45 results on '"Lee, Man K S"'
Search Results
2. Cardiovascular characterisation of a novel mouse model that combines hypertension and diabetes co-morbidities
- Author
-
Sharma, Arpeeta, Choi, Judy S. Y., Watson, Anna M. D., Li, Leila, Sonntag, Thomas, Lee, Man K. S., Murphy, Andrew J., De Blasio, Miles, Head, Geoffrey A., Ritchie, Rebecca H., and de Haan, Judy B.
- Published
- 2023
- Full Text
- View/download PDF
3. Diastolic dysfunction in a pre-clinical model of diabetes is associated with changes in the cardiac non-myocyte cellular composition
- Author
-
Cohen, Charles D., De Blasio, Miles J., Lee, Man K. S., Farrugia, Gabriella E., Prakoso, Darnel, Krstevski, Crisdion, Deo, Minh, Donner, Daniel G., Kiriazis, Helen, Flynn, Michelle C., Gaynor, Taylah L., Murphy, Andrew J., Drummond, Grant R., Pinto, Alexander R., and Ritchie, Rebecca H.
- Published
- 2021
- Full Text
- View/download PDF
4. The Multiparametric Analysis of Mitochondrial Dynamics in T Cells from Cryopreserved Peripheral Blood Mononuclear Cells (PBMCs)
- Author
-
Masson, Jesse J. R., primary, Ostrowski, Matias, additional, Duette, Gabriel, additional, Lee, Man K. S., additional, Murphy, Andrew J., additional, Crowe, Suzanne M., additional, and Palmer, Clovis S., additional
- Published
- 2020
- Full Text
- View/download PDF
5. A high-salt diet promotes atherosclerosis by altering haematopoiesis
- Author
-
Lee, Man K. S., primary and Murphy, Andrew J., additional
- Published
- 2023
- Full Text
- View/download PDF
6. High-density lipoprotein inhibits human M1 macrophage polarization through redistribution of caveolin-1
- Author
-
Lee, Man K S, Moore, Xiao-Lei, Fu, Yi, Al-Sharea, Annas, Dragoljevic, Dragana, Fernandez-Rojo, Manuel A, Parton, Robert, Sviridov, Dmitri, Murphy, Andrew J, and Chin-Dusting, Jaye P F
- Published
- 2016
- Full Text
- View/download PDF
7. Additional file 1 of Diastolic dysfunction in a pre-clinical model of diabetes is associated with changes in the cardiac non-myocyte cellular composition
- Author
-
Cohen, Charles D., De Blasio, Miles J., Lee, Man K. S., Farrugia, Gabriella E., Prakoso, Darnel, Krstevski, Crisdion, Deo, Minh, Donner, Daniel G., Kiriazis, Helen, Flynn, Michelle C., Gaynor, Taylah L., Murphy, Andrew J., Drummond, Grant R., Pinto, Alexander R., and Ritchie, Rebecca H.
- Abstract
Additional file 1: Table S1. Physiological endpoint characteristics of STZ-HFD-induced murine diabetes. Table S2. Endpoint M-Mode echocardiography for assessing cardiac systolic function in murine diabetes. Table S3. Organ weights. Table S4. Flow cytometry antibody panel utilised in whole blood from mice. Table S5. Flow cytometry antibody panel utilised in myocardium from mice. Figure S1. (A) Representative images of transmitral annular blood flow via Doppler echocardiography. Quantified Doppler flow; (B) Anaesthetised heart rate (HR), (C) peak E-wave velocity, (D) A-wave velocity (E) E:A ratio, (F) deceleration time (DT) and (G) isovolumic relaxation time (IVRT). (H) Shows representative images for tissue Doppler echocardiography, quantified in figures I-L. (I) Peak e' velocity (P=0.054), (J) peak a' velocity (P = 0.072), (K) e':a' ratio, (L) E:e' ratio. ND = non-diabetic, DM = diabetes mellitus. Data presented as mean ± SEM and individual data points, and analysed using an unpaired t-test. Statistical significance was assumed at P
- Published
- 2021
- Full Text
- View/download PDF
8. Diastolic Dysfunction in a Pre-clinical Model of Diabetes Is Associated With Changes in the Cardiac Non-myocyte Cellular Composition
- Author
-
Cohen, Charles D., primary, Blasio, Miles J. De, additional, Lee, Man K. S., additional, Farrugia, Gabriella E., additional, Prakoso, Darnel, additional, Krstevski, Crisdion, additional, Deo, Minh, additional, Donner, Daniel G., additional, Kiriazis, Helen, additional, Flynn, Michelle C., additional, Gaynor, Taylah L., additional, Murphy, Andrew J., additional, Drummond, Grant R., additional, Pinto, Alexander R., additional, and Ritchie, Rebecca Helen, additional
- Published
- 2021
- Full Text
- View/download PDF
9. Healthy Gut, Healthy Bones: Targeting the Gut Microbiome to Promote Bone Health
- Author
-
Cooney, Olivia D., primary, Nagareddy, Prabhakar R., additional, Murphy, Andrew J., additional, and Lee, Man K. S., additional
- Published
- 2021
- Full Text
- View/download PDF
10. Glycolysis Is Required for LPS-Induced Activation and Adhesion of Human CD14+CD16− Monocytes
- Author
-
Lee, Man K. S., primary, Al-Sharea, Annas, additional, Shihata, Waled A., additional, Bertuzzo Veiga, Camilla, additional, Cooney, Olivia D., additional, Fleetwood, Andrew J., additional, Flynn, Michelle C., additional, Claeson, Ellen, additional, Palmer, Clovis S., additional, Lancaster, Graeme I., additional, Henstridge, Darren C., additional, Hamilton, John A., additional, and Murphy, Andrew J., additional
- Published
- 2019
- Full Text
- View/download PDF
11. Cardioprotective Actions of the Annexin-A1 N-Terminal Peptide, Ac2-26, Against Myocardial Infarction
- Author
-
Qin, Cheng Xue, primary, Rosli, Sarah, additional, Deo, Minh, additional, Cao, Nga, additional, Walsh, Jesse, additional, Tate, Mitchel, additional, Alexander, Amy E., additional, Donner, Daniel, additional, Horlock, Duncan, additional, Li, Renming, additional, Kiriazis, Helen, additional, Lee, Man K. S., additional, Bourke, Jane E., additional, Yang, Yuan, additional, Murphy, Andrew J., additional, Du, Xiao-Jun, additional, Gao, Xiao Ming, additional, and Ritchie, Rebecca H., additional
- Published
- 2019
- Full Text
- View/download PDF
12. Glycolysis Is Required for LPS-Induced Activation and Adhesion of Human CD14(+)CD16(-) Monocytes
- Author
-
Lee, Man K. S., Al-Sharea, Annas, Shihata, Waled A., Veiga, Camilla Bertuzzo, Cooney, Olivia D., Fleetwood, Andrew J., Flynn, Michelle C., Claeson, Ellen, Palmer, Clovis S., Lancaster, Graeme I., Henstridge, Darren C., Hamilton, John A., Murphy, Andrew J., Lee, Man K. S., Al-Sharea, Annas, Shihata, Waled A., Veiga, Camilla Bertuzzo, Cooney, Olivia D., Fleetwood, Andrew J., Flynn, Michelle C., Claeson, Ellen, Palmer, Clovis S., Lancaster, Graeme I., Henstridge, Darren C., Hamilton, John A., and Murphy, Andrew J.
