steinwachs S, Esser C, Menzel R, Giani Tagliabue S, Bonati L, Cox F, Ale-Agha N, Jakobs P, Altschmied J, Haendeler J, Coumoul X, Ventura N. Aryl Hydrocarbon Receptor-Dependent and -Independent Pathways Mediate Curcumin Anti-Aging Effects. Antioxidants (Basel) 2022; 11:613. [PMID: 35453298 PMCID: PMC9024831 DOI: 10.3390/antiox11040613] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 03/18/2022] [Accepted: 03/19/2022] [Indexed: 02/04/2023] Open
Abstract
The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor whose activity can be modulated by polyphenols, such as curcumin. AhR and curcumin have evolutionarily conserved effects on aging. Here, we investigated whether and how the AhR mediates the anti-aging effects of curcumin across species. Using a combination of in vivo, in vitro, and in silico analyses, we demonstrated that curcumin has AhR-dependent or -independent effects in a context-specific manner. We found that in Caenorhabditis elegans, AhR mediates curcumin-induced lifespan extension, most likely through a ligand-independent inhibitory mechanism related to its antioxidant activity. Curcumin also showed AhR-independent anti-aging activities, such as protection against aggregation-prone proteins and oxidative stress in C. elegans and promotion of the migratory capacity of human primary endothelial cells. These AhR-independent effects are largely mediated by the Nrf2/SKN-1 pathway.
Collapse
Affiliation(s)
- Vanessa Brinkmann
- Institute of Clinical Chemistry and Laboratory Diagnostic, Medical Faculty, Heinrich Heine University Düsseldorf, Moorenstr 5, 40225 Düsseldorf, Germany; (V.B.); (M.R.); (A.S.); (F.C.); (N.A.-A.); (P.J.); (J.A.); (J.H.)
- IUF—Leibniz Research Institute for Environmental Medicine, Auf’m Hennekamp 50, 40225 Düsseldorf, Germany; (A.H.); (L.T.); (J.K.); (S.S.); (C.E.)
| | - Margherita Romeo
- Institute of Clinical Chemistry and Laboratory Diagnostic, Medical Faculty, Heinrich Heine University Düsseldorf, Moorenstr 5, 40225 Düsseldorf, Germany; (V.B.); (M.R.); (A.S.); (F.C.); (N.A.-A.); (P.J.); (J.A.); (J.H.)
- IUF—Leibniz Research Institute for Environmental Medicine, Auf’m Hennekamp 50, 40225 Düsseldorf, Germany; (A.H.); (L.T.); (J.K.); (S.S.); (C.E.)
| | - Lucie Larigot
- Faculté des Sciences Fondamentales et Biomédicales, Université de Paris, 45 Rue des Saints-Pères, F-75006 Paris, France; (L.L.); (X.C.)
| | - Anne Hemmers
- IUF—Leibniz Research Institute for Environmental Medicine, Auf’m Hennekamp 50, 40225 Düsseldorf, Germany; (A.H.); (L.T.); (J.K.); (S.S.); (C.E.)
| | - Lisa Tschage
- IUF—Leibniz Research Institute for Environmental Medicine, Auf’m Hennekamp 50, 40225 Düsseldorf, Germany; (A.H.); (L.T.); (J.K.); (S.S.); (C.E.)
| | - Jennifer Kleinjohann
- IUF—Leibniz Research Institute for Environmental Medicine, Auf’m Hennekamp 50, 40225 Düsseldorf, Germany; (A.H.); (L.T.); (J.K.); (S.S.); (C.E.)
| | - Alfonso Schiavi
- Institute of Clinical Chemistry and Laboratory Diagnostic, Medical Faculty, Heinrich Heine University Düsseldorf, Moorenstr 5, 40225 Düsseldorf, Germany; (V.B.); (M.R.); (A.S.); (F.C.); (N.A.-A.); (P.J.); (J.A.); (J.H.)
- IUF—Leibniz Research Institute for Environmental Medicine, Auf’m Hennekamp 50, 40225 Düsseldorf, Germany; (A.H.); (L.T.); (J.K.); (S.S.); (C.E.)
| | - Swantje Steinwachs
- IUF—Leibniz Research Institute for Environmental Medicine, Auf’m Hennekamp 50, 40225 Düsseldorf, Germany; (A.H.); (L.T.); (J.K.); (S.S.); (C.E.)
| | - Charlotte Esser
- IUF—Leibniz Research Institute for Environmental Medicine, Auf’m Hennekamp 50, 40225 Düsseldorf, Germany; (A.H.); (L.T.); (J.K.); (S.S.); (C.E.)
| | - Ralph Menzel
- Institute of Biology, Humboldt-University Berlin, Philippstr. 13, 10115 Berlin, Germany;
| | - Sara Giani Tagliabue
- Department of Earth and Environmental Sciences, University of Milano-Bicocca, Piazza della Scienza 1, 20126 Milano, Italy; (S.G.T.); (L.B.)
| | - Laura Bonati
- Department of Earth and Environmental Sciences, University of Milano-Bicocca, Piazza della Scienza 1, 20126 Milano, Italy; (S.G.T.); (L.B.)
| | - Fiona Cox
- Institute of Clinical Chemistry and Laboratory Diagnostic, Medical Faculty, Heinrich Heine University Düsseldorf, Moorenstr 5, 40225 Düsseldorf, Germany; (V.B.); (M.R.); (A.S.); (F.C.); (N.A.-A.); (P.J.); (J.A.); (J.H.)
- Institute of Clinical Pharmacology and Pharmacology, Medical Faculty, University Hospital and Heinrich Heine University Düsseldorf, Moorenstr 5, 40225 Düsseldorf, Germany
| | - Niloofar Ale-Agha
- Institute of Clinical Chemistry and Laboratory Diagnostic, Medical Faculty, Heinrich Heine University Düsseldorf, Moorenstr 5, 40225 Düsseldorf, Germany; (V.B.); (M.R.); (A.S.); (F.C.); (N.A.-A.); (P.J.); (J.A.); (J.H.)
| | - Philipp Jakobs
- Institute of Clinical Chemistry and Laboratory Diagnostic, Medical Faculty, Heinrich Heine University Düsseldorf, Moorenstr 5, 40225 Düsseldorf, Germany; (V.B.); (M.R.); (A.S.); (F.C.); (N.A.-A.); (P.J.); (J.A.); (J.H.)
| | - Joachim Altschmied
- Institute of Clinical Chemistry and Laboratory Diagnostic, Medical Faculty, Heinrich Heine University Düsseldorf, Moorenstr 5, 40225 Düsseldorf, Germany; (V.B.); (M.R.); (A.S.); (F.C.); (N.A.-A.); (P.J.); (J.A.); (J.H.)
- IUF—Leibniz Research Institute for Environmental Medicine, Auf’m Hennekamp 50, 40225 Düsseldorf, Germany; (A.H.); (L.T.); (J.K.); (S.S.); (C.E.)
| | - Judith Haendeler
- Institute of Clinical Chemistry and Laboratory Diagnostic, Medical Faculty, Heinrich Heine University Düsseldorf, Moorenstr 5, 40225 Düsseldorf, Germany; (V.B.); (M.R.); (A.S.); (F.C.); (N.A.-A.); (P.J.); (J.A.); (J.H.)
| | - Xavier Coumoul
- Faculté des Sciences Fondamentales et Biomédicales, Université de Paris, 45 Rue des Saints-Pères, F-75006 Paris, France; (L.L.); (X.C.)
| | - Natascia Ventura
- Institute of Clinical Chemistry and Laboratory Diagnostic, Medical Faculty, Heinrich Heine University Düsseldorf, Moorenstr 5, 40225 Düsseldorf, Germany; (V.B.); (M.R.); (A.S.); (F.C.); (N.A.-A.); (P.J.); (J.A.); (J.H.)
- IUF—Leibniz Research Institute for Environmental Medicine, Auf’m Hennekamp 50, 40225 Düsseldorf, Germany; (A.H.); (L.T.); (J.K.); (S.S.); (C.E.)
| |
Collapse
|
60
|
Gallego I, Villate-Beitia I, Saenz-Del-Burgo L, Puras G, Pedraz JL. Therapeutic Opportunities and Delivery Strategies for Brain Revascularization in Stroke, Neurodegeneration, and Aging. Pharmacol Rev 2022; 74:439-461. [PMID: 35302047 DOI: 10.1124/pharmrev.121.000418] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 01/18/2022] [Accepted: 01/22/2022] [Indexed: 12/25/2022] Open
Abstract
Central nervous system (CNS) diseases, especially acute ischemic events and neurodegenerative disorders, constitute a public health problem with no effective treatments to allow a persistent solution. Failed therapies targeting neuronal recovery have revealed the multifactorial and intricate pathophysiology underlying such CNS disorders as ischemic stroke, Alzheimeŕs disease, amyotrophic lateral sclerosis, vascular Parkisonism, vascular dementia, and aging, in which cerebral microvasculature impairment seems to play a key role. In fact, a reduction in vessel density and cerebral blood flow occurs in these scenarios, contributing to neuronal dysfunction and leading to loss of cognitive function. In this review, we provide an overview of healthy brain microvasculature structure and function in health and the effect of the aforementioned cerebral CNS diseases. We discuss the emerging new therapeutic opportunities, and their delivery approaches, aimed at recovering brain vascularization in this context. SIGNIFICANCE STATEMENT: The lack of effective treatments, mainly focused on neuron recovery, has prompted the search of other therapies to treat cerebral central nervous system diseases. The disruption and degeneration of cerebral microvasculature has been evidenced in neurodegenerative diseases, stroke, and aging, constituting a potential target for restoring vascularization, neuronal functioning, and cognitive capacities by the development of therapeutic pro-angiogenic strategies.
Collapse
Affiliation(s)
- Idoia Gallego
- NanoBioCel Research Group, Laboratory of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain (I.G., I.V.-B., L.S.-B., G.P., J.L.P); Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine, Institute of Health Carlos III, Madrid, Spain (I.G., I.V.-B., L.S.-B., G.P., J.L.P.); and Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain (I.G., I.V.-B., L.S.-B., G.P., J.L.P.)
| | - Ilia Villate-Beitia
- NanoBioCel Research Group, Laboratory of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain (I.G., I.V.-B., L.S.-B., G.P., J.L.P); Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine, Institute of Health Carlos III, Madrid, Spain (I.G., I.V.-B., L.S.-B., G.P., J.L.P.); and Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain (I.G., I.V.-B., L.S.-B., G.P., J.L.P.)
| | - Laura Saenz-Del-Burgo
- NanoBioCel Research Group, Laboratory of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain (I.G., I.V.-B., L.S.-B., G.P., J.L.P); Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine, Institute of Health Carlos III, Madrid, Spain (I.G., I.V.-B., L.S.-B., G.P., J.L.P.); and Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain (I.G., I.V.-B., L.S.-B., G.P., J.L.P.)
| | - Gustavo Puras
- NanoBioCel Research Group, Laboratory of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain (I.G., I.V.-B., L.S.-B., G.P., J.L.P); Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine, Institute of Health Carlos III, Madrid, Spain (I.G., I.V.-B., L.S.-B., G.P., J.L.P.); and Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain (I.G., I.V.-B., L.S.-B., G.P., J.L.P.)
| | - José Luis Pedraz
- NanoBioCel Research Group, Laboratory of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain (I.G., I.V.-B., L.S.-B., G.P., J.L.P); Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine, Institute of Health Carlos III, Madrid, Spain (I.G., I.V.-B., L.S.-B., G.P., J.L.P.); and Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain (I.G., I.V.-B., L.S.-B., G.P., J.L.P.)
| |
Collapse
|
61
|
Wang Y, Chen F, Zhang Y, Zheng X, Liu S, Tang M, Wang Z, Wang P, Bao Y, Li D. Biphasic effect of sulforaphane on angiogenesis in hypoxia via modulation of both Nrf2 and mitochondrial dynamics. Food Funct 2022; 13:2884-2898. [PMID: 35179529 DOI: 10.1039/d1fo04112f] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Sulforaphane (SFN) is an isothiocyanate (ITC) derived from a glucosinolate, glucoraphinin found in cruciferous vegetables. There are few studies that focus on the role of SFN in angiogenesis under hypoxic conditions. The effect of SFN on angiogenesis and the underlying mechanisms including the roles of Nrf2 and mitochondrial dynamics were investigated using cultured human umbilical vein endothelial cells (HUVECs) in hypoxia. SFN at low doses (1.25-5 μM) increased hypoxia-induced HUVEC migration and tube formation, and alleviated hypoxia-induced retarded proliferation, but high doses (≥10 μM) exhibited an opposite effect. Under hypoxia, the expression of Nrf2 and heme oxygenase-1 was up-regulated by SFN treatment. Nrf2 knockdown abrogated SFN (2.5 μM)-induced tube formation and further potentiated the inhibitory effect of SFN (10 μM) on angiogenesis. Meanwhile, the mitochondrial function, morphology and expression of dynamic-related proteins suggested that low-dose SFN protected against hypoxia-induced mitochondrial injury and alleviated hypoxia-induced fission Nrf2-dependently without affecting the expression of key effector proteins (Drp1, Fis1, Mfn1/2 and Opa1), while high concentrations (≥10 μM SFN) aggravated hypoxia-induced mitochondrial injury, fission and Drp1 expression, and inhibited Mfn1/2 expression. These findings suggest that SFN biphasically affected the angiogenic capacity of hypoxia challenged HUVECs potentially via mechanisms involving an integrated modulation of Nrf2 and mitochondrial dynamics.
