1
|
Li C, Wang X, Tian M, Zhang M, Zhang X, Fu Q, Liu L, Zhang L, Wang H. The JNK-associated leucine zipper protein exerts a protective effect on renal parenchymal injury by limiting the inflammatory secretome in tubular cells. Cell Signal 2024; 124:111428. [PMID: 39307375 DOI: 10.1016/j.cellsig.2024.111428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 09/01/2024] [Accepted: 09/18/2024] [Indexed: 09/27/2024]
Abstract
JNK-associated leucine zipper protein (JLP) is a newly identified renal endogenous anti-fibrotic factor that is selectively enriched in renal tubular epithelial cells (TECs). The loss of JLP by TECs is a landmark event that heralds the progression of kidney fibrosis. JLP deficiency ensues a series of pathogenetic cellular processes that are conducive to fibrotic injury. Inflammatory injury is functionally relevant in fibrotic kidneys, and TECs play an essential role in fueling inflammation through aberrant secretions. It is speculated that the loss of JLP in TECs is associated with the relentless inflammation during the development of kidney fibrosis. This study examined the alteration of a panel of inflammatory signatures in TECs under kidney fibrotic circumstances using a Jlp gene-modified unilateral ureteral obstruction (UUO) mouse model or cultured HK-2 cells. It was found that a deficiency of JLP in TECs led to a significant increase in the secretion of inflammatory cytokines including interleukin-1β (IL-1β), tumor necrosis factor (TNF-α), and monocyte chemotactic protein-1 (MCP-1), overactivation of the nuclear factor (NF)-κB/c-Jun N-terminal kinase (JNK) pathway, as well as nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3) inflammasome-mediated pyroptosis in response to pro-fibrotic damage. Additionally, the absence of JLP resulted in enhanced macrophage migration and fibroblast activation as paracrine effects elicited by injured TECs. In conclusion, the loss of JLP in TECs catalyses inflammatory injuries in the development of kidney fibrosis.
Collapse
Affiliation(s)
- Chen Li
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaofei Wang
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Maoqing Tian
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Meng Zhang
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xin Zhang
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qiang Fu
- Paediatric Department, Central Hospital of Jingzhou City, Jingzhou, China
| | - Lunzhi Liu
- Hubei Provincial Clinical Medical Research Center for Nephropathy, Minda Hospital of Hubei Minzu University, Enshi, China.
| | - Lu Zhang
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China.
| | - Huiming Wang
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
2
|
Qin T, Ma TY, Huang K, Lu SJ, Zhong JH, Li JJ. Lipoprotein (a)-Related Inflammatory Imbalance: A Novel Horizon for the Development of Atherosclerosis. Curr Atheroscler Rep 2024; 26:383-394. [PMID: 38878139 PMCID: PMC11236888 DOI: 10.1007/s11883-024-01215-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/15/2024] [Indexed: 07/11/2024]
Abstract
PURPOSE OF REVIEW The primary objective of this review is to explore the pathophysiological roles and clinical implications of lipoprotein(a) [Lp(a)] in the context of atherosclerotic cardiovascular disease (ASCVD). We seek to understand how Lp(a) contributes to inflammation and arteriosclerosis, aiming to provide new insights into the mechanisms of ASCVD progression. RECENT FINDINGS Recent research highlights Lp(a) as an independent risk factor for ASCVD. Studies show that Lp(a) not only promotes the inflammatory processes but also interacts with various cellular components, leading to endothelial dysfunction and smooth muscle cell proliferation. The dual role of Lp(a) in both instigating and, under certain conditions, mitigating inflammation is particularly noteworthy. This review finds that Lp(a) plays a complex role in the development of ASCVD through its involvement in inflammatory pathways. The interplay between Lp(a) levels and inflammatory responses highlights its potential as a target for therapeutic intervention. These insights could pave the way for novel approaches in managing and preventing ASCVD, urging further investigation into Lp(a) as a therapeutic target.
Collapse
Affiliation(s)
- Ting Qin
- Department of Cardiology, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Hainan, 570208, China
| | - Tian-Yi Ma
- Department of Cardiology, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Hainan, 570208, China
| | - Kang Huang
- Department of Cardiology, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Hainan, 570208, China
| | - Shi-Juan Lu
- Department of Cardiology, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Hainan, 570208, China.
| | - Jiang-Hua Zhong
- Department of Cardiology, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Hainan, 570208, China.
| | - Jian-Jun Li
- Cadiometabolic Center, State Key Laboratory of Cardiovascular Diseases, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China.
| |
Collapse
|
3
|
Chehab O, Abdollahi A, Whelton SP, Wu CO, Ambale-Venkatesh B, Post WS, Bluemke DA, Tsai MY, Lima JAC. Association of Lipoprotein(a) Levels With Myocardial Fibrosis in the Multi-Ethnic Study of Atherosclerosis. J Am Coll Cardiol 2023; 82:2280-2291. [PMID: 38057070 PMCID: PMC11730445 DOI: 10.1016/j.jacc.2023.10.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/21/2023] [Accepted: 10/04/2023] [Indexed: 12/08/2023]
Abstract
BACKGROUND Lipoprotein(a) (Lp[a]) has been identified as an emerging risk factor for adverse cardiovascular (CV) outcomes, including heart failure. However, the connections among Lp(a), myocardial fibrosis (interstitial and replacement), and cardiac remodeling as pathways to CV diseases remains unclear. OBJECTIVES This study investigated the relationship between Lp(a) levels and myocardial fibrosis by cardiac magnetic resonance (CMR) T1 mapping and late gadolinium enhancement, as well as cardiac remodeling by cine CMR, in the MESA (Multi-Ethnic Study of Atherosclerosis) cohort. METHODS The study included 2,040 participants with baseline Lp(a) measurements and T1 mapping for interstitial myocardial fibrosis (IMF) evaluation in 2010. Lp(a) was analyzed as a continuous variable (per log unit) and using clinical cutoff values of 30 and 50 mg/dL. Multivariate linear and logistic regression were used to assess the associations of Lp(a) with CMR measures of extracellular volume (ECV fraction [ECV%]), native T1 time, and myocardial scar, as well as parameters of cardiac remodeling, in 2,826 participants. RESULTS Higher Lp(a) levels were associated with increased ECV% (per log-unit Lp[a]; β = 0.2%; P = 0.007) and native T1 time (per log-unit Lp[a]; β = 4%; P < 0.001). Similar relationships were observed between elevated Lp(a) levels and a higher risk of clinically significant IMF defined by prognostic thresholds per log-unit Lp(a) of ECV% (OR: 1.20; 95% CI: 1.04-1.43) and native T1 (OR: 1.2; 95% CI: 1.1-1.4) equal to 30% and 955 ms, respectively. Clinically used Lp(a) cutoffs (30 and 50 mg/dL) were associated with greater prevalence of myocardial scar (OR: 1.85; 95% CI: 1.1-3.2 and OR: 1.9; 95% CI: 1.1-3.4, respectively). Finally, higher Lp(a) levels were associated with left atrial enlargement and dysfunction. CONCLUSIONS Elevated Lp(a) levels are linked to greater subclinical IMF, increased myocardial scar prevalence, and left atrial remodeling.
Collapse
Affiliation(s)
- Omar Chehab
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Ashkan Abdollahi
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Seamus P Whelton
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Colin O Wu
- Office of Biostatistics Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Wendy S Post
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - David A Bluemke
- Department of Radiology, University of Wisconsin School of Medicine and Public Heath, Madison, Wisconsin, USA
| | - Michael Y Tsai
- Department of Pathology, University of Minnesota, Saint Paul-Minneapolis, Minneapolis, Minnesota, USA
| | - João A C Lima
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA; Department of Radiology, Johns Hopkins University, Baltimore, Maryland, USA.
| |
Collapse
|
4
|
Gao G, Kang D, Zhang J, Jiang Z, He X, Wu Y. Risk factors and a prediction model for unruptured intracranial aneurysms in patients with ischemic stroke using carotid intima-media thickness and systemic atherosclerosis. Front Neurol 2023; 14:1227673. [PMID: 37706007 PMCID: PMC10495834 DOI: 10.3389/fneur.2023.1227673] [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: 05/23/2023] [Accepted: 07/28/2023] [Indexed: 09/15/2023] Open
Abstract
Background Systemic atherosclerosis and carotid intima-media thickness (IMT) have been widely used in clinical practice for ischemic stroke; however, little is known about the risk factors for unruptured intracranial aneurysms (UIAs) in patients with ischemic stroke (IS). Therefore, we performed this study to identify the risk factors and construct a prediction model for UIA in patients with IS. Methods Data were retrospectively collected from patients with IS from 2015 to 2022 at the First Hospital of Quanzhou City, Quanzhou, Fujian, China. Risk factors for UIA in patients with IS were identified using a multivariate logistic regression model, and a receiver operating characteristic (ROC) curve was applied to construct the prediction model. Results Out of the 122 patients with IS, 52 who presented with UIA (ISUIA) were categorized into the study group and the remaining 70 IS patients without UIA into the control group. Patients in the ISUIA group had lower carotid IMT and carotid artery plaque scores than those in the IS group (P < 0.05). Multivariate analyses found that aspirin use (OR: 12.987; P = 0.031), elevated C-reactive protein (CRP) level (OR: 1.019; P = 0.004), and carotid IMT > 0.09 mm (OR: 0.218; P < 0.001) were significantly associated with the risk of UIA in patients with IS. However, UIA in patients with IS was unaffected by the carotid artery plaque score (P = 0.114). The constricted prediction model based on the abovementioned factors for UIA in IS patients was 0.79 (95% CI: 0.71-0.87). Conclusion The findings revealed that the risk factors for UIA in patients with IS included aspirin use, elevated CRP level, and smaller carotid IMT, and the predictive value of the prediction model was relatively better.
Collapse
Affiliation(s)
- Gaiming Gao
- Department of Neurosurgery, First Hospital of Quanzhou, Quanzhou, Fujian, China
| | - Dezhi Kang
- Department of Neurosurgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Jinning Zhang
- Department of Neurosurgery, First Hospital of Quanzhou, Quanzhou, Fujian, China
| | - Zhixian Jiang
- Department of Neurosurgery, First Hospital of Quanzhou, Quanzhou, Fujian, China
| | - Xueyang He
- Department of Neurosurgery, First Hospital of Quanzhou, Quanzhou, Fujian, China
| | - Yanyu Wu
- Department of Anesthesiology, First Hospital of Quanzhou, Quanzhou, Fujian, China
| |
Collapse
|
5
|
Lampsas S, Xenou M, Oikonomou E, Pantelidis P, Lysandrou A, Sarantos S, Goliopoulou A, Kalogeras K, Tsigkou V, Kalpis A, Paschou SA, Theofilis P, Vavuranakis M, Tousoulis D, Siasos G. Lipoprotein(a) in Atherosclerotic Diseases: From Pathophysiology to Diagnosis and Treatment. Molecules 2023; 28:969. [PMID: 36770634 PMCID: PMC9918959 DOI: 10.3390/molecules28030969] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 01/12/2023] [Accepted: 01/17/2023] [Indexed: 01/21/2023] Open
Abstract
Lipoprotein(a) (Lp(a)) is a low-density lipoprotein (LDL) cholesterol-like particle bound to apolipoprotein(a). Increased Lp(a) levels are an independent, heritable causal risk factor for atherosclerotic cardiovascular disease (ASCVD) as they are largely determined by variations in the Lp(a) gene (LPA) locus encoding apo(a). Lp(a) is the preferential lipoprotein carrier for oxidized phospholipids (OxPL), and its role adversely affects vascular inflammation, atherosclerotic lesions, endothelial function and thrombogenicity, which pathophysiologically leads to cardiovascular (CV) events. Despite this crucial role of Lp(a), its measurement lacks a globally unified method, and, between different laboratories, results need standardization. Standard antilipidemic therapies, such as statins, fibrates and ezetimibe, have a mediocre effect on Lp(a) levels, although it is not yet clear whether such treatments can affect CV events and prognosis. This narrative review aims to summarize knowledge regarding the mechanisms mediating the effect of Lp(a) on inflammation, atherosclerosis and thrombosis and discuss current diagnostic and therapeutic potentials.