- Abstract
Monocytes in humans consist of 3 subsets; CD14(+)CD16(-) (classical), CD14(+)CD16(+) (intermediate) and CD14(dim)CD16+ (non-classical), which exhibit distinct and heterogeneous responses to activation. During acute inflammation CD14(+)CD16(-) monocytes are significantly elevated and migrate to the sites of injury via the adhesion cascade. The field of immunometabolism has begun to elucidate the importance of the engagement of specific metabolic pathways in immune cell function. Yet, little is known about monocyte metabolism and the role of metabolism in mediating monocyte activation and adherence to vessels. Accordingly, we aimed to determine whether manipulating the metabolism of CD14(+)CD16(-) monocytes alters their ability to become activated and adhere. We discovered that LPS stimulation increased the rate of glycolysis in human CD14(+)CD16(-) monocytes. Inhibition of glycolysis with 2-deoxy-D-glucose blunted LPS-induced activation and adhesion of monocytes. Mechanistically, we found that increased glycolysis was regulated by mTOR-induced glucose transporter (GLUT)- 1. Furthermore, enhanced glycolysis increased accumulation of reactive oxygen species (ROS) and activation of p38 MAPK, which lead to activation and adhesion of monocytes. These findings reveal that glycolytic metabolism is critical for the activation of CD14(+)CD16(-) monocytes and contributes to our understanding of the interplay between metabolic substrate preference and immune cell function., Funding Agencies|NHMRCNational Health and Medical Research Council of Australia [APP1142398]; Centenary Award from CSL; National Heart foundationNational Heart Foundation of Australia [101951]
- Published
- 2019
- Full Text
- View/download PDF
13. Apoptotic Ablation of Platelets Reduces Atherosclerosis in Mice With Diabetes.
- Author
-
Lee, Man K. S., Kraakman, Michael J., Dragoljevic, Dragana, Hanssen, Nordin M. J., Flynn, Michelle C., Al-Sharea, Annas, Sreejit, Gopalkrishna, Bertuzzo-Veiga, Camilla, Cooney, Olivia D., Baig, Fatima, Morriss, Elizabeth, Cooper, Mark E., Josefsson, Emma C., Kile, Benjamin T., Nagareddy, Prabhakara R., and Murphy, Andrew J.
- Published
- 2021
- Full Text
- View/download PDF
14. Chronic sympathetic driven hypertension promotes atherosclerosis by enhancing hematopoiesis
- Author
-
Al-Sharea, Annas, primary, Lee, Man K. S., additional, Whillas, Alexandra, additional, Michell, Danielle L., additional, Shihata, Waled A., additional, Nicholls, Alyce J., additional, Cooney, Olivia D., additional, Kraakman, Michael J., additional, Veiga, Camilla Bertuzzo, additional, Jefferis, Ann-Maree, additional, Jackson, Kristy, additional, Nagareddy, Prabhakara R., additional, Lambert, Gavin, additional, Wong, Connie H. Y., additional, Andrews, Karen L., additional, Head, Geoff A., additional, Chin-Dusting, Jaye, additional, and Murphy, Andrew J., additional
- Published
- 2018
- Full Text
- View/download PDF
15. Effects of high‐ and low‐dose aspirin on adaptive immunity and hypertension in the stroke‐prone spontaneously hypertensive rat
- Author
-
Khan, Shanzana I., primary, Shihata, Waled A., additional, Andrews, Karen L., additional, Lee, Man K. S., additional, Moore, Xiao-Lei, additional, Jefferis, Ann-Maree, additional, Vinh, Antony, additional, Gaspari, Tracey, additional, Dragoljevic, Dragana, additional, Jennings, Garry L., additional, Murphy, Andrew J., additional, and Chin-Dusting, Jaye P. F., additional
- Published
- 2018
- Full Text
- View/download PDF
16. Y‐chromosome lineage determines cardiovascular organ T‐cell infiltration in the stroke‐prone spontaneously hypertensive rat
- Author
-
Khan, Shanzana I., primary, Andrews, Karen L., additional, Jackson, Kristy L., additional, Memon, Basimah, additional, Jefferis, Ann‐Maree, additional, Lee, Man K. S., additional, Diep, Henry, additional, Wei, Zihui, additional, Drummond, Grant R., additional, Head, Geoffrey A., additional, Jennings, Garry L., additional, Murphy, Andrew J., additional, Vinh, Antony, additional, Sampson, Amanda K., additional, and Chin‐Dusting, Jaye P. F., additional
- Published
- 2018
- Full Text
- View/download PDF
17. Assessment of metabolic and mitochondrial dynamics in CD4+ and CD8+ T cells in virologically suppressed HIV-positive individuals on combination antiretroviral therapy
- Author
-
Masson, Jesse J. R., primary, Murphy, Andrew J., additional, Lee, Man K. S., additional, Ostrowski, Matias, additional, Crowe, Suzanne M., additional, and Palmer, Clovis S., additional
- Published
- 2017
- Full Text
- View/download PDF
18. High‐density lipoprotein inhibits human M1 macrophage polarization through redistribution of caveolin‐1
- Author
-
Lee, Man K S, Moore, Xiao‐Lei, Fu, Yi, Al‐Sharea, Annas, Dragoljevic, Dragana, Fernandez‐Rojo, Manuel A, Parton, Robert, Sviridov, Dmitri, Murphy, Andrew J, and Chin‐Dusting, Jaye P F
- Subjects
Mice, Knockout ,Mice ,Phenotype ,Macrophages ,Caveolin 1 ,Animals ,Humans ,Themed Section: Research Papers ,Lipoproteins, HDL ,Reactive Oxygen Species - Abstract
Monocyte-derived macrophages are critical in the development of atherosclerosis and can adopt a wide range of functional phenotypes depending on their surrounding milieu. High-density lipoproteins (HDLs) have many cardio-protective properties including potent anti-inflammatory effects. We investigated the effects of HDL on human macrophage phenotype and the mechanisms by which these occur.Human blood monocytes were differentiated into macrophages in the presence or absence of HDL and were then induced to either an inflammatory macrophage (M1) or anti-inflammatory macrophage (M2) phenotype using LPS and IFN-γ or IL-4, respectively.HDL inhibited the induction of macrophages to an M1-phenotype, as evidenced by a decrease in the expression of M1-specific cell surface markers CD192 and CD64, as well as M1-associated inflammatory genes TNF-α, IL-6 and MCP-1 (CCL2). HDL also inhibited M1 function by reducing the production of ROS. In contrast, HDL had no effect on macrophage induction to the M2-phenotype. Similarly, methyl-β-cyclodextrin, a non-specific cholesterol acceptor also suppressed the induction of M1 suggesting that cholesterol efflux is important in this process. Furthermore, HDL decreased membrane caveolin-1 in M1 macrophages. We confirmed that caveolin-1 is required for HDL to inhibit M1 induction as bone marrow-derived macrophages from caveolin-1 knockout mice continued to polarize into M1-phenotype despite the presence of HDL. Moreover, HDL decreased ERK1/2 and STAT3 phosphorylation in M1 macrophages.We concluded that HDL reduces the induction of macrophages to the inflammatory M1-phenotype via redistribution of caveolin-1, preventing the activation of ERK1/2 and STAT3.
- Published
- 2015
19. Cardioprotective Actions of the Annexin-A1 N-Terminal Peptide, Ac2-26, Against Myocardial Infarction.
- Author
-
Qin, Cheng Xue, Rosli, Sarah, Deo, Minh, Cao, Nga, Walsh, Jesse, Tate, Mitchel, Alexander, Amy E., Donner, Daniel, Horlock, Duncan, Li, Renming, Kiriazis, Helen, Lee, Man K. S., Bourke, Jane E., Yang, Yuan, Murphy, Andrew J., Du, Xiao-Jun, Gao, Xiao Ming, and Ritchie, Rebecca H.
- Subjects
HEART fibrosis ,PEPTIDE receptors ,REPERFUSION ,MYOCARDIAL infarction ,CORONARY disease - Abstract
The anti-inflammatory, pro-resolving annexin-A1 protein acts as an endogenous brake against exaggerated cardiac necrosis, inflammation, and fibrosis following myocardial infarction (MI) in vivo. Little is known, however, regarding the cardioprotective actions of the N-terminal-derived peptide of annexin A1, Ac
2-26 , particularly beyond its anti-necrotic actions in the first few hours after an ischemic insult. In this study, we tested the hypothesis that exogenous Ac2-26 limits cardiac injury in vitro and in vivo. Firstly, we demonstrated that Ac2-26 limits cardiomyocyte death both in vitro and in mice subjected to ischemia-reperfusion (I-R) injury in vivo (Ac2-26, 1 mg/kg, i.v. just prior to post-ischemic reperfusion). Further, Ac2-26 (1 mg/kg i.v.) reduced cardiac inflammation (after 48 h reperfusion), as well as both cardiac fibrosis and apoptosis (after 7-days reperfusion). Lastly, we investigated whether Ac2-26 preserved cardiac function after MI. Ac2-26 (1 mg/kg/day s.c., osmotic pump) delayed early cardiac dysfunction 1 week post MI, but elicited no further improvement 4 weeks after MI. Taken together, our data demonstrate the first evidence that Ac2-26 not only preserves cardiomyocyte survival in vitro , but also offers cardioprotection beyond the first few hours after an ischemic insult in vivo. Annexin-A1 mimetics thus represent a potential new therapy to improve cardiac outcomes after MI. [ABSTRACT FROM AUTHOR]- Published
- 2019
- Full Text
- View/download PDF
20. Attack of the NETs! NETosis primes IL-1β-mediated inflammation in diabetic foot ulcers.
- Author
-
Lee, Man K. S., Sreejit, Gopalkrishna, Nagareddy, Prabhakara R., and Murphy, Andrew J.