Collapse
Affiliation(s)
- Yaqian Wang
- Department of Nutrition, School of Public Health, Sun Yat-Sen University (Northern Campus), Guangzhou, Guangdong Province 510080, P. R. China. .,Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, Guangdong Province 510080, P. R. China.,Guangdong Engineering Technology Center of Nutrition Transformation, Guangzhou, Guangdong Province 510080, P. R. China
| | - Fangfang Chen
- Department of Nutrition, School of Public Health, Sun Yat-Sen University (Northern Campus), Guangzhou, Guangdong Province 510080, P. R. China. .,Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, Guangdong Province 510080, P. R. China.,Guangdong Engineering Technology Center of Nutrition Transformation, Guangzhou, Guangdong Province 510080, P. R. China
| | - Yuan Zhang
- Department of Geriatrics, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province 510150, P. R. China
| | - Xiangyu Zheng
- Department of Nutrition, School of Public Health, Sun Yat-Sen University (Northern Campus), Guangzhou, Guangdong Province 510080, P. R. China. .,Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, Guangdong Province 510080, P. R. China.,Guangdong Engineering Technology Center of Nutrition Transformation, Guangzhou, Guangdong Province 510080, P. R. China
| | - Shiyan Liu
- Department of Nutrition, School of Public Health, Sun Yat-Sen University (Northern Campus), Guangzhou, Guangdong Province 510080, P. R. China. .,Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, Guangdong Province 510080, P. R. China.,Guangdong Engineering Technology Center of Nutrition Transformation, Guangzhou, Guangdong Province 510080, P. R. China
| | - Meijuan Tang
- Department of Nutrition, School of Public Health, Sun Yat-Sen University (Northern Campus), Guangzhou, Guangdong Province 510080, P. R. China. .,Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, Guangdong Province 510080, P. R. China.,Guangdong Engineering Technology Center of Nutrition Transformation, Guangzhou, Guangdong Province 510080, P. R. China
| | - Ziling Wang
- Department of Nutrition, School of Public Health, Sun Yat-Sen University (Northern Campus), Guangzhou, Guangdong Province 510080, P. R. China. .,Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, Guangdong Province 510080, P. R. China.,Guangdong Engineering Technology Center of Nutrition Transformation, Guangzhou, Guangdong Province 510080, P. R. China
| | - Pan Wang
- Department of Nutrition, School of Public Health, Sun Yat-Sen University (Northern Campus), Guangzhou, Guangdong Province 510080, P. R. China. .,Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, Guangdong Province 510080, P. R. China.,Guangdong Engineering Technology Center of Nutrition Transformation, Guangzhou, Guangdong Province 510080, P. R. China
| | - Yongping Bao
- Norwich Medical School, University of East Anglia, Norwich, Norfolk NR4 7UQ, UK.
| | - Dan Li
- Department of Nutrition, School of Public Health, Sun Yat-Sen University (Northern Campus), Guangzhou, Guangdong Province 510080, P. R. China. .,Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, Guangdong Province 510080, P. R. China.,Guangdong Engineering Technology Center of Nutrition Transformation, Guangzhou, Guangdong Province 510080, P. R. China
| |
Collapse
|
62
|
Blood-Brain Barrier Transporters: Opportunities for Therapeutic Development in Ischemic Stroke. Int J Mol Sci 2022; 23:ijms23031898. [PMID: 35163820 PMCID: PMC8836701 DOI: 10.3390/ijms23031898] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/02/2022] [Accepted: 02/04/2022] [Indexed: 12/20/2022] Open
Abstract
Globally, stroke is a leading cause of death and long-term disability. Over the past decades, several efforts have attempted to discover new drugs or repurpose existing therapeutics to promote post-stroke neurological recovery. Preclinical stroke studies have reported successes in identifying novel neuroprotective agents; however, none of these compounds have advanced beyond a phase III clinical trial. One reason for these failures is the lack of consideration of blood-brain barrier (BBB) transport mechanisms that can enable these drugs to achieve efficacious concentrations in ischemic brain tissue. Despite the knowledge that drugs with neuroprotective properties (i.e., statins, memantine, metformin) are substrates for endogenous BBB transporters, preclinical stroke research has not extensively studied the role of transporters in central nervous system (CNS) drug delivery. Here, we review current knowledge on specific BBB uptake transporters (i.e., organic anion transporting polypeptides (OATPs in humans; Oatps in rodents); organic cation transporters (OCTs in humans; Octs in rodents) that can be targeted for improved neuroprotective drug delivery. Additionally, we provide state-of-the-art perspectives on how transporter pharmacology can be integrated into preclinical stroke research. Specifically, we discuss the utility of in vivo stroke models to transporter studies and considerations (i.e., species selection, co-morbid conditions) that will optimize the translational success of stroke pharmacotherapeutic experiments.
Collapse
|
63
|
Potential of Polyphenols to Restore SIRT1 and NAD+ Metabolism in Renal Disease. Nutrients 2022; 14:nu14030653. [PMID: 35277012 PMCID: PMC8837945 DOI: 10.3390/nu14030653] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/18/2022] [Accepted: 01/29/2022] [Indexed: 11/17/2022] Open
Abstract
SIRT1 is an NAD+-dependent class III histone deacetylase that is abundantly expressed in the kidney, where it modulates gene expression, apoptosis, energy homeostasis, autophagy, acute stress responses, and mitochondrial biogenesis. Alterations in SIRT1 activity and NAD+ metabolism are frequently observed in acute and chronic kidney diseases of diverse origins, including obesity and diabetes. Nevertheless, in vitro and in vivo studies and clinical trials with humans show that the SIRT1-activating compounds derived from natural sources, such as polyphenols found in fruits, vegetables, and plants, including resveratrol, quercetin, and isoflavones, can prevent disease and be part of treatments for a wide variety of diseases. Here, we summarize the roles of SIRT1 and NAD+ metabolism in renal pathophysiology and provide an overview of polyphenols that have the potential to restore SIRT1 and NAD+ metabolism in renal diseases.
Collapse
|
64
|
Wu J, Miller E, Davidson C, Walker BR, Hadoke PWF. Enhanced Angiogenesis by 11βHSD1 Blockage Is Insufficient to Improve Reperfusion Following Hindlimb Ischaemia. Front Cardiovasc Med 2022; 8:795823. [PMID: 35097015 PMCID: PMC8790072 DOI: 10.3389/fcvm.2021.795823] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 12/13/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Critical limb ischaemia (CLI), which is estimated to affect 2 million people in the United States, reduces quality of life, is associated with high morbidity and mortality, and has limited treatment options. Direct stimulation of angiogenesis using proangiogenic growth factors has been investigated as a therapeutic strategy to improve reperfusion in the ischaemic leg. Despite positive outcomes in animal studies, there has been little success in clinical translation. This investigation addressed the hypothesis that angiogenesis could be stimulated indirectly in the ischaemic hindlimb by blocking 11β-hydroxysteroid dehydrogenase 1 (11βHSD1)-mediated reactivation of anti-angiogenic glucocorticoids. Method and Results: Corticosterone suppressed ex vivo angiogenesis in the mouse aortic ring assay. 11βHSD1 deletion (Hsd11b1Del1/Del1) or pharmacological inhibition (with 300 nM UE2316) which block the reactivation of glucocorticoid (i.e., the conversion of 11-dehydrocorticosterone (11DHC) to bioactive corticosterone) significantly reduced 11DHC-induced suppression of angiogenesis. In a sponge implantation model, 11βHSD1 deletion, but not pharmacological inhibition, enhanced inflammation-induced angiogenesis. By contrast, in the mouse hindlimb ischaemia model, post-ischaemic reperfusion and vascular density were not affected by either deletion or pharmacological inhibition of 11βHSD1 in young or aged mice. 3D vascular imaging suggested that hind limb reperfusion in the 1st week following induction of ischaemia may be driven by the rapid expansion of collateral arteries rather than by angiogenesis. Conclusion: 11βHSD1-mediated glucocorticoid reactivation suppressed angiogenesis ex vivo and in vivo. However, regulation of angiogenesis alone was insufficient to promote reperfusion in hindlimb ischaemia. Future investigation of post-ischaemic reperfusion should include other aspects of systemic vascular remodeling including arteriogenesis and collateral formation.
Collapse
Affiliation(s)
- Junxi Wu
- The Queen's Medical Research Institute, University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom,Department of Biomedical Engineering, University of Strathclyde, Glasgow, United Kingdom
| | - Eileen Miller
- The Queen's Medical Research Institute, University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Callam Davidson
- The Queen's Medical Research Institute, University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Brian R. Walker
- The Queen's Medical Research Institute, University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Patrick W. F. Hadoke
- The Queen's Medical Research Institute, University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom,*Correspondence: Patrick W. F. Hadoke
| |
Collapse
|
65
|
Noh B, Blasco-Conesa MP, Lai YJ, Ganesh BP, Urayama A, Moreno-Gonzalez I, Marrelli SP, McCullough LD, Moruno-Manchon JF. G-quadruplexes Stabilization Upregulates CCN1 and Accelerates Aging in Cultured Cerebral Endothelial Cells. FRONTIERS IN AGING 2022; 2:797562. [PMID: 35822045 PMCID: PMC9261356 DOI: 10.3389/fragi.2021.797562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 12/07/2021] [Indexed: 11/27/2022]
Abstract
Senescence in the cerebral endothelium has been proposed as a mechanism that can drive dysfunction of the cerebral vasculature, which precedes vascular dementia. Cysteine-rich angiogenic inducer 61 (Cyr61/CCN1) is a matricellular protein secreted by cerebral endothelial cells (CEC). CCN1 induces senescence in fibroblasts. However, whether CCN1 contributes to senescence in CEC and how this is regulated requires further study. Aging has been associated with the formation of four-stranded Guanine-quadruplexes (G4s) in G-rich motifs of DNA and RNA. Stabilization of the G4 structures regulates transcription and translation either by upregulation or downregulation depending on the gene target. Previously, we showed that aged mice treated with a G4-stabilizing compound had enhanced senescence-associated (SA) phenotypes in their brains, and these mice exhibited enhanced cognitive deficits. A sequence in the 3'-UTR of the human CCN1 mRNA has the ability to fold into G4s in vitro. We hypothesize that G4 stabilization regulates CCN1 in cultured primary CEC and induces endothelial senescence. We used cerebral microvessel fractions and cultured primary CEC from young (4-months old, m/o) and aged (18-m/o) mice to determine CCN1 levels. SA phenotypes were determined by high-resolution fluorescence microscopy in cultured primary CEC, and we used Thioflavin T to recognize RNA-G4s for fluorescence spectra. We found that cultured CEC from aged mice exhibited enhanced levels of SA phenotypes, and higher levels of CCN1 and G4 stabilization. In cultured CEC, CCN1 induced SA phenotypes, such as SA β-galactosidase activity, and double-strand DNA damage. Furthermore, CCN1 levels were upregulated by a G4 ligand, and a G-rich motif in the 3'-UTR of the Ccn1 mRNA was folded into a G4. In conclusion, we demonstrate that CCN1 can induce senescence in cultured primary CEC, and we provide evidence that G4 stabilization is a novel mechanism regulating the SASP component CCN1.
Collapse
Affiliation(s)
- Brian Noh
- Department of Neurology, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Maria P. Blasco-Conesa
- Department of Neurology, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Yun-Ju Lai
- Department of Neurology, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, United States
- Solomont School of Nursing, Zuckerberg College of Health Sciences, University of Massachusetts Lowell, Lowell, MA, United States
| | - Bhanu Priya Ganesh
- Department of Neurology, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Akihiko Urayama
- Department of Neurology, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Ines Moreno-Gonzalez
- Department of Neurology, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, United States
- Department of Cell Biology, Faculty of Sciences, Instituto de Investigacion Biomedica de Malaga-IBIMA, Malaga University, Malaga, Spain
- Networking Biomedical Research Networking Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Sean P. Marrelli
- Department of Neurology, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Louise D. McCullough
- Department of Neurology, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Jose Felix Moruno-Manchon
- Department of Neurology, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
66
|
Brandt MM, Cheng C, Merkus D, Duncker DJ, Sorop O. Mechanobiology of Microvascular Function and Structure in Health and Disease: Focus on the Coronary Circulation. Front Physiol 2022; 12:771960. [PMID: 35002759 PMCID: PMC8733629 DOI: 10.3389/fphys.2021.771960] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 11/11/2021] [Indexed: 12/19/2022] Open
Abstract
The coronary microvasculature plays a key role in regulating the tight coupling between myocardial perfusion and myocardial oxygen demand across a wide range of cardiac activity. Short-term regulation of coronary blood flow in response to metabolic stimuli is achieved via adjustment of vascular diameter in different segments of the microvasculature in conjunction with mechanical forces eliciting myogenic and flow-mediated vasodilation. In contrast, chronic adjustments in flow regulation also involve microvascular structural modifications, termed remodeling. Vascular remodeling encompasses changes in microvascular diameter and/or density being largely modulated by mechanical forces acting on the endothelium and vascular smooth muscle cells. Whereas in recent years, substantial knowledge has been gathered regarding the molecular mechanisms controlling microvascular tone and how these are altered in various diseases, the structural adaptations in response to pathologic situations are less well understood. In this article, we review the factors involved in coronary microvascular functional and structural alterations in obstructive and non-obstructive coronary artery disease and the molecular mechanisms involved therein with a focus on mechanobiology. Cardiovascular risk factors including metabolic dysregulation, hypercholesterolemia, hypertension and aging have been shown to induce microvascular (endothelial) dysfunction and vascular remodeling. Additionally, alterations in biomechanical forces produced by a coronary artery stenosis are associated with microvascular functional and structural alterations. Future studies should be directed at further unraveling the mechanisms underlying the coronary microvascular functional and structural alterations in disease; a deeper understanding of these mechanisms is critical for the identification of potential new targets for the treatment of ischemic heart disease.
Collapse
Affiliation(s)
- Maarten M Brandt
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Caroline Cheng
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands.,Division of Internal Medicine and Dermatology, Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, Netherlands
| | - Daphne Merkus
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands.,Walter Brendel Center of Experimental Medicine (WBex), LMU Munich, Munich, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance (MHA), Munich, Germany
| | - Dirk J Duncker
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Oana Sorop
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| |
Collapse
|
67
|
The essential role for endothelial cell sprouting in coronary collateral growth. J Mol Cell Cardiol 2022; 165:158-171. [PMID: 35074317 PMCID: PMC8940680 DOI: 10.1016/j.yjmcc.2022.01.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 01/11/2022] [Accepted: 01/16/2022] [Indexed: 12/11/2022]
Abstract
RATIONALE Coronary collateral growth is a natural bypass for ischemic heart diseases. It offers tremendous therapeutic benefit, but the process of coronary collateral growth isincompletely understood due to limited preclinical murine models that would enable interrogation of its mechanisms and processes via genetic modification and lineage tracing. Understanding the processes by which coronary collaterals develop can unlock new therapeutic strategies for ischemic heart disease. OBJECTIVE To develop a murine model of coronary collateral growth by repetitive ischemia and investigate whether capillary endothelial cells could contribute to the coronary collateral formation in an adult mouse heart after repetitive ischemia by lineage tracing. METHODS AND RESULTS A murine model of coronary collateral growth was developed using short episodes of repetitive ischemia. Repetitive ischemia stimulation resulted in robust collateral growth in adult mouse hearts, validated by high-resolution micro-computed tomography. Repetitive ischemia-induced collateral formation compensated ischemia caused by occlusion of the left anterior descending artery. Cardiac function improved during ischemia after repetitive ischemia, suggesting the improvement of coronary blood flow. A capillary-specific Cre driver (Apln-CreER) was used for lineage tracing capillary endothelial cells. ROSA mT/mG reporter mice crossed with the Apln-CreER transgene mice underwent a 17 days' repetitive ischemia protocol for coronary collateral growth. Two-photon and confocal microscopy imaging of heart slices revealed repetitive ischemia-induced coronary collateral growth initiated from sprouting Apelin+ endothelial cells. Newly formed capillaries in the collateral-dependent zone expanded in diameter upon repetitive ischemia stimulation and arterialized with smooth muscle cell recruitment, forming mature coronary arteries. Notably, pre-existing coronary arteries and arterioles were not Apelin+, and all Apelin+ collaterals arose from sprouting capillaries. Cxcr4, Vegfr2, Jag1, Mcp1, and Hif1⍺ mRNA levels in the repetitive ischemia-induced hearts were also upregulated at the early stage of coronary collateral growth, suggesting angiogenic signaling pathways are activated for coronary collaterals formation during repetitive ischemia. CONCLUSIONS We developed a murine model of coronary collateral growth induced by repetitive ischemia. Our lineage tracing study shows that sprouting endothelial cells contribute to coronary collateral growth in adult mouse hearts. For the first time, sprouting angiogenesis is shown to give rise to mature coronary arteries in response to repetitive ischemia in the adult mouse hearts.