Collapse
Affiliation(s)
- Stamatios Lampsas
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Maria Xenou
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Evangelos Oikonomou
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Panteleimon Pantelidis
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Antonios Lysandrou
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Savvas Sarantos
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Athina Goliopoulou
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Konstantinos Kalogeras
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, 11527 Athens, Greece
- 1st Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Hippokration General Hospital, 11527 Athens, Greece
| | - Vasiliki Tsigkou
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Athanasios Kalpis
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Stavroula A. Paschou
- 1st Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Hippokration General Hospital, 11527 Athens, Greece
| | - Panagiotis Theofilis
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Manolis Vavuranakis
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Dimitris Tousoulis
- 1st Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Hippokration General Hospital, 11527 Athens, Greece
| | - Gerasimos Siasos
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, 11527 Athens, Greece
- Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
6
|
Meroni M, Longo M, Lombardi R, Paolini E, Macchi C, Corsini A, Sirtori CR, Fracanzani AL, Ruscica M, Dongiovanni P. Low Lipoprotein(a) Levels Predict Hepatic Fibrosis in Patients With Nonalcoholic Fatty Liver Disease. Hepatol Commun 2022; 6:535-549. [PMID: 34677008 PMCID: PMC8870034 DOI: 10.1002/hep4.1830] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 08/30/2021] [Accepted: 09/08/2021] [Indexed: 12/21/2022] Open
Abstract
Dyslipidemia and cardiovascular complications are comorbidities of nonalcoholic fatty liver disease (NAFLD), which ranges from simple steatosis to nonalcoholic steatohepatitis, fibrosis, and cirrhosis up to hepatocellular carcinoma. Lipoprotein(a) (Lp(a)) has been associated with cardiovascular risk and metabolic abnormalities, but its impact on the severity of liver damage in patients with NAFLD remains to be clarified. Circulating Lp(a) levels were assessed in 600 patients with biopsy-proven NAFLD. The association of Lp(a) with liver damage was explored by categorizing serum Lp(a) into quartiles. The receiver operating characteristic curve was used to analyze the accuracy of serum Lp(a) in hepatic fibrosis prediction. Hepatic expression of lipoprotein A (LPA) and of genes involved in lipid metabolism and fibrogenic processes were evaluated by RNA sequencing in a subset of patients with NAFLD for whom Lp(a) dosage was available (n = 183). In patients with NAFLD, elevated Lp(a) levels were modestly associated with circulating lipids, carotid plaques, and hypertension (P < 0.05). Conversely, patients with low serum Lp(a) displayed insulin resistance (P < 0.05), transaminase elevation (P < 0.05), and increased risk of developing severe fibrosis (P = 0.007) and cirrhosis (P = 0.002). In addition, the diagnostic accuracy of Lp(a) in predicting fibrosis increased by combining it with transaminases (area under the curve fibrosis stage 4, 0.87; P < 0.0001). Hepatic LPA expression reflected serum Lp(a) levels (P = 0.018), and both were reduced with the progression of NAFLD (P < 0.05). Hepatic LPA messenger RNA levels correlated with those of genes involved in lipoprotein release, lipid synthesis, and fibrogenesis (P < 0.05). Finally, transmembrane 6 superfamily member 2 (TM6SF2) rs58542926, apolipoprotein E (ApoE) rs445925, and proprotein convertase subtilisin/kexin type 9 (PCSK9) rs7552841, known variants that modulate circulating lipids, may influence serum Lp(a) levels (P < 0.05). Conclusion: Circulating Lp(a) combined with transaminases may represent a novel noninvasive biomarker to predict advanced fibrosis in patients with NAFLD.
Collapse
Affiliation(s)
- Marica Meroni
- General Medicine and Metabolic DiseasesFondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Cà Granda Ospedale Maggiore PoliclinicoMilanItaly
| | - Miriam Longo
- General Medicine and Metabolic DiseasesFondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Cà Granda Ospedale Maggiore PoliclinicoMilanItaly.,Department of Clinical Sciences and Community HealthUniversità degli Studi di MilanoMilanItaly
| | - Rosa Lombardi
- General Medicine and Metabolic DiseasesFondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Cà Granda Ospedale Maggiore PoliclinicoMilanItaly.,Department of Pathophysiology and TransplantationUniversità degli Studi di MilanoMilanItaly
| | - Erika Paolini
- General Medicine and Metabolic DiseasesFondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Cà Granda Ospedale Maggiore PoliclinicoMilanItaly.,Department of Pharmacological and Biomolecular SciencesUniversità degli Studi di MilanoMilanItaly
| | - Chiara Macchi
- Department of Pharmacological and Biomolecular SciencesUniversità degli Studi di MilanoMilanItaly
| | - Alberto Corsini
- Department of Pharmacological and Biomolecular SciencesUniversità degli Studi di MilanoMilanItaly.,Multimedica IRCCSSesto San GiovanniMilanItaly
| | - Cesare R Sirtori
- Department of Pharmacological and Biomolecular SciencesUniversità degli Studi di MilanoMilanItaly
| | - Anna Ludovica Fracanzani
- General Medicine and Metabolic DiseasesFondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Cà Granda Ospedale Maggiore PoliclinicoMilanItaly.,Department of Pathophysiology and TransplantationUniversità degli Studi di MilanoMilanItaly
| | - Massimiliano Ruscica
- Department of Pharmacological and Biomolecular SciencesUniversità degli Studi di MilanoMilanItaly
| | - Paola Dongiovanni
- General Medicine and Metabolic DiseasesFondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Cà Granda Ospedale Maggiore PoliclinicoMilanItaly
| |
Collapse
|
7
|
Brosolo G, Da Porto A, Bulfone L, Vacca A, Bertin N, Colussi G, Cavarape A, Sechi LA, Catena C. Plasma Lipoprotein(a) Levels as Determinants of Arterial Stiffening in Hypertension. Biomedicines 2021; 9:biomedicines9111510. [PMID: 34829739 PMCID: PMC8615029 DOI: 10.3390/biomedicines9111510] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 10/12/2021] [Accepted: 10/18/2021] [Indexed: 12/24/2022] Open
Abstract
Previous studies have shown that plasma lipoprotein(a) (Lp(a)) plays an important role in the development of hypertensive organ damage. The aim of the present study was to investigate the relationship of Lp(a) with markers of arterial stiffening in hypertension. In 138 essential hypertensive patients free of diabetes, renal failure and cardiovascular complications, we measured plasma lipids and assessed vascular stiffness through the use of pulse wave analysis and calculation of the brachial augmentation index (AIx), and measured the pulse wave velocity (PWV). Plasma Lp(a) levels were significantly and directly related to both AIx (r = 0.490; p < 0.001) and PWV (r = 0.212; p = 0.013). Multiple regression analysis showed that AIx was independently correlated with age, C-reactive protein, and plasma Lp(a) (beta 0.326; p < 0.001), while PWV was independently and directly correlated with age, and inversely with HDL, but not with plasma Lp(a). Logistic regression indicated that plasma Lp(a) could predict an AIx value above the median for the distribution (p = 0.026). Thus, in a highly selective group of patients with hypertension, plasma Lp(a) levels were significantly and directly related to markers of vascular stiffening. Because of the relevance of vascular stiffening to cardiovascular risk, the reduction of Lp(a) levels might be beneficial for cardiovascular protection in patients with hypertension.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Cristiana Catena
- Correspondence: (L.A.S.); (C.C.); Tel.: +39-0432-559804 (L.A.S.)
| |
Collapse
|
8
|
Liu J, Cao Y, Sheng Z. Effect of lipoprotein α on coronary stents expansion and its risk factors. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2021; 46:249-256. [PMID: 33927071 PMCID: PMC10929932 DOI: 10.11817/j.issn.1672-7347.2021.200398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Indexed: 11/03/2022]
Abstract
OBJECTIVES To analyze the effect of hyperlipoproteinemia (α) on immediate expansion after coronary stent implantation guided by intravascular ultrasound (IVUS). METHODS A total of 160 patients (175 lesions) with coronary heart disease diagnosed by coronary artery angiography, who were performed percutaneous intervention guided by IVUS in the Department of Cardiology, Third Xiangya Hospital, Central South University, were enrolled retrospectively.According to the concentration of lipoproteina, the patients were divided into 2 groups: a hyperlipoproteinemia (α) group and a control group. Cardiac ejection fraction was measured with echocardiography. Logistic regression was used to analyze the influential factors for hyperlipoproteinemia (α). The target vessel was examined by IVUS to analyze the immediate expansion effect of hyperlipoproteinemia (α) after stent implantation. RESULTS The mean stent expansion index, lesion length, stent number, stent symmetry index and posterior balloon diameter were (94.73±18.9)%, (52.92±29.1) mm, (2.11±0.85), (83.62±13.07)%, and (9.46±2.00) mm in the hyperlipoproteinemia (α) group, respectively. Compared with the control group, there were significantly difference (all P<0.05). Multivariable regression analysis showed that the decreased creatinine clearance rate was an independent risk factor for hyperlipoproteinemia (α) (P<0.05). CONCLUSIONS Hyperlipoproteinemia (α) appears to be a predictor of stent underexpansion, and the decreased creatinine clearance rate is an independent risk factor for hyperlipoproteinemia (α).
Collapse
Affiliation(s)
- Jiajia Liu
- Department of Cardiology, Third Xiangya Hospital, Central South University, Changsha 410013, China.
| | - Yu Cao
- Department of Cardiology, Third Xiangya Hospital, Central South University, Changsha 410013, China.
| | - Zhe Sheng
- Department of Cardiology, Third Xiangya Hospital, Central South University, Changsha 410013, China
| |
Collapse
|
9
|
Ahn G, Kim YH, Ahn JY. Multifaceted effects of milk-exosomes (Mi-Exo) as a modulator of scar-free wound healing. NANOSCALE ADVANCES 2021; 3:528-537. [PMID: 36131751 PMCID: PMC9419160 DOI: 10.1039/d0na00665c] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 11/26/2020] [Indexed: 05/09/2023]
Abstract
Scar-free treatment is complex involving many cells in the human body but a very elaborate reaction. This process demands regulation of various growth factors on behalf of TGFβ3 around the damaged tissue, and it is also important to protect cells from inflammatory reactions and oxidative stress to avoid abnormalities. Here, we focused on bovine derived milk exosomes (Mi-Exo) and their scar-free healing potential. The physiological properties (size and shape), biological markers (TSG101 and Bta-miR2478) and stability on storage of Mi-Exo were analyzed. Mi-Exo exhibited significant NP (number of Mi-Exo particles)-dependent scavenging activity in ABTS assay. In addition, Mi-Exo suppressed the expression of pro-inflammatory mediators, IL-6 and TNFα, and pro-inflammatory chemokines, COX-2 and iNOS. This study showed that cell migration was significantly inhibited in a Mi-Exo NP-dependent manner. We also evaluated the expression of TGFβ1 and TGFβ3 on the basis of mRNA and protein levels. Furthermore, the role of functional behavior of Mi-Exo in TGFβ1 maturation was explored. This is the first study to demonstrate that Mi-Exo may target the TGFβ signaling pathway, which plays important roles in scar-free wound healing.