- Subjects
- *
DIABETIC foot , *INFLAMMATION , *WOUND healing , *MACROPHAGE activation - Abstract
In volume 133 issue 4 of Clinical Science, Liu et al. showed that neutrophils release extracellular traps (NETs) in the setting of diabetes which acts as a stimulus for NLRP3 inflammasome activation in macrophages to promote IL1ß-dependent exacerbation of inflammation. They also provide evidence to show that degrading NETs improves the wound healing process. These findings provide an insight into how NETs communicate with other cells in the vicinity (e.g. macrophages) to exacerbate the inflammatory response. Most importantly, they provide novel avenues to improve wound healing process such as diabetic foot ulcers (DFUs) by targeting NETs. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
21. Effects of high- and low-dose aspirin on adaptive immunity and hypertension in the stroke-prone spontaneously hypertensive rat.
- Author
-
Khan, Shanzana I., Shihata, Waled A., Andrews, Karen L., Lee, Man K. S., Xiao-Lei Moore, Jefferis, Ann-Maree, Vinh, Antony, Gaspari, Tracey, Dragoljevic, Dragana, Jennings, Garry L., Murphy, Andrew J., and Chin-Dusting, Jaye P. F.
- Published
- 2019
- Full Text
- View/download PDF
22. High-density lipoprotein inhibits human M1 macrophage polarization through redistribution of caveolin-1
- Author
-
Lee, Man K S, primary, Moore, Xiao-Lei, additional, Fu, Yi, additional, Al-Sharea, Annas, additional, Dragoljevic, Dragana, additional, Fernandez-Rojo, Manuel A, additional, Parton, Robert, additional, Sviridov, Dmitri, additional, Murphy, Andrew J, additional, and Chin-Dusting, Jaye P F, additional
- Published
- 2015
- Full Text
- View/download PDF
23. Caveolin-1 Plays a Critical Role in the Differentiation of Monocytes into Macrophages
- Author
-
Fu, Yi, primary, Moore, Xiao-Lei, additional, Lee, Man K. S., additional, Fernández-Rojo, Manuel A., additional, Parat, Marie-Odile, additional, Parton, Robert G., additional, Meikle, Peter J., additional, Sviridov, Dmitri, additional, and Chin-Dusting, Jaye P. F., additional
- Published
- 2012
- Full Text
- View/download PDF
24. Autocrine IFN-I inhibits isocitrate dehydrogenase in the TCA cycle of LPS-stimulated macrophages.
- Author
-
De Souza, David P., Achuthan, Adrian, Lee, Man K. S., Binger, Katrina J., Ming-Chin Lee, Davidson, Sophia, Tull, Dedreia L., McConville, Malcolm J., Cook, Andrew D., Murphy, Andrew J., Hamilton, John A., Fleetwood, Andrew J., Lee, Man Ks, and Lee, Ming-Chin
- Subjects
- *
ISOCITRATE dehydrogenase , *MACROPHAGES , *MACROPHAGE activation , *ANIMAL experimentation , *CELL physiology , *CELL receptors , *COMPARATIVE studies , *ENERGY metabolism , *IMMUNITY , *INTERFERONS , *INTERLEUKINS , *RESEARCH methodology , *MEDICAL cooperation , *MICE , *OXIDOREDUCTASES , *RESEARCH , *EVALUATION research , *ACYCLIC acids , *LIPOPOLYSACCHARIDES , *CHEMICAL inhibitors - Abstract
Macrophage activation in response to LPS is coupled to profound metabolic changes, typified by accumulation of the TCA cycle intermediates citrate, itaconate, and succinate. We have identified that endogenous type I IFN controls the cellular citrate/α-ketoglutarate ratio and inhibits expression and activity of isocitrate dehydrogenase (IDH); and, via 13C-labeling studies, demonstrated that autocrine type I IFN controls carbon flow through IDH in LPS-activated macrophages. We also found that type I IFN-driven IL-10 contributes to inhibition of IDH activity and itaconate synthesis in LPS-stimulated macrophages. Our findings have identified the autocrine type I IFN pathway as being responsible for the inhibition of IDH in LPS-stimulated macrophages. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
25. HIV-1-DNA/RNA and immunometabolism in monocytes: contribution to the chronic immune activation and inflammation in people with HIV-1.
- Author
-
Muñoz-Muela E, Trujillo-Rodríguez M, Serna-Gallego A, Saborido-Alconchel A, Gasca-Capote C, Álvarez-Ríos A, Ruiz-Mateos E, Sviridov D, Murphy AJ, Lee MKS, López-Cortés LF, and Gutiérrez-Valencia A
- Subjects
- Humans, Male, Female, Adult, Middle Aged, Cross-Sectional Studies, Viral Load, Immunophenotyping, Proviruses genetics, Biomarkers, HIV Infections immunology, HIV Infections virology, HIV Infections metabolism, Monocytes metabolism, Monocytes immunology, HIV-1, DNA, Viral, RNA, Viral metabolism, Inflammation metabolism, Inflammation immunology
- Abstract
Background: Among people living with HIV-1 (PHIV), immunological non-responders (INR) experience incomplete immune recovery despite suppressive antiretroviral treatment (ART), facing more severe non-AIDS events than immunological responders (IR) due to higher chronic immune activation and inflammation (cIA/I). We analyzed the HIV-1 reservoir and immunometabolism in monocytes as a source of cIA/I., Methods: Cross-sectional study in which 110 participants were enrolled: 25 treatment-naïve; 35 INR; 40 IR; and 10 healthy controls. Cell-associated HIV-1-DNA (HIV-DNA) and -RNA (HIV-RNA) were measured in FACS-isolated monocytes using digital droplet PCR. Intact, 5' deleted, and 3' deleted proviruses were quantified by the intact proviral DNA assay. Systemic inflammation, monocyte immunophenotype, and immunometabolism were characterized by immunoassays, flow cytometry, and real-time cellular bioenergetics measurements, respectively., Findings: Monocytes from INR harbor higher HIV-RNA and HIV-DNA levels than IR. HIV-RNA was found in 14/21 treatment-naïve [2512 copies/10
6 TBP (331-4666)], 17/33 INR [240 (148-589)], and 15/28 IR [144 (15-309)], correlating directly with sCD163, IP-10, GLUT1high cells and glucose uptake, and inversely with the CD4+ /CD8+ ratio. HIV-DNA was identified in all participants with detectable HIV-RNA, with intact provirus in 9/12 treatment-naïve [13 copies/106 monocytes (7-44)], 8/14 INR [46 (18-67)], and 9/13 IR [9 (7-24)]. INR presented glucose metabolism alterations and mitochondrial impairment; decreased coupling efficiency and BHI, and increased mitochondrial dysfunction inversely correlating with the CD4+ /CD8+ ratio., Interpretation: HIV-RNA, more than HIV-DNA, in monocytes and their altered metabolism are factors associated with the higher cIA/I that characterize INR., Funding: This work was supported by the European Regional Development Fund, ISCIII, grant PI20/01646. Other funding sources: Instituto de Salud Carlos III through the Subprogram Miguel Servet (CP19/00159) to AGV, PFIS contracts (FI19/00304) to EMM, (FI21/00165) to ASA, and (FI19/00083) to CGC, and a mobility grant (MV21/00103) to EMM, from the Ministerio de Ciencia e Innovación, Spain. AJM was granted by a CSL Centenary Award., Competing Interests: Declaration of interests The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Crown Copyright © 2024. Published by Elsevier B.V. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
26. The Role of Activator Protein-1 Complex in Diabetes-Associated Atherosclerosis: Insights From Single-Cell RNA Sequencing.