Collapse
|
68
|
El Hadri K, Smith R, Duplus E, El Amri C. Inflammation, Oxidative Stress, Senescence in Atherosclerosis: Thioredoxine-1 as an Emerging Therapeutic Target. Int J Mol Sci 2021; 23:ijms23010077. [PMID: 35008500 PMCID: PMC8744732 DOI: 10.3390/ijms23010077] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/19/2021] [Accepted: 12/19/2021] [Indexed: 02/07/2023] Open
Abstract
Atherosclerosis is a leading cause of cardiovascular diseases (CVD) worldwide and intimately linked to aging. This pathology is characterized by chronic inflammation, oxidative stress, gradual accumulation of low-density lipoproteins (LDL) particles and fibrous elements in focal areas of large and medium arteries. These fibrofatty lesions in the artery wall become progressively unstable and thrombogenic leading to heart attack, stroke or other severe heart ischemic syndromes. Elevated blood levels of LDL are major triggering events for atherosclerosis. A cascade of molecular and cellular events results in the atherosclerotic plaque formation, evolution, and rupture. Moreover, the senescence of multiple cell types present in the vasculature were reported to contribute to atherosclerotic plaque progression and destabilization. Classical therapeutic interventions consist of lipid-lowering drugs, anti-inflammatory and life style dispositions. Moreover, targeting oxidative stress by developing innovative antioxidant agents or boosting antioxidant systems is also a well-established strategy. Accumulation of senescent cells (SC) is also another important feature of atherosclerosis and was detected in various models. Hence, targeting SCs appears as an emerging therapeutic option, since senolytic agents favorably disturb atherosclerotic plaques. In this review, we propose a survey of the impact of inflammation, oxidative stress, and senescence in atherosclerosis; and the emerging therapeutic options, including thioredoxin-based approaches such as anti-oxidant, anti-inflammatory, and anti-atherogenic strategy with promising potential of senomodulation.
Collapse
|
69
|
Mao L, Mostafa R, Ibili E, Fert-Bober J. Role of protein deimination in cardiovascular diseases: potential new avenues for diagnostic and prognostic biomarkers. Expert Rev Proteomics 2021; 18:1059-1071. [PMID: 34929115 DOI: 10.1080/14789450.2021.2018303] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Arginine deimination (citrullination) is a post-translational modification catalyzed by a family of peptidyl arginine deiminase (PAD) enzymes. Cell-based functional studies and animal models have manifested the key role of PADs in various cardiovascular diseases (CVDs). AREA COVERED This review summarizes the latest developments in the role of PADs in CVD pathogenesis. It focuses on the PAD functions and diverse citrullinated proteins in cardiovascular conditions like deep vein thrombosis, ischemia/reperfusion, and atherosclerosis. Identification of PAD isoforms and citrullinated targets are essential for directing diagnosis and clinical intervention. Finally, anti-citrullinated protein antibodies (ACPAs) are addressed as an independent risk factor for cardiovascular events. A search of PubMed biomedical literature from the past ten years was performed with a combination of the following keywords: PAD/PADI, deimination/citrullination, autoimmune, fibrosis, NET, neutrophil, macrophage, inflammation, inflammasome, cardiovascular, heart disease, myocardial infarction, ischemia, atherosclerosis, thrombosis, and aging. Additional papers from retrieved articles were also considered. EXPERT OPINION PADs are unique family of enzymes that converts peptidyl-arginine to -citrulline in protein permanently. Overexpression or increased activity of PAD has been observed in various CVDs with acute and chronic inflammation as the background. Importantly, far beyond being simply involved in forming neutrophil extracellular traps (NETs), accumulating evidence indicated PAD activation as a trigger for numerous processes, such as transcriptional regulation, endothelial dysfunction, and thrombus formation. In summary, the findings so far have testified the important role of deimination in cardiovascular biology, while more basic and translational studies are essential to further exploration.
Collapse
Affiliation(s)
- Liqun Mao
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Advanced Clinical Biosystems Institute, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Rowann Mostafa
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Advanced Clinical Biosystems Institute, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Esra Ibili
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Advanced Clinical Biosystems Institute, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Justyna Fert-Bober
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Advanced Clinical Biosystems Institute, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
70
|
Yuan M, Wang Y, Wang S, Huang Z, Jin F, Zou Q, Li J, Pu Y, Cai Z. Bioenergetic Impairment in the Neuro-Glia-Vascular Unit: An Emerging Physiopathology during Aging. Aging Dis 2021; 12:2080-2095. [PMID: 34881087 PMCID: PMC8612602 DOI: 10.14336/ad.2021.04017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 04/17/2021] [Indexed: 12/28/2022] Open
Abstract
An emerging concept termed the "neuro-glia-vascular unit" (NGVU) has been established in recent years to understand the complicated mechanism of multicellular interactions among vascular cells, glial cells, and neurons. It has been proverbially reported that the NGVU is significantly associated with neurodegenerative disorders, such as Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS). Physiological aging is an inevitable progression associated with oxidative damage, bioenergetic alterations, mitochondrial dysfunction, and neuroinflammation, which is partially similar to the pathology of AD. Thus, senescence is regarded as the background for the development of neurodegenerative diseases. With the exacerbation of global aging, senescence is an increasingly serious problem in the medical field. In this review, the coupling of each component, including neurons, glial cells, and vascular cells, in the NGVU is described in detail. Then, various mechanisms of age-dependent impairment in each part of the NGVU are discussed. Moreover, the potential bioenergetic alterations between different cell types in the NGVU are highlighted, which seems to be an emerging physiopathology associated with the aged brain. Bioenergetic intervention in the NGVU may be a new direction for studies on delaying or diminishing aging in the future.
Collapse
Affiliation(s)
- Minghao Yuan
- 1Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, 400013, Chongqing, China.,2Chongqing School, University of Chinese Academy of Sciences, Chongqing, China.,3Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, Chongqing, China.,4Chongqing Medical University, Chongqing, China
| | - Yangyang Wang
- 1Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, 400013, Chongqing, China.,3Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, Chongqing, China
| | - Shengyuan Wang
- 1Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, 400013, Chongqing, China.,2Chongqing School, University of Chinese Academy of Sciences, Chongqing, China.,3Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, Chongqing, China.,4Chongqing Medical University, Chongqing, China
| | - Zhenting Huang
- 1Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, 400013, Chongqing, China.,3Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, Chongqing, China
| | - Feng Jin
- 1Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, 400013, Chongqing, China.,2Chongqing School, University of Chinese Academy of Sciences, Chongqing, China.,3Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, Chongqing, China
| | - Qian Zou
- 1Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, 400013, Chongqing, China.,3Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, Chongqing, China
| | - Jing Li
- 1Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, 400013, Chongqing, China.,3Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, Chongqing, China
| | - Yinshuang Pu
- 1Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, 400013, Chongqing, China.,3Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, Chongqing, China
| | - Zhiyou Cai
- 1Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, 400013, Chongqing, China.,2Chongqing School, University of Chinese Academy of Sciences, Chongqing, China.,3Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, Chongqing, China.,4Chongqing Medical University, Chongqing, China
| |
Collapse
|
71
|
Yeo HS, Lim JY. Effects of different types of exercise training on angiogenic responses in the left ventricular muscle of aged rats. Exp Gerontol 2021; 158:111650. [PMID: 34890715 DOI: 10.1016/j.exger.2021.111650] [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: 07/24/2021] [Revised: 11/09/2021] [Accepted: 12/01/2021] [Indexed: 11/16/2022]
Abstract
BACKGROUND We evaluated angiogenic responses in the left ventricular muscle and aerobic capacity according to exercise type (aerobic, resistance, combined) in aged rats. METHODS In total, 24 male Sprague-Dawley rats (100 weeks old) were used. To investigate the effect of regular training, the rats were divided into non-exercise (NE), aerobic exercise (AE), resistance exercise (RE), and combined exercise (CE) groups (six rats per group). Regular training tailored to each exercise type was performed for 8 weeks (five times a week, 1 h per day). After 8 weeks of training, aerobic capacity was evaluated by a treadmill running test. Left ventricular muscle tissue was collected and the protein levels of angiogenesis indicators (eNOS, HIF-1α, PGC-1α, VEGF, FLK-1, Ang-1, Ang-2) were analyzed by Western blotting. Capillaries were observed by immunohistochemical staining for CD31. RESULTS Body weight, heart weight, and heart/body weight ratio showed no difference among the groups. The AE and CE groups showed higher treadmill running capacity than the NE and RE groups. The eNOS, VEGF, HIF-1α, PGC-1α, and Ang-2 protein levels were significantly higher in the AE than NE group. The PGC-1α and FLK-1 protein levels were significantly higher in the RE than NE group. In addition, in the CE group, the eNOS, FLK-1, and PGC-1α protein levels were significantly higher than in the NE group. Expression of CD31 in cardiac tissue was higher in the AE and CE groups than in the other groups. CONCLUSIONS Taken together, the results suggest that regular exercise training, irrespective of exercise type, might improve cardiovascular function by inducing angiogenic responses in the aged myocardium; however, AE may be the most effective.
Collapse
Affiliation(s)
- Hyo-Seong Yeo
- Department of Rehabilitation Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea; Seoul National University Institute on Aging, Seoul, South Korea; Aging & Mobility Biophysics Laboratory, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Jae-Young Lim
- Department of Rehabilitation Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea; Seoul National University Institute on Aging, Seoul, South Korea; Aging & Mobility Biophysics Laboratory, Seoul National University Bundang Hospital, Seongnam, South Korea.
| |
Collapse
|
72
|
Lu B, Huang L, Cao J, Li L, Wu W, Chen X, Ding C. Adipose tissue macrophages in aging-associated adipose tissue function. J Physiol Sci 2021; 71:38. [PMID: 34863096 PMCID: PMC10717320 DOI: 10.1186/s12576-021-00820-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 11/04/2021] [Indexed: 12/29/2022]
Abstract
"Inflammaging" refers to the chronic, low-grade inflammation that characterizes aging. Aging, like obesity, is associated with visceral adiposity and insulin resistance. Adipose tissue macrophages (ATMs) have played a major role in obesity-associated inflammation and insulin resistance. Macrophages are elevated in adipose tissue in aging. However, the changes and also possibly functions of ATMs in aging and aging-related diseases are unclear. In this review, we will summarize recent advances in research on the role of adipose tissue macrophages with aging-associated insulin resistance and discuss their potential therapeutic targets for preventing and treating aging and aging-related diseases.
Collapse
Affiliation(s)
- Bangchao Lu
- Department of Geriatrics, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangshu, China
| | - Liang Huang
- Department of Geriatrics, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangshu, China
| | - Juan Cao
- Department of Geriatrics, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangshu, China
| | - Lingling Li
- Department of Geriatrics, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangshu, China
| | - Wenhui Wu
- Department of Geriatrics, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangshu, China
| | - Xiaolin Chen
- Department of Geriatrics, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangshu, China
| | - Congzhu Ding
- Department of Geriatrics, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangshu, China.
| |
Collapse
|
73
|
Qi X, Ricart K, Ahmed KA, Patel RP, Boulton ME. Supplemental nitrite increases choroidal neovascularization in mice. Nitric Oxide 2021; 117:7-15. [PMID: 34537345 DOI: 10.1016/j.niox.2021.09.005] [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: 07/24/2021] [Revised: 09/09/2021] [Accepted: 09/14/2021] [Indexed: 10/20/2022]
Abstract
Low doses of nitrite, close to physiological levels, increase blood flow in normal and ischemic tissues through a nitric oxide (NO) dependent mechanism. Given that nitrite therapy and dietary supplementation with vegetables high in nitrate (e.g. beets) are gaining popularity we decided to determine if low doses of nitrite impact the development of choroidal neovascularization (CNV), a key feature of wet age related macular degeneration (AMD). Sodium nitrite (at 50 mg/L, 150 mg/L, and 300 mg/L), nitrate (1 g/L) or water alone were provided in the drinking water of C57BL/6 J mice aged 2 or 12 months. Mice were allowed to drink ad libitum for 1 week at which time laser-induced choroidal neovascularization (L-CNV) was induced. The mice continued to drink the supplemented water ad libitum for a further 14 days at which point optical coherence tomography (OCT) was performed to determine the volume of the CNV lesion. Blood was drawn to determine nitrite and nitrate levels and eyes taken for histology. CNV volume was 2.86 × 107 μm3 (±0.4 × 107) in young mice on water alone but CNV volume more than doubled to >6.9 × 107 μm3 (±0.8 × 107) in mice receiving 300 mg/L nitrite or 7.34 × 107 μm3 (±1.4 × 107) in 1 g/L nitrate (p < 0.01). A similar trend was observed in older mice. CNV volume was 5.3 × 107 μm3 (±0.5 × 107) in older mice on water alone but CNV volume almost doubled to approximately 9.3 × 107 μm3 (±1.1 × 107) in mice receiving 300 mg/L nitrite or 8.7 × 107 μm3 (±0.9 × 107) 1 g/L nitrate (p < 0.01). Plasma nitrite levels were highest in young mice receiving 150 mg/L in the drinking water with no changes in plasma nitrate observed. In older mice, drinking water nitrite did not significantly change plasma nitrite, but plasma nitrate was increased. Plasma nitrate was elevated in both young and old mice provided with nitrate supplemented drinking water. Our data demonstrate that the CNV lesion is larger in older mice compared to young and that therapeutic levels of oral nitrite increase the volume of CNV lesions in both young and older mice. Therapeutic nitrite or nitrate supplementation should be used with caution in the elderly population prone to CNV.
