1
|
Zhou B, Pathania A, Pant D, Halpern D, Gaudu P, Trieu-Cuot P, Dias-Leao A, Pagot C, Solgadi A, Gruss A, Gloux K. Prophages divert Staphylococcus aureus defenses against host lipids. J Lipid Res 2024; 65:100693. [PMID: 39505263 PMCID: PMC11721228 DOI: 10.1016/j.jlr.2024.100693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 10/23/2024] [Accepted: 10/29/2024] [Indexed: 11/08/2024] Open
Abstract
Phages are ubiquitous in bacteria, including clinical Staphylococcus aureus, where Sfi 21/Sa3 phages often integrate into the hlb gene, which encodes Hlb sphingomyelinase. This integration acts as a rapid regulatory switch for Hlb production. Our findings suggest that Sfi 21/Sa3 prophages and Hlb activity influence S. aureus fitness by modulating the incorporation of the toxic linoleic acid (C18:2) from serum into the bacterial membrane. This process relies on C18:2 derived from 1,3-diglyceride, facilitated by the FakB1 kinase subunit. Palmitic acid (C16), primarily released from serum through Hlb activity, competes with C18:2 for FakB1. This mechanism contributes to adaptation to AFN-1252, an antibiotic inhibiting the fatty acid synthesis pathway (anti-FASII). Since S. aureus relies on exogenous fatty acids for growth, AFN-1252 treatment leads to an increased proportion of C18:2 in the membrane. Furthermore, Hlb inhibition, whether by prophage insertion, gene inactivation, or enzyme inhibition, delays S. aureus adaptation, resulting in a higher proportion of C18:2 in the membrane. This study sheds light on the role of lipid environments in infections and may contribute to the accurate prediction of infection risks and therapeutic efficacy. Moreover, since both anti-FASII agent and Hlb inhibitor enhance C18:2 incorporation, they represent potential candidates for combined strategies against S. aureus.
Collapse
Affiliation(s)
- Biyang Zhou
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy en Josas, France
| | - Amit Pathania
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy en Josas, France
| | - Deepak Pant
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy en Josas, France
| | - David Halpern
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy en Josas, France
| | - Philippe Gaudu
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy en Josas, France
| | - Patrick Trieu-Cuot
- Institut Pasteur, Université Paris Cité, CNRS UMR 2001, Unité de Biologie des Bactéries Pathogènes à Gram-Positif, Paris, France
| | - Andressa Dias-Leao
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy en Josas, France
| | - Charlotte Pagot
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy en Josas, France
| | - Audrey Solgadi
- UMS-IPSIT SAMM Facility, Université Paris-Saclay, Inserm, CNRS, Ingénierie et Plateformes au Service de l'Innovation Thérapeutique, Paris-Saclay, France
| | - Alexandra Gruss
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy en Josas, France
| | - Karine Gloux
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy en Josas, France.
| |
Collapse
|
2
|
Dong R, Xue H, Chen L, Jin W, Luo Z, Shen C, Huang L, Shao J, Wang J. Associations of Lipid Profiles With the Onset of HEV-Related Acute Liver Failure: A Multicenter Cohort Study. J Med Virol 2024; 96:e70033. [PMID: 39529488 DOI: 10.1002/jmv.70033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/21/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024]
Abstract
Hepatitis E virus (HEV) is one of the major etiologies for acute liver failure. This multicenter retrospective cohort study aimed to investigate the associations of lipid profiles with the risk of HEV-related acute liver failure (HEV-ALF) among hospitalized patients with acute hepatitis E. A total of 1061 participants were obtained from three tertiary medical centers in Jiangsu, China, between February 2018 and May 2024. Univariate and multivariate Cox regression models were constructed to assess the associations between lipid profiles and the risk of HEV-ALF onset. The time-dependent area under the receiver-operating-characteristic curve (AUROC) and decision curve analysis were used to further evaluate the predictive value of blood lipids. After adjusting for potential confounders, total cholesterol (HR = 0.535, 95% CI: 0.437-0.656, p < 0.001), high-density lipoprotein-cholesterol (HR = 0.065, 95% CI: 0.027-0.154, p < 0.001), low-density lipoprotein-cholesterol (HR = 0.653, 95% CI: 0.512-0.833, p = 0.001), and apolipoprotein A (ApoA) (HR = 0.006, 95% CI: 0.002-0.020, p < 0.001) were significantly associated with a reduced risk of HEV-ALF. Moreover, blood ApoA exhibited excellent discrimination ability and net benefit for predicting 7-day (AUROC = 82.47%, 95% CI: 77.92-87.02) and 14-day (AUROC = 78.81%, 95% CI: 74.13-83.49) HEV-ALF onset. The findings may provide further evidence on the progression of HEV infection and future risk prediction.
Collapse
Affiliation(s)
- Rui Dong
- Department of Fundamental and Community Nursing, School of Nursing, Nanjing Medical University, Nanjing, China
| | - Hong Xue
- Department of Liver Disease, The Third Affiliated Hospital of Nantong University, Nantong, China
| | - Lin Chen
- Nantong Institute of Liver Disease, The Third Affiliated Hospital of Nantong University, Nantong, China
| | - Wenjuan Jin
- Department of Infectious Disease, The Affiliated Suzhou Ninth People's Hospital of Soochow University, Suzhou, China
| | - Zhenghan Luo
- Department of Infectious Disease Prevention and Control, Huadong Research Institute for Medicine and Biotechniques, Nanjing, China
| | - Chao Shen
- Department of Immunization Program, Nanjing Municipal Center for Disease Control and Prevention, Nanjing, China
| | - Lili Huang
- NHC Key Laboratory of Contraceptives Vigilance and Fertility Surveillance/Jiangsu Health Development Research Center, Nanjing, China
| | - Jianguo Shao
- Department of Gastroenterology, The Third Affiliated Hospital of Nantong University, Nantong, China
| | - Jie Wang
- Department of Fundamental and Community Nursing, School of Nursing, Nanjing Medical University, Nanjing, China
| |
Collapse
|
3
|
Ren D, Li Y, Zhang G, Li T, Liu Z. Lipid metabolic profiling and diagnostic model development for hyperlipidemic acute pancreatitis. Front Physiol 2024; 15:1457349. [PMID: 39512473 PMCID: PMC11540618 DOI: 10.3389/fphys.2024.1457349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 09/23/2024] [Indexed: 11/15/2024] Open
Abstract
Introduction Hyperlipidemic acute pancreatitis (HLAP) is a form of pancreatitis induced by hyperlipidemia, posing significant diagnostic challenges due to its complex lipid metabolism disturbances. Methods This study compared the serum lipid profiles of HLAP patients with those of a healthy cohort using ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS). Orthogonal partial least squares discriminant analysis (OPLS-DA) was applied to identify distinct lipid metabolites. Logistic regression and LASSO regression were used to develop a diagnostic model based on the lipid molecules identified. Results A total of 393 distinct lipid metabolites were detected, impacting critical pathways such as fatty acid, sphingolipid, and glycerophospholipid metabolism. Five specific lipid molecules were selected to construct a diagnostic model, which achieved an area under the curve (AUC) of 1 in the receiver operating characteristic (ROC) analysis, indicating outstanding diagnostic accuracy. Discussion These findings highlight the importance of lipid metabolism disturbances in HLAP. The identified lipid molecules could serve as valuable biomarkers for HLAP diagnosis, offering potential for more accurate and early detection.
Collapse
Affiliation(s)
- Dongmei Ren
- Department of Hepatobiliary Surgery II, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Yong Li
- Department of Hepatobiliary Surgery II, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Guangnian Zhang
- Department of Hepatobiliary Surgery II, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Tiantian Li
- Department of Hepatobiliary Surgery II, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Zhenglong Liu
- School of Basic Medical Sciences and Forensic Medicine, North Sichuan Medical College, Nanchong, China
| |
Collapse
|
4
|
Ward C, Schlichtholz B. Post-Acute Sequelae and Mitochondrial Aberration in SARS-CoV-2 Infection. Int J Mol Sci 2024; 25:9050. [PMID: 39201736 PMCID: PMC11354507 DOI: 10.3390/ijms25169050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/29/2024] [Accepted: 08/16/2024] [Indexed: 09/03/2024] Open
Abstract
This review investigates links between post-acute sequelae of SARS-CoV-2 infection (PASC), post-infection viral persistence, mitochondrial involvement and aberrant innate immune response and cellular metabolism during SARS-CoV-2 infection. Advancement of proteomic and metabolomic studies now allows deeper investigation of alterations to cellular metabolism, autophagic processes and mitochondrial dysfunction caused by SARS-CoV-2 infection, while computational biology and machine learning have advanced methodologies of predicting virus-host gene and protein interactions. Particular focus is given to the interaction between viral genes and proteins with mitochondrial function and that of the innate immune system. Finally, the authors hypothesise that viral persistence may be a function of mitochondrial involvement in the sequestration of viral genetic material. While further work is necessary to understand the mechanisms definitively, a number of studies now point to the resolution of questions regarding the pathogenesis of PASC.
Collapse
Affiliation(s)
| | - Beata Schlichtholz
- Department of Biochemistry, Gdańsk University of Medicine, 80-210 Gdańsk, Poland;
| |
Collapse
|
5
|
Feng Y, Luo S, Fang C, Ma S, Fan D, Chen Y, Chen Z, Zheng X, Tang Y, Duan X, Liu X, Ruan X, Guo X. ANGPTL8 deficiency attenuates lipopolysaccharide-induced liver injury by improving lipid metabolic dysregulation. J Lipid Res 2024; 65:100595. [PMID: 39019343 PMCID: PMC11364043 DOI: 10.1016/j.jlr.2024.100595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 06/18/2024] [Accepted: 07/02/2024] [Indexed: 07/19/2024] Open
Abstract
Liver injury is closely related to poor outcomes in sepsis patients. Current studies indicate that sepsis is accompanied by metabolic disorders, especially those related to lipid metabolism. It is highly important to explore the mechanism of abnormal liver lipid metabolism during sepsis. As a key regulator of glucose and lipid metabolism, angiopoietin-like 8 (ANGPTL8) is involved in the regulation of multiple chronic metabolic diseases. In the present study, severe liver lipid deposition and lipid peroxidation were observed in the early stages of lipopolysaccharide (LPS) induced liver injury. LPS promotes the expression of ANGPTL8 both in vivo and in vitro. Knockout of Angptl8 reduced hepatic lipid accumulation and lipid peroxidation, improved fatty acid oxidation and liver function, and increased the survival rate of septic mice by activating the PGC1α/PPARα pathway. We also found that the expression of ANGPTL8 induced by LPS depends on TNF-α, and that inhibiting the TNF-α pathway reduces LPS-induced hepatic lipid deposition and lipid peroxidation. However, knocking out Angptl8 improved the survival rate of septic mice better than inhibiting the TNF-α pathway. Taken together, the results of our study suggest that ANGPTL8 functions as a novel cytokine in LPS-induced liver injury by suppressing the PGC1α/PPARα signaling pathway. Therefore, targeting ANGPTL8 to improve liver lipid metabolism represents an attractive strategy for the management of sepsis patients.