- Author
-
Khan AW, Aziz M, Sourris KC, Lee MKS, Dai A, Watson AMD, Maxwell S, Sharma A, Zhou Y, Cooper ME, Calkin AC, Murphy AJ, Baratchi S, and Jandeleit-Dahm KAM
- Subjects
- Animals, Humans, Mice, Male, Endothelial Cells metabolism, Sequence Analysis, RNA, Atherosclerosis metabolism, Atherosclerosis genetics, Transcription Factor AP-1 metabolism, Transcription Factor AP-1 genetics, Single-Cell Analysis, Diabetes Mellitus, Experimental metabolism, Diabetes Mellitus, Experimental genetics, Diabetes Mellitus, Experimental complications
- Abstract
Despite advances in treatment, atherosclerotic cardiovascular disease remains the leading cause of death in patients with diabetes. Even when risk factors are mitigated, the disease progresses, and thus, newer targets need to be identified that directly inhibit the underlying pathobiology of atherosclerosis in diabetes. A single-cell sequencing approach was used to distinguish the proatherogenic transcriptional profile in aortic cells in diabetes using a streptozotocin-induced diabetic Apoe-/- mouse model. Human carotid endarterectomy specimens from individuals with and without diabetes were also evaluated via immunohistochemical analysis. Further mechanistic studies were performed in human aortic endothelial cells (HAECs) and human THP-1-derived macrophages. We then performed a preclinical study using an activator protein-1 (AP-1) inhibitor in a diabetic Apoe-/- mouse model. Single-cell RNA sequencing analysis identified the AP-1 complex as a novel target in diabetes-associated atherosclerosis. AP-1 levels were elevated in carotid endarterectomy specimens from individuals with diabetes compared with those without diabetes. AP-1 was validated as a mechanosensitive transcription factor via immunofluorescence staining for regional heterogeneity of endothelial cells of the aortic region exposed to turbulent blood flow and by performing microfluidics experiments in HAECs. AP-1 inhibition with T-5224 blunted endothelial cell activation as assessed by a monocyte adhesion assay and expression of genes relevant to endothelial function. Furthermore, AP-1 inhibition attenuated foam cell formation. Critically, treatment with T-5224 attenuated atherosclerosis development in diabetic Apoe-/- mice. This study has identified the AP-1 complex as a novel target, the inhibition of which treats the underlying pathobiology of atherosclerosis in diabetes., (© 2024 by the American Diabetes Association.)
- Published
- 2024
- Full Text
- View/download PDF
27. Defective AMPK regulation of cholesterol metabolism accelerates atherosclerosis by promoting HSPC mobilization and myelopoiesis.
- Author
-
Lee MKS, Cooney OD, Lin X, Nadarajah S, Dragoljevic D, Huynh K, Onda DA, Galic S, Meikle PJ, Edlund T, Fullerton MD, Kemp BE, Murphy AJ, and Loh K
- Subjects
- AMP-Activated Protein Kinases metabolism, Animals, Apolipoproteins E genetics, Cholesterol, Female, Male, Mice, Atherosclerosis metabolism, Myelopoiesis
- Abstract
Objectives: Dysregulation of cholesterol metabolism in the liver and hematopoietic stem and progenitor cells (HSPCs) promotes atherosclerosis development. Previously, it has been shown that HMG-CoA-Reductase (HMGCR), the rate-limiting enzyme in the mevalonate pathway, can be phosphorylated and inactivated by the metabolic stress sensor AMP-activated protein kinase (AMPK). However, the physiological significance of AMPK regulation of HMGCR to atherogenesis has yet to be elucidated. The aim of this study was to determine the role of AMPK/HMGCR axis in the development of atherosclerosis., Methods: We have generated a novel atherosclerotic-prone mouse model with defects in the AMPK regulation of HMGCR (Apoe
-/- /Hmgcr KI mice). Atherosclerotic lesion size, plaque composition, immune cell and lipid profiles were assessed in Apoe-/- and Apoe-/- /Hmgcr KI mice., Results: In this study, we showed that both male and female atherosclerotic-prone mice with a disruption of HMGCR regulation by AMPK (Apoe-/- /Hmgcr KI mice) display increased aortic lesion size concomitant with an increase in plaque-associated macrophages and lipid accumulation. Consistent with this, Apoe-/- /Hmgcr KI mice exhibited an increase in total circulating cholesterol and atherogenic monocytes, Ly6-Chi subset. Mechanistically, increased circulating atherogenic monocytes in Apoe-/- /Hmgcr KI mice was associated with enhanced egress of bone marrow HSPCs and extramedullary myelopoiesis, driven by a combination of elevated circulating 27-hydroxycholesterol and intracellular cholesterol in HSPCs., Conclusions: Our results uncovered a novel signalling pathway involving AMPK-HMGCR axis in the regulation of cholesterol homeostasis in HSPCs, and that inhibition of this regulatory mechanism accelerates the development and progression of atherosclerosis. These findings provide a molecular basis to support the use of AMPK activators that currently undergoing Phase II clinical trial such as O-3O4 and PXL 770 for reducing atherosclerotic cardiovascular disease risks., (Copyright © 2022 The Author(s). Published by Elsevier GmbH.. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
28. Type I interferon antagonism of the JMJD3-IRF4 pathway modulates macrophage activation and polarization.
- Author
-
Ming-Chin Lee K, Achuthan AA, De Souza DP, Lupancu TJ, Binger KJ, Lee MKS, Xu Y, McConville MJ, de Weerd NA, Dragoljevic D, Hertzog PJ, Murphy AJ, Hamilton JA, and Fleetwood AJ
- Subjects
- Granulocyte-Macrophage Colony-Stimulating Factor genetics, Ketoglutaric Acids metabolism, Ketoglutaric Acids pharmacology, Succinic Acid, Interferon Type I, Macrophage Activation
- Abstract
Metabolic adaptations can directly influence the scope and scale of macrophage activation and polarization. Here we explore the impact of type I interferon (IFNβ) on macrophage metabolism and its broader impact on cytokine signaling pathways. We find that IFNβ simultaneously increased the expression of immune-responsive gene 1 and itaconate production while inhibiting isocitrate dehydrogenase activity and restricting α-ketoglutarate accumulation. IFNβ also increased the flux of glutamine-derived carbon into the tricarboxylic acid cycle to boost succinate levels. Combined, we identify that IFNβ controls the cellular α-ketoglutarate/succinate ratio. We show that by lowering the α-ketoglutarate/succinate ratio, IFNβ potently blocks the JMJD3-IRF4-dependent pathway in GM-CSF and IL-4 activated macrophages. The suppressive effects of IFNβ on JMJD3-IRF4-dependent responses, including M2 polarization and GM-CSF-induced inflammatory pain, were reversed by supplementation with α-ketoglutarate. These results reveal that IFNβ modulates macrophage activation and polarization through control of the cellular α-ketoglutarate/succinate ratio., Competing Interests: Declaration of interests The authors declare no conflict of interests., (Copyright © 2022 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
29. Retention of the NLRP3 Inflammasome-Primed Neutrophils in the Bone Marrow Is Essential for Myocardial Infarction-Induced Granulopoiesis.
- Author
-
Sreejit G, Nooti SK, Jaggers RM, Athmanathan B, Ho Park K, Al-Sharea A, Johnson J, Dahdah A, Lee MKS, Ma J, Murphy AJ, and Nagareddy PR
- Subjects
- Animals, Humans, Mice, Signal Transduction, Granulocytes metabolism, Inflammasomes metabolism, Myocardial Infarction complications, NLR Family, Pyrin Domain-Containing 3 Protein metabolism, Neutrophils metabolism
- Abstract
Background: Acute myocardial infarction (MI) results in overzealous production and infiltration of neutrophils to the ischemic heart. This is mediated in part by granulopoiesis induced by the S100A8/A9-NLRP3-IL-1β signaling axis in injury-exposed neutrophils. Despite the transcriptional upregulation of the NLRP3 (Nod Like Receptor Family Pyrin Domain-Containing 3) inflammasome and associated signaling components in neutrophils, the serum levels of IL-1β (interleukin-1β), the effector molecule in granulopoiesis, were not affected by MI, suggesting that IL-1β is not released systemically. We hypothesize that IL-1β is released locally within the bone marrow (BM) by inflammasome-primed and reverse-migrating neutrophils., Methods: Using a combination of time-dependent parabiosis and flow cytometry techniques, we first characterized the migration patterns of different blood cell types across the parabiotic barrier. We next induced MI in parabiotic mice by permanent ligation of the left anterior descending artery and examined the ability of injury-exposed neutrophils to permeate the parabiotic barrier and induce granulopoiesis in noninfarcted parabionts. Last, using multiple neutrophil adoptive and BM transplant studies, we studied the molecular mechanisms that govern reverse migration and retention of the primed neutrophils, IL-1β secretion, and granulopoiesis. Cardiac function was assessed by echocardiography., Results: MI promoted greater accumulation of the inflammasome-primed neutrophils in the BM. Introducing a time-dependent parabiotic barrier to the free movement of neutrophils inhibited their ability to stimulate granulopoiesis in the noninfarcted parabionts. Previous priming of the NLRP3 inflammasome is not a prerequisite, but the presence of a functional CXCR4 (C-X-C-motif chemokine receptor 4) on the primed-neutrophils and elevated serum S100A8/A9 levels are necessary for homing and retention of the reverse-migrating neutrophils. In the BM, the primed-neutrophils secrete IL-1β through formation of gasdermin D pores and promote granulopoiesis. Pharmacological and genetic strategies aimed at the inhibition of neutrophil homing or release of IL-1β in the BM markedly suppressed MI-induced granulopoiesis and improved cardiac function., Conclusions: Our data reveal a new paradigm of how circulatory cells establish a direct communication between organs by delivering signaling molecules (eg, IL-1β) directly at the sites of action rather through systemic release. We suggest that this pathway may exist to limit the off-target effects of systemic IL-1β release.