Collapse
Affiliation(s)
- Gna Ahn
- School of Biological Sciences, Chungbuk National University 1 Chungdae-Ro, Seowon-Gu Cheongju 28644 South Korea
| | - Yang-Hoon Kim
- School of Biological Sciences, Chungbuk National University 1 Chungdae-Ro, Seowon-Gu Cheongju 28644 South Korea
| | - Ji-Young Ahn
- School of Biological Sciences, Chungbuk National University 1 Chungdae-Ro, Seowon-Gu Cheongju 28644 South Korea
| |
Collapse
|
10
|
Emanuele E, Carlin MV, D'Angelo A, Peros E, Barale F, Geroldi D, Politi P. Elevated plasma levels of lipoprotein(a) in psychiatric patients: a possible contribution to increased vascular risk. Eur Psychiatry 2020; 21:129-33. [PMID: 16516110 DOI: 10.1016/j.eurpsy.2004.10.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2004] [Accepted: 10/29/2004] [Indexed: 11/19/2022] Open
Abstract
AbstractAn increased incidence of adverse cardiovascular events has been reported in psychiatric patients, but the exact mechanisms underlying this association are still uncertain. Elevated plasma level of lipoprotein(a) [Lp(a)] is an independent risk factor for atherothrombotic disease in the general population. To study the implications of Lp(a) in psychiatric patients, we measured the plasma levels of Lp(a) in 74 patients with psychiatric disorders (39 schizophrenia, 10 major depression, 13 bipolar disorder and 12 personality disorder) and 74 healthy controls. The Lp(a) levels of the patient groups with schizophrenia, major depression and bipolar disorder were significantly higher than that of the control group. The median Lp(a) value of these diagnostic groups was comparable with those reported in patients with prior atherothrombotic events. On the other hand, no differences were found among personality disorder and controls. Our findings suggest that the elevation of plasma Lp(a) may contribute to increased cardiovascular risk in several patients with psychiatric disorders.
Collapse
Affiliation(s)
- Enzo Emanuele
- Molecular Medicine Laboratory, Clinica Medica 2, IRCCS Policlinico San Matteo, University of Pavia, Piazzale Golgi, 2, 27100 Pavia, Italy.
| | | | | | | | | | | | | |
Collapse
|
11
|
Später T, Menger MM, Nickels RM, Menger MD, Laschke MW. Macrophages promote network formation and maturation of transplanted adipose tissue-derived microvascular fragments. J Tissue Eng 2020; 11:2041731420911816. [PMID: 32313616 PMCID: PMC7153185 DOI: 10.1177/2041731420911816] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 02/18/2020] [Indexed: 12/12/2022] Open
Abstract
Adipose tissue–derived microvascular fragments rapidly reassemble into microvascular networks within implanted scaffolds. Herein, we analyzed the contribution of macrophages to this process. C57BL/6 mice received clodronate (clo)-containing liposomes for macrophage depletion, whereas animals treated with phosphate-buffered-saline-containing liposomes served as controls. Microvascular fragments were isolated from clo- and phosphate-buffered-saline-treated donor mice and seeded onto collagen–glycosaminoglycan matrices, which were implanted into dorsal skinfold chambers of clo- and phosphate-buffered-saline-treated recipient mice. The implants’ vascularization and incorporation were analyzed by stereomicroscopy, intravital fluorescence microscopy, histology, and immunohistochemistry. Compared to controls, matrices within clo-treated animals exhibited a significantly reduced functional microvessel density. Moreover, they contained a lower fraction of microvessels with an α-smooth muscle actin (SMA)+ cell layer, indicating impaired vessel maturation. This was associated with a deteriorated implant incorporation. These findings demonstrate that macrophages not only promote the reassembly of microvascular fragments into microvascular networks, but also improve their maturation during this process.
Collapse
Affiliation(s)
- Thomas Später
- Institute for Clinical & Experimental Surgery, Saarland University, Homburg, Germany
| | - Maximilian M Menger
- Institute for Clinical & Experimental Surgery, Saarland University, Homburg, Germany.,Department of Trauma, Hand and Reconstructive Surgery, Saarland University, Homburg, Germany
| | - Ruth M Nickels
- Institute for Clinical & Experimental Surgery, Saarland University, Homburg, Germany
| | - Michael D Menger
- Institute for Clinical & Experimental Surgery, Saarland University, Homburg, Germany
| | - Matthias W Laschke
- Institute for Clinical & Experimental Surgery, Saarland University, Homburg, Germany
| |
Collapse
|
12
|
Sumarjaya IDGD, Nadha IKB, Lestari AAW. High Lipoprotein(a) Levels as a Predictor of Major Adverse Cardiovascular Events in Hospitalized-Acute Myocardial Infarction Patients. Vasc Health Risk Manag 2020; 16:125-132. [PMID: 32308405 PMCID: PMC7152732 DOI: 10.2147/vhrm.s233503] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Accepted: 03/03/2020] [Indexed: 11/23/2022] Open
Abstract
Background Risk stratification models with incorporation of biochemical markers have received attention recently. In acute myocardial infarction (AMI) one such marker is lipoprotein(a) (Lp(a)). Lp(a) has prothrombotic and proinflammatory properties. High levels of Lp(a) probably contribute to the additional adverse effects in AMI, as it enhances the damaging effect of acute thrombosis. This study aimed to evaluate serum Lp(a) as a predictor of major adverse cardiovascular events (MACE) in hospitalized-acute myocardial infarction patients. Methods A prospective cohort study was conducted at Sanglah Hospital, Denpasar, during June–August 2018, among 66 people by consecutive sampling. Samples that met the inclusion and exclusion criteria were examined for serum Lp(a) at the time of admission and the occurrence of MACE during hospitalization was observed. Data regarding serum Lp(a), demography, smoking history, dyslipidemia, hypertension, diabetes mellitus, and MACE were collected. Log rank test and Cox proportional hazards regression were conducted with SPSS version 20 for Windows. Results During observation, MACE occurred in 25 (38%) patients, including cardiogenic shock in 7 (10.6%) patients, heart failure in 20 (30.3%) patients, cardiovascular death in 5 (7, 6%) patients, malignant arrhythmias in 5 (7.6%) patients, and postinfarction angina in 5 (7.6%) patients. After the Log rank test, a significant difference in survival was observed (p = 0.001) between groups of high Lp(a) (survival rate of 60.6 hours; 95% CI 43.3–77.9) and low Lp(a) (average survival of 104.3 hours, 95% CI 91.4–117.2). The hazard ratio of high Lp(a) against MACE was 4.63 (p=0.002), and it increased to 4.69 in multivariate analysis with Cox proportional hazards regression test (p=0.003). Conclusion The high level of Lp(a) in AMI patients was a risk factor for the occurrence of MACE during hospitalization. Patients with high Lp(a) also had worse survival compared to patients with low Lp(a). ![]()
Point your SmartPhone at the code above. If you have a QR code reader the video abstract will appear. Or use: https://youtu.be/LUESRF3Zdcc
Collapse
Affiliation(s)
- I Dewa Gde Dwi Sumarjaya
- Specialist Program of Cardiology and Vascular Medicine, Faculty of Medicine, Universitas Udayana, Sanglah General Hospital, Bali, Indonesia
| | - I Ketut Badjra Nadha
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Universitas Udayana, Sanglah General Hospital, Bali, Indonesia
| | - Anak Agung Wiradewi Lestari
- Department of Clinical Pathology, Faculty of Medicine, Universitas Udayana, Sanglah General Hospital, Bali, Indonesia
| |
Collapse
|
13
|
Jawi MM, Frohlich J, Chan SY. Lipoprotein(a) the Insurgent: A New Insight into the Structure, Function, Metabolism, Pathogenicity, and Medications Affecting Lipoprotein(a) Molecule. J Lipids 2020; 2020:3491764. [PMID: 32099678 PMCID: PMC7016456 DOI: 10.1155/2020/3491764] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 08/17/2019] [Indexed: 12/15/2022] Open
Abstract
Lipoprotein(a) [Lp(a)], aka "Lp little a", was discovered in the 1960s in the lab of the Norwegian physician Kåre Berg. Since then, we have greatly improved our knowledge of lipids and cardiovascular disease (CVD). Lp(a) is an enigmatic class of lipoprotein that is exclusively formed in the liver and comprises two main components, a single copy of apolipoprotein (apo) B-100 (apo-B100) tethered to a single copy of a protein denoted as apolipoprotein(a) apo(a). Plasma levels of Lp(a) increase soon after birth to a steady concentration within a few months of life. In adults, Lp(a) levels range widely from <2 to 2500 mg/L. Evidence that elevated Lp(a) levels >300 mg/L contribute to CVD is significant. The improvement of isoform-independent assays, together with the insight from epidemiologic studies, meta-analyses, genome-wide association studies, and Mendelian randomization studies, has established Lp(a) as the single most common independent genetically inherited causal risk factor for CVD. This breakthrough elevated Lp(a) from a biomarker of atherosclerotic risk to a target of therapy. With the emergence of promising second-generation antisense therapy, we hope that we can answer the question of whether Lp(a) is ready for prime-time clinic use. In this review, we present an update on the metabolism, pathophysiology, and current/future medical interventions for high levels of Lp(a).
Collapse
Affiliation(s)
- Motasim M. Jawi
- Healthy Heart Program, St. Paul's Hospital, Vancouver V6Z 1Y6, Canada
- Division of Experimental Medicine, Department of Medicine, Faculty of Medicine, University of British Columbia, Vancouver V5Z 1M9, Canada
- Department of Clinical PhysiologyCorrection: Department of Physiology, University of Jeddah, P.O. Box: 24, Jeddah 21959, Saudi Arabia
| | - Jiri Frohlich
- Healthy Heart Program, St. Paul's Hospital, Vancouver V6Z 1Y6, Canada
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia V6T 2B5, Canada
| | - Sammy Y. Chan
- Healthy Heart Program, St. Paul's Hospital, Vancouver V6Z 1Y6, Canada
- Department of Medicine, Division of Cardiology, University of British Columbia, Vancouver V5Z 1M9, Canada
| |
Collapse
|
14
|
Soto ME, Huesca-Gómez C, Torres-Paz Y, Fuentevilla-Álvarez G, Gamboa R. Lack of Association between Cytokine Genetic Polymorphisms in Takayasu's Arteritis in Mexican Patients. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:ijerph16234863. [PMID: 31816847 PMCID: PMC6926720 DOI: 10.3390/ijerph16234863] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 11/25/2019] [Accepted: 11/27/2019] [Indexed: 05/21/2023]
Abstract
Aim: To investigate the relation between polymorphisms in the interleukin 10 (IL)-10, tumor necrosis factor (TNF)-α, transforming growth factor (TGF)-β and interferon (IFN)-γ genes and Takayasu's arteritis in the Mexican population. Methods: A case-control study was performed to investigate the associations of IL-10, TNF-α, TGF-β and IFN-γ polymorphisms in a sample of 52 Takayasu's arteritis patients, diagnosed according to the criteria of the American College of Rheumatology and EULAR PRINTO criteria when the patients were under 18 years of age; 60 clinically healthy unrelated Mexican individuals by the 5' exonuclease TaqMan polymerase chain reaction. Polymorphic haplotypes were constructed after linkage disequilibrium analysis. Results: Significant differences were not found in the distribution for genotype and allele frequencies of the polymorphisms studied between healthy controls and Takayasu´s arteritis patients. Likewise, significant associations were not detected in the haplotype analysis with the different genes studied. Conclusions: These findings suggest that the polymorphisms in IL-10, TNF-α, TGF-β and IFN-γ might not contribute to the susceptibility of Takayasu´s arteritis in the Mexican population.