Collapse
Affiliation(s)
- Ying Feng
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, School of Basic Medical Sciences, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Shan Luo
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, School of Basic Medical Sciences, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China; Department of Endocrine rheumatology, Taihe Hospital, Shiyan, Hubei, China
| | - Chen Fang
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, School of Basic Medical Sciences, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Shinan Ma
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, School of Basic Medical Sciences, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China; Biomedical Research Institute, Hubei University of Medicine, Shiyan, China
| | - Dandan Fan
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, School of Basic Medical Sciences, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China; School of Biomedical Engineering, Hubei University of Medicine, Shiyan, Hubei, China
| | - Yanghui Chen
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, School of Basic Medical Sciences, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Zhuo Chen
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, School of Basic Medical Sciences, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China; Department of Neurology, Wuhan NO.1 Hospital, Wuhan, China
| | - Xiang Zheng
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, School of Basic Medical Sciences, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China; Department of Critical Care Medicine, Taihe Hospital, Shiyan, Hubei, China
| | - Yijun Tang
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, School of Basic Medical Sciences, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Xiaobei Duan
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, School of Basic Medical Sciences, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China; School of Biomedical Engineering, Hubei University of Medicine, Shiyan, Hubei, China
| | - Xingling Liu
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, China
| | - Xuzhi Ruan
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, School of Basic Medical Sciences, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China.
| | - Xingrong Guo
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, School of Basic Medical Sciences, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China.
| |
Collapse
|
6
|
Mousa SO, Afifi MF, Hassuna NA, Yassa MF, Moness HM. Assessment of serum proprotein convertase subtilisin/kexin type 9 in pediatric sepsis syndrome. Sci Rep 2024; 14:15634. [PMID: 38972879 PMCID: PMC11228027 DOI: 10.1038/s41598-024-65609-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 06/21/2024] [Indexed: 07/09/2024] Open
Abstract
Sepsis is a life-threatening condition that arises when the body's response to infection causes injury to its tissues and organs. Proprotein convertase subtilisin/kexin type 9 (PCSK9) is an enzyme released in response to the drop in cholesterol level occurring in sepsis. Our study aimed to evaluate the prognostic role of serum Proprotein convertase subtilisin/kexin type 9 (PCSK9) level in children with sepsis and severe sepsis. Sixty children were included in this study. They were divided into two groups: 30 children in the sepsis group and 30 in the severe sepsis group. Another 30 apparently healthy children were included as a control group. Blood samples were withdrawn from all included children for complete blood count (CBC), renal function tests (RFT), liver function tests (LFT), LDL-cholesterol (LDL-C), blood culture, and serum PCSK9. In this study, PCSK9 and LDL-C were higher in the two sepsis groups than in the control group (p < 0.05). They were also higher in the severe sepsis group than the sepsis group and in the non-survivors than in the survivors (p < 0.05). PCSK9 was positively correlated with length of hospital stay in surviving children (r = 0.67, p = 0.001) and had predicted significant hematological dysfunction (adjusted B = - 96.95, p = 0.03). In conclusion, the PCSK9 assay can be used as a biomarker for bad prognosis in children suffering from clinical sepsis.
Collapse
Affiliation(s)
- Suzan Omar Mousa
- Pediatric Department, Minia University Children Hospital, Faculty of Medicine, Minia University, El-Minya, 61111, Egypt.
| | - Mohamed Farouk Afifi
- Pediatric Department, Minia University Children Hospital, Faculty of Medicine, Minia University, El-Minya, 61111, Egypt
| | - Noha Anwar Hassuna
- Medical Microbiology and Immunology Department, Faculty of Medicine, Minia University, El-Minya, Egypt
| | - Michael Fekry Yassa
- Pediatric Department, Minia University Children Hospital, Faculty of Medicine, Minia University, El-Minya, 61111, Egypt
| | - Hend Mohamed Moness
- Clinical Pathology Department, Faculty of Medicine, Minia University Hospitals, Minia University, El-Minya, Egypt
| |
Collapse
|
7
|
Mohammadi K, Sleeman MW, Boyapati A, Bigdelou P, Geba GP, Fazio S. Effect of IL-6R blockade on plasma lipids and clinical outcomes among hospitalized patients with COVID-19 infection. J Lipid Res 2024; 65:100568. [PMID: 38795859 PMCID: PMC11237931 DOI: 10.1016/j.jlr.2024.100568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 05/16/2024] [Accepted: 05/19/2024] [Indexed: 05/28/2024] Open
Abstract
Plasma lipid levels are modulated by systemic infection and inflammation; it is unknown whether these changes reflect inflammatory responses or caused directly by pathogen presence. We explored the hypothesis that anti-inflammatory intervention via interleukin 6 receptor (IL-6R) blockade would influence plasma lipid levels during severe infection and evaluated the association of plasma lipid changes with clinical outcomes. Sarilumab (monoclonal antibody blocking IL-6R) efficacy was previously assessed in patients with coronavirus disease 2019 (COVID-19) (NCT04315298). This analysis determined whether strong inflammatory reduction by sarilumab in patients with COVID-19 pneumonia of increasing severity (severe, critical, multisystem organ dysfunction) affected plasma lipid changes between day 1 and day 7 of study therapy. Baseline lipid levels reflected the presence of acute systemic infection, characterized by very low HDL-C, low LDL-C, and moderately elevated triglycerides (TGs). Disease severity was associated with progressively more abnormal lipid levels. At day 7, median lipid levels increased more in the sarilumab versus placebo group (HDL-C +10.3%, LDL-C +54.7%, TG +32% vs. HDL-C +1.7%, LDL-C +15.4%, TG +8.8%, respectively). No significant association between lipid changes and clinical outcomes was observed. In conclusion, severe-to-critical COVID-19 pneumonia causes profound HDL-C depression that is only modestly responsive to strong anti-IL-6R inflammatory intervention. Conversely, LDL-C depression is strongly responsive to IL-6R blockade, with LDL-C levels likely returning to the predisease set point. These results advance our understanding of the complex relationship between serum lipids and infection/inflammation and suggest that HDL-C depression during acute contagious disease is driven by infection and not IL-6-mediated inflammation.
Collapse
Affiliation(s)
| | | | | | | | | | - Sergio Fazio
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA.
| |
Collapse
|
8
|
Deng H, Liang WY, Chen LQ, Yuen TH, Sahin B, Vasilescu DM, Trinder M, Walley K, Rensen PC, Boyd JH, Brunham LR. CETP inhibition enhances monocyte activation and bacterial clearance and reduces streptococcus pneumonia-associated mortality in mice. JCI Insight 2024; 9:e173205. [PMID: 38646937 PMCID: PMC11141867 DOI: 10.1172/jci.insight.173205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 03/05/2024] [Indexed: 04/25/2024] Open
Abstract
Sepsis is a leading cause of mortality worldwide, and pneumonia is the most common cause of sepsis in humans. Low levels of high-density lipoprotein cholesterol (HDL-C) levels are associated with an increased risk of death from sepsis, and increasing levels of HDL-C by inhibition of cholesteryl ester transfer protein (CETP) decreases mortality from intraabdominal polymicrobial sepsis in APOE*3-Leiden.CETP mice. Here, we show that treatment with the CETP inhibitor (CETPi) anacetrapib reduced mortality from Streptococcus pneumoniae-induced sepsis in APOE*3-Leiden.CETP and APOA1.CETP mice. Mechanistically, CETP inhibition reduced the host proinflammatory response via attenuation of proinflammatory cytokine transcription and release. This effect was dependent on the presence of HDL, leading to attenuation of immune-mediated organ damage. In addition, CETP inhibition promoted monocyte activation in the blood prior to the onset of sepsis, resulting in accelerated macrophage recruitment to the lung and liver. In vitro experiments demonstrated that CETP inhibition significantly promoted the activation of proinflammatory signaling in peripheral blood mononuclear cells and THP1 cells in the absence of HDL; this may represent a mechanism responsible for improved bacterial clearance during sepsis. These findings provide evidence that CETP inhibition represents a potential approach to reduce mortality from pneumosepsis.
Collapse
Affiliation(s)
- Haoyu Deng
- Department of Medicine, Faculty of Medicine
- Centre for Heart and Lung Innovation, St. Paul’s Hospital
| | - Wan Yi Liang
- Department of Microbiology and Immunology, Faculty of Science, and
| | - Le Qi Chen
- Department of Microbiology and Immunology, Faculty of Science, and
| | - Tin Ho Yuen
- Department of Microbiology and Immunology, Faculty of Science, and
| | - Basak Sahin
- Centre for Heart and Lung Innovation, St. Paul’s Hospital
| | | | - Mark Trinder
- Centre for Heart and Lung Innovation, St. Paul’s Hospital
- Department of Experimental Medicine, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Keith Walley
- Centre for Heart and Lung Innovation, St. Paul’s Hospital
| | - Patrick C.N. Rensen
- Department of Medicine, Division of Endocrinology, and
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - John H. Boyd
- Department of Medicine, Faculty of Medicine
- Centre for Heart and Lung Innovation, St. Paul’s Hospital
| | - Liam R. Brunham
- Department of Medicine, Faculty of Medicine
- Centre for Heart and Lung Innovation, St. Paul’s Hospital
| |
Collapse
|
9
|
Szakmany T, Fitzgerald E, Garlant HN, Whitehouse T, Molnar T, Shah S, Tong D, Hall JE, Ball GR, Kempsell KE. The 'analysis of gene expression and biomarkers for point-of-care decision support in Sepsis' study; temporal clinical parameter analysis and validation of early diagnostic biomarker signatures for severe inflammation andsepsis-SIRS discrimination. Front Immunol 2024; 14:1308530. [PMID: 38332914 PMCID: PMC10850284 DOI: 10.3389/fimmu.2023.1308530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 12/26/2023] [Indexed: 02/10/2024] Open
Abstract
Introduction Early diagnosis of sepsis and discrimination from SIRS is crucial for clinicians to provide appropriate care, management and treatment to critically ill patients. We describe identification of mRNA biomarkers from peripheral blood leukocytes, able to identify severe, systemic inflammation (irrespective of origin) and differentiate Sepsis from SIRS, in adult patients within a multi-center clinical study. Methods Participants were recruited in Intensive Care Units (ICUs) from multiple UK hospitals, including fifty-nine patients with abdominal sepsis, eighty-four patients with pulmonary sepsis, forty-two SIRS patients with Out-of-Hospital Cardiac Arrest (OOHCA), sampled at four time points, in addition to thirty healthy control donors. Multiple clinical parameters were measured, including SOFA score, with many differences observed between SIRS and sepsis groups. Differential gene expression analyses were performed using microarray hybridization and data analyzed using a combination of parametric and non-parametric statistical tools. Results Nineteen high-performance, differentially expressed mRNA biomarkers were identified between control and combined SIRS/Sepsis groups (FC>20.0, p<0.05), termed 'indicators of inflammation' (I°I), including CD177, FAM20A and OLAH. Best-performing minimal signatures e.g. FAM20A/OLAH showed good accuracy for determination of severe, systemic inflammation (AUC>0.99). Twenty entities, termed 'SIRS or Sepsis' (S°S) biomarkers, were differentially expressed between sepsis and SIRS (FC>2·0, p-value<0.05). Discussion The best performing signature for discriminating sepsis from SIRS was CMTM5/CETP/PLA2G7/MIA/MPP3 (AUC=0.9758). The I°I and S°S signatures performed variably in other independent gene expression datasets, this may be due to technical variation in the study/assay platform.