- Published
- 2022
- Full Text
- View/download PDF
30. A spontaneously hypertensive diet-induced atherosclerosis-prone mouse model of metabolic syndrome.
- Author
-
Dragoljevic D, Veiga CB, Michell DL, Shihata WA, Al-Sharea A, Head GA, Murphy AJ, Kraakman MJ, and Lee MKS
- Subjects
- Animals, Atherosclerosis blood, Atherosclerosis metabolism, Blood Glucose metabolism, Cholesterol blood, Disease Models, Animal, Female, Glucose Intolerance blood, Glucose Intolerance metabolism, Glucose Intolerance pathology, Hypercholesterolemia blood, Hypercholesterolemia metabolism, Hypercholesterolemia pathology, Hyperglycemia blood, Hyperglycemia metabolism, Hyperglycemia pathology, Hyperlipidemias blood, Hyperlipidemias metabolism, Hyperlipidemias pathology, Hypertension blood, Hypertension metabolism, Insulin Resistance physiology, Metabolic Syndrome blood, Metabolic Syndrome metabolism, Mice, Mice, Inbred C57BL, Obesity blood, Obesity metabolism, Obesity pathology, Triglycerides blood, Atherosclerosis pathology, Diet adverse effects, Hypertension pathology, Metabolic Syndrome pathology
- Abstract
Metabolic Syndrome (MetS) is a complex and multifactorial condition often characterised by obesity, hypertension, hyperlipidaemia, insulin resistance, glucose intolerance and fasting hyperglycaemia. Collectively, MetS can increase the risk of atherosclerotic-cardiovascular disease, which is the leading cause of death worldwide. However, no animal model currently exists to study MetS in the context of atherosclerosis. In this study we developed a pre-clinical mouse model that recapitulates the spectrum of MetS features while developing atherosclerosis. When BPHx mice were placed on a western type diet for 16 weeks, all the classical characteristics of MetS were observed. Comprehensive metabolic analyses and atherosclerotic imaging revealed BPHx mice to be obese and hypertensive, with elevated total plasma cholesterol and triglyceride levels, that accelerated atherosclerosis. Altogether, we demonstrate that the BPHx mouse has all the major components of MetS, and accelerates the development of atherosclerosis., (Copyright © 2021. Published by Elsevier Masson SAS.)
- Published
- 2021
- Full Text
- View/download PDF
31. Shark liver oil supplementation enriches endogenous plasmalogens and reduces markers of dyslipidemia and inflammation.
- Author
-
Paul S, Smith AAT, Culham K, Gunawan KA, Weir JM, Cinel MA, Jayawardana KS, Mellett NA, Lee MKS, Murphy AJ, Lancaster GI, Nestel PJ, Kingwell BA, and Meikle PJ
- Subjects
- Adult, Animals, Biomarkers blood, Cross-Over Studies, Dietary Supplements, Double-Blind Method, Dyslipidemias metabolism, Fish Oils administration & dosage, Humans, Inflammation metabolism, Male, Middle Aged, Plasmalogens blood, Sharks, Dyslipidemias drug therapy, Fish Oils pharmacology, Inflammation drug therapy, Plasmalogens metabolism
- Abstract
Plasmalogens are membrane glycerophospholipids with diverse biological functions. Reduced plasmalogen levels have been observed in metabolic diseases; hence, increasing their levels might be beneficial in ameliorating these conditions. Shark liver oil (SLO) is a rich source of alkylglycerols that can be metabolized into plasmalogens. This study was designed to evaluate the impact of SLO supplementation on endogenous plasmalogen levels in individuals with features of metabolic disease. In this randomized, double-blind, placebo-controlled cross-over study, the participants (10 overweight or obese males) received 4-g Alkyrol® (purified SLO) or placebo (methylcellulose) per day for 3 weeks followed by a 3-week washout phase and were then crossed over to 3 weeks of the alternate placebo/Alkyrol® treatment. SLO supplementation led to significant changes in plasma and circulatory white blood cell lipidomes, notably increased levels of plasmalogens and other ether lipids. In addition, SLO supplementation significantly decreased the plasma levels of total free cholesterol, triglycerides, and C-reactive protein. These findings suggest that SLO supplementation can enrich plasma and cellular plasmalogens and this enrichment may provide protection against obesity-related dyslipidemia and inflammation., Competing Interests: Conflict of interest The authors declare that they have no conflicts of interest with the contents of this article., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
32. Transient Intermittent Hyperglycemia Accelerates Atherosclerosis by Promoting Myelopoiesis.
- Author
-
Flynn MC, Kraakman MJ, Tikellis C, Lee MKS, Hanssen NMJ, Kammoun HL, Pickering RJ, Dragoljevic D, Al-Sharea A, Barrett TJ, Hortle F, Byrne FL, Olzomer E, McCarthy DA, Schalkwijk CG, Forbes JM, Hoehn K, Makowski L, Lancaster GI, El-Osta A, Fisher EA, Goldberg IJ, Cooper ME, Nagareddy PR, Thomas MC, and Murphy AJ
- Subjects
- Animals, Atherosclerosis genetics, Atherosclerosis metabolism, Atherosclerosis pathology, Biomarkers blood, Calgranulin A genetics, Calgranulin A metabolism, Calgranulin B genetics, Calgranulin B metabolism, Diet, High-Fat, Disease Models, Animal, Glucose Transporter Type 1 genetics, Glucose Transporter Type 1 metabolism, Glycolysis, Hyperglycemia blood, Macrophages metabolism, Macrophages pathology, Male, Mice, Inbred C57BL, Mice, Knockout, ApoE, Monocytes pathology, Neutrophils pathology, Plaque, Atherosclerotic, Receptor for Advanced Glycation End Products genetics, Receptor for Advanced Glycation End Products metabolism, Signal Transduction, Atherosclerosis etiology, Blood Glucose metabolism, Hyperglycemia complications, Monocytes metabolism, Myelopoiesis, Neutrophils metabolism
- Abstract
Rationale: Treatment efficacy for diabetes mellitus is largely determined by assessment of HbA1c (glycated hemoglobin A1c) levels, which poorly reflects direct glucose variation. People with prediabetes and diabetes mellitus spend >50% of their time outside the optimal glucose range. These glucose variations, termed transient intermittent hyperglycemia (TIH), appear to be an independent risk factor for cardiovascular disease, but the pathological basis for this association is unclear., Objective: To determine whether TIH per se promotes myelopoiesis to produce more monocytes and consequently adversely affects atherosclerosis., Methods and Results: To create a mouse model of TIH, we administered 4 bolus doses of glucose at 2-hour intervals intraperitoneally once to WT (wild type) or once weekly to atherosclerotic prone mice. TIH accelerated atherogenesis without an increase in plasma cholesterol, seen in traditional models of diabetes mellitus. TIH promoted myelopoiesis in the bone marrow, resulting in increased circulating monocytes, particularly the inflammatory Ly6-C
hi subset, and neutrophils. Hematopoietic-restricted deletion of S100a9 , S100a8 , or its cognate receptor Rage prevented monocytosis. Mechanistically, glucose uptake via GLUT (glucose transporter)-1 and enhanced glycolysis in neutrophils promoted the production of S100A8/A9. Myeloid-restricted deletion of Slc2a1 (GLUT-1) or pharmacological inhibition of S100A8/A9 reduced TIH-induced myelopoiesis and atherosclerosis., Conclusions: Together, these data provide a mechanism as to how TIH, prevalent in people with impaired glucose metabolism, contributes to cardiovascular disease. These findings provide a rationale for continual glucose control in these patients and may also suggest that strategies aimed at targeting the S100A8/A9-RAGE (receptor for advanced glycation end products) axis could represent a viable approach to protect the vulnerable blood vessels in diabetes mellitus. Graphic Abstract: A graphic abstract is available for this article.- Published