Collapse
Affiliation(s)
- Xiaoping Qi
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, AL, 35294, USA
| | - Karina Ricart
- Department of Pathology and Center for Free Radical Biology, University of Alabama at Birmingham, AL, 35294, USA
| | - Khandaker A Ahmed
- Department of Pathology and Center for Free Radical Biology, University of Alabama at Birmingham, AL, 35294, USA
| | - Rakesh P Patel
- Department of Pathology and Center for Free Radical Biology, University of Alabama at Birmingham, AL, 35294, USA.
| | - Michael E Boulton
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, AL, 35294, USA.
| |
Collapse
|
74
|
Wang G, Han B, Zhang R, Liu Q, Wang X, Huang X, Liu D, Qiao W, Yang M, Luo X, Hou J, Yu B. C1q/TNF-Related Protein 9 Attenuates Atherosclerosis by Inhibiting Hyperglycemia-Induced Endothelial Cell Senescence Through the AMPKα/KLF4 Signaling Pathway. Front Pharmacol 2021; 12:758792. [PMID: 34744738 PMCID: PMC8569937 DOI: 10.3389/fphar.2021.758792] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Accepted: 10/07/2021] [Indexed: 12/18/2022] Open
Abstract
Hyperglycemia-induced endothelial cell senescence has been widely reported to be involved in the pathogenesis of type 2 diabetes mellitus‒accelerated atherosclerosis. Thus, understanding the underlying mechanisms and identifying potential therapeutic targets for endothelial cell senescence are valuable for attenuating atherosclerosis progression. C1q/tumor necrosis factor-related protein 9 (CTRP9), an emerging potential cardiokine, exerts a significant protective effect with respect to atherosclerosis, particularly in endothelial cells. However, the exact mechanism by which CTRP9 prevents endothelial cells from hyperglycemia-induced senescence remains unclear. This study aimed to investigate the effects of CTRP9 on hyperglycemia-induced endothelial cell senescence and atherosclerotic plaque formation in diabetic apolipoprotein E knockout (ApoE KO) mice. Human umbilical vein endothelial cells (HUVECs) were cultured in normal glucose (5.5 mM) and high glucose (40 mM) with or without recombinant human CTRP9 protein (3 μg/ml) for 48 h. Purified lentiviruses overexpressing CTRP9 (Lv-CTRP9) and control vectors containing green fluorescent protein (Lv-GFP) were injected via the tail vein into streptozotocin-induced diabetic ApoE KO mice. Results revealed that exposure of HUVECs to HG significantly increased the expression of Krüppel-like factor 4 (KLF4) and cyclin-dependent kinase inhibitor p21 (p21) and decreased that of telomerase reverse transcriptase (TERT). Treatment with recombinant human CTRP9 protein protected HUVECs from HG-induced premature senescence and dysfunction. CTRP9 promoted the phosphorylation of AMP-activated kinase (AMPK), attenuated the expression of KLF4 and p21 induced by HG, and increased the expression of TERT in HUVECs. Furthermore, in the background of AMPKα knockdown or KLF4 activation, the protective effects of CTRP9 were abolished. In-vivo experiments showed that the overexpression of CTRP9 inhibited vascular senescence and reduced atherosclerotic plaque formation in ApoE KO mice with diabetes. In conclusion, we demonstrate that KLF4 upregulation plays a crucial role in HG-induced endothelial senescence. This anti-atherosclerotic effect of CTRP9 may be partly attributed to the inhibition of HG-induced endothelial senescence through an AMPKα/KLF4-dependent mechanism, suggesting that CTRP9 could benefit further therapeutic approaches for type 2 diabetes mellitus‒accelerated atherosclerosis.
Collapse
Affiliation(s)
- Gang Wang
- The Key Laboratory of Myocardial Ischemia Organization, Chinese Ministry of Education, Harbin, China.,Department of Cardiology Organization, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Baihe Han
- The Key Laboratory of Myocardial Ischemia Organization, Chinese Ministry of Education, Harbin, China.,Department of Cardiology Organization, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ruoxi Zhang
- Department of Cardiology, Harbin Yinghua Hospital, Harbin, China
| | - Qi Liu
- The Key Laboratory of Myocardial Ischemia Organization, Chinese Ministry of Education, Harbin, China.,Department of Cardiology Organization, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xuedong Wang
- The Key Laboratory of Myocardial Ischemia Organization, Chinese Ministry of Education, Harbin, China.,Department of Cardiology Organization, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xingtao Huang
- The Key Laboratory of Myocardial Ischemia Organization, Chinese Ministry of Education, Harbin, China.,Department of Cardiology Organization, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Dandan Liu
- The Key Laboratory of Myocardial Ischemia Organization, Chinese Ministry of Education, Harbin, China.,Department of Cardiology Organization, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Weishen Qiao
- The Key Laboratory of Myocardial Ischemia Organization, Chinese Ministry of Education, Harbin, China.,Department of Cardiology Organization, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Mengyue Yang
- The Key Laboratory of Myocardial Ischemia Organization, Chinese Ministry of Education, Harbin, China.,Department of Cardiology Organization, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xing Luo
- The Key Laboratory of Myocardial Ischemia Organization, Chinese Ministry of Education, Harbin, China.,Department of Cardiology Organization, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jingbo Hou
- The Key Laboratory of Myocardial Ischemia Organization, Chinese Ministry of Education, Harbin, China.,Department of Cardiology Organization, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bo Yu
- The Key Laboratory of Myocardial Ischemia Organization, Chinese Ministry of Education, Harbin, China.,Department of Cardiology Organization, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
75
|
Naqvi N, Iismaa SE, Graham RM, Husain A. Mechanism-Based Cardiac Regeneration Strategies in Mammals. Front Cell Dev Biol 2021; 9:747842. [PMID: 34708043 PMCID: PMC8542766 DOI: 10.3389/fcell.2021.747842] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 09/06/2021] [Indexed: 12/11/2022] Open
Abstract
Heart failure in adults is a leading cause of morbidity and mortality worldwide. It can arise from a variety of diseases, with most resulting in a loss of cardiomyocytes that cannot be replaced due to their inability to replicate, as well as to a lack of resident cardiomyocyte progenitor cells in the adult heart. Identifying and exploiting mechanisms underlying loss of developmental cardiomyocyte replicative capacity has proved to be useful in developing therapeutics to effect adult cardiac regeneration. Of course, effective regeneration of myocardium after injury requires not just expansion of cardiomyocytes, but also neovascularization to allow appropriate perfusion and resolution of injury-induced inflammation and interstitial fibrosis, but also reversal of adverse left ventricular remodeling. In addition to overcoming these challenges, a regenerative therapy needs to be safe and easily translatable. Failure to address these critical issues will delay the translation of regenerative approaches. This review critically analyzes current regenerative approaches while also providing a framework for future experimental studies aimed at enhancing success in regenerating the injured heart.
Collapse
Affiliation(s)
- Nawazish Naqvi
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States
| | - Siiri E Iismaa
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
| | - Robert M Graham
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
| | - Ahsan Husain
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
76
|
Palmer A, Epton S, Crawley E, Straface M, Gammon L, Edgar MM, Xu Y, Elahi S, Chin-Aleong J, Martin JE, Bishop CL, Knowles CH, Sanger GJ. Expression of p16 Within Myenteric Neurons of the Aged Colon: A Potential Marker of Declining Function. Front Neurosci 2021; 15:747067. [PMID: 34690683 PMCID: PMC8529329 DOI: 10.3389/fnins.2021.747067] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 09/14/2021] [Indexed: 01/31/2023] Open
Abstract
Human colonic neuromuscular functions decline among the elderly. The aim was to explore the involvement of senescence. A preliminary PCR study looked for age-dependent differences in expression of CDKN1A (encoding the senescence-related p21 protein) and CDKN2A (encoding p16 and p14) in human ascending and descending colon (without mucosa) from 39 (approximately 50: 50 male: female) adult (aged 27–60 years) and elderly donors (70–89 years). Other genes from different aging pathways (e.g., inflammation, oxidative stress, autophagy) and cell-types (e.g., neurons, neuron axonal transport) were also examined. Unlike CDKN1A, CDKN2A (using primers for p16 and p14 but not when using p14-specific primers) was upregulated in both regions of colon. Compared with the number of genes appearing to upregulate in association with temporal age, more genes positively associated with increased CDKN2A expression (respectively, 16 and five of 44 genes studied for ascending and descending colon). Confirmation of increased expression of CDKN2A was sought by immunostaining for p16 in the myenteric plexus of colon from 52 patients, using a semi-automated software protocol. The results showed increased staining not within the glial cells (S100 stained), but in the cytoplasm of myenteric nerve cell bodies (MAP2 stained, with identified nucleus) of ascending, but not descending colon of the elderly, and not in the cell nucleus of either region or age group (5,710 neurons analyzed: n = 12–14 for each group). It was concluded that increased p16 staining within the cytoplasm of myenteric nerve cell bodies of elderly ascending (but not descending) colon, suggests a region-dependent, post-mitotic cellular senescence-like activity, perhaps involved with aging of enteric neurons within the colon.
Collapse
Affiliation(s)
- Alexandra Palmer
- Center for Neuroscience, Surgery and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Sarah Epton
- Barts Health NHS Trust, Department of Colorectal Surgery and Pathology, The Royal London Hospital, London, United Kingdom
| | - Ellie Crawley
- Center for Neuroscience, Surgery and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Marilisa Straface
- Center for Neuroscience, Surgery and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Luke Gammon
- Center for Cell Biology and Cutaneous Research, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Meghan M Edgar
- Gastroenterology Drug Discovery Unit, Takeda Pharmaceuticals, San Diego, CA, United States
| | - Yichen Xu
- Center for Neuroscience, Surgery and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Shezan Elahi
- Center for Neuroscience, Surgery and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Joanne Chin-Aleong
- Barts Health NHS Trust, Department of Colorectal Surgery and Pathology, The Royal London Hospital, London, United Kingdom
| | - Joanne E Martin
- Center for Neuroscience, Surgery and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom.,Barts Health NHS Trust, Department of Colorectal Surgery and Pathology, The Royal London Hospital, London, United Kingdom
| | - Cleo L Bishop
- Center for Cell Biology and Cutaneous Research, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom.,Center for Inflammation and Therapeutic Innovation Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Charles H Knowles
- Center for Neuroscience, Surgery and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom.,Barts Health NHS Trust, Department of Colorectal Surgery and Pathology, The Royal London Hospital, London, United Kingdom
| | - Gareth J Sanger
- Center for Neuroscience, Surgery and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
77
|
Forestalling age-impaired angiogenesis and blood flow by targeting NOX: Interplay of NOX1, IL-6, and SASP in propagating cell senescence. Proc Natl Acad Sci U S A 2021; 118:2015666118. [PMID: 34654740 DOI: 10.1073/pnas.2015666118] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/20/2021] [Indexed: 12/29/2022] Open
Abstract
In an aging population, intense interest has shifted toward prolonging health span. Mounting evidence suggests that cellular reactive species are propagators of cell damage, inflammation, and cellular senescence. Thus, such species have emerged as putative provocateurs and targets for senolysis, and a clearer understanding of their molecular origin and regulation is of paramount importance. In an inquiry into signaling triggered by aging and proxy instigator, hyperglycemia, we show that NADPH Oxidase (NOX) drives cell DNA damage and alters nuclear envelope integrity, inflammation, tissue dysfunction, and cellular senescence in mice and humans with similar causality. Most notably, selective NOX1 inhibition rescues age-impaired blood flow and angiogenesis, vasodilation, and the endothelial cell wound response. Indeed, NOX1i delivery in vivo completely reversed age-impaired hind-limb blood flow and angiogenesis while disrupting a NOX1-IL-6 senescence-associated secretory phenotype (SASP) proinflammatory signaling loop. Relevant to its comorbidity with age, clinical samples from diabetic versus nondiabetic subjects reveal as operant this NOX1-mediated vascular senescence and inflammation in humans. On a mechanistic level, our findings support a previously unidentified role for IL-6 in this feedforward inflammatory loop and peroxisome proliferator-activated receptor gamma (PPARγ) down-regulation as inversely modulating p65-mediated NOX1 transcription. Targeting this previously unidentified NOX1-SASP signaling axis in aging is predicted to be an effective strategy for mitigating senescence in the vasculature and other organ systems.
Collapse
|
78
|
Dadwal UC, Bhatti FUR, Awosanya OD, de Andrade Staut C, Nagaraj RU, Perugini AJ, Tewari NP, Valuch CR, Sun S, Mendenhall SK, Zhou D, Mostardo SL, Blosser RJ, Li J, Kacena MA. The Effects of SRT1720 Treatment on Endothelial Cells Derived from the Lung and Bone Marrow of Young and Aged, Male and Female Mice. Int J Mol Sci 2021; 22:11097. [PMID: 34681756 PMCID: PMC8540697 DOI: 10.3390/ijms222011097] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/10/2021] [Accepted: 10/10/2021] [Indexed: 02/07/2023] Open
Abstract
Angiogenesis is critical for successful fracture healing. Age-related alterations in endothelial cells (ECs) may cause impaired bone healing. Therefore, examining therapeutic treatments to improve angiogenesis in aging may enhance bone healing. Sirtuin 1 (SIRT1) is highly expressed in ECs and its activation is known to counteract aging. Here, we examined the effects of SRT1720 treatment (SIRT1 activator) on the growth and function of bone marrow and lung ECs (BMECs and LECs, respectively), derived from young (3-4 month) and old (20-24 month) mice. While aging did not alter EC proliferation, treatment with SRT1720 significantly increased proliferation of all LECs. However, SRT1720 only increased proliferation of old female BMECs. Vessel-like tube assays showed similar vessel-like structures between young and old LECs and BMECs from both male and female mice. SRT1720 significantly improved vessel-like structures in all LECs. No age, sex, or treatment differences were found in migration related parameters of LECs. In males, old BMECs had greater migration rates than young BMECs, whereas in females, old BMECs had lower migration rates than young BMECs. Collectively, our data suggest that treatment with SRT1720 appears to enhance the angiogenic potential of LECs irrespective of age or sex. However, its role in BMECs is sex- and age-dependent.
Collapse
Affiliation(s)
- Ushashi Chand Dadwal
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (U.C.D.); (F.U.R.B.); (O.D.A.); (C.d.A.S.); (R.U.N.); (A.J.P.III); (N.P.T.); (S.S.); (S.K.M.); (D.Z.); (S.L.M.); (R.J.B.)