Collapse
Affiliation(s)
- Tamas Szakmany
- Department of Anaesthesia, Intensive Care and Pain Medicine, Division of Population Medicine, Cardiff University, Cardiff, United Kingdom
- Anaesthesia, Critical Care and Theatres Directorate, Cwm Taf Morgannwg University Health Board, Royal Glamorgan Hospital, Llantrisant, United Kingdom
| | | | | | - Tony Whitehouse
- NIHR Surgical Reconstruction and Microbiology Research Centre, Queen Elizabeth Hospital, Mindelsohn Way Edgbaston, Birmingham, United Kingdom
| | - Tamas Molnar
- Critical Care Directorate, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, United Kingdom
| | - Sanjoy Shah
- Critical Care Directorate, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, United Kingdom
| | - Dong Ling Tong
- Faculty of Information and Communication Technology, Universiti Tunku Abdul Rahman, Kampar, Perak, Malaysia
| | - Judith E. Hall
- Department of Anaesthesia, Intensive Care and Pain Medicine, Division of Population Medicine, Cardiff University, Cardiff, United Kingdom
| | - Graham R. Ball
- Medical Technology Research Facility, Anglia Ruskin University, Essex, United Kingdom
| | | |
Collapse
|
10
|
Bao X, Liang Y, Chang H, Cai T, Feng B, Gordon K, Zhu Y, Shi H, He Y, Xie L. Targeting proprotein convertase subtilisin/kexin type 9 (PCSK9): from bench to bedside. Signal Transduct Target Ther 2024; 9:13. [PMID: 38185721 PMCID: PMC10772138 DOI: 10.1038/s41392-023-01690-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 09/27/2023] [Accepted: 10/27/2023] [Indexed: 01/09/2024] Open
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) has evolved as a pivotal enzyme in lipid metabolism and a revolutionary therapeutic target for hypercholesterolemia and its related cardiovascular diseases (CVD). This comprehensive review delineates the intricate roles and wide-ranging implications of PCSK9, extending beyond CVD to emphasize its significance in diverse physiological and pathological states, including liver diseases, infectious diseases, autoimmune disorders, and notably, cancer. Our exploration offers insights into the interaction between PCSK9 and low-density lipoprotein receptors (LDLRs), elucidating its substantial impact on cholesterol homeostasis and cardiovascular health. It also details the evolution of PCSK9-targeted therapies, translating foundational bench discoveries into bedside applications for optimized patient care. The advent and clinical approval of innovative PCSK9 inhibitory therapies (PCSK9-iTs), including three monoclonal antibodies (Evolocumab, Alirocumab, and Tafolecimab) and one small interfering RNA (siRNA, Inclisiran), have marked a significant breakthrough in cardiovascular medicine. These therapies have demonstrated unparalleled efficacy in mitigating hypercholesterolemia, reducing cardiovascular risks, and have showcased profound value in clinical applications, offering novel therapeutic avenues and a promising future in personalized medicine for cardiovascular disorders. Furthermore, emerging research, inclusive of our findings, unveils PCSK9's potential role as a pivotal indicator for cancer prognosis and its prospective application as a transformative target for cancer treatment. This review also highlights PCSK9's aberrant expression in various cancer forms, its association with cancer prognosis, and its crucial roles in carcinogenesis and cancer immunity. In conclusion, this synthesized review integrates existing knowledge and novel insights on PCSK9, providing a holistic perspective on its transformative impact in reshaping therapeutic paradigms across various disorders. It emphasizes the clinical value and effect of PCSK9-iT, underscoring its potential in advancing the landscape of biomedical research and its capabilities in heralding new eras in personalized medicine.
Collapse
Affiliation(s)
- Xuhui Bao
- Institute of Therapeutic Cancer Vaccines, Fudan University Pudong Medical Center, Shanghai, China.
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, China.
- Department of Oncology, Fudan University Pudong Medical Center, Shanghai, China.
- Center for Clinical Research, Fudan University Pudong Medical Center, Shanghai, China.
- Clinical Research Center for Cell-based Immunotherapy, Fudan University, Shanghai, China.
- Department of Pathology, Duke University Medical Center, Durham, NC, USA.
| | - Yongjun Liang
- Center for Medical Research and Innovation, Fudan University Pudong Medical Center, Shanghai, China
| | - Hanman Chang
- Institute for Food Safety and Health, Illinois Institute of Technology, Chicago, IL, USA
| | - Tianji Cai
- Department of Sociology, University of Macau, Taipa, Macau, China
| | - Baijie Feng
- Department of Oncology, Fudan University Pudong Medical Center, Shanghai, China
| | - Konstantin Gordon
- Medical Institute, Peoples' Friendship University of Russia, Moscow, Russia
- A. Tsyb Medical Radiological Research Center, Obninsk, Russia
| | - Yuekun Zhu
- Department of Colorectal Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Hailian Shi
- Shanghai Key Laboratory of Compound Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Zhangjiang Hi-tech Park, Shanghai, China
| | - Yundong He
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, China.
| | - Liyi Xie
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
11
|
Mink S, Saely CH, Frick M, Leiherer A, Drexel H, Fraunberger P. Association between Lipid Levels, Anti-SARS-CoV-2 Spike Antibodies and COVID-19 Mortality: A Prospective Cohort Study. J Clin Med 2023; 12:5068. [PMID: 37568470 PMCID: PMC10420155 DOI: 10.3390/jcm12155068] [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: 07/07/2023] [Revised: 07/21/2023] [Accepted: 07/30/2023] [Indexed: 08/13/2023] Open
Abstract
BACKGROUND Recent studies suggest that both lipid levels and anti-severe-acute-respiratory-syndrome-coronavirus-2 (SARS-CoV-2) antibody levels are associated with outcome in coronavirus disease 2019 (COVID-19). While both parameters have separately been implicated in the neutralization and clearance of pathogens during severe infections, it is currently unclear whether the interplay of these parameters affects outcome in COVID-19. We therefore aimed to determine whether there was a relationship between lipoproteins, anti-SARS-CoV-2 antibodies, and COVID-19 mortality. METHODS In this prospective, multicenter cohort study, we recruited 1152 hospitalized patients with COVID-19 from five hospitals. Total cholesterol (TC), LDL-C, HDL-C, triglycerides, and anti-SARS-CoV-2 spike antibodies were measured on hospital admission. The investigated endpoint was in-hospital mortality. RESULTS LDL-C, HDL-C, and TC were significantly lower in non-survivors than in survivors (mg/dL, 95%CI; 56.1, 50.4-61.8 vs. 72.6, 70.2-75.0, p < 0.001; 34.2, 31.7-36.8 vs. 38.1, 37.2-39.1, p = 0.025; 139.3, 130.9-147.7 vs. 157.4, 54.1-160.6, p = 0.002). Mortality risk increased progressively with lower levels of LDL-C, HDL-C, and TC (aOR 1.73, 1.30-2.31, p < 0.001; 1.44, 1.10-1.88, p = 0.008; 1.49, 1.14-1.94, p < 0.001). Mortality rates varied between 2.1% for high levels of both LDL-C and anti-SARS-CoV-2 antibodies and 16.3% for low levels of LDL-C and anti-SARS-CoV-2 antibodies (aOR 9.14, 95%CI 3.17-26.34, p < 0.001). Accordingly, for total cholesterol and anti-SARS-CoV-2 antibodies, mortality rates varied between 2.1% and 15.0% (aOR 8.01, 95%CI 2.77-23.18, p < 0.001). CONCLUSION The combination of serum lipid levels and anti-SARS-CoV-2 antibodies is strongly associated with in-hospital mortality of patients with COVID-19. Patients with low levels of LDL-C and total cholesterol combined with low levels of anti-SARS-CoV-2 antibodies exhibited the highest mortality rates.
Collapse
Affiliation(s)
- Sylvia Mink
- Central Medical Laboratories, 6800 Feldkirch, Austria
- Medical-Scientific Faculty, Private University of the Principality of Liechtenstein, 9495 Triesen, Liechtenstein
| | - Christoph H. Saely
- Medical-Scientific Faculty, Private University of the Principality of Liechtenstein, 9495 Triesen, Liechtenstein
- VIVIT Institute, Academic Teaching Hospital Feldkirch, 6800 Feldkirch, Austria
| | - Matthias Frick
- Department of Internal Medicine, Academic Teaching Hospital Feldkirch, 6800 Feldkirch, Austria
| | - Andreas Leiherer
- Central Medical Laboratories, 6800 Feldkirch, Austria
- Medical-Scientific Faculty, Private University of the Principality of Liechtenstein, 9495 Triesen, Liechtenstein
- VIVIT Institute, Academic Teaching Hospital Feldkirch, 6800 Feldkirch, Austria
| | - Heinz Drexel
- Medical-Scientific Faculty, Private University of the Principality of Liechtenstein, 9495 Triesen, Liechtenstein
- VIVIT Institute, Academic Teaching Hospital Feldkirch, 6800 Feldkirch, Austria
- Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Peter Fraunberger
- Central Medical Laboratories, 6800 Feldkirch, Austria
- Medical-Scientific Faculty, Private University of the Principality of Liechtenstein, 9495 Triesen, Liechtenstein
| |
Collapse
|
12
|
Patients with Bacterial Sepsis Are Heterogeneous with Regard to Their Systemic Lipidomic Profiles. Metabolites 2022; 13:metabo13010052. [PMID: 36676977 PMCID: PMC9864715 DOI: 10.3390/metabo13010052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/22/2022] [Accepted: 12/23/2022] [Indexed: 12/31/2022] Open
Abstract
Sepsis is defined as life-threatening organ dysfunction caused by a dysregulated host response to infection. In the present study, we investigated the systemic/serum lipidomic profile at the time of hospital admission for patients with bacterial sepsis. The study included 60 patients; 35 patients fulfilled the most recent 2016 Sepsis-3 criteria (referred to as Sepsis-3) whereas the remaining 25 patients had sepsis only according to the previous Sepsis-2 definition and could be classified as having Systemic Inflammatory Response Syndrome (SIRS). A total of 966 lipid metabolites were identified. Patients fulfilling the Sepsis-3 criteria differed from the Sepsis-2 patients with regard to only 15 lipid metabolites, and especially sphingolipids metabolism differed between these patient subsets. A total of only 43 metabolites differed between patients with and without bacteremia, including 12 lysophosphatidylcholines and 18 triacylglycerols (15 C18/C20 fatty acid metabolites decreased and three C14 myristate acid metabolites that were increased in bacteremia). Unsupervised hierarchical clustering analyses based on the identified sphingolipids, phosphatidylcholine and triacylglycerols showed that (i) the majority of Sepsis-3 patients differed from SIRS patients especially with regard to lysophosphatidylcholine levels; (ii) the minority of Sepsis-3 patients that clustered together with the majority of SIRS patients showed lower Sequential Organ Failure Assessment (SOFA) scores than the other Sepsis-3 patients; and (iii) the variation between the patients in the identified/altered sphingolipid and triacylglycerol metabolites further increased the heterogeneity of Sepsis-3 patients with regard to their systemic lipidomic profile at the time of diagnosis. To conclude, patients fulfilling the Sepsis-3 criteria differ with regard to their metabolic profile, and this variation depends on disease severity.