- 2020
- Full Text
- View/download PDF
33. Interplay between Clonal Hematopoiesis of Indeterminate Potential and Metabolism.
- Author
-
Lee MKS, Dragoljevic D, Bertuzzo Veiga C, Wang N, Yvan-Charvet L, and Murphy AJ
- Subjects
- Animals, Clonal Hematopoiesis genetics, Humans, Mutation genetics, Cardiovascular Diseases metabolism, Cardiovascular Diseases physiopathology, Clonal Hematopoiesis physiology, Diabetes Mellitus metabolism, Diabetes Mellitus physiopathology
- Abstract
Clonal hematopoiesis of indeterminate potential (CHIP), defined as a clone of hematopoietic cells consisting of a single acquired mutation during a lifetime, has recently been discovered to be a major risk factor for atherosclerotic cardiovascular disease (CVD). As such, this phenomenon has sparked interest into the role that these single mutations may play in CVD. Atherosclerotic CVD is a complex disease and we have previously shown that atherosclerosis can be accelerated by metabolic- or autoimmune-related risk factors such as diabetes, obesity, and rheumatoid arthritis. In this review, we discuss the role of CHIP, the interplay between CHIP and metabolic diseases, as well as how metabolism of hematopoietic stem cells (HSCs) could regulate CHIP-related HSC fate., (Copyright © 2020 Elsevier Ltd. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
34. The Multiparametric Analysis of Mitochondrial Dynamics in T Cells from Cryopreserved Peripheral Blood Mononuclear Cells (PBMCs).
- Author
-
Masson JJR, Ostrowski M, Duette G, Lee MKS, Murphy AJ, Crowe SM, and Palmer CS
- Subjects
- Antibodies metabolism, CD4-Positive T-Lymphocytes metabolism, CD8-Positive T-Lymphocytes metabolism, Cell Survival physiology, Cryopreservation methods, Humans, Leukocytes, Mononuclear metabolism, Membrane Potential, Mitochondrial physiology, Mitochondria metabolism, Reactive Oxygen Species metabolism, CD4-Positive T-Lymphocytes physiology, CD8-Positive T-Lymphocytes physiology, Leukocytes, Mononuclear physiology, Mitochondria physiology, Mitochondrial Dynamics physiology
- Abstract
The analysis of mitochondrial dynamics within immune cells allows us to understand how fundamental metabolism influences immune cell functions, and how dysregulated immunometabolic processes impact biology and disease pathogenesis. For example, during infections, mitochondrial fission and fusion coincide with effector and memory T-cell differentiation, respectively, resulting in metabolic reprogramming. As frozen cells are generally not optimal for immunometabolic analyses, and given the logistic difficulties of analysis on cells within a few hours of blood collection, we have optimized and validated a simple cryopreservation protocol for peripheral blood mononuclear cells, yielding >95% cellular viability, as well as preserved metabolic and immunologic properties. Combining fluorescent dyes with cell surface antibodies, we demonstrate how to analyze mitochondrial density, membrane potential, and reactive oxygen species production in CD4 and CD8 T cells from cryopreserved clinical samples.
- Published
- 2020
- Full Text
- View/download PDF
35. Glycolysis Is Required for LPS-Induced Activation and Adhesion of Human CD14 + CD16 - Monocytes.
- Author
-
Lee MKS, Al-Sharea A, Shihata WA, Bertuzzo Veiga C, Cooney OD, Fleetwood AJ, Flynn MC, Claeson E, Palmer CS, Lancaster GI, Henstridge DC, Hamilton JA, and Murphy AJ
- Subjects
- Cell Adhesion, Cells, Cultured, Deoxyglucose metabolism, Glucose Transporter Type 1 metabolism, Glycolysis, Humans, Immunophenotyping, Lipopolysaccharide Receptors metabolism, Lipopolysaccharides metabolism, MAP Kinase Signaling System, Monocytes immunology, Receptors, IgG metabolism, TOR Serine-Threonine Kinases metabolism, Inflammation immunology, Monocytes metabolism, Reactive Oxygen Species metabolism
- Abstract
Monocytes in humans consist of 3 subsets; CD14
+ CD16- (classical), CD14+ CD16+ (intermediate) and CD14dim CD16+ (non-classical), which exhibit distinct and heterogeneous responses to activation. During acute inflammation CD14+ CD16- monocytes are significantly elevated and migrate to the sites of injury via the adhesion cascade. The field of immunometabolism has begun to elucidate the importance of the engagement of specific metabolic pathways in immune cell function. Yet, little is known about monocyte metabolism and the role of metabolism in mediating monocyte activation and adherence to vessels. Accordingly, we aimed to determine whether manipulating the metabolism of CD14+ CD16- monocytes alters their ability to become activated and adhere. We discovered that LPS stimulation increased the rate of glycolysis in human CD14+ CD16- monocytes. Inhibition of glycolysis with 2-deoxy-D-glucose blunted LPS-induced activation and adhesion of monocytes. Mechanistically, we found that increased glycolysis was regulated by mTOR-induced glucose transporter (GLUT)-1. Furthermore, enhanced glycolysis increased accumulation of reactive oxygen species (ROS) and activation of p38 MAPK, which lead to activation and adhesion of monocytes. These findings reveal that glycolytic metabolism is critical for the activation of CD14+ CD16- monocytes and contributes to our understanding of the interplay between metabolic substrate preference and immune cell function.- Published
- 2019
- Full Text
- View/download PDF
36. Cardioprotective Actions of the Annexin-A1 N-Terminal Peptide, Ac 2-26 , Against Myocardial Infarction.
- Author
-
Qin CX, Rosli S, Deo M, Cao N, Walsh J, Tate M, Alexander AE, Donner D, Horlock D, Li R, Kiriazis H, Lee MKS, Bourke JE, Yang Y, Murphy AJ, Du XJ, Gao XM, and Ritchie RH
- Abstract
The anti-inflammatory, pro-resolving annexin-A1 protein acts as an endogenous brake against exaggerated cardiac necrosis, inflammation, and fibrosis following myocardial infarction (MI) in vivo . Little is known, however, regarding the cardioprotective actions of the N-terminal-derived peptide of annexin A1, Ac
2-26 , particularly beyond its anti-necrotic actions in the first few hours after an ischemic insult. In this study, we tested the hypothesis that exogenous Ac2-26 limits cardiac injury in vitro and in vivo. Firstly, we demonstrated that Ac2-26 limits cardiomyocyte death both in vitro and in mice subjected to ischemia-reperfusion (I-R) injury in vivo (Ac2-26, 1 mg/kg, i.v. just prior to post-ischemic reperfusion). Further, Ac2-26 (1 mg/kg i.v.) reduced cardiac inflammation (after 48 h reperfusion), as well as both cardiac fibrosis and apoptosis (after 7-days reperfusion). Lastly, we investigated whether Ac2-26 preserved cardiac function after MI. Ac2-26 (1 mg/kg/day s.c., osmotic pump) delayed early cardiac dysfunction 1 week post MI, but elicited no further improvement 4 weeks after MI. Taken together, our data demonstrate the first evidence that Ac2-26 not only preserves cardiomyocyte survival in vitro , but also offers cardioprotection beyond the first few hours after an ischemic insult in vivo . Annexin-A1 mimetics thus represent a potential new therapy to improve cardiac outcomes after MI.- Published