Collapse
Affiliation(s)
- María Elena Soto
- Immunology Department, Instituto Nacional de Cardiología “Ignacio Chávez”. Juan Badiano No. 1, Col. Sección XVI, Tlalpan, México City 14080, Mexico;
| | - Claudia Huesca-Gómez
- Physiology Department, Instituto Nacional de Cardiología “Ignacio Chávez”, Juan Badiano No. 1, Col. Sección XVI, Tlalpan, Mexico City 14080, Mexico; (C.H.-G.); (Y.T.-P.); (G.F.-Á.)
| | - Yazmín Torres-Paz
- Physiology Department, Instituto Nacional de Cardiología “Ignacio Chávez”, Juan Badiano No. 1, Col. Sección XVI, Tlalpan, Mexico City 14080, Mexico; (C.H.-G.); (Y.T.-P.); (G.F.-Á.)
| | - Giovanny Fuentevilla-Álvarez
- Physiology Department, Instituto Nacional de Cardiología “Ignacio Chávez”, Juan Badiano No. 1, Col. Sección XVI, Tlalpan, Mexico City 14080, Mexico; (C.H.-G.); (Y.T.-P.); (G.F.-Á.)
| | - Ricardo Gamboa
- Physiology Department, Instituto Nacional de Cardiología “Ignacio Chávez”, Juan Badiano No. 1, Col. Sección XVI, Tlalpan, Mexico City 14080, Mexico; (C.H.-G.); (Y.T.-P.); (G.F.-Á.)
- Correspondence: ; Tel.: +52-(55)-55-73-29-11 (ext. 25402)
| |
Collapse
|
15
|
Labudovic D, Kostovska I, Tosheska Trajkovska K, Cekovska S, Brezovska Kavrakova J, Topuzovska S. Lipoprotein(a) – Link between Atherogenesis and Thrombosis. Prague Med Rep 2019; 120:39-51. [DOI: 10.14712/23362936.2019.9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Lipoprotein(a) – Lp(a) – is an independent risk factor for cardiovascular disease (CVD). Indeed, individuals with plasma concentrations of Lp(a) > 200 mg/l carry an increased risk of developing CVD. Circulating levels of Lp(a) are remarkably resistant to common lipid lowering therapies, currently available treatment for reduction of Lp(a) is plasma apheresis, which is costly and labour intensive. The Lp(a) molecule is composed of two parts: LDL/apoB-100 core and glycoprotein, apolipoprotein(a) – Apo(a), both of them can interact with components of the coagulation cascade, inflammatory pathways and blood vessel cells (smooth muscle cells and endothelial cells). Therefore, it is very important to determine the molecular pathways by which Lp(a) affect the vascular system in order to design therapeutics for targeting the Lp(a) cellular effects. This paper summarises the cellular effects and molecular mechanisms by which Lp(a) participate in atherogenesis, thrombogenesis, inflammation and development of cardiovascular diseases.
Collapse
|
16
|
Tada H, Takamura M, Kawashiri MA. Lipoprotein(a) as an Old and New Causal Risk Factor of Atherosclerotic Cardiovascular Disease. J Atheroscler Thromb 2019; 26:583-591. [PMID: 31061262 PMCID: PMC6629747 DOI: 10.5551/jat.rv17034] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Accepted: 04/02/2019] [Indexed: 12/16/2022] Open
Abstract
Lipoprotein(a) [Lp(a)], discovered in 1963, has been associated with atherosclerotic cardiovascular disease (ASCVD) independent of other traditional risk factors, including LDL cholesterol. Lp(a) is an apolipoprotein B (apoB)-containing lipoprotein, which contains an LDL-like particle. Unlike LDL, which is a primary therapeutic target to decrease ASCVD, current guidelines recommend measuring Lp(a) for risk assessments because there is no clear evidence demonstrating the clinical benefit of decreasing Lp(a) using classical drugs such as niacin. However, recent Mendelian randomization studies indicate that Lp(a) causally correlates with ASCVD. In addition, novel drugs, including PCSK9 inhibitors, as well as antisense oligonucleotide for apo(a), have exhibited efficacy in decreasing Lp(a) substantially, invigorating a discussion whether Lp(a) could be a novel therapeutic target for further ASCVD risk reduction. This review aims to provide current understanding, and future perspectives, of Lp(a), which is currently considered a mere biomarker but may emerge as a novel therapeutic target in future clinical settings.
Collapse
Affiliation(s)
- Hayato Tada
- Department of Cardiology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Masayuki Takamura
- Department of Cardiology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Masa-aki Kawashiri
- Department of Cardiology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| |
Collapse
|
17
|
Cao YX, Liu HH, Li S, Li JJ. A Meta-Analysis of the Effect of PCSK9-Monoclonal Antibodies on Circulating Lipoprotein (a) Levels. Am J Cardiovasc Drugs 2019; 19:87-97. [PMID: 30229525 DOI: 10.1007/s40256-018-0303-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Lipoprotein (a) [Lp(a)] is an atherogenic lipoprotein. While no effective therapy for Lp(a) is currently available, recently, several pooled analyses with small sample sizes have suggested that proprotein convertase subtilisin/kexin type 9 monoclonal antibodies (PCSK9-mAbs) could reduce circulating Lp(a) levels. This meta-analysis was performed to comprehensively investigate the efficacy of PCSK9-mAbs with respect to serum Lp(a) concentrations. METHODS PubMed, MEDLINE, Embase, ClinicalTrials.gov, Cochrane CENTRAL, Web of Science and recent conferences up to July 2018 were searched. Randomized clinical trials evaluating the effect of PCSK9-mAbs and control treatment on plasma Lp(a) concentrations were included. Mean differences and odds ratios with 95% confidence intervals (CIs) were used. RESULTS Twenty-seven randomized clinical trials with a total of 11,864 participants were included. PCSK9-mAbs showed a significant efficacy in reducing Lp(a) (- 21.9%, 95% CI - 24.3 to - 19.5), irrespective of PCSK9-mAb types, treatment duration, participant characteristics, treatment methods, differences of control treatment, baseline Lp(a) levels, and test methods. The greatest reduction was achieved with 150 mg alirocumab biweekly (- 24.6%, 95% CI - 28.0 to - 21.2) and 140 mg evolocumab monthly (- 26.8%, 95% CI - 31.6 to - 21.9). Meta-regression analyses found that the more intense low-density lipoprotein cholesterol levels declined during PCSK9-mAb treatment, the greater the reduction in Lp(a) levels. Safety was in accordance with previous reports. CONCLUSIONS The results of this analysis suggested that PCSK9-mAbs could significantly reduce circulating Lp(a) levels. Long-term studies may be needed to confirm the effect of PCSK9-mAbs on Lp(a) in the future.
Collapse
Affiliation(s)
- Ye-Xuan Cao
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, BeiLiShi Road 167, Beijing, 100037, China
| | - Hui-Hui Liu
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, BeiLiShi Road 167, Beijing, 100037, China
| | - Sha Li
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, BeiLiShi Road 167, Beijing, 100037, China
| | - Jian-Jun Li
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, BeiLiShi Road 167, Beijing, 100037, China.
| |
Collapse
|
18
|
Ojiaku CA, Yoo EJ, Panettieri RA. Transforming Growth Factor β1 Function in Airway Remodeling and Hyperresponsiveness. The Missing Link? Am J Respir Cell Mol Biol 2017; 56:432-442. [PMID: 27854509 DOI: 10.1165/rcmb.2016-0307tr] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The pathogenesis of asthma includes a complex interplay among airway inflammation, hyperresponsiveness, and remodeling. Current evidence suggests that airway structural cells, including bronchial smooth muscle cells, myofibroblasts, fibroblasts, and epithelial cells, mediate all three aspects of asthma pathogenesis. Although studies show a connection between airway remodeling and changes in bronchomotor tone, the relationship between the two remains unclear. Transforming growth factor β1 (TGF-β1), a growth factor elevated in the airway of patients with asthma, plays a role in airway remodeling and in the shortening of various airway structural cells. However, the role of TGF-β1 in mediating airway hyperresponsiveness remains unclear. In this review, we summarize the literature addressing the role of TGF-β1 in airway remodeling and shortening. Through our review, we aim to further elucidate the role of TGF-β1 in asthma pathogenesis and the link between airway remodeling and airway hyperresponsiveness in asthma and to define TGF-β1 as a potential therapeutic target for reducing asthma morbidity and mortality.
Collapse
Affiliation(s)
- Christie A Ojiaku
- 1 Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; and.,2 Rutgers Institute for Translational Medicine and Science, Child Health Institute, Rutgers University, New Brunswick, New Jersey
| | - Edwin J Yoo
- 1 Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; and.,2 Rutgers Institute for Translational Medicine and Science, Child Health Institute, Rutgers University, New Brunswick, New Jersey
| | - Reynold A Panettieri
- 2 Rutgers Institute for Translational Medicine and Science, Child Health Institute, Rutgers University, New Brunswick, New Jersey
| |
Collapse
|
19
|
Pirro M, Bianconi V, Paciullo F, Mannarino MR, Bagaglia F, Sahebkar A. Lipoprotein(a) and inflammation: A dangerous duet leading to endothelial loss of integrity. Pharmacol Res 2017; 119:178-187. [DOI: 10.1016/j.phrs.2017.02.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 01/08/2017] [Accepted: 02/02/2017] [Indexed: 12/15/2022]
|
20
|
Almontashiri NAM. The 9p21.3 risk locus for coronary artery disease: A 10-year search for its mechanism. J Taibah Univ Med Sci 2017; 12:199-204. [PMID: 31435240 PMCID: PMC6694924 DOI: 10.1016/j.jtumed.2017.03.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2017] [Revised: 03/03/2017] [Accepted: 03/05/2017] [Indexed: 01/07/2023] Open
Abstract
The 9p21.3 risk locus is the first locus to be associated with an increased risk of coronary artery disease (CAD)-related events and many other phenotypes. This locus contains 59 single nucleotide polymorphisms (SNPs) in a region with multiple long range enhancers and long non-coding RNAs (lncRNAs) that affect the expression of neighbouring genes, cyclin-dependent kinase 2A and 2B (CDKN2A and CDKN2B), which are required for controlling vascular smooth muscle cell proliferation and ageing. Several studies have attempted to identify the precise mechanism by which this locus exerts its pathogenic effect to increase the risk of CAD-related events. In this review, we will highlight the major advances in our understanding of the genotype–phenotype correlation at the mechanistic and phenotypic levels. The high population attributable risk of the 9p21.3 risk locus, mechanistic knowledge acquired thus far, and ongoing research efforts could facilitate the design of novel therapeutic molecules to reduce the risk of CAD and its related events.