Collapse
|
13
|
Liu C, Zou Q, Tang H, Liu J, Zhang S, Fan C, Zhang J, Liu R, Liu Y, Liu R, Zhao Y, Wu Q, Qi Z, Shen Y. Melanin nanoparticles alleviate sepsis-induced myocardial injury by suppressing ferroptosis and inflammation. Bioact Mater 2022; 24:313-321. [PMID: 36632502 PMCID: PMC9813528 DOI: 10.1016/j.bioactmat.2022.12.026] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 12/23/2022] [Accepted: 12/23/2022] [Indexed: 12/28/2022] Open
Abstract
Myocardial injury as one of the severe complications leads to the increasing morbidity and mortality in patients with sepsis. Recent studies reported that reactive oxygen species (ROS)-mediated ferroptosis plays a critical role in the development of heart diseases. Therefore, we hypothesized that anti-ferroptosis agent might be a novel potential therapeutic strategy for sepsis-induced cardiac injury. Herein, we demonstrated that a small biocompatible and MRI-visible melanin nanoparticles (MMPP) improves myocardial function by inhibiting ROS-related ferroptosis signaling pathway. In LPS-induced murine sepsis model, after a single dose intravenously injection of MMPP treatment, MMPP markedly alleviated the myocardial injury including cardiac function and heart structure disorder through suppressing iron-accumulation induced ferroptosis. In vitro, MMPP inhibited cardiomyocyte death by attenuating oxidative stress, inflammation and maintaining mitochondrial homeostasis. Collectively, our findings demonstrated that MMPP protected heart against sepsis-induced myocardial injury via inhibiting ferroptosis and inflammation, which might be a novel therapeutic approach in future.
Collapse
Affiliation(s)
- Chang Liu
- School of Medical Technology, Tianjin Medical University, Tianjin, 300203, China,Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, 571199, China
| | - Quan Zou
- Department of Radiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Huixin Tang
- School of Medical Technology, Tianjin Medical University, Tianjin, 300203, China
| | - Jia Liu
- School of Medical Technology, Tianjin Medical University, Tianjin, 300203, China
| | - Shiqi Zhang
- School of Medical Technology, Tianjin Medical University, Tianjin, 300203, China
| | - Caihong Fan
- School of Medical Technology, Tianjin Medical University, Tianjin, 300203, China
| | - Junwei Zhang
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, 300071, China
| | - Ruiqing Liu
- School of Medical Technology, Tianjin Medical University, Tianjin, 300203, China
| | - Yashan Liu
- School of Medical Technology, Tianjin Medical University, Tianjin, 300203, China
| | - Ruiyan Liu
- School of Medical Technology, Tianjin Medical University, Tianjin, 300203, China
| | - Yan Zhao
- School of Medical Technology, Tianjin Medical University, Tianjin, 300203, China
| | - Qiang Wu
- Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, 571199, China
| | - Zhi Qi
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, 300071, China,Tianjin Key Laboratory of General Surgery in Construction, Tianjin Union Medical Center, Tianjin, 300000, China,Xinjiang Production and Construction Corps Hospital, Xinjiang, 830092, China,Corresponding author. Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, 300071, China.
| | - Yanna Shen
- School of Medical Technology, Tianjin Medical University, Tianjin, 300203, China,Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, 571199, China,Corresponding author. School of Medical Technology, Tianjin Medical University, Tianjin, 300203, China.
| |
Collapse
|
14
|
Horn P, Radtke S, Metzing UB, Steidl R, Sponholz C, Sommerfeld O, Roth J, Claus RA, Birkenfeld AL, Settmacher U, Rauchfuß F, von Loeffelholz C. Associations of Betatrophin/ANGPTL8 with Septic Dyslipidemia in Human Peritonitis: An Explorative Analysis. Biomedicines 2022; 10:biomedicines10123151. [PMID: 36551906 PMCID: PMC9775570 DOI: 10.3390/biomedicines10123151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 11/17/2022] [Accepted: 11/28/2022] [Indexed: 12/12/2022] Open
Abstract
Sepsis is defined by life-threatening organ dysfunction mediated by the host’s response to infection. This can result in septic dyslipidemia, which is involved in the neutralization of pathogen-related lipids. Knowledge of the regulatory mechanisms of septic dyslipidemia is incomplete. The cytokine betatrophin/Angiopoietin-like protein 8 (ANGPTL8) plays a role in the regulation of triacylglyceride metabolism, though its function in septic dyslipidemia remains unknown. Sixty-six patients were enrolled in a cross-sectional study. Circulating concentrations and adipose tissue (AT) mRNA expression of betatrophin/ANGPTL8 were studied in patients suffering from peritoneal sepsis. Insulin-resistant individuals and subjects without metabolic derangement/systemic inflammation were enrolled as controls. All underwent open abdominal surgery. Circulating betatrophin/ANGPTL8 was analyzed by an enzyme-linked immunosorbent assay and AT mRNA expression levels were assessed by real-time PCR. Standard laboratory analyses including lipid electrophoresis were evaluated. Sepsis patients showed pronounced septic dyslipidemia (p < 0.05 for all major lipid classes). Despite comparable betatrophin/ANGPTL8 mRNA expression in AT (p = 0.24), we found significantly increased circulating betatrophin/ANGPTL8 with septic dyslipidemia (p = 0.009). Expression levels of betatrophin/ANGPTL8 in AT correlated with circulating concentrations in both control groups (r = 0.61; p = 0.008 and r = 0.43; p = 0.034), while this association was undetectable in sepsis. After stratification, betatrophin/ANGPTL8 remained associated with hypertriacylglyceridemia (p < 0.05).
Collapse
Affiliation(s)
- Paul Horn
- Department of Internal Medicine IV, Gastroenterology, Hepatology and Infectious Diseases, Jena University Hospital, 07747 Jena, Germany
| | - Sascha Radtke
- Department of Anaesthesiology and Intensive Care, Jena University Hospital, Friedrich Schiller University, Am Klinikum 1, 07747 Jena, Germany
| | - Uta Barbara Metzing
- Department of Trauma, Hand and Reconstructive Surgery, Jena University Hospital, Friedrich Schiller University, Am Klinikum 1, 07747 Jena, Germany
| | - Ricardo Steidl
- Department of Anaesthesiology and Intensive Care, Jena University Hospital, Friedrich Schiller University, Am Klinikum 1, 07747 Jena, Germany
| | - Christoph Sponholz
- Department of Anaesthesiology and Intensive Care, Jena University Hospital, Friedrich Schiller University, Am Klinikum 1, 07747 Jena, Germany
| | - Oliver Sommerfeld
- Department of Anaesthesiology and Intensive Care, Jena University Hospital, Friedrich Schiller University, Am Klinikum 1, 07747 Jena, Germany
| | - Johannes Roth
- Department of Anaesthesiology and Intensive Care, Jena University Hospital, Friedrich Schiller University, Am Klinikum 1, 07747 Jena, Germany
| | - Ralf A. Claus
- Department of Anaesthesiology and Intensive Care, Jena University Hospital, Friedrich Schiller University, Am Klinikum 1, 07747 Jena, Germany
| | - Andreas L. Birkenfeld
- Department of Diabetology Endocrinology and Nephrology, University Hospital Tübingen, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
- Department of Therapy of Diabetes, Institute of Diabetes Research and Metabolic Diseases in the Helmholtz Center Munich, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
- Division of Diabetes and Nutritional Sciences, Rayne Institute, King’s College London, London SE5 9RJ, UK
| | - Utz Settmacher
- Department of General, Visceral and Vascular Surgery, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany
| | - Falk Rauchfuß
- Department of General, Visceral and Vascular Surgery, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany
| | - Christian von Loeffelholz
- Department of Anaesthesiology and Intensive Care, Jena University Hospital, Friedrich Schiller University, Am Klinikum 1, 07747 Jena, Germany
- Correspondence: ; Tel.: +49-3641-9323-277; Fax: +49-3641-9323-102
| |
Collapse
|
15
|
von Eckardstein A, Nordestgaard BG, Remaley AT, Catapano AL. High-density lipoprotein revisited: biological functions and clinical relevance. Eur Heart J 2022; 44:1394-1407. [PMID: 36337032 PMCID: PMC10119031 DOI: 10.1093/eurheartj/ehac605] [Citation(s) in RCA: 137] [Impact Index Per Article: 45.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 09/16/2022] [Accepted: 10/10/2022] [Indexed: 11/09/2022] Open
Abstract
Abstract
Previous interest in high-density lipoproteins (HDLs) focused on their possible protective role in atherosclerotic cardiovascular disease (ASCVD). Evidence from genetic studies and randomized trials, however, questioned that the inverse association of HDL-cholesterol (HDL-C) is causal. This review aims to provide an update on the role of HDL in health and disease, also beyond ASCVD. Through evolution from invertebrates, HDLs are the principal lipoproteins, while apolipoprotein B-containing lipoproteins first developed in vertebrates. HDLs transport cholesterol and other lipids between different cells like a reusable ferry, but serve many other functions including communication with cells and the inactivation of biohazards like bacterial lipopolysaccharides. These functions are exerted by entire HDL particles or distinct proteins or lipids carried by HDL rather than by its cholesterol cargo measured as HDL-C. Neither does HDL-C measurement reflect the efficiency of reverse cholesterol transport. Recent studies indicate that functional measures of HDL, notably cholesterol efflux capacity, numbers of HDL particles, or distinct HDL proteins are better predictors of ASCVD events than HDL-C. Low HDL-C levels are related observationally, but also genetically, to increased risks of infectious diseases, death during sepsis, diabetes mellitus, and chronic kidney disease. Additional, but only observational, data indicate associations of low HDL-C with various autoimmune diseases, and cancers, as well as all-cause mortality. Conversely, extremely high HDL-C levels are associated with an increased risk of age-related macular degeneration (also genetically), infectious disease, and all-cause mortality. HDL encompasses dynamic multimolecular and multifunctional lipoproteins that likely emerged during evolution to serve several physiological roles and prevent or heal pathologies beyond ASCVD. For any clinical exploitation of HDL, the indirect marker HDL-C must be replaced by direct biomarkers reflecting the causal role of HDL in the respective disease.
Collapse
Affiliation(s)
- Arnold von Eckardstein
- Institute of Clinical Chemistry, University Hospital Zurich and University of Zurich , Zurich , Switzerland
| | - Børge G Nordestgaard
- Department of Clinical Biochemistry, Copenhagen University Hospital, Herlev and Gentofte Hospital , Herlev , Denmark
- The Copenhagen General Population Study, Copenhagen University Hospital, Herlev and Gentofte Hospital , Herlev , Denmark
- Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Alan T Remaley
- Lipoprotein Metabolism Section, Translational Vascular Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health , Bethesda, MD , USA
| | - Alberico L Catapano
- Department of Pharmacological and Biomolecular Sciences, University of Milan , Milan , Italy
- IRCCS MultiMedica, Sesto S. Giovanni , Milan , Italy
| |
Collapse
|
16
|
Owen A, Patel JM, Parekh D, Bangash MN. Mechanisms of Post-critical Illness Cardiovascular Disease. Front Cardiovasc Med 2022; 9:854421. [PMID: 35911546 PMCID: PMC9334745 DOI: 10.3389/fcvm.2022.854421] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 06/22/2022] [Indexed: 11/13/2022] Open
Abstract
Prolonged critical care stays commonly follow trauma, severe burn injury, sepsis, ARDS, and complications of major surgery. Although patients leave critical care following homeostatic recovery, significant additional diseases affect these patients during and beyond the convalescent phase. New cardiovascular and renal disease is commonly seen and roughly one third of all deaths in the year following discharge from critical care may come from this cluster of diseases. During prolonged critical care stays, the immunometabolic, inflammatory and neurohumoral response to severe illness in conjunction with resuscitative treatments primes the immune system and parenchymal tissues to develop a long-lived pro-inflammatory and immunosenescent state. This state is perpetuated by persistent Toll-like receptor signaling, free radical mediated isolevuglandin protein adduct formation and presentation by antigen presenting cells, abnormal circulating HDL and LDL isoforms, redox and metabolite mediated epigenetic reprogramming of the innate immune arm (trained immunity), and the development of immunosenescence through T-cell exhaustion/anergy through epigenetic modification of the T-cell genome. Under this state, tissue remodeling in the vascular, cardiac, and renal parenchymal beds occurs through the activation of pro-fibrotic cellular signaling pathways, causing vascular dysfunction and atherosclerosis, adverse cardiac remodeling and dysfunction, and proteinuria and accelerated chronic kidney disease.