- 2019
- Full Text
- View/download PDF
37. Chronic sympathetic driven hypertension promotes atherosclerosis by enhancing hematopoiesis.
- Author
-
Al-Sharea A, Lee MKS, Whillas A, Michell DL, Shihata WA, Nicholls AJ, Cooney OD, Kraakman MJ, Veiga CB, Jefferis AM, Jackson K, Nagareddy PR, Lambert G, Wong CHY, Andrews KL, Head GA, Chin-Dusting J, and Murphy AJ
- Subjects
- Animals, Atherosclerosis pathology, Autonomic Nerve Block, Biomarkers, Biopsy, Bone Marrow metabolism, Bone Marrow pathology, Disease Models, Animal, Disease Susceptibility, Hematopoietic Stem Cells cytology, Hematopoietic Stem Cells metabolism, Immunohistochemistry, Mice, Mice, Knockout, Myelopoiesis, Phenotype, Signal Transduction drug effects, Stem Cell Niche, Atherosclerosis blood, Atherosclerosis etiology, Hematopoiesis, Hypertension complications, Hypertension etiology, Sympathetic Nervous System physiopathology
- Abstract
Hypertension is a major, independent risk factor for atherosclerotic cardiovascular disease. However, this pathology can arise through multiple pathways, which could influence vascular disease through distinct mechanisms. An overactive sympathetic nervous system is a dominant pathway that can precipitate in elevated blood pressure. We aimed to determine how the sympathetic nervous system directly promotes atherosclerosis in the setting of hypertension. We used a mouse model of sympathetic nervous system-driven hypertension on the atherosclerotic-prone apolipoprotein E-deficient background. When mice were placed on a western type diet for 16 weeks, we showed the evolution of unstable atherosclerotic lesions. Fortuitously, the changes in lesion composition were independent of endothelial dysfunction, allowing for the discovery of alternative mechanisms. With the use of flow cytometry and bone marrow imaging, we found that sympathetic activation caused deterioration of the hematopoietic stem and progenitor cell niche in the bone marrow, promoting the liberation of these cells into the circulation and extramedullary hematopoiesis in the spleen. Specifically, sympathetic activation reduced the abundance of key hematopoietic stem and progenitor cell niche cells, sinusoidal endothelial cells and osteoblasts. Additionally, sympathetic bone marrow activity prompted neutrophils to secrete proteases to cleave the hematopoietic stem and progenitor cell surface receptor CXCR4. All these effects could be reversed using the β-blocker propranolol during the feeding period. These findings suggest that elevated blood pressure driven by the sympathetic nervous system can influence mechanisms that modulate the hematopoietic system to promote atherosclerosis and contribute to cardiovascular events., (Copyright© 2019 Ferrata Storti Foundation.)
- Published
- 2019
- Full Text
- View/download PDF
38. Hand of FATe: lipid metabolism in hematopoietic stem cells.
- Author
-
Lee MKS, Al-Sharea A, Dragoljevic D, and Murphy AJ
- Subjects
- Adipocytes cytology, Adipocytes metabolism, Animals, Hematopoietic Stem Cells cytology, Humans, Bone Marrow metabolism, Glycolysis physiology, Hematopoiesis physiology, Hematopoietic Stem Cells metabolism, Lipid Metabolism physiology
- Abstract
Purpose of Review: Hematopoietic stem cells (HSCs) reside in the bone marrow and are important in replenishing all cells in the blood through a process termed hematopoiesis. One of the defining characteristics of HSCs is that they must be able to balance their self-renewal capacity with their differentiation into committed blood cells in various blood lineages. For these events to occur, HSCs must be tightly regulated in the bone marrow by intrinsic and extrinsic factors to maintain steady hematopoiesis., Recent Findings: Recently, the effect on how metabolism regulates HSC function has received a great amount of attention. In particular, lipids have been found to participate in mitochondrial activity to maintain HSCs, a role previously overlooked due to HSCs being thought of as mostly glycolytic. Moreover, there has been a re-emergence of how adipocytes in the bone marrow can regulate HSCs., Summary: As these areas evolve, more studies are required to determine the exact contribution of lipids toward HSC maintenance. These studies will allow newer therapeutic targets to help reduce abnormal hematopoiesis such as myelopoiesis, which contributes to many metabolic diseases.
- Published
- 2018
- Full Text
- View/download PDF
39. Evidence that TLR4 Is Not a Receptor for Saturated Fatty Acids but Mediates Lipid-Induced Inflammation by Reprogramming Macrophage Metabolism.
- Author
-
Lancaster GI, Langley KG, Berglund NA, Kammoun HL, Reibe S, Estevez E, Weir J, Mellett NA, Pernes G, Conway JRW, Lee MKS, Timpson P, Murphy AJ, Masters SL, Gerondakis S, Bartonicek N, Kaczorowski DC, Dinger ME, Meikle PJ, Bond PJ, and Febbraio MA
- Subjects
- Animals, Humans, Inflammation etiology, Macrophages cytology, Mice, Mice, Inbred C3H, Mice, Inbred C57BL, Obesity complications, Signal Transduction, Inflammation metabolism, Macrophages metabolism, Obesity metabolism, Palmitates metabolism, Toll-Like Receptor 4 metabolism
- Abstract
Chronic inflammation is a hallmark of obesity and is linked to the development of numerous diseases. The activation of toll-like receptor 4 (TLR4) by long-chain saturated fatty acids (lcSFAs) is an important process in understanding how obesity initiates inflammation. While experimental evidence supports an important role for TLR4 in obesity-induced inflammation in vivo, via a mechanism thought to involve direct binding to and activation of TLR4 by lcSFAs, several lines of evidence argue against lcSFAs being direct TLR4 agonists. Using multiple orthogonal approaches, we herein provide evidence that while loss-of-function models confirm that TLR4 does, indeed, regulate lcSFA-induced inflammation, TLR4 is not a receptor for lcSFAs. Rather, we show that TLR4-dependent priming alters cellular metabolism, gene expression, lipid metabolic pathways, and membrane lipid composition, changes that are necessary for lcSFA-induced inflammation. These results reconcile previous discordant observations and challenge the prevailing view of TLR4's role in initiating obesity-induced inflammation., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
40. Leptin-deficient obesity prolongs survival in a murine model of myelodysplastic syndrome.
- Author
-
Kraakman MJ, Kammoun HL, Dragoljevic D, Al-Sharea A, Lee MKS, Flynn MC, Stolz CJ, Guirguis AA, Lancaster GI, Chin-Dusting J, Curtis DJ, and Murphy AJ
- Subjects
- Animals, Bone Marrow chemistry, Bone Marrow Transplantation, Disease Models, Animal, Homeodomain Proteins, Leptin deficiency, Leukemia, Myeloid, Acute etiology, Mice, Mice, Transgenic, Myelodysplastic Syndromes pathology, Myeloid Cells, Nuclear Pore Complex Proteins, Survival Rate, Transcription Factors, Myelodysplastic Syndromes mortality, Obesity
- Abstract
Obesity enhances the risk of developing myelodysplastic syndromes. However, the effect of obesity on survival is unclear. Obese people present with monocytosis due to inflammatory signals emanating from obese adipose tissue. We hypothesized that obesity-induced myelopoiesis would promote the transition of myelodysplastic syndrome to acute myeloid leukemia and accelerate mortality in obesity. Obese Ob/Ob mice or their lean littermate controls received a bone marrow transplant from NUP98-HOXD13 transgenic mice, a model of myelodysplastic syndrome. The metabolic parameters of the mice were examined throughout the course of the study, as were blood leukocytes. Myeloid cells were analyzed in the bone, spleen, liver and adipose tissue by flow cytometry halfway through the disease progression and at the endpoint. Survival curves were also calculated. Contrary to our hypothesis, transplantation of NUP98-HOXD13 bone marrow into obese recipient mice significantly increased survival time compared with lean recipient controls. While monocyte skewing was exacerbated in obese mice receiving NUP98-HOXD13 bone marrow, transformation to acute myeloid leukemia was not enhanced. Increased survival of obese mice was associated with a preservation of fat mass as well as increased myeloid cell deposition within the adipose tissue, and a concomitant reduction in detrimental myeloid cell accumulation within other organs. The study herein revealed that obesity increases survival in animals with myelodysplastic syndrome. This may be due to the greater fat mass of Ob/Ob mice, which acts as a sink for myeloid cells, preventing their accumulation in other key organs, such as the liver., (Copyright© 2018 Ferrata Storti Foundation.)
- Published
- 2018
- Full Text
- View/download PDF
41. Nicotinic acetylcholine receptor alpha 7 stimulation dampens splenic myelopoiesis and inhibits atherogenesis in Apoe -/- mice.