- Richard L. Roudebush VA Medical Center, Indianapolis, IN 46202, USA
| | - Fazal Ur Rehman Bhatti
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (U.C.D.); (F.U.R.B.); (O.D.A.); (C.d.A.S.); (R.U.N.); (A.J.P.III); (N.P.T.); (S.S.); (S.K.M.); (D.Z.); (S.L.M.); (R.J.B.)
| | - Olatundun Dupe Awosanya
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (U.C.D.); (F.U.R.B.); (O.D.A.); (C.d.A.S.); (R.U.N.); (A.J.P.III); (N.P.T.); (S.S.); (S.K.M.); (D.Z.); (S.L.M.); (R.J.B.)
| | - Caio de Andrade Staut
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (U.C.D.); (F.U.R.B.); (O.D.A.); (C.d.A.S.); (R.U.N.); (A.J.P.III); (N.P.T.); (S.S.); (S.K.M.); (D.Z.); (S.L.M.); (R.J.B.)
| | - Rohit U. Nagaraj
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (U.C.D.); (F.U.R.B.); (O.D.A.); (C.d.A.S.); (R.U.N.); (A.J.P.III); (N.P.T.); (S.S.); (S.K.M.); (D.Z.); (S.L.M.); (R.J.B.)
| | - Anthony Joseph Perugini
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (U.C.D.); (F.U.R.B.); (O.D.A.); (C.d.A.S.); (R.U.N.); (A.J.P.III); (N.P.T.); (S.S.); (S.K.M.); (D.Z.); (S.L.M.); (R.J.B.)
| | - Nikhil Prasad Tewari
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (U.C.D.); (F.U.R.B.); (O.D.A.); (C.d.A.S.); (R.U.N.); (A.J.P.III); (N.P.T.); (S.S.); (S.K.M.); (D.Z.); (S.L.M.); (R.J.B.)
| | - Conner Riley Valuch
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA; (C.R.V.); (J.L.)
| | - Seungyup Sun
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (U.C.D.); (F.U.R.B.); (O.D.A.); (C.d.A.S.); (R.U.N.); (A.J.P.III); (N.P.T.); (S.S.); (S.K.M.); (D.Z.); (S.L.M.); (R.J.B.)
| | - Stephen Kyle Mendenhall
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (U.C.D.); (F.U.R.B.); (O.D.A.); (C.d.A.S.); (R.U.N.); (A.J.P.III); (N.P.T.); (S.S.); (S.K.M.); (D.Z.); (S.L.M.); (R.J.B.)
| | - Donghui Zhou
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (U.C.D.); (F.U.R.B.); (O.D.A.); (C.d.A.S.); (R.U.N.); (A.J.P.III); (N.P.T.); (S.S.); (S.K.M.); (D.Z.); (S.L.M.); (R.J.B.)
| | - Sarah Lyn Mostardo
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (U.C.D.); (F.U.R.B.); (O.D.A.); (C.d.A.S.); (R.U.N.); (A.J.P.III); (N.P.T.); (S.S.); (S.K.M.); (D.Z.); (S.L.M.); (R.J.B.)
| | - Rachel Jean Blosser
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (U.C.D.); (F.U.R.B.); (O.D.A.); (C.d.A.S.); (R.U.N.); (A.J.P.III); (N.P.T.); (S.S.); (S.K.M.); (D.Z.); (S.L.M.); (R.J.B.)
- Richard L. Roudebush VA Medical Center, Indianapolis, IN 46202, USA
| | - Jiliang Li
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA; (C.R.V.); (J.L.)
| | - Melissa Ann Kacena
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (U.C.D.); (F.U.R.B.); (O.D.A.); (C.d.A.S.); (R.U.N.); (A.J.P.III); (N.P.T.); (S.S.); (S.K.M.); (D.Z.); (S.L.M.); (R.J.B.)
- Richard L. Roudebush VA Medical Center, Indianapolis, IN 46202, USA
| |
Collapse
|
79
|
Li B, Chen K, Liu F, Zhang J, Chen X, Chen T, Chen Q, Yao Y, Hu W, Wang L, Wu Y. Developmental Angiogenesis Requires the Mitochondrial Phenylalanyl-tRNA Synthetase. Front Cardiovasc Med 2021; 8:724846. [PMID: 34540921 PMCID: PMC8440837 DOI: 10.3389/fcvm.2021.724846] [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: 06/14/2021] [Accepted: 08/04/2021] [Indexed: 12/03/2022] Open
Abstract
Background: Mitochondrial aminoacyl-tRNA synthetases (mtARSs) catalyze the binding of specific amino acids to their cognate tRNAs and play an essential role in the synthesis of proteins encoded by mitochondrial DNA. Defects in mtARSs have been linked to human diseases, but their tissue-specific pathophysiology remains elusive. Here we examined the role of mitochondrial phenylalanyl-tRNA synthetase (FARS2) in developmental angiogenesis and its potential contribution to the pathogenesis of cardiovascular disease. Methods: Morpholinos were injected into fertilized zebrafish ova to establish an in vivo fars2 knock-down model. A visualization of the vasculature was achieved by using Tg (fli1: EGFP)y1 transgenic zebrafish. In addition, small interference RNAs (siRNAs) were transferred into human umbilical vein endothelial cells (HUVECs) to establish an in vitro FARS2 knock-down model. Cell motility, proliferation, and tubulogenesis were determined using scratch-wound CCK8, transwell-based migration, and tube formation assays. In addition, mitochondria- and non-mitochondria-related respiration were evaluated using a Seahorse XF24 analyzer and flow cytometry assays. Analyses of the expression levels of transcripts and proteins were performed using qRT-PCR and western blotting, respectively. Results: The knock-down of fars2 hampered the embryonic development in zebrafish and delayed the formation of the vasculature in Tg (fli1: EGFP)y1 transgenic zebrafish. In addition, the siRNA-mediated knock-down of FARS2 impaired angiogenesis in HUVECs as indicated by decreased cell motility and tube formation capacity. The knock-down of FARS2 also produced variable decreases in mitochondrial- and non-mitochondrial respiration in HUVECs and disrupted the regulatory pathways of angiogenesis in both HUVECs and zebrafish. Conclusion: Our current work offers novel insights into angiogenesis defects and cardiovascular diseases induced by FARS2 deficiency.
Collapse
Affiliation(s)
- Bowen Li
- Department of Biochemistry and Molecular Biology, Air Force Medical University, Xi'an, China.,Shaanxi Provincial Key Laboratory of Clinic Genetics, Air Force Medical University, Xi'an, China
| | - Kun Chen
- Department of Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, Air Force Medical University, Xi'an, China
| | - Fangfang Liu
- Department of Neurosciences, Air Force Medical University, Xi'an, China
| | - Juan Zhang
- Department of Biochemistry and Molecular Biology, College of Life Sciences, Northwest University, Xi'an, China
| | - Xihui Chen
- Department of Biochemistry and Molecular Biology, Air Force Medical University, Xi'an, China.,Shaanxi Provincial Key Laboratory of Clinic Genetics, Air Force Medical University, Xi'an, China
| | - Tangdong Chen
- Department of Biochemistry and Molecular Biology, Air Force Medical University, Xi'an, China.,Shaanxi Provincial Key Laboratory of Clinic Genetics, Air Force Medical University, Xi'an, China
| | - Qi Chen
- Department of Biochemistry and Molecular Biology, Air Force Medical University, Xi'an, China.,Shaanxi Provincial Key Laboratory of Clinic Genetics, Air Force Medical University, Xi'an, China
| | - Yan Yao
- Department of Clinical Medicine, Yan'an University, Yan'an, China
| | - Weihong Hu
- Department of Clinical Medicine, Yan'an University, Yan'an, China
| | - Li Wang
- Department of Biochemistry and Molecular Biology, Air Force Medical University, Xi'an, China.,Shaanxi Provincial Key Laboratory of Clinic Genetics, Air Force Medical University, Xi'an, China.,School of Aerospace Medicine, Air Force Medical University, Xi'an, China
| | - Yuanming Wu
- Department of Biochemistry and Molecular Biology, Air Force Medical University, Xi'an, China.,Shaanxi Provincial Key Laboratory of Clinic Genetics, Air Force Medical University, Xi'an, China
| |
Collapse
|
80
|
Arisaka Y, Masuda H, Yoda T, Yui N. Delayed Senescence of Human Vascular Endothelial Cells by Molecular Mobility of Supramolecular Biointerfaces. Macromol Biosci 2021; 21:e2100216. [PMID: 34390172 DOI: 10.1002/mabi.202100216] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 07/16/2021] [Indexed: 01/10/2023]
Abstract
Yes-associated protein (YAP), a transcriptional coactivator of the Hippo signaling pathway, has been widely implicated in vascular aging and diseases. For preventing vascular endothelial cell senescence, the design and development of biomaterials to regulate YAP activity are required. This study prepares polyrotaxane-coated surfaces with molecular mobility and clarifies the role of the mobility on vascular endothelial cell senescence through Hippo-YAP signaling. The polyrotaxane surface with high mobility induces cytoplasmic YAP localization in endothelial cells, whereas the surface with low mobility induces nuclear YAP localization. After serial cultivation of endothelial cells using polyrotaxane surfaces with different mobilities for 35 d, the endothelial cells aged on the polyrotaxane surface with high mobility exhibit higher proliferative potential, smaller spreading size, and lower activity of senescence-associated β-galactosidase than those aged on the surface with low mobility. These findings suggest that cellular senescence can be delayed by modulating the molecular mobility on polyrotaxane surfaces.
Collapse
Affiliation(s)
- Yoshinori Arisaka
- Department of Organic Biomaterials, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), 2-3-10 Kanda-Surugadai, Chiyoda, Tokyo, 101-0062, Japan
| | - Hiroki Masuda
- Department of Maxillofacial Surgery, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo, Tokyo, 113-8549, Japan
| | - Tetsuya Yoda
- Department of Maxillofacial Surgery, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo, Tokyo, 113-8549, Japan
| | - Nobuhiko Yui
- Department of Organic Biomaterials, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), 2-3-10 Kanda-Surugadai, Chiyoda, Tokyo, 101-0062, Japan
| |
Collapse
|
81
|
Qureshi AM, Muthusami P, Krings T, Amirabadi A, Radovanovic I, Dirks P, Shroff M, Armstrong D, terBrugge K, Pereira VM. Clinical and Angioarchitectural Features of Hemorrhagic Brain Arterio-Venous Malformations in Adults and Children: Contrasts and Implications on Outcome. Neurosurgery 2021; 89:645-652. [PMID: 34270753 DOI: 10.1093/neuros/nyab251] [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: 12/08/2020] [Accepted: 05/08/2021] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Hemorrhage from brain arteriovenous malformations (bAVMs) is estimated at 3% per annum. Features influencing risk of hemorrhage include perforator/posterior circulation supply, associated aneurysms, and deep drainage. Children are more likely to present with bAVM bleeds. OBJECTIVE To analyze differences in bAVM angioarchitecture between children and adults and describe predictors of poor outcome. METHODS Data were collected from adult and pediatric tertiary referral hospitals. Demographic data, bleed location, treatment, and follow-up modified Rankin Scale (mRS) were collected. Angioarchitectural assessment included aneurysm presence, nidus morphology, perinidal angiogenesis, intranidal shunting, steal phenomenon, venous ectasia, venous stenosis, venous reflux, and pseudophlebitic pattern. Regression analyses conducted to determine predictors of mRS > 2. RESULTS A total of 270 adult and 135 pediatric ruptured bAVMs were assessed. Median age was 42 (adults) and 10.9 (children) yr. Intranidal aneurysms were more frequent in children (P = .012), whereas prenidal aneurysms were more common in adults (P < .01). Children demonstrated more perinidal angiogenesis (P = .04), whereas steal phenomenon was commoner in adults (P < .01). Venous ectasia (P < .01), reflux (P < .01), and pseudophlebitic pattern (P = .012) were more frequent in adults. Children had better outcome (mRS score ≤ 2) (P < .01). Older age (odds ratio [OR] = 1.02), eloquent location (OR = 2.5), multicompartmental hemorrhage (OR = 1.98), venous reflux (OR = 2.5), diffuse nidus (OR = 1.83), pseudophlebitic pattern (OR = 1.96), intranidal shunts (OR = 2), and no treatment (OR = 3.68) were significant predictors of mRS > 2. CONCLUSION Children are more likely to have intranidal aneurysms and perinidal angiogenesis, whereas adults have more prenidal aneurysms, venous ectasia, corticovenous reflux, and pseudophlebitic pattern. Eloquent location, diffuse nidus, intranidal shunts, venous reflux, and pseudophlebitic pattern predict poorer outcome.
Collapse
Affiliation(s)
- Ayman M Qureshi
- Department of Medical Imaging, Toronto Western Hospital, University Health Network, Toronto, Canada.,Department of Diagnostic Imaging & Image Guided Therapy, Hospital for Sick Children, University of Toronto, Toronto, Canada
| | - Prakash Muthusami
- Department of Diagnostic Imaging & Image Guided Therapy, Hospital for Sick Children, University of Toronto, Toronto, Canada
| | - Timo Krings
- Department of Medical Imaging, Toronto Western Hospital, University Health Network, Toronto, Canada
| | - Afsaneh Amirabadi
- Department of Diagnostic Imaging & Image Guided Therapy, Hospital for Sick Children, University of Toronto, Toronto, Canada
| | - Ivan Radovanovic
- Division of Neurosurgery, Toronto Western Hospital, University Health Network, Toronto, Canada
| | - Peter Dirks
- Division of Neurosurgery, Hospital for Sick Children, University of Toronto, Toronto, Canada
| | - Manohar Shroff
- Department of Diagnostic Imaging & Image Guided Therapy, Hospital for Sick Children, University of Toronto, Toronto, Canada
| | - Derek Armstrong
- Department of Diagnostic Imaging & Image Guided Therapy, Hospital for Sick Children, University of Toronto, Toronto, Canada
| | - Karel terBrugge
- Department of Medical Imaging, Toronto Western Hospital, University Health Network, Toronto, Canada
| | - Vitor M Pereira
- Department of Medical Imaging, Toronto Western Hospital, University Health Network, Toronto, Canada
| |
Collapse
|
82
|
Govindasamy V, Rajendran A, Lee ZX, Ooi GC, Then KY, Then KL, Gayathri M, Kumar Das A, Cheong SK. The potential role of mesenchymal stem cells in modulating antiageing process. Cell Biol Int 2021; 45:1999-2016. [PMID: 34245637 DOI: 10.1002/cbin.11652] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 05/24/2021] [Accepted: 06/17/2021] [Indexed: 12/19/2022]
Abstract
Ageing and age-related diseases share some basic origin that largely converges on inflammation. Precisely, it boils down to a common pathway characterised by the appearance of a fair amount of proinflammatory cytokines known as inflammageing. Among the proposed treatment for antiageing, MSCs gained attention in recent years. Since mesenchymal stem cells (MSCs) can differentiate itself into a myriad of terminal cells, previously it was believed that these cells migrate to the site of injury and perform their therapeutic effect. However, with the more recent discovery of huge amounts of paracrine factors secreted by MSCs, it is now widely accepted that these cells do not engraft upon transplantation but rather unveil their benefits through excretion of bioactive molecules namely those involved in inflammatory and immunomodulatory activities. Conversely, the true function of these paracrine changes has not been thoroughly investigated all these years. Hence, this review will describe in detail on ways MSCs may capitalize its paracrine properties in modulating antiageing process. Through a comprehensive literature search various elements in the antiageing process, we aim to provide a novel treatment perspective of MSCs in antiageing related clinical conditions.