Collapse
Affiliation(s)
- Andrew Owen
- Department of Critical Care, Queen Elizabeth Hospital, University Hospitals Birmingham, Birmingham, United Kingdom
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Jaimin M. Patel
- Department of Critical Care, Queen Elizabeth Hospital, University Hospitals Birmingham, Birmingham, United Kingdom
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Dhruv Parekh
- Department of Critical Care, Queen Elizabeth Hospital, University Hospitals Birmingham, Birmingham, United Kingdom
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Mansoor N. Bangash
- Department of Critical Care, Queen Elizabeth Hospital, University Hospitals Birmingham, Birmingham, United Kingdom
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
- *Correspondence: Mansoor N. Bangash
| |
Collapse
|
17
|
Potential Role of Certain Biomarkers Such as Vitamin B12, ROS, Albumin, as Early Predictors for Prognosis of COVID-19 Outcomes. MEDICINES (BASEL, SWITZERLAND) 2022; 9:medicines9060036. [PMID: 35736249 PMCID: PMC9229029 DOI: 10.3390/medicines9060036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/07/2022] [Accepted: 06/09/2022] [Indexed: 01/10/2023]
Abstract
COVID-19 disease is still a major global concern because of its morbidity and its mortality in severe disease. Certain biomarkers including Reactive Oxygen Species (ROS), vitamins, and trace elements are known to play a crucial role in the pathophysiology of the disease. The aim of our study was to evaluate how certain biomarkers, such as ROS, biochemical indicators, trace elements in serum blood of 139 COVID-19 hospitalized patients, and 60 non-COVID cases according to age and sex variations, can serve as the predictors for prognosis of COVID-19 outcome. An attempt of correlating these biomarkers with the severity of the disease as well as with each other is represented. All subjects were hospitalized from April 2021 until June 2021. A statistically significant increase of B12 levels (p = 0.0029) and ROS levels (p < 0.0001) as well as a decrease in albumin and Total Protein (T.P.) levels (p < 0.001) was observed especially in the early stage of the disease before CRP and ferritin elevation. Additionally, a statistically significant increase in ferritin (p = 0.007), B12 (p = 0.035, sALT p = 0.069, Glucose p = 0.012 and urea p = 0.096 and a decrease in Ca p = 0.005, T.P p = 0.052 albumin p = 0.046 between stage B (CRP values 6−30 mg/L) and C (CRP values 30−100 mg/L) was evident. Thus, this study concludes that clinicians could successfully employ biomarkers such as vitamin B12, ROS and albumin as possible prognosis tools for an early diagnosis. In addition, the total biochemical profile can assist in the understanding of the severity of COVID-19 disease, and could potentially lead to a better diet or early pharmaceutical treatment to prevent some of the more acute symptoms.
Collapse
|
18
|
Abstract
This article reviews the discovery of PCSK9, its structure-function characteristics, and its presently known and proposed novel biological functions. The major critical function of PCSK9 deduced from human and mouse studies, as well as cellular and structural analyses, is its role in increasing the levels of circulating low-density lipoprotein (LDL)-cholesterol (LDLc), via its ability to enhance the sorting and escort of the cell surface LDL receptor (LDLR) to lysosomes. This implicates the binding of the catalytic domain of PCSK9 to the EGF-A domain of the LDLR. This also requires the presence of the C-terminal Cys/His-rich domain, its binding to the secreted cytosolic cyclase associated protein 1, and possibly another membrane-bound "protein X". Curiously, in PCSK9-deficient mice, an alternative to the downregulation of the surface levels of the LDLR by PCSK9 is taking place in the liver of female mice in a 17β-estradiol-dependent manner by still an unknown mechanism. Recent studies have extended our understanding of the biological functions of PCSK9, namely its implication in septic shock, vascular inflammation, viral infections (Dengue; SARS-CoV-2) or immune checkpoint modulation in cancer via the regulation of the cell surface levels of the T-cell receptor and MHC-I, which govern the antitumoral activity of CD8+ T cells. Because PCSK9 inhibition may be advantageous in these processes, the availability of injectable safe PCSK9 inhibitors that reduces by 50% to 60% LDLc above the effect of statins is highly valuable. Indeed, injectable PCSK9 monoclonal antibody or small interfering RNA could be added to current immunotherapies in cancer/metastasis.
Collapse
Affiliation(s)
- Nabil G Seidah
- Laboratory of Biochemical Neuroendocrinology, Montreal Clinical Research Institute (IRCM, affiliated to the University of Montreal), Montreal, QC, Canada
| | - Annik Prat
- Laboratory of Biochemical Neuroendocrinology, Montreal Clinical Research Institute (IRCM, affiliated to the University of Montreal), Montreal, QC, Canada
| |
Collapse
|
19
|
Grewal T, Buechler C. Emerging Insights on the Diverse Roles of Proprotein Convertase Subtilisin/Kexin Type 9 (PCSK9) in Chronic Liver Diseases: Cholesterol Metabolism and Beyond. Int J Mol Sci 2022; 23:ijms23031070. [PMID: 35162992 PMCID: PMC8834914 DOI: 10.3390/ijms23031070] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/07/2022] [Accepted: 01/17/2022] [Indexed: 02/05/2023] Open
Abstract
Chronic liver diseases are commonly associated with dysregulated cholesterol metabolism. Proprotein convertase subtilisin/kexin type 9 (PCSK9) is a serine protease of the proprotein convertase family that is mainly synthetized and secreted by the liver, and represents one of the key regulators of circulating low-density lipoprotein (LDL) cholesterol levels. Its ability to bind and induce LDL-receptor degradation, in particular in the liver, increases circulating LDL-cholesterol levels in the blood. Hence, inhibition of PCSK9 has become a very potent tool for the treatment of hypercholesterolemia. Besides PCSK9 limiting entry of LDL-derived cholesterol, affecting multiple cholesterol-related functions in cells, more recent studies have associated PCSK9 with various other cellular processes, including inflammation, fatty acid metabolism, cancerogenesis and visceral adiposity. It is increasingly becoming evident that additional roles for PCSK9 beyond cholesterol homeostasis are crucial for liver physiology in health and disease, often contributing to pathophysiology. This review will summarize studies analyzing circulating and hepatic PCSK9 levels in patients with chronic liver diseases. The factors affecting PCSK9 levels in the circulation and in hepatocytes, clinically relevant studies and the pathophysiological role of PCSK9 in chronic liver injury are discussed.
Collapse
Affiliation(s)
- Thomas Grewal
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia;
| | - Christa Buechler
- Department of Internal Medicine I, Regensburg University Hospital, 93053 Regensburg, Germany
- Correspondence:
| |
Collapse
|
20
|
Park HJ, Jung JH, Han K, Shin J, Lee Y, Chang Y, Park K, Cho YJ, Choi YS, Kim SM, Nam GE. Association between metabolic syndrome and mortality in patients with COVID-19: A nationwide cohort study. Obes Res Clin Pract 2022; 16:484-490. [PMID: 36335025 PMCID: PMC9618429 DOI: 10.1016/j.orcp.2022.10.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 10/19/2022] [Accepted: 10/21/2022] [Indexed: 12/02/2022]
Abstract
OBJECTIVES We investigated the association between metabolic syndrome (MetS) and mortality among coronavirus disease 2019 (COVID-19) patients in Korea. METHODS We analyzed 3876 individuals aged ≥ 20 years who were confirmed with COVID-19 from January 1 to June 4, 2020 based on the Korea National Health Insurance Service (NHIS)-COVID-19 database and had undergone health examination by NHIS between 2015 and 2017. Multivariable Cox proportional hazard regression analyses were performed. RESULTS Of total participants, the prevalence of MetS was 21.0% (n = 815). During 58.6 days of mean follow-up, 3.1 % (n = 120) of the participants died. Compared to individuals without MetS, COVID-19 patients with MetS had a significantly increased mortality risk after adjusting for confounders in total participants (hazard ratio [HR]: 1.68, 95 % confidence interval [CI]: 1.14-2.47) and women (HR: 2.41, 95 % CI: 1.17-4.96). A low high-density lipoprotein cholesterol level in total participants (HR: 1.63, 95 % CI: 1.12-2.37) and hyperglycemia in women (HR: 1.97, 95 % CI: 1.01-3.84) was associated with higher mortality risk. The mortality risk increased as the number of MetS components increased among total participants and women (P for trend = 0.009 and 0.016, respectively). In addition, MetS groups had higher mortality risk in aged ≥ 60 years (HR: 1.60, 95 % CI: 1.07-2.39), and never-smokers (2.08, 1.21-3.59). CONCLUSIONS The presence of MetS and greater number of its components were associated with increased mortality risks particularly in female patients with COVID-19. Managing MetS may contribute to better outcomes of COVID-19.
Collapse
Affiliation(s)
- Hyo Jin Park
- Department of Family Medicine, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Jin-Hyung Jung
- Department of Biostatistics, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Kyungdo Han
- Department of Statistics and Actuarial Science, Soongsil University, Seoul, Republic of Korea
| | - Jean Shin
- Department of Family Medicine, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Yoojeong Lee
- Department of Family Medicine, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Yujin Chang
- Department of Family Medicine, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Kyeyeung Park
- Department of Family Medicine, Hanyang University College of Medicine, Seoul, Republic of Korea
| | - Yoon Jeong Cho
- Department of Family Medicine, Daegu Catholic University School of Medicine, Daegu, Republic of Korea
| | - Youn Seon Choi
- Department of Family Medicine, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Seon Mee Kim
- Department of Family Medicine, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea,Corresponding authors
| | - Ga Eun Nam
- Department of Family Medicine, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea,Corresponding authors
| |
Collapse
|
21
|
Jacome Sanz D, Saralahti AK, Pekkarinen M, Kesseli J, Nykter M, Rämet M, Ojanen MJT, Pesu M. Proprotein convertase subtilisin/kexin type 9 regulates the production of acute-phase reactants from the liver. Liver Int 2021; 41:2511-2522. [PMID: 34174143 DOI: 10.1111/liv.14993] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 06/07/2021] [Accepted: 06/24/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Proprotein convertase subtilisin/kexin type 9 (PCSK9) controls blood cholesterol levels by fostering the LDL receptor (LDLR) degradation in hepatocytes. Additionally, PCSK9 has been suggested to participate in immunoregulation by modulating cytokine production. We studied the immunological role of PCSK9 in Streptococcus pneumoniae bacteraemia in vivo and in a human hepatocyte cell line. METHODS CRISPR/Cas9 mutagenesis was utilized to create pcsk9 knock-out (KO) zebrafish, which were infected with S pneumoniae to assess the role of PCSK9 for the survival of the fish and in the transcriptomic response of the liver. The direct effects of PCSK9 on the expression of acute-phase reaction (APR) genes were studied in HepG2 cells. RESULTS The pcsk9 KO zebrafish lines (pcsk9tpu-13 and pcsk9tpu-2,+15 ) did not show developmental defects or gross phenotypical differences. In the S pneumoniae infected zebrafish, the mortality of pcsk9 KOs was similar to the controls. A liver-specific gene expression analysis revealed that a pneumococcal challenge upregulated pcsk9, and that the pcsk9 deletion reduced the expression of APR genes, including hepcidin antimicrobial peptide (hamp) and complement component 7b (c7b). Accordingly, silencing PCSK9 in vitro in HepG2 cells using small interfering RNAs (siRNAs) decreased HAMP expression. CONCLUSIONS We demonstrate that PCSK9 is not critical for zebrafish survival in a systemic pneumococcal infection. However, PCSK9 deficiency was associated with the lower expression of APR genes in zebrafish and altered the expression of innate immunity genes in a human hepatocyte cell line. Overall, our data suggest an evolutionarily conserved function for PCSK9 in APR in the liver.