Collapse
Affiliation(s)
- Naif A M Almontashiri
- Department of Pathology, Massachusetts General Hospital, Harvard University, Boston, USA
| |
Collapse
|
21
|
Nakaya Y, Fukuda D, Oyamada T, Ogawa K, Harada N, Nakagami H, Morishita R, Sata M, Sakaue H. A novel lipoprotein (a) lowering drug, D-47, decreases neointima thickening after vascular injury. THE JOURNAL OF MEDICAL INVESTIGATION 2017; 64:64-67. [PMID: 28373630 DOI: 10.2152/jmi.64.64] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Although Lp(a) have been thought to be a cardiovascular risk factor, it is unclear whether lowering Lp(a) levels reduces the risk of cardiovascular diseases. No pharmacological agents which selectively reduce serum Lp(a) levels, and Lp(a) is present in primate but absent in common laboratory animals such as mice and pigs. In the present study we used transgenic mice of human Lp(a) and tested effect a novel Lp(a) lowering drug D-47 on neointima formation after vascular injury. D-47 successfully decreased plasma levels of Lp(a) and possibly inhibited neointima formation in Lp(a) transgenic mice. The results indicate that we can modulate plasma Lp(a) levels by pharmacologic agents and inhibit atherogenic properties of Lp(a) by reducing plasma levels of Lp(a). J. Med. Invest. 64: 64-67, February, 2017.
Collapse
Affiliation(s)
- Yutaka Nakaya
- Department of Nutrition and Metabolism, Institute of Biomedical sciences, Tokushima University Graduate School
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Xie H, Chen L, Liu H, Cui Y, Zhang Z, Cui L. Long-Term Prognostic Value of Lipoprotein(a) in Symptomatic Patients With Nonobstructive Coronary Artery Disease. Am J Cardiol 2017; 119:945-950. [PMID: 28139221 DOI: 10.1016/j.amjcard.2016.11.053] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2016] [Revised: 11/29/2016] [Accepted: 11/29/2016] [Indexed: 11/17/2022]
Abstract
Our study aimed to evaluate the association of lipoprotein(a) [Lp(a)] and prognosis in patients with nonobstructive coronary artery disease (CAD). A total of 4,254 symptomatic patients with suspected CAD referred for coronary angiography were analyzed and 451 patients (250 women, average age 58 ± 9 years) with nonobstructive CAD (defined as no angiographic stenosis ≥50% in any major epicardial artery) were finally included in our cohort. Subjects were categorized into tertile groups according to Lp(a) levels on admission. The primary end point was major adverse cardiovascular events (MACEs), defined as cardiac death and incident acute coronary syndrome. Over a mean follow-up of 32 ± 22 months, 37 (8%) MACE (15 cases of cardiac death and 22 cases of acute coronary syndrome) occurred. Kaplan-Meier analysis revealed that elevated Lp(a) level was associated with worse prognosis (p = 0.001). After Cox multivariate adjustment for other clinical confounders, an elevated Lp(a) level remained an independent predictor of MACE either as a continuous variable (hazard ratio 1.031, 95% confidence interval 1.019 to 1.043, p <0.001) or as a categorical variable (hazard ratio 3.155, 95% confidence interval 1.599 to 6.229, p = 0.001). Furthermore, addition of Lp(a) to established coronary risk factors significantly improved the predictive value of reference models for MACE. In conclusion, an elevated Lp(a) level is independently associated with worse prognosis and may provide useful risk stratification in symptomatic patients with nonobstructive CAD.
Collapse
Affiliation(s)
- Hao Xie
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Liming Chen
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China.
| | - Hang Liu
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Yuqi Cui
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Zhun Zhang
- Department of Breast and Thyroid Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Lianqun Cui
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China.
| |
Collapse
|
23
|
Wirtwein M, Melander O, Sjőgren M, Hoffmann M, Narkiewicz K, Gruchala M, Sobiczewski W. Elevated ambulatory systolic-diastolic pressure regression index is genetically determined in hypertensive patients with coronary heart disease. Blood Press 2017; 26:174-180. [PMID: 28092973 DOI: 10.1080/08037051.2016.1273741] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
OBJECTIVES Ambulatory systolic-diastolic pressure regression index (ASDPRI) as a composite marker of cardiovascular (CV) properties is related to CV complications. However, genetic determinants of ASDPRI are not known. The aim of this study is to report the relationship between certain single nucleotide polymorphisms (SNP) and ASDPRI in hypertensive patients with CAD confirmed by coronary angiography. METHODS A total of 1345 hypertensive subjects with CAD were included. SNPs were selected from genome-wide association studies. SNPs were reported to be associated with coronary artery disease risk. There were significant differences in 24 h and daytime and nighttime ASDPRIs for PHCTR1, LPA and ADAMTS7 polymorphisms. Genetic risk score (GRS18) was constructed to evaluate additive effect of 18 SNPs for ASDPRI. RESULTS Analysis of covariance revealed a significant relationship between the PPAB2B (β - 0.85; 95 CI -1.85--0.16, p < 0.02), WDR12 (β - 1.31; 95 CI -2.19--0.43, p < 0.01) polymorphisms and nighttime ASDPRI dipping. Analysis of covariance revealed a significant relationship between GRS 18 and 24-h ASDPRI (β 0.34; 95 CI 0.16-0.31, p < 0.01). CONCLUSIONS In conclusion, ADAMTS7 and LPA polymorphisms are related to 24-h ASDPRI but PPAB2B and WDR12 gene polymorphisms are associated with nighttime ASDPRI dipping. A total of 24-h ASDPRI is determined by GRS18.
Collapse
Affiliation(s)
- Marcin Wirtwein
- a Department of Pharmacology , Medical University of Gdansk , Gdansk , Poland
| | - Olle Melander
- b Department of Clinical Sciences , Lund University , Malmö , Sweden
| | - Marketa Sjőgren
- b Department of Clinical Sciences , Lund University , Malmö , Sweden
| | - Michal Hoffmann
- c Department of Hypertension and Diabetology , Medical University of Gdansk , Gdansk , Poland
| | - Krzysztof Narkiewicz
- c Department of Hypertension and Diabetology , Medical University of Gdansk , Gdansk , Poland
| | - Marcin Gruchala
- d 1st Department of Cardiology , Medical University of Gdansk , Gdansk , Poland
| | | |
Collapse
|
24
|
Kumar P, Misra S, Kumar A, Faruq M, Shakya S, Vardhan G, Vivekanandhan S, Srivastava AK, Prasad K. Transforming growth factor-β1 (C509T, G800A, and T869C) gene polymorphisms and risk of ischemic stroke in North Indian population: A hospital-based case-control study. Ann Indian Acad Neurol 2017; 20:5-12. [PMID: 28298836 PMCID: PMC5341267 DOI: 10.4103/0972-2327.199910] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Background: Transforming growth factor-beta 1 (TGF-β1) is a multifunctional pleiotropic cytokine involved in inflammation and pathogenesis of cerebrovascular diseases. There is limited information on the association between variations within the TGF-β1 gene polymorphisms and risk of ischemic stroke (IS). The aim of this study was to investigate the association of the TGF-β1 gene (C509T, G800A, and T869C) polymorphisms, and their haplotypes with the risk of IS in North Indian population. Methods: A total of 250 IS patients and 250 age- and sex-matched controls were studied. IS was classified using the Trial of Org 10172 in Acute Stroke Treatment classification. Conditional logistic regression analysis was used to calculate the strength of association between TGF-β1 gene polymorphisms and risk of IS. Genotyping was performed using SNaPshot method. Results: Hypertension, diabetes, dyslipidemia, alcohol, smoking, family history of stroke, sedentary lifestyle, and low socioeconomic status were found to be associated with the risk of IS. The distribution of C509T, G800A and T869C genotypes was consistent with Hardy-Weinberg Equilibrium in the IS and control groups. Adjusted conditional logistic regression analysis showed a significant association of TGF-β1 C509T (odds ratio [OR], 2.1; 95% CI; 1.2–3.8; P = 0.006), G800A (OR, 4.4; 95% CI; 2.1–9.3; P < 0.001) and T869C (OR, 2.6; 95% CI; 1.5–4.5; P = 0.001) with the risk of IS under dominant model. Haplotype analysis showed that C509-A800-T869 and T509-G800-C869 haplotypes were significantly associated with the increased risk of IS. C509T and T869C were in strong linkage disequilibrium (D' =0.51, r2 = 0.23). Conclusion: Our results suggest that TGF-β1 polymorphisms and their haplotypes are significantly associated with the risk of IS in North Indian population.
Collapse
Affiliation(s)
- Pradeep Kumar
- Department of Neurology, All India Institute of Medical Sciences, New Delhi, India
| | - Shubham Misra
- Department of Neurology, All India Institute of Medical Sciences, New Delhi, India
| | - Amit Kumar
- Department of Neurology, All India Institute of Medical Sciences, New Delhi, India
| | - Mohammad Faruq
- Department of Functional Genomics, Institutes of Genomics and Integrative Biology, New Delhi, India
| | - Sunil Shakya
- Department of Neurology, All India Institute of Medical Sciences, New Delhi, India
| | - Gyan Vardhan
- Department of Neurology, All India Institute of Medical Sciences, New Delhi, India
| | - Subiah Vivekanandhan
- Department of Neurobiochemistry, All India Institute of Medical Sciences, New Delhi, India
| | | | - Kameshwar Prasad
- Department of Neurology, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
25
|
Sureshbabu A, Muhsin SA, Choi ME. TGF-β signaling in the kidney: profibrotic and protective effects. Am J Physiol Renal Physiol 2016; 310:F596-F606. [PMID: 26739888 DOI: 10.1152/ajprenal.00365.2015] [Citation(s) in RCA: 203] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 01/04/2016] [Indexed: 12/31/2022] Open
Abstract
Transforming growth factor-β (TGF-β) is generally considered as a central mediator of fibrotic diseases. Indeed, much focus has been placed on inhibiting TGF-β and its downstream targets as ideal therapeutic strategies. However, pharmacological blockade of TGF-β has not yet translated into successful therapy for humans, which may be due to pleiotropic effects of TGF-β signaling. Equally, TGF-β signaling as a protective response in kidney injury has been relatively underexplored. An emerging body of evidence from experimental kidney disease models indicates multifunctionality of TGF-β capable of inducing profibrotic and protective effects. This review discusses recent advances highlighting the diverse roles of TGF-β in promoting not only renal fibrosis but also protective responses of TGF-β signaling. We review, in particular, growing evidence that supports protective effects of TGF-β by mechanisms which include inhibiting inflammation and induction of autophagy. Additional detailed studies are required to fully understand the diverse mechanisms of TGF-β actions in renal fibrosis and inflammation that will likely direct toward effective antifibrotic therapies.