Collapse
Affiliation(s)
- Vijayendran Govindasamy
- Research and Development Department, CryoCord Sdn Bhd, Bio-X Centre, Cyberjaya, Selangor, Malaysia
| | - Abilashini Rajendran
- Research and Development Department, CryoCord Sdn Bhd, Bio-X Centre, Cyberjaya, Selangor, Malaysia
| | - Zhi-Xin Lee
- Research and Development Department, CryoCord Sdn Bhd, Bio-X Centre, Cyberjaya, Selangor, Malaysia
| | - Ghee-Chien Ooi
- Research and Development Department, CryoCord Sdn Bhd, Bio-X Centre, Cyberjaya, Selangor, Malaysia
| | - Kong-Yong Then
- Research and Development Department, CryoCord Sdn Bhd, Bio-X Centre, Cyberjaya, Selangor, Malaysia.,Brighton Healthcare (Bio-X Healthcare Sdn Bhd), Bio-X Centre, Cyberjaya, Selangor, Malaysia
| | - Khong-Lek Then
- Research and Development Department, CryoCord Sdn Bhd, Bio-X Centre, Cyberjaya, Selangor, Malaysia
| | - Merilynn Gayathri
- Brighton Healthcare (Bio-X Healthcare Sdn Bhd), Bio-X Centre, Cyberjaya, Selangor, Malaysia
| | - Anjan Kumar Das
- Deparment of Surgery, IQ City Medical College, Durgapur, West Bengal, India
| | - Soon-Keng Cheong
- Department of Medicine, Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman (UTAR), Kajang, Selangor, Malaysia
| |
Collapse
|
83
|
Emechebe U, Nelson JW, Alkayed NJ, Kaul S, Adey AC, Barnes AP. Age-dependent transcriptional alterations in cardiac endothelial cells. Physiol Genomics 2021; 53:295-308. [PMID: 34097533 PMCID: PMC8321782 DOI: 10.1152/physiolgenomics.00037.2021] [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: 04/14/2021] [Revised: 05/14/2021] [Accepted: 05/14/2021] [Indexed: 02/08/2023] Open
Abstract
Aging is a significant risk factor for cardiovascular disease. Despite the fact that endothelial cells play critical roles in cardiovascular function and disease, the molecular impact of aging on this cell population in many organ systems remains unknown. In this study, we sought to determine age-associated transcriptional alterations in cardiac endothelial cells. Highly enriched populations of endothelial cells (ECs) isolated from the heart, brain, and kidney of young (3 mo) and aged (24 mo) C57/BL6 mice were profiled for RNA expression via bulk RNA sequencing. Approximately 700 cardiac endothelial transcripts significantly differ by age. Gene set enrichment analysis indicated similar patterns for cellular pathway perturbations. Receptor-ligand comparisons indicated parallel alterations in age-affected circulating factors and cardiac endothelial-expressed receptors. Gene and pathway enrichment analyses show that age-related transcriptional response of cardiac endothelial cells is distinct from that of endothelial cells derived from the brain or kidney vascular bed. Furthermore, single-cell analysis identified nine distinct EC subtypes and shows that the Apelin Receptor-enriched subtype is reduced with age in mouse heart. Finally, we identify age-dysregulated genes in specific aged cardiac endothelial subtypes.
Collapse
Affiliation(s)
- Uchenna Emechebe
- The Knight Cardiovascular Institute, Oregon Health and Science University, Portland, Oregon
| | - Jonathan W Nelson
- The Knight Cardiovascular Institute, Oregon Health and Science University, Portland, Oregon
| | - Nabil J Alkayed
- The Knight Cardiovascular Institute, Oregon Health and Science University, Portland, Oregon
- Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, Oregon
| | - Sanjiv Kaul
- The Knight Cardiovascular Institute, Oregon Health and Science University, Portland, Oregon
| | - Andrew C Adey
- The Knight Cardiovascular Institute, Oregon Health and Science University, Portland, Oregon
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, Oregon
- Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon
- Cancer Early Detection Advanced Research Institute, Oregon Health and Science University, Portland, Oregon
| | - Anthony P Barnes
- The Knight Cardiovascular Institute, Oregon Health and Science University, Portland, Oregon
- Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, Oregon
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, Oregon
| |
Collapse
|
84
|
Gete YG, Koblan LW, Mao X, Trappio M, Mahadik B, Fisher JP, Liu DR, Cao K. Mechanisms of angiogenic incompetence in Hutchinson-Gilford progeria via downregulation of endothelial NOS. Aging Cell 2021; 20:e13388. [PMID: 34086398 PMCID: PMC8282277 DOI: 10.1111/acel.13388] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 04/12/2021] [Accepted: 05/08/2021] [Indexed: 12/22/2022] Open
Abstract
Hutchinson–Gilford progeria syndrome (HGPS) is a rare genetic disorder with features of accelerated aging. The majority of HGPS cases are caused by a de novo point mutation in the LMNA gene (c.1824C>T; p.G608G) resulting in progerin, a toxic lamin A protein variant. Children with HGPS typically die from coronary artery diseases or strokes at an average age of 14.6 years. Endothelial dysfunction is a known driver of cardiovascular pathogenesis; however, it is currently unknown how progerin antagonizes normal angiogenic function in HGPS. Here, we use human iPSC‐derived endothelial cell (iPSC‐EC) models to study angiogenesis in HGPS. We cultured normal and HGPS iPSC‐ECs under both static and fluidic culture conditions. HGPS iPSC‐ECs show reduced endothelial nitric oxide synthase (eNOS) expression and activity compared with normal controls and concomitant decreases in intracellular nitric oxide (NO) level, which result in deficits in capillary‐like microvascular network formation. Furthermore, the expression of matrix metalloproteinase 9 (MMP‐9) was reduced in HGPS iPSC‐ECs, while the expression of tissue inhibitor metalloproteinases 1 and 2 (TIMP1 and TIMP2) was upregulated relative to healthy controls. Finally, we used an adenine base editor (ABE7.10max‐VRQR) to correct the pathogenic c.1824C>T allele in HGPS iPSC‐ECs. Remarkably, ABE7.10max‐VRQR correction of the HGPS mutation significantly reduced progerin expression to a basal level, rescued nuclear blebbing, increased intracellular NO level, normalized the misregulated TIMPs, and restored angiogenic competence in HGPS iPSC‐ECs. Together, these results provide molecular insights of endothelial dysfunction in HGPS and suggest that ABE could be a promising therapeutic approach for correcting HGPS‐related cardiovascular phenotypes.
Collapse
Affiliation(s)
- Yantenew G. Gete
- Department of Cell Biology and Molecular Genetics University of Maryland College Park MD USA
| | - Luke W. Koblan
- Merkin Institute of Transformative Technologies in Healthcare Broad Institute of Harvard and MIT Cambridge MA USA
- Department of Chemistry and Chemical Biology Harvard University Cambridge MA USA
- Howard Hughes Medical Institute Harvard University Cambridge MA USA
| | - Xiaojing Mao
- Department of Cell Biology and Molecular Genetics University of Maryland College Park MD USA
| | - Mason Trappio
- Department of Cell Biology and Molecular Genetics University of Maryland College Park MD USA
| | - Bhushan Mahadik
- Fischell Department of Bioengineering University of Maryland College Park MD USA
| | - John P. Fisher
- Fischell Department of Bioengineering University of Maryland College Park MD USA
| | - David R. Liu
- Merkin Institute of Transformative Technologies in Healthcare Broad Institute of Harvard and MIT Cambridge MA USA
- Department of Chemistry and Chemical Biology Harvard University Cambridge MA USA
- Howard Hughes Medical Institute Harvard University Cambridge MA USA
| | - Kan Cao
- Department of Cell Biology and Molecular Genetics University of Maryland College Park MD USA
| |
Collapse
|
85
|
Lupan AM, Rusu EG, Preda MB, Marinescu CI, Ivan C, Burlacu A. miRNAs generated from Meg3-Mirg locus are downregulated during aging. Aging (Albany NY) 2021; 13:15875-15897. [PMID: 34156971 PMCID: PMC8266327 DOI: 10.18632/aging.203208] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 06/02/2021] [Indexed: 12/13/2022]
Abstract
Aging determines a multilevel functional decline and increases the risk for cardiovascular pathologies. MicroRNAs are recognized as fine tuners of all cellular functions, being involved in various cardiac diseases. The heart is one of the most affected organs in aged individuals, however little is known about the extent and robustness to which miRNA profiles are modulated in cardiac cells during aging. This paper provides a comprehensive characterization of the aging-associated miRNA profile in the murine cardiac fibroblasts, which are increasingly recognized for their active involvement in the cardiac physiology and pathology. Next-generation sequencing of cardiac fibroblasts isolated from young and old mice revealed that an important fraction of the miRNAs generated by the Meg3-Mirg locus was downregulated during aging. To address the specificity of this repression, four miRNAs selected as representative for this locus were further assessed in other cells and organs isolated from aged mice. The results suggested that the repression of miRNAs generated by the Meg3-Mirg locus was a general feature of aging in multiple organs. Bioinformatic analysis of the predicted target genes identified Integrin Beta-2 as an aged-upregulated gene, which was thereafter confirmed in multiple mouse organs. In conclusion, our study provides new data concerning the mechanisms of natural aging and highlights the robustness of the miRNA modulation during this process.
Collapse
Affiliation(s)
- Ana-Mihaela Lupan
- Laboratory of Stem Cell Biology, Institute of Cellular Biology and Pathology “Nicolae Simionescu”, Bucharest 050568, Romania
| | - Evelyn-Gabriela Rusu
- Laboratory of Stem Cell Biology, Institute of Cellular Biology and Pathology “Nicolae Simionescu”, Bucharest 050568, Romania
| | - Mihai Bogdan Preda
- Laboratory of Stem Cell Biology, Institute of Cellular Biology and Pathology “Nicolae Simionescu”, Bucharest 050568, Romania
| | - Catalina Iolanda Marinescu
- Laboratory of Stem Cell Biology, Institute of Cellular Biology and Pathology “Nicolae Simionescu”, Bucharest 050568, Romania
| | - Cristina Ivan
- Department of Experimental Therapeutics, Division of Basic Science Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Alexandrina Burlacu
- Laboratory of Stem Cell Biology, Institute of Cellular Biology and Pathology “Nicolae Simionescu”, Bucharest 050568, Romania
| |
Collapse
|
86
|
UNC5B Promotes Vascular Endothelial Cell Senescence via the ROS-Mediated P53 Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5546711. [PMID: 34239689 PMCID: PMC8238614 DOI: 10.1155/2021/5546711] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 05/23/2021] [Accepted: 06/01/2021] [Indexed: 11/17/2022]
Abstract
Vascular endothelial cell senescence is involved in human aging and age-related vascular disorders. Guidance receptor UNC5B is implicated in oxidative stress and angiogenesis. Nonetheless, little is known about the role of UNC5B in endothelial cell senescence. Here, we cultured primary human umbilical vein endothelial cells to young and senescent phases. Subsequently, the expression of UNC5B was identified in replicative senescent cells, and then, its effect on endothelial cell senescence was confirmed by UNC5B-overexpressing lentiviral vectors and RNA interference. Overexpression of UNC5B in young endothelial cells significantly increased senescence-associated β-galactosidase-positive cells, upregulated the mRNAs expression of the senescence-associated secretory phenotype genes, reduced total cell number, and inhibited the potential for cell proliferation. Furthermore, overexpression of UNC5B promoted the generation of intracellular reactive oxygen species (ROS) and activated the P53 pathway. Besides, overexpression of UNC5B disturbed endothelial function by inhibiting cell migration and tube formation. Nevertheless, silencing UNC5B generated conflicting outcomes. Blocking ROS production or inhibiting the function of P53 rescued endothelial cell senescence induced by UNC5B. These findings suggest that UNC5B promotes endothelial cell senescence, potentially by activating the ROS-P53 pathway. Therefore, inhibiting UNC5B might reduce endothelial cell senescence and hinder age-related vascular disorders.
Collapse
|
87
|
Characteristics of Moyamoya Disease in the Older Population: Is It Possible to Define a Typical Presentation and Optimal Therapeutical Management? J Clin Med 2021; 10:jcm10112287. [PMID: 34070336 PMCID: PMC8197522 DOI: 10.3390/jcm10112287] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 05/17/2021] [Accepted: 05/21/2021] [Indexed: 12/17/2022] Open
Abstract
Whereas several studies have been so far presented about the surgical outcomes in terms of mortality and perioperative complications for elderly patients submitted to neurosurgical treatments, the management of elderly moyamoya patients is unclear. This review aims to explore the available data about the clinical manifestation, characteristics, and outcome after surgery of older patients with moyamoya arteriopathy (MA). We found only two articles strictly concerning elderly patients with MA. We have also evaluated other reported adult series of moyamoya patients, including elderly cases in their analysis. Patients with MA above 50 years old may be considered a peculiar subset in which patients are often presenting with ischemic symptoms and a higher Suzuki grade. Conservative treatment may be proposed in asymptomatic or stable cases due to their fragility and possible increase of post-operative complications, while the best surgical options in symptomatic cases are still under investigation, although we believe that a minimal invasive superficial temporal artery—middle cerebral artery bypass could be considered the treatment of choice for the immediate effect on brain perfusion with a limited rate of post-operative complications.
Collapse
|
88
|
Stojanović SD, Fiedler J, Bauersachs J, Thum T, Sedding DG. Senescence-induced inflammation: an important player and key therapeutic target in atherosclerosis. Eur Heart J 2021; 41:2983-2996. [PMID: 31898722 PMCID: PMC7453834 DOI: 10.1093/eurheartj/ehz919] [Citation(s) in RCA: 96] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 09/13/2019] [Accepted: 12/12/2019] [Indexed: 12/21/2022] Open
Abstract
Inflammation is a hallmark and potent driver of pathological vascular remodelling in atherosclerosis. However, current anti-inflammatory therapeutic strategies have shown mixed results. As an alternative perspective on the conundrum of chronic inflammation emerging evidence points towards a small subset of senescent cells as a critical player and central node driving atherosclerosis. Senescent cells belonging to various cell types are a dominant and chronic source of a large array of pro-inflammatory cytokines and various additional plaque destabilizing factors, being involved with various aspects of atherosclerosis pathogenesis. Antagonizing these key agitators of local chronic inflammation and plaque instability may provide a causative and multi-purpose therapeutic strategy to treat atherosclerosis. Anti-senescence treatment options with translational potential are currently in development. However, several questions and challenges remain to be addressed before these novel treatment approaches may enter the clinical setting.