- Author
-
Al-Sharea A, Lee MKS, Whillas A, Flynn MC, Chin-Dusting J, and Murphy AJ
- Subjects
- Animals, Aorta, Thoracic metabolism, Aorta, Thoracic pathology, Aortic Diseases genetics, Aortic Diseases metabolism, Aortic Diseases pathology, Atherosclerosis genetics, Atherosclerosis metabolism, Atherosclerosis pathology, Diet, Western, Disease Models, Animal, Male, Mice, Knockout, ApoE, Plaque, Atherosclerotic, Spleen metabolism, alpha7 Nicotinic Acetylcholine Receptor metabolism, Aorta, Thoracic drug effects, Aortic Diseases prevention & control, Atherosclerosis prevention & control, Benzylidene Compounds pharmacology, Myelopoiesis drug effects, Nicotinic Agonists pharmacology, Pyridines pharmacology, Spleen drug effects, alpha7 Nicotinic Acetylcholine Receptor agonists
- Abstract
Background and Aims: Monocyte levels predict cardiovascular outcomes and play a causal role in atherogenesis. Monocytes can be produced in the spleen and track to the atherosclerotic lesion in significant numbers. The cholinergic system has been shown to have anti-inflammatory actions in the spleen. We aimed to explore whether therapeutic stimulation of the nicotinic acetylcholine receptor alpha 7 (nAChRα7) can suppress atherogenesis., Methods: Apoe
-/- mice were placed on a Western-type diet and treated with bi-daily injections of the nAChRα7 agonist GTS-21 or vehicle every 2-3 days for 8 weeks., Results: GTS-21 caused a reduction in atherosclerosis in the aortic arch and proximal aorta. This also resulted in less plaque macrophages. Moreover, GTS-21 reduced the abundance of blood monocytes, which was caused by inhibition of inflammatory cytokines and extramedullary hematopoiesis in the spleen, along with splenic monocytes., Conclusions: Stimulation of nAChRα7 with GTS-21 reduced atherosclerosis, which was associated with dampened splenic myelopoiesis., (Copyright © 2017 Elsevier B.V. All rights reserved.)- Published
- 2017
- Full Text
- View/download PDF
42. Metabolic Remodeling, Inflammasome Activation, and Pyroptosis in Macrophages Stimulated by Porphyromonas gingivalis and Its Outer Membrane Vesicles.
- Author
-
Fleetwood AJ, Lee MKS, Singleton W, Achuthan A, Lee MC, O'Brien-Simpson NM, Cook AD, Murphy AJ, Dashper SG, Reynolds EC, and Hamilton JA
- Subjects
- Animals, Caspase 1 metabolism, Cytokines metabolism, Gene Expression, Glycolysis, Humans, Inflammation, Interleukin-10 metabolism, Interleukin-12 metabolism, Interleukin-18, Interleukin-1beta metabolism, Interleukin-6 metabolism, Mice, Mice, Inbred C57BL, Mitochondria metabolism, Nitric Oxide metabolism, Oxidative Phosphorylation, Oxygen Consumption, Porphyromonas gingivalis pathogenicity, Tumor Necrosis Factor-alpha metabolism, Extracellular Vesicles metabolism, Inflammasomes immunology, Macrophages metabolism, Macrophages microbiology, Porphyromonas gingivalis immunology, Pyroptosis
- Abstract
Porphyromonas gingivalis is one of the bacterial species most closely associated with periodontitis and can shed large numbers of outer membrane vesicles (OMVs), which are increasingly thought to play a significant role in bacterial virulence and pathogenicity. Macrophages are amongst the first immune cells to respond to bacteria and their products, so we sought to directly compare the response of macrophages to P. gingivalis or its purified OMVs. Macrophages stimulated with OMVs produced large amounts of TNFα, IL-12p70, IL-6, IL-10, IFNβ, and nitric oxide compared to cells infected with P. gingivalis , which produced very low levels of these mediators. Both P. gingivalis and OMVs induced a shift in macrophage metabolism from oxidative phosphorylation (OXPHOS) to glycolysis, which was supported by enhanced lactate release, decreased mitochondrial oxygen consumption with reduced spare respiratory capacity, as well as increased mitochondrial reactive oxygen species (ROS) production. Corresponding to this metabolic shift, gene expression analysis of macrophages infected with P. gingivalis or stimulated with OMVs revealed a broad transcriptional upregulation of genes critical to glycolysis and a downregulation of genes associated with the TCA cycle. Upon examination of inflammasome signaling and pyroptosis it was found that P. gingivalis did not activate the inflammasome in macrophages as the mature forms of caspase-1, IL-1β, and IL-18 were not detected and there was no extracellular release of lactate dehydrogenase (LDH) or 7-AAD staining. In comparison, macrophages stimulated with OMVs potently activated caspase-1, produced large amounts of IL-1β, IL-18, released LDH, and were positive for 7-AAD indicative of pyroptotic cell death. These data directly quantitate the distinct effects of P. gingivalis and its OMVs on macrophage inflammatory phenotype, mitochondrial function, inflammasome activation, and pyroptotic cell death that may have potential implications for their roles in chronic periodontitis.
- Published
- 2017
- Full Text
- View/download PDF
43. The iPSC Awakens ANGPTL3 in Tangier Disease.
- Author
-
Lee MKS and Murphy AJ
- Subjects
- Angiopoietin-Like Protein 3, Angiopoietin-like Proteins, Atherosclerosis, Humans, Induced Pluripotent Stem Cells, Tangier Disease
- Published
- 2017
- Full Text
- View/download PDF
44. The modern interleukin-1 superfamily: Divergent roles in obesity.
- Author
-
Lee MK, Yvan-Charvet L, Masters SL, and Murphy AJ
- Subjects
- Adipose Tissue immunology, Adipose Tissue metabolism, Animals, Cytokines genetics, Cytokines metabolism, Fatty Acids metabolism, Humans, Immune System cytology, Immune System immunology, Immune System metabolism, Immunomodulation, Inflammasomes metabolism, Inflammation etiology, Inflammation metabolism, Interleukin-1 chemistry, Interleukin-18 genetics, Interleukin-18 metabolism, Interleukin-33 genetics, Interleukin-33 metabolism, Multigene Family, NLR Family, Pyrin Domain-Containing 3 Protein metabolism, Obesity complications, Oxidation-Reduction, Signal Transduction, Interleukin-1 genetics, Interleukin-1 metabolism, Obesity etiology, Obesity metabolism
- Abstract
Obesity is now recognised as a chronic, low-grade inflammatory disease contributing to insulin resistance, type 2 diabetes (T2D) and cardiovascular disease (CVD). Multiple mechanisms leading to the low grade inflammation in this setting have been suggested. Due to the complexity and interconnection of inflammatory and metabolic responses, there also remains a need to fully elucidate the inflammatory mechanisms that control obesity and associated metabolic disorders. One important avenue in the field that has gained great attention is the interleukin (IL)-1 superfamily of cytokines that consist of IL-1β, IL-18 and IL-33. IL-1β is well known for its contribution as an inflammatory mediator in obesity contributing to insulin resistance and T2D, whereas the IL-18 and IL-33 cytokines have been shown to oppose metabolic dysregulation. This review will focus on the current understanding of the IL-1 superfamily of cytokines in the setting of obesity and discuss how endogenous feedback loops can be exploited for therapeutic approaches to fight obesity and subsequent cardiometabolic disorders., (Copyright © 2016 Elsevier Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
45. IL-18 Production from the NLRP1 Inflammasome Prevents Obesity and Metabolic Syndrome.
- Author
-
Murphy AJ, Kraakman MJ, Kammoun HL, Dragoljevic D, Lee MK, Lawlor KE, Wentworth JM, Vasanthakumar A, Gerlic M, Whitehead LW, DiRago L, Cengia L, Lane RM, Metcalf D, Vince JE, Harrison LC, Kallies A, Kile BT, Croker BA, Febbraio MA, and Masters SL
- Subjects
- Adaptor Proteins, Signal Transducing genetics, Animals, Apoptosis Regulatory Proteins genetics, Body Weight, Diet, High-Fat adverse effects, Interleukin-18 genetics, Liver metabolism, Liver pathology, Male, Metabolic Syndrome prevention & control, Mice, Knockout, Obesity etiology, Obesity prevention & control, Adaptor Proteins, Signal Transducing metabolism, Apoptosis Regulatory Proteins metabolism, Inflammasomes metabolism, Interleukin-18 biosynthesis, Metabolic Syndrome metabolism, Obesity metabolism
- Abstract
Interleukin-18 (IL-18) is activated by Caspase-1 in inflammasome complexes and has anti-obesity effects; however, it is not known which inflammasome regulates this process. We found that mice lacking the NLRP1 inflammasome phenocopy mice lacking IL-18, with spontaneous obesity due to intrinsic lipid accumulation. This is exacerbated when the mice are fed a high-fat diet (HFD) or a high-protein diet, but not when mice are fed a HFD with low energy density (high fiber). Furthermore, mice with an activating mutation in NLRP1, and hence increased IL-18, have decreased adiposity and are resistant to diet-induced metabolic dysfunction. Feeding these mice a HFD further increased plasma IL-18 concentrations and strikingly resulted in loss of adipose tissue mass and fatal cachexia, which could be prevented by genetic deletion of IL-18. Thus, NLRP1 is an innate immune sensor that functions in the context of metabolic stress to produce IL-18, preventing obesity and metabolic syndrome., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.