Collapse
Affiliation(s)
- Angara Sureshbabu
- Division of Nephrology and Hypertension, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, New York; and
| | - Saif A Muhsin
- New York-Presbyterian Hospital-Weill Cornell Medical Center, New York, New York
| | - Mary E Choi
- Division of Nephrology and Hypertension, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, New York; and .,New York-Presbyterian Hospital-Weill Cornell Medical Center, New York, New York
| |
Collapse
|
26
|
Kazemian MR, Solouk A, Tan A, Seifalian AM. Preventing in-stent restenosis using lipoprotein (a), lipid and cholesterol adsorbent materials. Med Hypotheses 2015; 85:986-8. [DOI: 10.1016/j.mehy.2015.08.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 08/31/2015] [Indexed: 11/16/2022]
|
27
|
Curtis A, Smith T, Ziganshin BA, Elefteriades JA. Ascending Aortic Proaneurysmal Genetic Mutations with Antiatherogenic Effects. Int J Angiol 2015; 24:189-97. [PMID: 26417187 DOI: 10.1055/s-0035-1556075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Thoracic aortic aneurysms are common and are associated with a high morbidity and mortality. Despite this lethal diagnosis, there is an increasing body of evidence to suggest that the diagnosis of an aneurysm, specifically in the ascending thoracic aorta, may significantly reduce the risk of developing systemic atherosclerosis. Clinical observations in the operating room have shown pristine blood vessels in patients undergoing surgery for thoracic aortic aneurysms. There is now evidence that both the carotid intima-media thickness and arterial calcification, which are early and late signs of atherosclerosis respectively, are decreased in those with thoracic aortic aneurysms. These clinical studies are supported by molecular, genetic, and pharmacological evidence. Two principle mechanisms have been identified to explain the relationship of a proaneurysmal state conferring protection from atherosclerosis. These include an excess proteolytic balance of matrix metalloproteinase activity, leading to fragmentation of elastic lamellae and disordered collagen deposition. In addition, transforming growth factor β modulates vascular smooth muscle cells, extracellular matrix, and leukocytes. This confers protection from the initial plaque formation and, later provides stability to the plaque possibly through alteration of the types I and II transforming growth factor β receptor ratio. Furthermore, studies are now beginning to establish an important role for statins and estradiol in modulating these complex pathways. In the future, as our understanding of these complex mechanisms underlying aneurysmal protection against atherosclerosis increases, corresponding therapies may be developed to offer protection from atherosclerosis.
Collapse
Affiliation(s)
- Alexander Curtis
- Aortic Institute at Yale-New Haven, Yale University School of Medicine, New Haven, Connecticut
| | - Tanya Smith
- Aortic Institute at Yale-New Haven, Yale University School of Medicine, New Haven, Connecticut
| | - Bulat A Ziganshin
- Aortic Institute at Yale-New Haven, Yale University School of Medicine, New Haven, Connecticut ; Department of Surgical Diseases No. 2, Kazan State Medical University, Kazan, Russia
| | - John A Elefteriades
- Aortic Institute at Yale-New Haven, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
28
|
Manocha A, Srivastava LM. Lipoprotein (a): a Unique Independent Risk Factor for Coronary Artery Disease. Indian J Clin Biochem 2015; 31:13-20. [PMID: 26855483 DOI: 10.1007/s12291-015-0483-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2015] [Accepted: 02/06/2015] [Indexed: 12/22/2022]
Abstract
The current epidemic affecting Indians is coronary artery disease (CAD), and is currently one of the most common causes of mortality and morbidity in developed and developing countries. The higher rate of CAD in Indians, as compared to people of other ethnic origin, may indicate a possible genetic susceptibility. Hence, Lp(a), an independent genetic risk marker for atherosclerosis and cardiovascular disease assumes great importance. Lp(a), an atherogenic lipoprotein, contains a cholesterol rich LDL particle, one molecule of apolipoprotein B-100 and a unique protein, apolipoprotein (a) which distinguishes it from LDL. Apo(a) is highly polymorphic and an inverse relationship between Lp(a) concentration and apo(a) isoform size has been observed. This is genetically controlled suggesting a functional diversity among the apo(a) isoforms. The LPA gene codes for apo(a) whose genetic heterogeneity is due to variations in its number of kringles. The exact pathogenic mechanism of Lp(a) is still not completely elucidated, but the structural homology of Lp(a) with LDL and plasmin is possibly responsible for its acting as a link between atherosclerosis and thrombosis. Upper limits of normal Lp(a) levels have not been defined for the Indian population. A cut off limit of 20 mg/dL has been suggested while for the Caucasian population it is 30 mg/dL. Though a variety of assays are available for its measurement, standardization of the analytical method is highly complicated as a majority of the methods are affected by the heterogeneity in apo(a) size. No therapeutic drug selectively targets Lp(a) but recently, new modifiers of apo(a) synthesis are being considered.
Collapse
Affiliation(s)
- Anjali Manocha
- Department of Biochemistry, Sir Ganga Ram Hospital, Rajender Nagar, New Delhi, 110060 India
| | - L M Srivastava
- Department of Biochemistry, Sir Ganga Ram Hospital, Rajender Nagar, New Delhi, 110060 India
| |
Collapse
|
29
|
Romagnuolo R, Marcovina SM, Boffa MB, Koschinsky ML. Inhibition of plasminogen activation by apo(a): role of carboxyl-terminal lysines and identification of inhibitory domains in apo(a). J Lipid Res 2014; 55:625-34. [PMID: 24478033 DOI: 10.1194/jlr.m036566] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Apo(a), the distinguishing protein component of lipoprotein(a) [Lp(a)], exhibits sequence similarity to plasminogen and can inhibit binding of plasminogen to cell surfaces. Plasmin generated on the surface of vascular cells plays a role in cell migration and proliferation, two of the fibroproliferative inflammatory events that underlie atherosclerosis. The ability of apo(a) to inhibit pericellular plasminogen activation on vascular cells was therefore evaluated. Two isoforms of apo(a), 12K and 17K, were found to significantly decrease tissue-type plasminogen activator-mediated plasminogen activation on human umbilical vein endothelial cells (HUVECs) and THP-1 monocytes and macrophages. Lp(a) purified from human plasma decreased plasminogen activation on THP-1 monocytes and HUVECs but not on THP-1 macrophages. Removal of kringle V or the strong lysine binding site in kringle IV10 completely abolished the inhibitory effect of apo(a). Treatment with carboxypeptidase B to assess the roles of carboxyl-terminal lysines in cellular receptors leads in most cases to decreases in plasminogen activation as well as plasminogen and apo(a) binding; however, inhibition of plasminogen activation by apo(a) was unaffected. Our findings directly demonstrate that apo(a) inhibits pericellular plasminogen activation in all three cell types, although binding of apo(a) to cell-surface receptors containing carboxyl-terminal lysines does not appear to play a major role in the inhibition mechanism.
Collapse
Affiliation(s)
- Rocco Romagnuolo
- Department of Chemistry & Biochemistry, University of Windsor, Windsor, ON, Canada; and
| | | | | | | |
Collapse
|
30
|
Chau KH, Bender JR, Elefteriades JA. Silver Lining in the Dark Cloud of Aneurysm Disease. Cardiology 2014; 128:327-32. [DOI: 10.1159/000358123] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Accepted: 12/18/2013] [Indexed: 11/19/2022]
|
31
|
The TGF-B1 and IL-10 gene polymorphisms are associated with risk of developing silent myocardial ischemia in the diabetic patients. Immunol Lett 2013; 156:18-22. [DOI: 10.1016/j.imlet.2013.09.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Revised: 09/11/2013] [Accepted: 09/12/2013] [Indexed: 01/27/2023]
|
32
|
Aguilera KY, Brekken RA. Recruitment and retention: factors that affect pericyte migration. Cell Mol Life Sci 2013; 71:299-309. [PMID: 23912898 DOI: 10.1007/s00018-013-1432-z] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Revised: 06/30/2013] [Accepted: 07/22/2013] [Indexed: 02/07/2023]
Abstract
Pericytes are critical for vascular morphogenesis and contribute to several pathologies, including cancer development and progression. The mechanisms governing pericyte migration and differentiation are complex and have not been fully established. Current literature suggests that platelet-derived growth factor/platelet-derived growth factor receptor-β, sphingosine 1-phosphate/endothelial differentiation gene-1, angiopoietin-1/tyrosine kinase with immunoglobulin-like and EGF-like domains 2, angiopoietin-2/tyrosine kinase with immunoglobulin-like and EGF-like domains 2, transforming growth factor β/activin receptor-like kinase 1, transforming growth factor β/activin receptor-like kinase 5, Semaphorin-3A/Neuropilin, and matrix metalloproteinase activity regulate the recruitment of pericytes to nascent vessels. Interestingly, many of these pathways are directly affected by secreted protein acidic and rich in cysteine (SPARC). Here, we summarize the function of these factors in pericyte migration and discuss if and how SPARC might influence these activities and thus provide an additional layer of control for the recruitment of vascular support cells. Additionally, the consequences of targeted inhibition of pericytes in tumors and the current understanding of pericyte recruitment in pathological environments are discussed.
Collapse
Affiliation(s)
- Kristina Y Aguilera
- Division of Surgical Oncology, Department of Surgery, Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX, 75390-8593, USA
| | | |
Collapse
|
33
|
Apolipoprotein(a) acts as a chemorepellent to human vascular smooth muscle cells via integrin αVβ3 and RhoA/ROCK-mediated mechanisms. Int J Biochem Cell Biol 2013; 45:1776-83. [PMID: 23726972 DOI: 10.1016/j.biocel.2013.05.021] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Revised: 04/24/2013] [Accepted: 05/21/2013] [Indexed: 01/31/2023]
Abstract
Lipoprotein(a) (Lp(a)) is an independent risk factor for the development of cardiovascular disease. Vascular smooth muscle cell (SMC) motility and plasticity, functions that are influenced by environmental cues, are vital to adaptation and remodelling in vascular physiology and pathophysiology. Lp(a) is reportedly damaging to SMC function via unknown molecular mechanisms. Apolipoprotein(a) (apo(a)), a unique glycoprotein moiety of Lp(a), has been demonstrated as its active component. The aims of this study were to determine functional effects of recombinant apo(a) on human vascular SMC motility and explore the underlying mechanism(s). Exposure of SMC to apo(a) in migration assays induced a potent, concentration-dependent chemorepulsion that was RhoA and integrin αVβ3-dependent, but transforming growth factor β-independent. SMC manipulation through RhoA gene silencing, Rho kinase inhibition, statin pre-treatment, αVβ3 neutralising antibody and tyrosine kinase inhibition all markedly inhibited apo(a)-mediated SMC migration. Our data reveal unique and potent activities of apo(a) that may negatively influence SMC remodelling in cardiovascular disease. Circulating levels of Lp(a) are resistant to lipid-lowering strategies and hence a greater understanding of the mechanisms underlying its functional effects on SMC may provide alternative therapeutic targets.
Collapse
|
34
|
Inhibition of neointima formation through DNA vaccination for apolipoprotein(a): a new therapeutic strategy for lipoprotein(a). Sci Rep 2013; 3:1600. [PMID: 23549288 PMCID: PMC3615337 DOI: 10.1038/srep01600] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Accepted: 03/18/2013] [Indexed: 11/13/2022] Open
Abstract
Lipoprotein(a) [Lp(a)] is an unique lipoprotein consisting of the glycoprotein apolipoprotein(a) [apo(a)] in low-density lipoprotein. Although Lp(a) is a well-known independent risk factor for cardiovascular disease; however, there is no drugs to decrease plasma Lp(a) level. Thus, to inhibit the biological activity of Lp(a), we developed DNA vaccine for apo(a) by the targeting to the selected 12 hydrophilic amino acids in the kringle-4 type 2 domain of apo(a). Hepatitis B virus core protein was used as an epitope carrier to enhance the immunogenicity. Intramuscular immunization with apo(a) vaccine resulted in the significant inhibition of neointima formation in carotid artery ligation model using Lp(a) transgenic mice, associated with anti-apo(a) antibody and decrease in vascular Lp(a) deposition. Overall, this study provided the first evidence that the pro-atherosclerotic actions of Lp(a) could be prevented by DNA vaccine directed against apo(a), suggesting a novel therapeutic strategy to treat cardiovascular diseases related to high Lp(a).