Collapse
Affiliation(s)
- Stevan D Stojanović
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany.,Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Jan Fiedler
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Daniel G Sedding
- Department of Internal Medicine III, Cardiology, Angiology and Intensive Care Medicine, Martin-Luther-University Halle (Saale), Ernst-Grube-Strasse 40, 06120 Halle (Saale), Germany
| |
Collapse
|
89
|
Callahan CM, Apostolova LG, Gao S, Risacher SL, Case J, Saykin AJ, Lane KA, Swinford CG, Yoder MC. Novel Markers of Angiogenesis in the Setting of Cognitive Impairment and Dementia. J Alzheimers Dis 2021; 75:959-969. [PMID: 32390626 DOI: 10.3233/jad-191293] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Aberrant angiogenesis may play a role in the development of Alzheimer's disease and related dementia. OBJECTIVE To explore the relationship between angiogenesis activity and evidence of neurodegeneration among older adults. METHODS Cross-sectional study of 49 older adults clinically characterized as cognitively normal, mild cognitive impairment, or early Alzheimer's disease. In addition to neuroimaging, we completed assays on peripheral blood, including: vascular endothelial growth factor, tumor necrosis factor, fibroblast growth factor, and amyloid-β peptide 40. We used advanced polychromatic flow cytometry to phenotype circulating mononuclear cells to assess angiogenesis activity. RESULTS Although we documented differences in cognitive performance, structural changes on neuroimaging, and burden of amyloid and tau on positron emission tomography, angiogenesis activity did not vary by group. Interestingly, VEGF levels were shown to be increased among subjects with mild cognitive impairment. In ANCOVA models controlling for age, sex, intracranial volume, and monocyte subpopulations, angiogenesis activity was correlated with increased white matter hyperintensities. CONCLUSION We demonstrate a significant association between angiogenesis activity and cerebrovascular disease. To better understand the potential of angiogenesis as an intervention target, longitudinal studies are needed.
Collapse
Affiliation(s)
- Christopher M Callahan
- Indiana University Center for Aging Research, Indianapolis, IN, USA.,Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA.,Regenstrief Institute, Inc., Indianapolis, IN, USA.,Eskenazi Health, Indianapolis, IN, USA.,Indiana Alzheimer Disease Center, Indianapolis, IN, USA
| | - Liana G Apostolova
- Indiana Alzheimer Disease Center, Indianapolis, IN, USA.,Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sujuan Gao
- Indiana Alzheimer Disease Center, Indianapolis, IN, USA.,Department of Biostatistics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Shannon L Risacher
- Indiana Alzheimer Disease Center, Indianapolis, IN, USA.,Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jamie Case
- Scripps Clinic Bio-Repository and Bio-Informatics Core, Scripps Clinic Medical Group, La Jolla, CA, USA
| | - Andrew J Saykin
- Indiana Alzheimer Disease Center, Indianapolis, IN, USA.,Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kathleen A Lane
- Department of Biostatistics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Cecily G Swinford
- Indiana Alzheimer Disease Center, Indianapolis, IN, USA.,Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Mervin C Yoder
- Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
90
|
Cytoprotective effects of erythropoietin: What about the lung? Biomed Pharmacother 2021; 139:111547. [PMID: 33831836 DOI: 10.1016/j.biopha.2021.111547] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 03/17/2021] [Accepted: 03/23/2021] [Indexed: 02/07/2023] Open
Abstract
Erythropoietin (Epo) is a pleiotropic cytokine, essential for erythropoiesis. Epo and its receptor (Epo-R) are produced by several tissues and it is now admitted that Epo displays other physiological functions than red blood cell synthesis. Indeed, Epo provides cytoprotective effects, which consist in prevention or fight against pathological processes. This perspective article reviews the various protective effects of Epo in several organs and tries to give a proof of concept about its effects in the lung. The tissue-protective effects of Epo could be a promising approach to limit the symptoms of acute and chronic lung diseases.
Collapse
|
91
|
Twist1 signaling in age-dependent decline in angiogenesis and lung regeneration. Aging (Albany NY) 2021; 13:7781-7799. [PMID: 33764901 PMCID: PMC8034921 DOI: 10.18632/aging.202875] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 03/14/2021] [Indexed: 12/11/2022]
Abstract
Angiogenesis – the formation of new blood capillaries- is impaired in aging animals and contributes to the pathogenesis of age-related diseases. A transcription factor, Twist1, contributes to the pathogenesis of age- and angiogenesis-related diseases such as pulmonary fibrosis and atherosclerosis. However, the mechanism by which Twist1 controls age-dependent decline in angiogenesis remains unclear. In this report, we have demonstrated that the levels of Twist1 are higher, while the expression of peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α) that stimulates angiogenesis, is lower in endothelial cells (ECs) isolated from aged human adipose tissues and mouse lungs compared to those from young tissues. Knockdown of Twist1 in aged human ECs increases the levels of PGC1α and angiogenic factor receptor, vascular endothelial growth factor receptor (VEGFR2), and restores EC proliferation and migration, while inhibition of PGC1α suppresses these effects. Knockdown of Twist1 in supplemented aged ECs also restores vascular networks in the subcutaneously implanted gel, while these effects are abrogated by knockdown of PGC1α. Age-dependent inhibition of post-pneumonectomy (PNX) lung growth is suppressed in Tie2-specific Twist1 conditional knockout mouse lungs, in which VEGFR2 expression increases after PNX. These results suggest that upregulation of endothelial Twist1 mediates age-dependent decline in angiogenesis and regenerative lung growth.
Collapse
|
92
|
Role of FoxO transcription factors in aging-associated cardiovascular diseases. VITAMINS AND HORMONES 2021; 115:449-475. [PMID: 33706958 DOI: 10.1016/bs.vh.2020.12.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Aging constitutes a major risk factor toward the development of cardiovascular diseases (CVDs). The aging heart undergoes several changes at the molecular, cellular and physiological levels, which diminishes its contractile function and weakens stress tolerance. Further, old age increases the exposure to risk factors such as hypertension, diabetes and hypercholesterolemia. Notably, research in the past decades have identified FoxO subfamily of the forkhead transcription factors as key players in regulating diverse cellular processes linked to cardiac aging and diseases. In the present chapter, we discuss the important role of FoxO in the development of various aging-associated cardiovascular complications such as cardiac hypertrophy, cardiac fibrosis, heart failure, vascular dysfunction, atherosclerosis, hypertension and myocardial ischemia. Besides, we will also discuss the role of FoxO in cardiometabolic alterations, autophagy and proteasomal degradation, which are implicated in aging-associated cardiac dysfunction.
Collapse
|
93
|
Aging and age-related diseases: from mechanisms to therapeutic strategies. Biogerontology 2021; 22:165-187. [PMID: 33502634 PMCID: PMC7838467 DOI: 10.1007/s10522-021-09910-5] [Citation(s) in RCA: 213] [Impact Index Per Article: 71.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 01/01/2021] [Indexed: 01/10/2023]
Abstract
Aging is a physiological process mediated by numerous biological and genetic pathways, which are directly linked to lifespan and are a driving force for all age-related diseases. Human life expectancy has greatly increased in the past few decades, but this has not been accompanied by a similar increase in their healthspan. At present, research on aging biology has focused on elucidating the biochemical and genetic pathways that contribute to aging over time. Several aging mechanisms have been identified, primarily including genomic instability, telomere shortening, and cellular senescence. Aging is a driving factor of various age-related diseases, including neurodegenerative diseases, cardiovascular diseases, cancer, immune system disorders, and musculoskeletal disorders. Efforts to find drugs that improve the healthspan by targeting the pathogenesis of aging have now become a hot topic in this field. In the present review, the status of aging research and the development of potential drugs for aging-related diseases, such as metformin, rapamycin, resveratrol, senolytics, as well as caloric restriction, are summarized. The feasibility, side effects, and future potential of these treatments are also discussed, which will provide a basis to develop novel anti-aging therapeutics for improving the healthspan and preventing aging-related diseases.
Collapse
|
94
|
Lan Y, Dong M, Li Y, Diao Y, Chen Z, Li Y. SIRT1-induced deacetylation of Akt expedites platelet phagocytosis and delays HEMEC aging. MOLECULAR THERAPY-NUCLEIC ACIDS 2021; 23:1323-1333. [PMID: 33717652 PMCID: PMC7920857 DOI: 10.1016/j.omtn.2021.01.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 01/20/2021] [Indexed: 12/25/2022]
Abstract
Maintaining the health of the endothelium is of critical importance to prevention against cell aging. The current study was performed to clarify the role of sirtuin1 (SIRT1) in platelet phagocytosis in cell aging and identified its downstream molecular mechanism. Platelet phagocytosis by human endometrial microvascular endothelial cells (HEMECs) was characterized by transmission electron and fluorescence microscopy. Functional experiments were conducted to examine platelet phagocytosis and cell aging using the overexpression or knockdown plasmids of SIRT1 and G alpha-interacting, vesicle-associated protein (GIRDIN) as well as Akt inhibitor and activator. It was found that SIRT1 facilitated platelet phagocytosis by HEMECs, contributing to inhibition of cell aging. Akt activation facilitated platelet phagocytosis and repressed cell aging. GIRDIN overexpression accelerated platelet phagocytosis by HEMECs, leading to a delay in cell aging. GIRDIN phosphorylation at Ser1417 was induced by Akt activation, while activation of Akt was induced by SIRT1-mediated deacetylation, consequently augmenting platelet phagocytosis and delaying cell aging. Taken together, SIRT1 delayed aging of HEMECs by deacetylating Akt, phosphorylating GIRDIN, and inducing platelet phagocytosis. The study highlights a possible target for the prevention of HEMEC aging.
Collapse
Affiliation(s)
- Yong Lan
- Department of Vascular Surgery, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, Beijing 100730, P.R. China
| | - Min Dong
- Department of Cardiology, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, Beijing 100730, P.R. China
| | - Yongjun Li
- Department of Vascular Surgery, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, Beijing 100730, P.R. China
| | - Yongpeng Diao
- Department of Vascular Surgery, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, Beijing 100730, P.R. China
| | - Zuoguang Chen
- Department of Vascular Surgery, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, Beijing 100730, P.R. China
| | - Yangfang Li
- Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, P.R. China
| |
Collapse
|
95
|
Toya T, Ahmad A, Attia Z, Cohen-Shelly M, Ozcan I, Noseworthy PA, Lopez-Jimenez F, Kapa S, Lerman LO, Friedman PA, Lerman A. Vascular Aging Detected by Peripheral Endothelial Dysfunction Is Associated With ECG-Derived Physiological Aging. J Am Heart Assoc 2021; 10:e018656. [PMID: 33455414 PMCID: PMC7955452 DOI: 10.1161/jaha.120.018656] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Background An artificial intelligence algorithm that detects age using the 12-lead ECG has been suggested to signal "physiologic age." This study aimed to investigate the association of peripheral microvascular endothelial function (PMEF) as an index of vascular aging, with accelerated physiologic aging gauged by ECG-derived artificial intelligence-estimated age. Methods and Results This study included 531 patients who underwent ECG and a noninvasive PMEF assessment using reactive hyperemia peripheral arterial tonometry. Abnormal PMEF was defined as reactive hyperemia peripheral arterial tonometry index ≤2.0. Accelerated or delayed physiologic aging was calculated by the Δ age (ECG-derived artificial intelligence-estimated age minus chronological age), and the association between Δ age and PMEF as well as its impact on composite major adverse cardiovascular events were investigated. Δ age was higher in patients with abnormal PMEF than in patients with normal PMEF (2.3±7.8 versus 0.5±7.7 years; P=0.01). Reactive hyperemia peripheral arterial tonometry index was negatively associated with Δ age after adjustment for cardiovascular risk factors (standardized β coefficient, -0.08; P=0.048). The highest quartile of Δ age was associated with an increased risk of major adverse cardiovascular events compared with the first quartile of Δ age in patients with abnormal PMEF, even after adjustment for cardiovascular risk factors (hazard ratio, 4.72; 95% CI, 1.24-17.91; P=0.02). Conclusions Vascular aging detected by endothelial function is associated with accelerated physiologic aging, as assessed by the artificial intelligence-ECG Δ age. Patients with endothelial dysfunction and the highest quartile of accelerated physiologic aging have a marked increase in risk for cardiovascular events.
Collapse
Affiliation(s)
- Takumi Toya
- Department of Cardiovascular Medicine Mayo Clinic Rochester MN.,Division of Cardiology National Defense Medical College Tokorozawa Saitama Japan
| | | | - Zachi Attia
- Department of Cardiovascular Medicine Mayo Clinic Rochester MN
| | | | - Ilke Ozcan
- Department of Cardiovascular Medicine Mayo Clinic Rochester MN
| | | | | | - Suraj Kapa
- Department of Cardiovascular Medicine Mayo Clinic Rochester MN
| | - Lilach O Lerman
- Division of Nephrology and Hypertension Mayo Clinic Rochester MN
| | - Paul A Friedman
- Department of Cardiovascular Medicine Mayo Clinic Rochester MN
| | - Amir Lerman
- Department of Cardiovascular Medicine Mayo Clinic Rochester MN
| |
Collapse
|
96
|
Sunohara T, Imamura H, Goto M, Fukumitsu R, Matsumoto S, Fukui N, Oomura Y, Akiyama T, Fukuda T, Go K, Kajiura S, Shigeyasu M, Asakura K, Horii R, Sakai C, Sakai N. Neck Location on the Outer Convexity is a Predictor of Incomplete Occlusion in Treatment with the Pipeline Embolization Device: Clinical and Angiographic Outcomes. AJNR Am J Neuroradiol 2021; 42:119-125. [PMID: 33184073 DOI: 10.3174/ajnr.a6859] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 08/11/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND PURPOSE With the increasing use of the Pipeline Embolization Device for the treatment of aneurysms, predictors of clinical and angiographic outcomes are needed. This study aimed to identify predictors of incomplete occlusion at last angiographic follow-up. MATERIALS AND METHODS In our retrospective, single-center cohort study, 105 ICA aneurysms in 89 subjects were treated with Pipeline Embolization Devices. Patients were followed per standardized protocol. Clinical and angiographic outcomes were analyzed. We introduced a new morphologic classification based on the included angle of the parent artery against the neck location: outer convexity type (included angle, <160°), inner convexity type (included angle, >200°), and lateral wall type (160° ≤ included angle ≤200°). This classification reflects the metal coverage rate and flow dynamics. RESULTS Imaging data were acquired in 95.3% of aneurysms persistent at 6 months. Complete occlusion was achieved in 70.5%, and incomplete occlusion, in 29.5% at last follow-up. Multivariable regression analysis revealed that 60 years of age or older (OR, 5.70; P = .001), aneurysms with the branching artery from the dome (OR, 10.56; P = .002), fusiform aneurysms (OR, 10.2; P = .009), and outer convexity-type saccular aneurysms (versus inner convexity type: OR, 30.3; P < .001; versus lateral wall type: OR, 9.71; P = .001) were independently associated with a higher rate of incomplete occlusion at the last follow-up. No permanent neurologic deficits or rupture were observed in the follow-up period. CONCLUSIONS The aneurysm neck located on the outer convexity is a new, incomplete occlusion predictor, joining older age, fusiform aneurysms, and aneurysms with the branching artery from the dome. No permanent neurologic deficits or rupture was observed in the follow-up, even with incomplete occlusion.