Collapse
Affiliation(s)
- Dafne Jacome Sanz
- Laboratory of Immunoregulation, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Anni K Saralahti
- Laboratory of Experimental Immunology, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Meeri Pekkarinen
- Laboratory of Computational Biology, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Juha Kesseli
- Laboratory of Computational Biology, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Matti Nykter
- Laboratory of Computational Biology, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Mika Rämet
- Laboratory of Experimental Immunology, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,Vaccine Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,PEDEGO Research Unit, Medical Research Center, University of Oulu, Oulu, Finland.,Department of Children and Adolescents, Oulu University Hospital, Oulu, Finland
| | - Markus J T Ojanen
- Laboratory of Immunoregulation, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Marko Pesu
- Laboratory of Immunoregulation, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,Fimlab laboratories Ltd, Tampere, Finland
| |
Collapse
|
22
|
Freitas RS, Roque CR, Matos GA, Belayev L, de Azevedo OGR, Alvarez-Leite JI, Guerrant RL, Oriá RB. Immunoinflammatory role of apolipoprotein E4 in malnutrition and enteric infections and the increased risk for chronic diseases under adverse environments. Nutr Rev 2021; 80:1001-1012. [PMID: 34406390 DOI: 10.1093/nutrit/nuab063] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Apolipoprotein E plays a crucial role in cholesterol metabolism. The immunomodulatory functions of the human polymorphic APOE gene have gained particular interest because APOE4, a well-recognized risk factor for late-onset Alzheimer's disease, has also been recently linked to increased risk of COVID-19 infection severity in a large UK biobank study. Although much is known about apoE functions in the nervous system, much less is known about APOE polymorphism effects on malnutrition and enteric infections and the consequences for later development in underprivileged environments. In this review, recent findings are summarized of apoE's effects on intestinal function in health and disease and the role of APOE4 in protecting against infection and malnutrition in children living in unfavorable settings, where poor sanitation and hygiene prevail, is highlighted. The potential impact of APOE4 on later development also is discussed and gaps in knowledge are identified that need to be addressed to protect children's development under adverse environments.
Collapse
Affiliation(s)
- Raul S Freitas
- Laboratory of the Biology of Tissue Healing, Ontogeny and Nutrition, Department of Morphology and Institute of Biomedicine, School of Medicine, Federal University of Ceara, Fortaleza, Ceará, Brazil
| | - Cássia R Roque
- Laboratory of the Biology of Tissue Healing, Ontogeny and Nutrition, Department of Morphology and Institute of Biomedicine, School of Medicine, Federal University of Ceara, Fortaleza, Ceará, Brazil
| | - Gabriella A Matos
- Laboratory of the Biology of Tissue Healing, Ontogeny and Nutrition, Department of Morphology and Institute of Biomedicine, School of Medicine, Federal University of Ceara, Fortaleza, Ceará, Brazil
| | - Ludmila Belayev
- Neuroscience Center of Excellence, School of Medicine, Health Sciences Center, Louisiana State University, Baton Rouge, Louisiana, United States
| | - Orleâncio G R de Azevedo
- Laboratory of the Biology of Tissue Healing, Ontogeny and Nutrition, Department of Morphology and Institute of Biomedicine, School of Medicine, Federal University of Ceara, Fortaleza, Ceará, Brazil
| | | | - Richard L Guerrant
- Center for Global Health, Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia, United States
| | - Reinaldo B Oriá
- Laboratory of the Biology of Tissue Healing, Ontogeny and Nutrition, Department of Morphology and Institute of Biomedicine, School of Medicine, Federal University of Ceara, Fortaleza, Ceará, Brazil
| |
Collapse
|
23
|
Su X, Zhang G, Cheng Y, Wang B. New insights into the emerging effects of inflammatory response on HDL particles structure and function. Mol Biol Rep 2021; 48:5723-5733. [PMID: 34319542 DOI: 10.1007/s11033-021-06553-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 07/08/2021] [Indexed: 12/11/2022]
Abstract
According to the increasing results, it has been well-demonstrated that the chronic inflammatory response, including systemic lupus erythematosus, rheumatoid arthritis, and inflammatory bowel disease are associated with an increased risk of atherosclerotic cardiovascular disease. The mechanism whereby inflammatory response up-regulates the risk of cardio-metabolic disorder disease is multifactorial; furthermore, the alterations in high density lipoprotein (HDL) structure and function which occur under the inflammatory response could play an important modulatory function. On the other hand, the serum concentrations of HDL cholesterol (HDL-C) have been shown to be reduced significantly under inflammatory status with remarked alterations in HDL particles. Nevertheless, the potential mechanism whereby the inflammatory response reduces serum HDL-C levels is not simply defined but reduces apolipoprotein A1 production. The alterations in HDL structure mediated by the inflammatory response has been also confirmed to decrease the ability of HDL particle to play an important role in reverse cholesterol transport and protect the LDL particles from oxidation. Recently, it has been shown that under the inflammatory condition, diverse alterations in HDL structure could be observed which lead to changes in HDL function. In the current review, the emerging effects of inflammatory response on HDL particles structure and function are well-summarized to elucidate the potential mechanism whereby different inflammatory status modulates the pathogenic development of dyslipidemia.
Collapse
Affiliation(s)
- Xin Su
- Department of Cardiology, The Xiamen Cardiovascular Hospital of Xiamen University, No. 2999 Jinshan Road, Xiamen, 361000, Fujian, China
| | - Guoming Zhang
- Department of Cardiology, The Xiamen Cardiovascular Hospital of Xiamen University, No. 2999 Jinshan Road, Xiamen, 361000, Fujian, China
| | - Ye Cheng
- Department of Cardiology, The Xiamen Cardiovascular Hospital of Xiamen University, No. 2999 Jinshan Road, Xiamen, 361000, Fujian, China.
| | - Bin Wang
- Department of Cardiology, The Xiamen Cardiovascular Hospital of Xiamen University, No. 2999 Jinshan Road, Xiamen, 361000, Fujian, China.
| |
Collapse
|
24
|
Tang R, Jia L, Li Y, Zheng J, Qi P. Narciclasine attenuates sepsis-induced myocardial injury by modulating autophagy. Aging (Albany NY) 2021; 13:15151-15163. [PMID: 34035183 PMCID: PMC8221305 DOI: 10.18632/aging.203078] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 04/29/2021] [Indexed: 12/22/2022]
Abstract
Acute myocardial injury (AMI) is often secondary to sepsis, which is a life-threatening disease associated with severe cardiac inflammation. Narciclasine, a plant alkaloid isolated from different members of the Amaryllidaceae family, has been extensively characterized as an antitumor and anti-inflammatory compound. In addition, autophagy is critical for sepsis-induced myocardial injury. However, the role and mechanism of autophagy by which narciclasine confers cardioprotection are still unclear. The present study aimed to investigate the underlying mechanism by which narciclasine affects the pathogenesis of sepsis-induced myocardial injury. Narciclasine effectively attenuated LPS-induced myocardial inflammation in vitro and in vivo. In addition, narciclasine protected cardiac function and suppressed the expression of inflammatory cytokines in LPS-induced heart tissue. Furthermore, narciclasine upregulated LPS-induced autophagic activity, and the autophagy inhibitor 3-MA abrogated narciclasine-mediated protection against LPS-induced AMI. Importantly, narciclasine exerted an inhibitory effect on the JNK signaling pathway, and JNK activity was tightly associated with narciclasine-induced autophagy and the consequent protective effects during AMI. Taken together, our findings indicate that narciclasine protects against LPS-induced AMI by inducing JNK-dependent autophagic flux; hence, narciclasine may be an effective and novel agent for the clinical treatment of sepsis-induced myocardial injury.
Collapse
Affiliation(s)
- Rong Tang
- Department of Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, Heilongjiang Province, China
| | - Liu Jia
- Department of Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, Heilongjiang Province, China
| | - Yunlong Li
- Department of Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, Heilongjiang Province, China
| | - Junbo Zheng
- Department of Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, Heilongjiang Province, China
| | - Pingping Qi
- Departments of Blood Transfusion, The First Affiliated Hospital of Harbin Medical University, Harbin 150086, Heilongjiang Province, China
| |
Collapse
|
25
|
Hu L, Wu C. In silico analysis suggests disruption of interactions between HAMP from hepatocytes and SLC40A1 from macrophages in hepatocellular carcinoma. BMC Med Genomics 2021; 14:128. [PMID: 34001107 PMCID: PMC8130390 DOI: 10.1186/s12920-021-00977-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 05/06/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Identification of factors associated with proliferation in the hepatocellular carcinoma (HCC) microenvironment aids in understanding the mechanisms of disease progression and provides druggable targets. Gene expression profiles of individual cells in HCC and para-carcinoma tissues can be effectively obtained using the single-cell RNA sequencing (scRNA-Seq) technique. Here, we aimed to identify proliferative hepatocytes from HCC and para-carcinoma tissues, detect differentially expressed genes between the two types of proliferative hepatocytes, and investigate their potential roles in aberrant proliferation. RESULTS Two respective gene signatures for proliferative cells and hepatocytes were established and used to identify proliferative hepatocytes from HCC and para-carcinoma tissues based on scRNA-Seq data. Gene expression profiles between the two types of proliferative hepatocytes were compared. Overall, 40 genes were upregulated in proliferative hepatocytes from para-carcinoma tissue, whereas no upregulated genes were detected in those from HCC tissue. Twelve of the genes, including HAMP, were specifically expressed in the liver tissue. Based on previous reports, we found that HAMP modulates cell proliferation through interaction with its receptor SLC40A1. Comprehensive analysis of cells in HCC and para-carcinoma tissues revealed that: (1) HAMP is specifically expressed in hepatocytes and significantly downregulated in malignant hepatocytes; (2) a subset of macrophages expressing SLC40A1 and genes reacting to various infections is present in para-carcinoma but not in HCC tissue. We independently validated the findings with scRNA-Seq and large-scale tissue bulk RNA-Seq/microarray analyses. CONCLUSION HAMP was significantly downregulated in malignant hepatocytes. In addition, a subset of macrophages expressing SLC40A1 and genes reacting to various infections was absent in HCC tissue. These findings support the involvement of HAMP-SLC40A1 signaling in aberrant hepatocyte proliferation in the HCC microenvironment. The collective data from our in silico analysis provide novel insights into the mechanisms underlying HCC progression and require further validation with wet laboratory experiments.