Collapse
|
35
|
Association between baseline lipoprotein (a) levels and restenosis after coronary stenting: Meta-analysis of 9 cohort studies. Atherosclerosis 2013; 227:360-6. [DOI: 10.1016/j.atherosclerosis.2013.01.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2012] [Revised: 12/30/2012] [Accepted: 01/10/2013] [Indexed: 11/17/2022]
|
36
|
Lipoprotein(a) in cardiovascular diseases. BIOMED RESEARCH INTERNATIONAL 2012; 2013:650989. [PMID: 23484137 PMCID: PMC3591100 DOI: 10.1155/2013/650989] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2012] [Revised: 11/06/2012] [Accepted: 11/08/2012] [Indexed: 11/18/2022]
Abstract
Lipoprotein(a) (Lp(a)) is an LDL-like molecule consisting of an apolipoprotein B-100 (apo(B-100)) particle attached by a disulphide bridge to apo(a). Many observations have pointed out that Lp(a) levels may be a risk factor for cardiovascular diseases. Lp(a) inhibits the activation of transforming growth factor (TGF) and contributes to the growth of arterial atherosclerotic lesions by promoting the proliferation of vascular smooth muscle cells and the migration of smooth muscle cells to endothelial cells. Moreover Lp(a) inhibits plasminogen binding to the surfaces of endothelial cells and decreases the activity of fibrin-dependent tissue-type plasminogen activator. Lp(a) may act as a proinflammatory mediator that augments the lesion formation in atherosclerotic plaques. Elevated serum Lp(a) is an independent predictor of coronary artery disease and myocardial infarction. Furthermore, Lp(a) levels should be a marker of restenosis after percutaneous transluminal coronary angioplasty, saphenous vein bypass graft atherosclerosis, and accelerated coronary atherosclerosis of cardiac transplantation. Finally, the possibility that Lp(a) may be a risk factor for ischemic stroke has been assessed in several studies. Recent findings suggest that Lp(a)-lowering therapy might be beneficial in patients with high Lp(a) levels. A future therapeutic approach could include apheresis in high-risk patients in order to reduce major coronary events.
Collapse
|
37
|
Lipoprotein(a): Cellular Effects and Molecular Mechanisms. CHOLESTEROL 2012; 2012:923289. [PMID: 22991657 PMCID: PMC3443569 DOI: 10.1155/2012/923289] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Accepted: 07/24/2012] [Indexed: 01/31/2023]
Abstract
Lipoprotein(a) (Lp(a)) is an independent risk factor for the development of cardiovascular disease (CVD). Indeed, individuals with plasma concentrations >20 mg/dL carry a 2-fold increased risk of developing CVD, accounting for ~25% of the population. Circulating levels of Lp(a) are remarkably resistant to common lipid lowering therapies, and there are currently no robust treatments available for reduction of Lp(a) apart from plasma apheresis, which is costly and labour intensive. The Lp(a) molecule is composed of two parts, an LDL/apoB-100 core and a unique glycoprotein, apolipoprotein(a) (apo(a)), both of which can interact with components of the coagulation cascade, inflammatory pathways, and cells of the blood vessel wall (smooth muscle cells (SMC) and endothelial cells (EC)). Therefore, it is of key importance to determine the molecular pathways by which Lp(a) exerts its influence on the vascular system in order to design therapeutics to target its cellular effects. This paper will summarise the role of Lp(a) in modulating cell behaviour in all aspects of the vascular system including platelets, monocytes, SMC, and EC.
Collapse
|
38
|
Ikonomidis I, Michalakeas CA, Parissis J, Paraskevaidis I, Ntai K, Papadakis I, Anastasiou-Nana M, Lekakis J. Inflammatory markers in coronary artery disease. Biofactors 2012; 38:320-8. [PMID: 22628054 DOI: 10.1002/biof.1024] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Accepted: 04/18/2012] [Indexed: 12/20/2022]
Abstract
Coronary artery disease (CAD) is one of the most common manifestations of atherosclerosis. Inflammation is considered one of the major processes that contribute to atherogenesis. Inflammation plays an important role not only on the initiation and progression of atherosclerosis but also on plaque rupture, an event that leads to acute vascular events. Various biomarkers express different pathways and pathophysiologic mechanisms of cardiovascular disease, and inflammatory biomarkers express different parts of the atherogenic process, regarding the initiation and progression of atherosclerosis or the destabilization of the atherosclerotic plaque. Therefore, inflammatory biomarkers may prove to be useful in the detection, staging, and prognosis of patients with CAD. Furthermore, the fact that inflammatory processes are essential steps in the course of the disease offers future therapeutic targets for the interruption of the atherogenic process or for the management of acute events.
Collapse
Affiliation(s)
- Ignatios Ikonomidis
- 2nd Cardiology Department, University of Athens, Attikon Hospital, Athens, Greece.
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Fragoso JM, Martínez-Ríos MA, Alvarez-León E, Vallejo M, Peña-Duque MA, Posadas-Sánchez R, Posadas-Romero C, Furuzawa-Carballeda J, Lima G, Llorente L, Vargas-Alarcón G. The T29C polymorphism of the transforming growth factor-β1 (TGF-β1) gene is associated with genetic susceptibility to acute coronary syndrome in Mexican patients. Cytokine 2012; 58:380-3. [PMID: 22465044 DOI: 10.1016/j.cyto.2012.03.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2011] [Revised: 01/20/2012] [Accepted: 03/05/2012] [Indexed: 11/16/2022]
Abstract
Inflammation plays an essential role in the development and progression of atherosclerotic lesions, and plaque disruption. The TGF-β1 plays an important role in the anti-inflammatory process. The aim of the present study was to evaluate the role of TGF-β1 gene polymorphisms as susceptibility markers for acute coronary syndrome (ACS). Two polymorphisms (TGF-β -509T>C and TGF-β T29C) of the TGF-β gene were analyzed by 5' exonuclease TaqMan genotyping assays in a group of 426 patients with coronary acute syndrome and 551 healthy unrelated controls. A significant difference was observed in the distribution of TGF-β T29C polymorphism between ACS patients and healthy controls (P<10(-3)). According to the co-dominant model, individuals with the TGF-β 29 TT genotype have a 2.5-fold increased risk of developing ACS (P<10(-3)). Multiple logistic analysis showed that the largest risk factor for developing ACS was given by smoking habit, diabetes, hypertension, dyslipidemia, and the TGF-β1 29 TT genotype. The analysis of linkage disequilibrium showed one haplotype (TT) with increased frequency and one haplotype (CC) with decreased frequency in ACS patients when compared to healthy controls. The results suggest that TGF-β1 T29C gene polymorphism could be involved in the risk of developing ACS in Mexican individuals.
Collapse
Affiliation(s)
- José Manuel Fragoso
- Department of Molecular Biology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Rivera LB, Brekken RA. SPARC promotes pericyte recruitment via inhibition of endoglin-dependent TGF-β1 activity. ACTA ACUST UNITED AC 2011; 193:1305-19. [PMID: 21708981 PMCID: PMC3216331 DOI: 10.1083/jcb.201011143] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
SPARC prevents endoglin association with αV integrin, which blocks the activation of TGF-β signaling and promotes pericyte migration to nascent blood vessels. Pericytes migrate to nascent vessels and promote vessel stability. Recently, we reported that secreted protein acidic and rich in cysteine (SPARC)–deficient mice exhibited decreased pericyte-associated vessels in an orthotopic model of pancreatic cancer, suggesting that SPARC influences pericyte behavior. In this paper, we report that SPARC promotes pericyte migration by regulating the function of endoglin, a TGF-β1 accessory receptor. Primary SPARC-deficient pericytes exhibited increased basal TGF-β1 activity and decreased cell migration, an effect blocked by inhibiting TGF-β1. Furthermore, TGF-β–mediated inhibition of pericyte migration was dependent on endoglin and αV integrin. SPARC interacted directly with endoglin and reduced endoglin interaction with αV integrin. SPARC deficiency resulted in endoglin-mediated blockade of pericyte migration, aberrant association of endoglin in focal complexes, an increase in αV integrins present in endoglin immunoprecipitates, and enhanced αV integrin–mediated activation of TGF-β. These results demonstrate that SPARC promotes pericyte migration by diminishing TGF-β activity and identify a novel function for endoglin in controlling pericyte behavior.
Collapse
Affiliation(s)
- Lee B Rivera
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | | |
Collapse
|
41
|
Sharma A, Kasim M, Joshi PH, Qian Z, Krivitsky E, Akram K, Rinehart S, Vazquez G, Miller J, Rohman MS, Voros S. Abnormal Lipoprotein(a) Levels Predict Coronary Artery Calcification in Southeast Asians but Not in Caucasians: Use of Noninvasive Imaging for Evaluation of an Emerging Risk Factor. J Cardiovasc Transl Res 2011; 4:470-6. [DOI: 10.1007/s12265-011-9273-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2011] [Accepted: 03/11/2011] [Indexed: 11/30/2022]
|
42
|
TGF-β and microvessel homeostasis. Microvasc Res 2010; 80:166-73. [DOI: 10.1016/j.mvr.2010.03.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2010] [Revised: 02/23/2010] [Accepted: 03/04/2010] [Indexed: 12/17/2022]
|
43
|
Xu S, Liu AC, Gotlieb AI. Common pathogenic features of atherosclerosis and calcific aortic stenosis: role of transforming growth factor-beta. Cardiovasc Pathol 2009; 19:236-47. [PMID: 19942455 DOI: 10.1016/j.carpath.2009.09.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2009] [Revised: 09/22/2009] [Accepted: 09/24/2009] [Indexed: 12/20/2022] Open
Abstract
Calcific aortic stenosis and atherosclerosis have been investigated separately in experimental in vitro and in vivo studies and in clinical studies. The similarities identified in both diseases suggest that similar pathogenic pathways are involved in both conditions. Most current therapeutic studies are focused on statins. The evidence suggests that statin effects on valves may, in large part, be independent of the lipid lowering effects of the drug. There are several molecules that play significant regulatory roles on the development and progression of valve sclerosis and calcification and on growth and complications of atherosclerotic plaques. The purpose of this review is to discuss the pathogenic features of the two conditions, highlight the important similarities, and then review the data that suggest that transforming growth factor-beta may play a key regulatory role in both diseases and that this is worthy of study as a potential therapeutic target for both conditions.