Collapse
Affiliation(s)
- T Sunohara
- From the Department of Neurosurgery, Kobe City Medical Center General Hospital, Kobe, Japan.
| | - H Imamura
- From the Department of Neurosurgery, Kobe City Medical Center General Hospital, Kobe, Japan
| | - M Goto
- From the Department of Neurosurgery, Kobe City Medical Center General Hospital, Kobe, Japan
| | - R Fukumitsu
- From the Department of Neurosurgery, Kobe City Medical Center General Hospital, Kobe, Japan
| | - S Matsumoto
- From the Department of Neurosurgery, Kobe City Medical Center General Hospital, Kobe, Japan
| | - N Fukui
- From the Department of Neurosurgery, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Y Oomura
- From the Department of Neurosurgery, Kobe City Medical Center General Hospital, Kobe, Japan
| | - T Akiyama
- From the Department of Neurosurgery, Kobe City Medical Center General Hospital, Kobe, Japan
| | - T Fukuda
- From the Department of Neurosurgery, Kobe City Medical Center General Hospital, Kobe, Japan
| | - K Go
- From the Department of Neurosurgery, Kobe City Medical Center General Hospital, Kobe, Japan
| | - S Kajiura
- From the Department of Neurosurgery, Kobe City Medical Center General Hospital, Kobe, Japan
| | - M Shigeyasu
- From the Department of Neurosurgery, Kobe City Medical Center General Hospital, Kobe, Japan
| | - K Asakura
- From the Department of Neurosurgery, Kobe City Medical Center General Hospital, Kobe, Japan
| | - R Horii
- From the Department of Neurosurgery, Kobe City Medical Center General Hospital, Kobe, Japan
| | - C Sakai
- From the Department of Neurosurgery, Kobe City Medical Center General Hospital, Kobe, Japan
| | - N Sakai
- From the Department of Neurosurgery, Kobe City Medical Center General Hospital, Kobe, Japan
| |
Collapse
|
97
|
Joshkon A, Heim X, Dubrou C, Bachelier R, Traboulsi W, Stalin J, Fayyad-Kazan H, Badran B, Foucault-Bertaud A, Leroyer AS, Bardin N, Blot-Chabaud M. Role of CD146 (MCAM) in Physiological and Pathological Angiogenesis-Contribution of New Antibodies for Therapy. Biomedicines 2020; 8:biomedicines8120633. [PMID: 33352759 PMCID: PMC7767164 DOI: 10.3390/biomedicines8120633] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/14/2020] [Accepted: 12/16/2020] [Indexed: 02/07/2023] Open
Abstract
The fundamental role of cell adhesion molecules in mediating various biological processes as angiogenesis has been well-documented. CD146, an adhesion molecule of the immunoglobulin superfamily, and its soluble form, constitute major players in both physiological and pathological angiogenesis. A growing body of evidence shows soluble CD146 to be significantly elevated in the serum or interstitial fluid of patients with pathologies related to deregulated angiogenesis, as autoimmune diseases, obstetric and ocular pathologies, and cancers. To block the undesirable effects of this molecule, therapeutic antibodies have been developed. Herein, we review the multifaceted functions of CD146 in physiological and pathological angiogenesis and summarize the interest of using monoclonal antibodies for therapeutic purposes.
Collapse
Affiliation(s)
- Ahmad Joshkon
- Hematology Department, Center for CardioVascular and Nutrition Research C2VN, Faculty of Pharmacy, Timone Campus, Aix-Marseille University, Institut National de la Santé et de la Recherche Médicale (INSERM), Institut National de Recherche Pour L’agriculture, L’alimentation et L’environnement (INRAE), 13005 Marseille, France; (X.H.); (C.D.); (R.B.); (W.T.); (J.S.); (A.F.-B.); (A.S.L.); (N.B.); (M.B.-C.)
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Science, Lebanese University, Hadath 1104, Lebanon; (H.F.-K.); (B.B.)
- Correspondence:
| | - Xavier Heim
- Hematology Department, Center for CardioVascular and Nutrition Research C2VN, Faculty of Pharmacy, Timone Campus, Aix-Marseille University, Institut National de la Santé et de la Recherche Médicale (INSERM), Institut National de Recherche Pour L’agriculture, L’alimentation et L’environnement (INRAE), 13005 Marseille, France; (X.H.); (C.D.); (R.B.); (W.T.); (J.S.); (A.F.-B.); (A.S.L.); (N.B.); (M.B.-C.)
- Service d’immunologie, Pôle de Biologie, Hôpital de la Conception, Assistance Publique Hôpitaux de Marseille (AP-HM), 13005 Marseille, France
| | - Cléa Dubrou
- Hematology Department, Center for CardioVascular and Nutrition Research C2VN, Faculty of Pharmacy, Timone Campus, Aix-Marseille University, Institut National de la Santé et de la Recherche Médicale (INSERM), Institut National de Recherche Pour L’agriculture, L’alimentation et L’environnement (INRAE), 13005 Marseille, France; (X.H.); (C.D.); (R.B.); (W.T.); (J.S.); (A.F.-B.); (A.S.L.); (N.B.); (M.B.-C.)
| | - Richard Bachelier
- Hematology Department, Center for CardioVascular and Nutrition Research C2VN, Faculty of Pharmacy, Timone Campus, Aix-Marseille University, Institut National de la Santé et de la Recherche Médicale (INSERM), Institut National de Recherche Pour L’agriculture, L’alimentation et L’environnement (INRAE), 13005 Marseille, France; (X.H.); (C.D.); (R.B.); (W.T.); (J.S.); (A.F.-B.); (A.S.L.); (N.B.); (M.B.-C.)
| | - Wael Traboulsi
- Hematology Department, Center for CardioVascular and Nutrition Research C2VN, Faculty of Pharmacy, Timone Campus, Aix-Marseille University, Institut National de la Santé et de la Recherche Médicale (INSERM), Institut National de Recherche Pour L’agriculture, L’alimentation et L’environnement (INRAE), 13005 Marseille, France; (X.H.); (C.D.); (R.B.); (W.T.); (J.S.); (A.F.-B.); (A.S.L.); (N.B.); (M.B.-C.)
| | - Jimmy Stalin
- Hematology Department, Center for CardioVascular and Nutrition Research C2VN, Faculty of Pharmacy, Timone Campus, Aix-Marseille University, Institut National de la Santé et de la Recherche Médicale (INSERM), Institut National de Recherche Pour L’agriculture, L’alimentation et L’environnement (INRAE), 13005 Marseille, France; (X.H.); (C.D.); (R.B.); (W.T.); (J.S.); (A.F.-B.); (A.S.L.); (N.B.); (M.B.-C.)
| | - Hussein Fayyad-Kazan
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Science, Lebanese University, Hadath 1104, Lebanon; (H.F.-K.); (B.B.)
| | - Bassam Badran
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Science, Lebanese University, Hadath 1104, Lebanon; (H.F.-K.); (B.B.)
| | - Alexandrine Foucault-Bertaud
- Hematology Department, Center for CardioVascular and Nutrition Research C2VN, Faculty of Pharmacy, Timone Campus, Aix-Marseille University, Institut National de la Santé et de la Recherche Médicale (INSERM), Institut National de Recherche Pour L’agriculture, L’alimentation et L’environnement (INRAE), 13005 Marseille, France; (X.H.); (C.D.); (R.B.); (W.T.); (J.S.); (A.F.-B.); (A.S.L.); (N.B.); (M.B.-C.)
| | - Aurelie S. Leroyer
- Hematology Department, Center for CardioVascular and Nutrition Research C2VN, Faculty of Pharmacy, Timone Campus, Aix-Marseille University, Institut National de la Santé et de la Recherche Médicale (INSERM), Institut National de Recherche Pour L’agriculture, L’alimentation et L’environnement (INRAE), 13005 Marseille, France; (X.H.); (C.D.); (R.B.); (W.T.); (J.S.); (A.F.-B.); (A.S.L.); (N.B.); (M.B.-C.)
| | - Nathalie Bardin
- Hematology Department, Center for CardioVascular and Nutrition Research C2VN, Faculty of Pharmacy, Timone Campus, Aix-Marseille University, Institut National de la Santé et de la Recherche Médicale (INSERM), Institut National de Recherche Pour L’agriculture, L’alimentation et L’environnement (INRAE), 13005 Marseille, France; (X.H.); (C.D.); (R.B.); (W.T.); (J.S.); (A.F.-B.); (A.S.L.); (N.B.); (M.B.-C.)
- Service d’immunologie, Pôle de Biologie, Hôpital de la Conception, Assistance Publique Hôpitaux de Marseille (AP-HM), 13005 Marseille, France
| | - Marcel Blot-Chabaud
- Hematology Department, Center for CardioVascular and Nutrition Research C2VN, Faculty of Pharmacy, Timone Campus, Aix-Marseille University, Institut National de la Santé et de la Recherche Médicale (INSERM), Institut National de Recherche Pour L’agriculture, L’alimentation et L’environnement (INRAE), 13005 Marseille, France; (X.H.); (C.D.); (R.B.); (W.T.); (J.S.); (A.F.-B.); (A.S.L.); (N.B.); (M.B.-C.)
| |
Collapse
|
98
|
Chehaitly A, Vessieres E, Guihot AL, Henrion D. Flow-mediated outward arterial remodeling in aging. Mech Ageing Dev 2020; 194:111416. [PMID: 33333130 DOI: 10.1016/j.mad.2020.111416] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 12/08/2020] [Accepted: 12/11/2020] [Indexed: 12/13/2022]
Abstract
The present review focuses on the effect of aging on flow-mediated outward remodeling (FMR) via alterations in estrogen metabolism, oxidative stress and inflammation. In ischemic disorders, the ability of the vasculature to adapt or remodel determines the quality of the recovery. FMR, which has a key role in revascularization, is a complex phenomenon that recruits endothelial and smooth muscle cells as well as the immune system. FMR becomes progressively less with age as a result of an increase in inflammation and oxidative stress, in part of mitochondrial origin. The alteration in FMR is greater in older individuals with risk factors and thus the therapy cannot merely amount to exercise with or without a mild vasodilating drug. Interestingly, the reduction in FMR occurs later in females. Estrogen and its alpha receptor (ERα) play a key role in FMR through the control of dilatory pathways including the angiotensin II type 2 receptor, thus providing possible tools to activate FMR in older subjects although only experimental data is available. Indeed, the main issue is the reversibility of the vascular damage induced over time, and to date promoting prevention and limiting exposure to the risk factors remain the best options in this regard.
Collapse
Affiliation(s)
- Ahmad Chehaitly
- MITOVASC Laboratory and CARFI Facility, INSERM U1083, CNRS UMR 6015, University of Angers, Angers, France
| | - Emilie Vessieres
- MITOVASC Laboratory and CARFI Facility, INSERM U1083, CNRS UMR 6015, University of Angers, Angers, France
| | - Anne-Laure Guihot
- MITOVASC Laboratory and CARFI Facility, INSERM U1083, CNRS UMR 6015, University of Angers, Angers, France
| | - Daniel Henrion
- MITOVASC Laboratory and CARFI Facility, INSERM U1083, CNRS UMR 6015, University of Angers, Angers, France.
| |
Collapse
|
99
|
Nunes KP, Webb RC. New insights into RhoA/Rho-kinase signaling: a key regulator of vascular contraction. Small GTPases 2020; 12:458-469. [PMID: 32970516 DOI: 10.1080/21541248.2020.1822721] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023] Open
Abstract
While Rho-signalling controlling vascular contraction is a canonical mechanism, with the modern approaches used in research, we are advancing our understanding and details into this pathway are often uncovered. RhoA-mediated Rho-kinase is the major regulator of vascular smooth muscle cells and a key player manoeuvring other functions in these cells. The discovery of new interactions, such as oxidative stress and hydrogen sulphide with Rho signalling are emerging addition not only in the physiology of the smooth muscle, but especially in the pathophysiology of vascular diseases. Likewise, the interplay between ageing and Rho-kinase in the vasculature has been recently considered. Importantly, in smooth muscle contraction, this pathway may also be affected by sex hormones, and consequently, sex-differences. This review provides an overview of Rho signalling mediating vascular contraction and focuses on recent topics discussed in the literature affecting this pathway such as ageing, sex differences and oxidative stress.
Collapse
Affiliation(s)
- Kenia Pedrosa Nunes
- Laboratory of Vascular Physiology, Department of Biomedical and Chemical Engineering and Sciences, Florida Institute of Technology, Melbourne, FL, USA
| | - R Clinton Webb
- Department of Cell Biology and Anatomy, University of South Carolina, Columbia, SC, USA
| |
Collapse
|
100
|
Establishment and Evaluation of an In Vitro System for Biophysical Stimulation of Human Osteoblasts. Cells 2020; 9:cells9091995. [PMID: 32872592 PMCID: PMC7564340 DOI: 10.3390/cells9091995] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 08/20/2020] [Accepted: 08/26/2020] [Indexed: 12/28/2022] Open
Abstract
While several studies investigated the effects of mechanical or electrical stimulation on osseointegration and bone fracture healing, little is known about the molecular and cellular impact of combined biophysical stimulation on peri-implant osseointegration. Therefore, we established an in vitro system, capable of applying shear stress and electric fields simultaneously. Capacitively coupled electric fields were used for electrical stimulation, while roughened Ti6Al4V bodies conducted harmonically oscillating micromotions on collagen scaffolds seeded with human osteoblasts. Different variations of single and combined stimulation were applied for three days, while samples loaded with Ti6Al4V bodies and untreated samples served as control. Metabolic activity, expression of osteogenic markers and bone remodeling markers were investigated. While combined stimulation showed no substantial benefit compared to sole mechanical stimulation, we observed that 25 µm micromotions applied by roughened Ti6Al4V bodies led to a significant increase in gene expression of osteocalcin and tissue inhibitor of metalloprotease 1. Additionally, we found an increase in metabolic activity and expression of bone remodeling markers with reduced procollagen type 1 synthesis after 100 mVRMS electrical stimulation. We were able to trigger specific cellular behaviors using different biophysical stimuli. In future studies, different variations of electrical stimulation will be combined with interfacial micromotions.
Collapse
|