Collapse
Affiliation(s)
- Liang Hu
- Department of Thyroid Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chao Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
26
|
Robert J, Osto E, von Eckardstein A. The Endothelium Is Both a Target and a Barrier of HDL's Protective Functions. Cells 2021; 10:1041. [PMID: 33924941 PMCID: PMC8146309 DOI: 10.3390/cells10051041] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/19/2021] [Accepted: 04/26/2021] [Indexed: 12/11/2022] Open
Abstract
The vascular endothelium serves as a barrier between the intravascular and extravascular compartments. High-density lipoproteins (HDL) have two kinds of interactions with this barrier. First, bloodborne HDL must pass the endothelium to access extravascular tissues, for example the arterial wall or the brain, to mediate cholesterol efflux from macrophages and other cells or exert other functions. To complete reverse cholesterol transport, HDL must even pass the endothelium a second time to re-enter circulation via the lymphatics. Transendothelial HDL transport is a regulated process involving scavenger receptor SR-BI, endothelial lipase, and ATP binding cassette transporters A1 and G1. Second, HDL helps to maintain the integrity of the endothelial barrier by (i) promoting junction closure as well as (ii) repair by stimulating the proliferation and migration of endothelial cells and their progenitor cells, and by preventing (iii) loss of glycocalix, (iv) apoptosis, as well as (v) transmigration of inflammatory cells. Additional vasoprotective functions of HDL include (vi) the induction of nitric oxide (NO) production and (vii) the inhibition of reactive oxygen species (ROS) production. These vasoprotective functions are exerted by the interactions of HDL particles with SR-BI as well as specific agonists carried by HDL, notably sphingosine-1-phophate (S1P), with their specific cellular counterparts, e.g., S1P receptors. Various diseases modify the protein and lipid composition and thereby the endothelial functionality of HDL. Thorough understanding of the structure-function relationships underlying the multiple interactions of HDL with endothelial cells is expected to elucidate new targets and strategies for the treatment or prevention of various diseases.
Collapse
Affiliation(s)
| | | | - Arnold von Eckardstein
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, 8091 Zurich, Switzerland; (J.R.); (E.O.)
| |
Collapse
|
27
|
Masana L, Correig E, Ibarretxe D, Anoro E, Arroyo JA, Jericó C, Guerrero C, Miret ML, Näf S, Pardo A, Perea V, Pérez-Bernalte R, Plana N, Ramírez-Montesinos R, Royuela M, Soler C, Urquizu-Padilla M, Zamora A, Pedro-Botet J. Low HDL and high triglycerides predict COVID-19 severity. Sci Rep 2021; 11:7217. [PMID: 33785815 PMCID: PMC8010012 DOI: 10.1038/s41598-021-86747-5] [Citation(s) in RCA: 120] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 03/15/2021] [Indexed: 12/13/2022] Open
Abstract
Lipids are indispensable in the SARS-CoV-2 infection process. The clinical significance of plasma lipid profile during COVID-19 has not been rigorously evaluated. We aim to ascertain the association of the plasma lipid profile with SARS-CoV-2 infection clinical evolution. Observational cross-sectional study including 1411 hospitalized patients with COVID-19 and an available standard lipid profile prior (n: 1305) or during hospitalization (n: 297). The usefulness of serum total, LDL, non-HDL and HDL cholesterol to predict the COVID-19 prognosis (severe vs mild) was analysed. Patients with severe COVID-19 evolution had lower HDL cholesterol and higher triglyceride levels before the infection. The lipid profile measured during hospitalization also showed that a severe outcome was associated with lower HDL cholesterol levels and higher triglycerides. HDL cholesterol and triglyceride concentrations were correlated with ferritin and D-dimer levels but not with CRP levels. The presence of atherogenic dyslipidaemia during the infection was strongly and independently associated with a worse COVID-19 infection prognosis. The low HDL cholesterol and high triglyceride concentrations measured before or during hospitalization are strong predictors of a severe course of the disease. The lipid profile should be considered as a sensitive marker of inflammation and should be measured in patients with COVID-19.
Collapse
Affiliation(s)
- Lluís Masana
- Facultat de Medicina, Universitat Rovira I Virgili, LIPIDCAS, University Hospital Sant Joan, IISPV, CIBERDEM, C/. Sant Llorenç, 21, 43201, Reus, Spain.
| | - Eudald Correig
- Statistics Department, Institut Investigació Sanitaria Pere Virgili, Universitat Rovira I Virgili, Reus, Spain
| | - Daiana Ibarretxe
- Facultat de Medicina, Universitat Rovira I Virgili, LIPIDCAS, University Hospital Sant Joan, IISPV, CIBERDEM, C/. Sant Llorenç, 21, 43201, Reus, Spain
| | - Eva Anoro
- LIPIDCAS, Pius Hospital Valls, Valls, Spain
| | - Juan Antonio Arroyo
- Lipid Unit, University Hospital Santa Creu I Sant Pau, Barcelona Autonomous University, Barcelona, Spain
| | - Carlos Jericó
- Lipid Unit, Hospital Moises Broggi, Consorci Sanitari Integral, Sant Joan Despí, Spain
| | - Carolina Guerrero
- Internal Medicine Department, Terrasa Hospital, Consorci Sanitari Terrassa, Barcelona, Spain
| | - Marcel la Miret
- LIPIDCAS, Endocrinology Department, Hospital Verge de La Cinta, Tortosa, Spain
| | - Silvia Näf
- LIPIDCAS, Endocrinology Department, University Hospital Joan XXIII, IISPV. CIBERDEM, Universitat Rovira I Virgili, Tarragona, Spain
| | - Anna Pardo
- Internal Medicine Department, Hospital Delfos, Barcelona, Spain
| | | | | | - Núria Plana
- Facultat de Medicina, Universitat Rovira I Virgili, LIPIDCAS, University Hospital Sant Joan, IISPV, CIBERDEM, C/. Sant Llorenç, 21, 43201, Reus, Spain
| | | | - Meritxell Royuela
- Lipid Unit, ALTHAIA, Xarxa Assistencial Universitària de Manresa, Barcelona, Spain
| | | | - Maria Urquizu-Padilla
- Lipid Unit, University Hospital Vall d'Hebron, Barcelona Autonomous University, Barcelona, Spain
| | - Alberto Zamora
- Lipid Unit, Corporació de Salut del Maresme I La Selva, Hospital de Blanes, Blanes, Spain
| | - Juan Pedro-Botet
- Lipid Unit, University Hospital del Mar, Barcelona Autonomous University, Barcelona, Spain
| | | |
Collapse
|
28
|
Madsen CM, Varbo A, Nordestgaard BG. Novel Insights From Human Studies on the Role of High-Density Lipoprotein in Mortality and Noncardiovascular Disease. Arterioscler Thromb Vasc Biol 2020; 41:128-140. [PMID: 33232200 DOI: 10.1161/atvbaha.120.314050] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The vast majority of research about HDL (high-density lipoprotein) has for decades revolved around the possible role of HDL in atherosclerosis and its therapeutic potential within cardiovascular disease prevention; however, failures with therapies aimed at increasing HDL cholesterol has left questions as to what the role and function of HDL in human health and disease is. Recent observational studies have further shown that extreme high HDL cholesterol is associated with high mortality leading to speculations that HDL could in some instances be harmful. In addition, evidence from observational, and to a lesser extent genetic studies has emerged indicating that HDL might be associated with the development of other major noncardiovascular diseases, such as infectious disease, autoimmune disease, cancer, type 2 diabetes, kidney disease, and lung disease. In this review, we discuss (1) the association between extreme high HDL cholesterol and mortality and (2) the emerging human evidence linking HDL to several major diseases outside the realm of cardiovascular disease.
Collapse
Affiliation(s)
- Christian M Madsen
- Department of Clinical Biochemistry (C.M.M., A.V., B.G.N.), Herlev and Gentofte Hospital, Copenhagen University Hospital, Denmark.,The Copenhagen General Population Study (C.M.M., A.V., B.G.N.), Herlev and Gentofte Hospital, Copenhagen University Hospital, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Denmark (C.M.M., A.V., B.G.N.)
| | - Anette Varbo
- Department of Clinical Biochemistry (C.M.M., A.V., B.G.N.), Herlev and Gentofte Hospital, Copenhagen University Hospital, Denmark.,The Copenhagen General Population Study (C.M.M., A.V., B.G.N.), Herlev and Gentofte Hospital, Copenhagen University Hospital, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Denmark (C.M.M., A.V., B.G.N.)
| | - Børge G Nordestgaard
- Department of Clinical Biochemistry (C.M.M., A.V., B.G.N.), Herlev and Gentofte Hospital, Copenhagen University Hospital, Denmark.,The Copenhagen General Population Study (C.M.M., A.V., B.G.N.), Herlev and Gentofte Hospital, Copenhagen University Hospital, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Denmark (C.M.M., A.V., B.G.N.).,The Copenhagen City Heart Study, Frederiksberg Hospital, Copenhagen University Hospital, Denmark (B.G.N.)
| |
Collapse
|
29
|
Trakaki A, Scharnagl H, Trieb M, Holzer M, Hinghofer-Szalkay H, Goswami N, Marsche G. Prolonged bedrest reduces plasma high-density lipoprotein levels linked to markedly suppressed cholesterol efflux capacity. Sci Rep 2020; 10:15001. [PMID: 32929107 PMCID: PMC7490699 DOI: 10.1038/s41598-020-71921-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 08/21/2020] [Indexed: 12/19/2022] Open
Abstract
Recent observations strongly connect high-density lipoproteins (HDL) function and levels with coronary heart disease outcomes and risk for infections and sepsis. To date, our knowledge of factors determining this connection is still very limited. The immobility associated with prolonged bedrest is detrimental to health, affecting several systems, including the cardiovascular, pulmonary, gastrointestinal, musculoskeletal and urinary. Effects of prolonged bedrest on the composition and functional properties of HDL remain elusive. We evaluated metrics of HDL composition and function in healthy male volunteers participating in a randomized, crossover head-down bedrest study. We observed that HDL cholesterol efflux capacity was profoundly decreased during bedrest, mediated by a bedrest associated reduction in plasma levels of HDL-cholesterol and major apolipoproteins (apo) apoA-I and apoA-II. Paraoxonase activity, plasma anti-oxidative capacity and the activities of lecithin-cholesterol acyltransferase and cholesteryl ester transfer protein were not affected. No change was observed in the content of HDL-associated serum amyloid A, a sensitive marker of inflammation. Resistive vibration exercise countermeasure during bedrest did not correct impaired cholesterol efflux capacity and only tended to increase arylesterase activity of HDL-associated paraoxonase. In conclusion, prolonged bedrest reduces plasma HDL levels linked to markedly suppressed HDL cholesterol efflux capacity. Resistive vibration exercise during bedrest did not correct HDL levels and impaired cholesterol efflux capacity.