Collapse
Affiliation(s)
- Songyi Xu
- Toronto General Research Institute and Laboratory Medicine Program, University Health Network, Toronto, ON, Canada
| | | | | |
Collapse
|
44
|
Krause BR, Sliskovic DR, Ma Bocan T. Section Review—Cardiovascular & Renal: Emerging Therapies in Atherosclerosis. Expert Opin Investig Drugs 2008. [DOI: 10.1517/13543784.4.5.353] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
45
|
Chen CL, Huang SS, Huang JS. Cholesterol modulates cellular TGF-beta responsiveness by altering TGF-beta binding to TGF-beta receptors. J Cell Physiol 2008; 215:223-33. [PMID: 17972267 DOI: 10.1002/jcp.21303] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Transforming growth factor-beta (TGF-beta) responsiveness in cultured cells can be modulated by TGF-beta partitioning between lipid raft/caveolae- and clathrin-mediated endocytosis pathways. The TbetaR-II/TbetaR-I binding ratio of TGF-beta on the cell surface has recently been found to be a signal that controls TGF-beta partitioning between these pathways. Since cholesterol is a structural component in lipid rafts/caveolae, we have studied the effects of cholesterol on TGF-beta binding to TGF-beta receptors and TGF-beta responsiveness in cultured cells and in animals. Here we demonstrate that treatment with cholesterol, alone or complexed in lipoproteins, decreases the TbetaR-II/TbetaR-I binding ratio of TGF-beta while treatment with cholesterol-lowering or cholesterol-depleting agents increases the TbetaR-II/TbetaR-I binding ratio of TGF-beta in all cell types studied. Among cholesterol derivatives and analogs examined, cholesterol is the most potent agent for decreasing the TbetaR-II/TbetaR-I binding ratio of TGF-beta. Cholesterol treatment increases accumulation of the TGF-beta receptors in lipid rafts/caveolae as determined by sucrose density gradient ultracentrifugation analysis of cell lysates. Cholesterol/LDL suppresses TGF-beta responsiveness and statins/beta-CD enhances it, as measured by the levels of P-Smad2 and PAI-1 expression in cells stimulated with TGF-beta. Furthermore, the cholesterol effects observed in cultured cells are also found in the aortic endothelium of atherosclerotic ApoE-null mice fed a high cholesterol diet. These results indicate that high plasma cholesterol levels may contribute to the pathogenesis of certain diseases (e.g., atherosclerosis) by suppressing TGF-beta responsiveness.
Collapse
Affiliation(s)
- Chun-Lin Chen
- Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, Missouri 63104, USA
| | | | | |
Collapse
|
46
|
Abstract
A reversal in the association between traditional and nontraditional risk factors and clinical outcomes is often encountered in patients with chronic illness, including among those with advanced chronic kidney disease (CKD) on maintenance hemodialysis (MHD). The effects of the malnutrition-inflammation complex syndrome (MICS) may play a significant role in the reversal of this risk factor-outcomes association. the MICS, this syndrome complex is not universal in its prevalence among MHD patients. The significant inter- and intra-individual differences in the prevalence of inflammation, oxidative stress, and malnutrition, indicates the influence of genetic factors in this variability. In recent years, enormous advancement in the field of molecular genetics, genomics and bioinformatics, have revolutionized studies of the genetic epidemiology of several diseases. However, genetic association studies are at a preliminary stage in the population with advanced CKD (Table 1). Preliminary studies of the impact of polyphisms in inflammation and oxidative stress-related genes and genes affecting body composition and metabolism suggest that genetic variation may indeed affect the phenotype of the MHD population. Further, some of these gene polymorphisms may also contribute to a reversal of the association between traditional risk factors, such as BMI, blood pressure, and cholesterol and clinical outcomes in this vulnerable patient population. Genetic studies in patients with advanced CKD pose enormous challenges, including recruitment of sufficient numbers of patients to achieve adequate statistical power, resolution of immense genotypic and phenotypic heterogeneity, and gene-environment and gene-gene interactions. However, well-designed adequately powered studies with carefully defined phenotypes may potentially allow definition of risk profiles characterized by combinations of relevant Single nucleotide polymorphisms in the setting of given environmental factors. Accurate risk stratification that takes into account genetic information would allow more informed targeting of pharmacologic intervention and better refined clinical trial methodologies.
Collapse
|
47
|
Cicek H, Bayil S, Zer Y, Celik A, Geyikli I. Comparison of Lipoprotein(a) levels between elderly and middle-aged men with coronary artery disease. Ann N Y Acad Sci 2007; 1100:179-84. [PMID: 17460176 DOI: 10.1196/annals.1395.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Lipoprotein(a) [Lp(a)] is known to be a risk factor for atherosclerotic disease in middle-aged men, but the role of Lp(a) in women and in the elderly is less clear. In most studies, excess Lp(a) is not associated with increased risk for persons >65 years of age. This study examined the strength of association of a number of risk factors to coronary artery disease (CAD) in groups of men <65 years (n = 108) and >65 of age (n = 66) with angiographically documented significant narrowing of coronary arteries. Serum Lp(a) concentrations were determined; elevated Lp(a) is positively associated with CAD for men <65 years (adjusted OR: 2.45, P <0.05) but not for men >65 of age (adjusted OR: 0.56, P = NS). For middle-aged men, elevated Lp(a) appears to be an independent risk factor for premature CAD, and the importance of Lp(a) as a risk factor appears to decrease with age. These data suggest that the utility of Lp(a) lipoprotein levels in predicting the risk of CAD in older men is limited. Factors, such as age; sex; levels of total cholesterol, low-density lipoprotein (LDL) cholesterol, and triglycerides; carotid-wall thickness; smoking status; the presence or absence of diabetes and systolic and diastolic hypertension; body mass index (BMI); and other traditional risk factors, must be evaluated together to determine the risk of CAD for the entire population.
Collapse
Affiliation(s)
- Hulya Cicek
- Department of Biochemistry and Clinical Biochemistry, Central Laboratory, Gaziantep University Hospital, 27600 Gaziantep, Turkey.
| | | | | | | | | |
Collapse
|
48
|
Wakabayashi I, Masuda H. Lipoprotein (a) as a determinant of arterial stiffness in elderly patients with type 2 diabetes mellitus. Clin Chim Acta 2006; 373:127-31. [PMID: 16820145 DOI: 10.1016/j.cca.2006.05.018] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2006] [Revised: 05/10/2006] [Accepted: 05/10/2006] [Indexed: 12/14/2022]
Abstract
BACKGROUND Lipoprotein (a) [Lp(a)] is known to be a risk factor for atherosclerotic disease. However, the relationship between Lp(a) and arterial stiffness has not been clarified. We investigated whether atherosclerotic risk factors, including serum Lp(a), are associated with aortic stiffness in elderly patients with type 2 diabetes mellitus. METHODS Aortic stiffness, evaluated by using aortic pulse wave velocity, and major atherosclerotic risk factors were measured in elderly (> or =65 years) patients with type 2 diabetes mellitus. Relationships between aortic pulse wave velocity, Lp(a) and other atherosclerotic risk factors were analyzed. RESULTS Among the atherosclerotic risk factors measured, age, pulse pressure, hemoglobin A1c (HbA1c), uric acid, fibrinogen, sialic acid and Lp(a) showed significant positive correlations with aortic pulse wave velocity. Lp(a) also showed significant positive correlations with pulse pressure, fibrinogen, sialic acid, apolipoprotein B and apolipoprotein B/apolipoprotein A-I ratio. The correlation between Lp(a) and aortic pulse wave velocity was independent of age, sex, blood hemoglobin A1c, uric acid and fibrinogen, history of diabetic nephropathy and therapy with lipid-lowering drugs. Apolipoprotein A-II showed a significant negative correlation with both aortic pulse wave velocity and Lp(a). The median level of Lp(a) in the highest tertile group of subjects divided by aortic pulse wave velocity was significantly higher than the median Lp(a) level in the lowest tertile. The middle and highest tertile groups of subjects divided by aortic pulse wave velocity showed significantly high odds ratios of high Lp(a) levels (> or =30 mg/dl) vs the lowest tertile. CONCLUSIONS Lp(a) is an independent determinant of aortic stiffness in elderly patients with type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Ichiro Wakabayashi
- Department of Hygiene and Preventive Medicine, Yamagata University School of Medicine, Iida-Nishi 2-2-2, Yamagata 990-9585, Japan.
| | | |
Collapse
|
49
|
Lazo-Langner A, Knoll GA, Wells PS, Carson N, Rodger MA. The risk of dialysis access thrombosis is related to the transforming growth factor-beta1 production haplotype and is modified by polymorphisms in the plasminogen activator inhibitor-type 1 gene. Blood 2006; 108:4052-8. [PMID: 16931622 DOI: 10.1182/blood-2006-06-028902] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Transforming growth factor-beta1 (TGF-beta1) and plasminogen activator inhibitor-type 1 (PAI-1) might play a role in the development of fibrosis and stenosis of hemodialysis vascular accesses. We studied polymorphisms in the TGFbeta1 (869T>C; 915G>C), and PAI-1 (4G/5G) genes in 416 hemodialysis patients (107 access thrombosis cases, 309 controls), to determine if they are related to vascular access thrombosis. Three TGF-beta1 production haplotypes (low, intermediate, and high) were defined according to the combination of polymorphisms found. The adjusted odds ratio (OR) and 95% confidence interval (CI) for access thrombosis in low TGF-beta1 producers was 7.31 (2.15-24.88; P = .001). The interaction between low TGF-beta1 production haplotype and the 4G/4G PAI-1 genotype was strongly associated with access thrombosis (adjusted OR 19.3; 95% CI 2.82-132.40; P = .003). Mean access thrombosis-free survival times in years (95% CI) were 14.65 (12.05-17.25), 11.96 (8.67-15.25), and 4.94 (3.06-6.83) in high, intermediate, and low TGF-beta1 producers, respectively (P = .044). Analysis of the synergy index and the case-only cross-product supported the presence of an interaction. We concluded that low TGF-beta1 production haplotype is a risk factor for hemodialysis access thrombosis and that in the presence of the 4G/4G PAI-1 genotype there is an additional increase in risk.
Collapse
Affiliation(s)
- Alejandro Lazo-Langner
- Division of Hematology, Department of Medicine, The Ottawa Hospital, University of Ottawa, Ottawa, Ontario Canada
| | | | | | | | | |
Collapse
|
50
|
Morita Y, Himeno H, Yakuwa H, Usui T. Serum lipoprotein(a) level and clinical coronary stenosis progression in patients with myocardial infarction: re-revascularization rate is high in patients with high-Lp(a). Circ J 2006; 70:156-62. [PMID: 16434808 DOI: 10.1253/circj.70.156] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND High serum lipoprotein(a) (Lp(a)) levels are associated with coronary artery disease. METHODS AND RESULTS The serum Lp(a) levels of 130 patients with acute myocardial infarction (AMI) who underwent direct percutaneous coronary intervention were investigated. On the basis of Lp(a) level at 1 month after the onset of AMI, the patients were classified into 2 groups (high-Lp(a) (> or =30 mg/dl) and low-Lp(a) (< 30 mg/dl)) for evaluation of the clinical coronary stenosis progression (CCSP) rate. CCSP is defined as either target lesion revascularization (TLR) or new lesion revascularization (NLR). The CCSP rate was significantly higher in the high-Lp(a) group than in the low-Lp(a) group (65.8% vs 29.3%, p<0.01). In patients who had coronary stents in the acute phase (n=79), the CCSP and NLR rates were significantly higher in the high-Lp(a) group than in the low-Lp(a) group (45.0% vs 20.3%, p<0.05; 35.0% vs 6.8%, p<0.01), but there was no significant difference in TLR rate between the 2 groups (10.0% vs 13.6%, p=0.858). CONCLUSIONS High serum Lp(a) level is a significant risk factor for CCSP, but does not influence restenosis after stenting.
Collapse
Affiliation(s)
- Yukiko Morita
- Department of Cardiology, Fujisawa City Hospital, 2-6-1 Fujisawa, Fujisawa 251-8550, Japan.
| | | | | | | |
Collapse
|