Collapse
Affiliation(s)
- Athina Trakaki
- Division of Pharmacology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Universitätsplatz 4, 8010, Graz, Austria
| | - Hubert Scharnagl
- Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria
| | - Markus Trieb
- Division of Pharmacology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Universitätsplatz 4, 8010, Graz, Austria.,BioTechMed Graz, Mozartgasse 12/II, 8010, Graz, Austria
| | - Michael Holzer
- Division of Pharmacology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Universitätsplatz 4, 8010, Graz, Austria
| | - Helmut Hinghofer-Szalkay
- Division of Physiology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Neue Stiftingtalstrasse 6/D-5, 8010, Graz, Austria
| | - Nandu Goswami
- Division of Physiology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Neue Stiftingtalstrasse 6/D-5, 8010, Graz, Austria.
| | - Gunther Marsche
- Division of Pharmacology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Universitätsplatz 4, 8010, Graz, Austria. .,BioTechMed Graz, Mozartgasse 12/II, 8010, Graz, Austria.
| |
Collapse
|
30
|
Cannabinoid receptor 2 activation alleviates septic lung injury by promoting autophagy via inhibition of inflammatory mediator release. Cell Signal 2020; 69:109556. [DOI: 10.1016/j.cellsig.2020.109556] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 02/01/2020] [Accepted: 02/03/2020] [Indexed: 12/14/2022]
|
31
|
Parolini C. A Compendium of the Biological Effects of Apolipoprotein A-I Milano. J Pharmacol Exp Ther 2020; 372:54-62. [PMID: 31649050 DOI: 10.1124/jpet.119.261719] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 10/22/2019] [Indexed: 03/08/2025] Open
Abstract
Obesity is a pathologic condition generated by an energy imbalance, that is, excess caloric consumption, leading to weight gain and metabolic disturbances characterized by adipose tissue inflammation and hyperglycemic conditions. In line with these observations, increasing evidence causally links inflammation, or the molecules and networks integral to inflammatory response, to the development of obesity and the complications that emerge from this pathology, such as cardiovascular, neurologic, respiratory, and metabolic illnesses, as well as sepsis and cancer. Not surprisingly, this chronic and abnormal metabolic background leads to constant derangements in innate and adaptive immunity. It is well known that high-density lipoprotein (HDL) possesses anti-inflammatory and antioxidant properties, and various studies have highlighted an emerging role of HDL in modulating immune function. The efficacy of synthetic HDL (sHDL) containing the recombinant form of apoA-IMilano (sHDL-apoA-IM), originating from the observation that carriers of this mutation have low levels of HDL cholesterol without increased atherosclerosis, has been largely proved in diverse animal models of atherosclerosis; however, the therapeutic use of sHDL-apoA-IM still needs clinical validation. One of the main limitations to the use of recombinant proteins in clinical studies lies in the unsustainable purification costs. Unpurified rice-milk-apoA-IM demonstrated anti-inflammatory and antiatherogenic properties in a mouse model, even though administrated by an unconventional way: by oral gavage. Additionally, recent data have uncovered new therapeutic applications for this sHDL-apoA-IM This review provides an overview of all potential application of sHDL-apoA-IM in some inflammatory-based diseases. SIGNIFICANCE STATEMENT: A recent study demonstrated that oral administration of rice-seed protein extracts containing the apoA-IM (i.e., the milk-apoA-IM) reduced atherosclerosis development in a mouse model. Moreover, the rice-milk-apoA-IM preserved both in vitro and in vivo anti-inflammatory properties, as observed when sHDL-apoA-IM was given by intravascular infusion. Besides, various studies suggested that sHDL-apoA-IM could positively affect other inflammatory-based diseases. Together, these data might represent a new starting point for "sHDL-apoA-IM-based therapies" in chronic degenerative disease.
Collapse
Affiliation(s)
- Cinzia Parolini
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milano, Italy
| |
Collapse
|
32
|
Roelands J, Garand M, Hinchcliff E, Ma Y, Shah P, Toufiq M, Alfaki M, Hendrickx W, Boughorbel S, Rinchai D, Jazaeri A, Bedognetti D, Chaussabel D. Long-Chain Acyl-CoA Synthetase 1 Role in Sepsis and Immunity: Perspectives From a Parallel Review of Public Transcriptome Datasets and of the Literature. Front Immunol 2019; 10:2410. [PMID: 31681299 PMCID: PMC6813721 DOI: 10.3389/fimmu.2019.02410] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Accepted: 09/26/2019] [Indexed: 12/21/2022] Open
Abstract
A potential role for the long-chain acyl-CoA synthetase family member 1 (ACSL1) in the immunobiology of sepsis was explored during a hands-on training workshop. Participants first assessed the robustness of the potential gap in biomedical knowledge identified via an initial screen of public transcriptome data and of the literature associated with ACSL1. Increase in ACSL1 transcript abundance during sepsis was confirmed in several independent datasets. Querying the ACSL1 literature also confirmed the absence of reports associating ACSL1 with sepsis. Inferences drawn from both the literature (via indirect associations) and public transcriptome data (via correlation) point to the likely participation of ACSL1 and ACSL4, another family member, in inflammasome activation in neutrophils during sepsis. Furthermore, available clinical data indicate that levels of ACSL1 and ACSL4 induction was significantly higher in fatal cases of sepsis. This denotes potential translational relevance and is consistent with involvement in pathways driving potentially deleterious systemic inflammation. Finally, while ACSL1 expression was induced in blood in vitro by a wide range of pathogen-derived factors as well as TNF, induction of ACSL4 appeared restricted to flagellated bacteria and pathogen-derived TLR5 agonists and IFNG. Taken together, this joint review of public literature and omics data records points to two members of the acyl-CoA synthetase family potentially playing a role in inflammasome activation in neutrophils. Translational relevance of these observations in the context of sepsis and other inflammatory conditions remain to be investigated.
Collapse
Affiliation(s)
- Jessica Roelands
- Sidra Medicine, Doha, Qatar.,Department of Surgery, Leiden University Medical Center, Leiden, Netherlands
| | | | - Emily Hinchcliff
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ying Ma
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Parin Shah
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | | | | | | | | | | | - Amir Jazaeri
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | | | | |
Collapse
|
33
|
Tanaka S, Diallo D, Delbosc S, Genève C, Zappella N, Yong-Sang J, Patche J, Harrois A, Hamada S, Denamur E, Montravers P, Duranteau J, Meilhac O. High-density lipoprotein (HDL) particle size and concentration changes in septic shock patients. Ann Intensive Care 2019; 9:68. [PMID: 31197574 PMCID: PMC6565796 DOI: 10.1186/s13613-019-0541-8] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 05/30/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Sepsis is associated with systemic inflammation that may impact lipoprotein function. In particular, high-density lipoproteins (HDLs) that display pleiotropic protective roles may be dysfunctional in septic conditions. The aim of this study was to evaluate the HDL profile and the inflammatory context in septic shock patients admitted to our intensive care unit (ICU). METHODS In this study, 20 septic shock patients and 20 controls (ICU patients without septic shock) were included. Plasma samples were collected on days 1, 2 and 7. Total cholesterol and lipoprotein concentrations were determined. HDL profiles were obtained using the Lipoprint® System (non-denaturing electrophoresis). Quantification of pro-inflammatory cytokines (interleukin 1b, 6 and 8), cell-free DNA and lipopolysaccharide-binding protein was also performed. RESULTS HDL concentration was statistically lower in septic shock patients than in controls. At days 1 and 2, septic patients had significantly more large-sized HDL than control patients. Patients recovered a normal lipid profile at day 7. CONCLUSIONS Our results emphasize that HDL levels are dramatically decreased in the acute phase of septic shock and that there is a shift toward large HDL particles, which may reflect a major dysfunction of these lipoproteins. Further mechanistic studies are required to explore this shift observed during sepsis.
Collapse
Affiliation(s)
- Sébastien Tanaka
- INSERM, UMR 1188 Diabète athérothombose Réunion Océan Indien (DéTROI), Université de La Réunion, 2 Rue Maxime Rivière, 97491 Sainte Clotilde, La Réunion, France
- AP-HP, Service d’Anesthésie-Réanimation, CHU Bichat-Claude Bernard, 46 Rue Henri Huchard, 75018 Paris, France
| | - Dévy Diallo
- Inserm U1148, Laboratory for Vascular, Translational Science Bichat Hospital, 46 Rue Henri Huchard, 75018 Paris, France
| | - Sandrine Delbosc
- Inserm U1148, Laboratory for Vascular, Translational Science Bichat Hospital, 46 Rue Henri Huchard, 75018 Paris, France
| | - Claire Genève
- AP-HP, Service d’Anesthésie-Réanimation, CHU Bichat-Claude Bernard, 46 Rue Henri Huchard, 75018 Paris, France
| | - Nathalie Zappella
- AP-HP, Service d’Anesthésie-Réanimation, CHU Bichat-Claude Bernard, 46 Rue Henri Huchard, 75018 Paris, France
| | - Jennyfer Yong-Sang
- INSERM, UMR 1188 Diabète athérothombose Réunion Océan Indien (DéTROI), Université de La Réunion, 2 Rue Maxime Rivière, 97491 Sainte Clotilde, La Réunion, France
| | - Jessica Patche
- INSERM, UMR 1188 Diabète athérothombose Réunion Océan Indien (DéTROI), Université de La Réunion, 2 Rue Maxime Rivière, 97491 Sainte Clotilde, La Réunion, France
| | - Anatole Harrois
- AP-HP, Service d’Anesthésie-Réanimation, Hôpitaux Universitaires Paris-Sud, Université Paris-Sud, Hôpital de Bicêtre, 78 Rue du Général Leclerc, 94270 Le Kremlin-Bicêtre, France
- Laboratoire d’étude de la Microcirculation, «Bio-CANVAS: Biomarkers in CardioNeuroVascular DISEASES» UMRS 942, Paris, France
| | - Sophie Hamada
- AP-HP, Service d’Anesthésie-Réanimation, Hôpitaux Universitaires Paris-Sud, Université Paris-Sud, Hôpital de Bicêtre, 78 Rue du Général Leclerc, 94270 Le Kremlin-Bicêtre, France
| | - Erick Denamur
- UMR1137 IAME, Inserm, Laboratoire de Génétique Moléculaire, Université Paris Diderot and AP-HP, Hôpital Bichat, Paris, France
| | - Philippe Montravers
- AP-HP, Service d’Anesthésie-Réanimation, CHU Bichat-Claude Bernard, 46 Rue Henri Huchard, 75018 Paris, France
- Inserm UMR1152, Physiopathologie et Epidémiologie des Maladies Respiratoires, Paris, France
| | - Jacques Duranteau
- AP-HP, Service d’Anesthésie-Réanimation, Hôpitaux Universitaires Paris-Sud, Université Paris-Sud, Hôpital de Bicêtre, 78 Rue du Général Leclerc, 94270 Le Kremlin-Bicêtre, France
- Laboratoire d’étude de la Microcirculation, «Bio-CANVAS: Biomarkers in CardioNeuroVascular DISEASES» UMRS 942, Paris, France
| | - Olivier Meilhac
- INSERM, UMR 1188 Diabète athérothombose Réunion Océan Indien (DéTROI), Université de La Réunion, 2 Rue Maxime Rivière, 97491 Sainte Clotilde, La Réunion, France
- CHU de La Réunion, Saint-Denis, France
- INSERM U1188 Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion au CYROI, 2, Rue Maxime Rivière, 97490 Sainte Clotilde, La Réunion, France
| |
Collapse
|
34
|
High-Density Lipoprotein-Based Therapeutics: Can a Novel Mechanism Succeed Where Previous Approaches Have Failed? Can J Cardiol 2019; 35:705-706. [PMID: 31151703 DOI: 10.1016/j.cjca.2019.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 03/16/2019] [Accepted: 03/17/2019] [Indexed: 11/20/2022] Open
|