1
|
Turkson V, Haller A, Jaeschke A, Hui DY. ApoE Receptor-2 R952Q Variant in Macrophages Elevates Soluble LRP1 to Potentiate Hyperlipidemia and Accelerate Atherosclerosis in Mice. Arterioscler Thromb Vasc Biol 2024. [PMID: 39508104 DOI: 10.1161/atvbaha.124.321748] [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: 08/22/2024] [Accepted: 10/24/2024] [Indexed: 11/08/2024]
Abstract
BACKGROUND apoER2 (apolipoprotein E receptor-2) is a transmembrane receptor in the low-density lipoprotein receptor (LDLR) family with unique tissue expression. A single-nucleotide polymorphism that encodes the R952Q sequence variant has been associated with elevated plasma cholesterol levels and increased myocardial infarction risk in humans. The objective of this study was to delineate the mechanism underlying the association between the apoER2 R952Q variant and increased atherosclerosis risk. METHODS An apoER2 R952Q mouse model was generated using a CRISPR/Cas9 strategy, intercrossed with LDLR knockout mice, followed by feeding a Western-type high-fat high-cholesterol diet for 16 weeks. Atherosclerosis was investigated by immunohistology. Plasma lipids and lipid distributions among the various lipoprotein classes were analyzed by colorimetric assay. Tissue-specific effects of the R952Q sequence variant on atherosclerosis were analyzed by bone marrow transplant studies. sLRP1 (soluble low-density lipoprotein receptor-related protein 1) was measured in plasma and conditioned media from bone marrow-derived macrophages by ELISA and GST-RAP (glutathione S-transferase-receptor-associated protein) pull-down, respectively. P38 MAPK (mitogen-activated protein kinase) phosphorylation in VLDL (very-low-density lipoprotein)-treated macrophages was determined by Western blot analysis. RESULTS Consistent with observations in humans with this sequence variant, the apoER2 R952Q mutation exacerbated diet-induced hypercholesterolemia, via impediment of plasma triglyceride-rich lipoprotein clearance, to accelerate atherosclerosis in Western diet-fed LDLR knockout mice. Reciprocal bone marrow transplant experiments revealed that the apoER2 R952Q mutation in bone marrow-derived cells instead of non-bone marrow-derived cells was responsible for the increase in hypercholesterolemia and atherosclerosis. Additional data showed that the apoER2 R952Q mutation in macrophages promotes VLDL-induced LRP1 (low-density lipoprotein receptor-related protein 1) shedding in a p38 MAPK-dependent manner. CONCLUSIONS The apoER2 R952Q mouse model recapitulates characteristics observed in human disease. The underlying mechanism is that the apoER2 R952Q mutation in macrophages exacerbates VLDL-stimulated sLRP1 production in a p38 MAPK-dependent manner, resulting in its competition with cell surface LRP1 to impede triglyceride-rich lipoprotein clearance, thereby resulting in increased hypercholesterolemia and accelerated atherosclerosis.
Collapse
Affiliation(s)
| | - April Haller
- Department of Pathology and Laboratory Medicine (A.H., A.J., D.Y.H.)
| | - Anja Jaeschke
- Department of Pathology and Laboratory Medicine (A.H., A.J., D.Y.H.)
| | - David Y Hui
- Department of Pathology and Laboratory Medicine (A.H., A.J., D.Y.H.)
| |
Collapse
|
2
|
Sun Z, Zhao T, Bai X, Li H, Gao J, Hao Y, Li Y, Xie Y, Hu A, Huang Q, Liu X, Zhang Y. Berberine Targets PKM2 to Activate the t-PA-Induced Fibrinolytic System and Improves Thrombosis. Pharmaceuticals (Basel) 2024; 17:1219. [PMID: 39338381 PMCID: PMC11434879 DOI: 10.3390/ph17091219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Arterial thrombosis, a condition in which thrombi form in arteries, can lead to various acute cardiovascular diseases and impact the quality of life and survival of patients. Berberine (BBR), a quaternary ammonium alkaloid, has been shown to treat these diseases. However, further exploration is needed to understand underlying mechanisms of BBR. METHODS AND RESULTS Rats were administered BBR via intramuscular injection. Then, an FeCl3-coated filter paper was applied to a carotid artery to induce thrombosis. The size of the thrombus and the blood flow velocity were evaluated by carotid ultrasound. The shape of the thrombus was observed using staining and microscopy. The expression levels of mRNA and proteins were verified. Additionally, mass spectrometry and single-cell RNA sequencing analysis were conducted. The administration of BBR resulted in a significant reduction in the thrombus area and an extension of the thrombus-clogging time. Furthermore, BBR administration effectively reversed the decreasing tissue-plasminogen activator (t-PA) expression and alterations in fibrinolysis system of model group. Additionally, the expression of PKM2 was suppressed following BBR administration, and the overexpression of PKM2 inhibited t-PA expression. CONCLUSIONS BBR ameliorates thrombosis by modulating expression of PKM2, subsequently impacting the expression of t-PA within fibrinolytic system. These preliminary findings suggest that BBR could be a potential preventive and therapeutic strategy for arterial thromboembolic diseases.
Collapse
Affiliation(s)
- Zeqi Sun
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin 150081, China
- State Key Laboratory-Province Key Laboratories of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin 150081, China
- Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin 150081, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin 150081, China
| | - Tong Zhao
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin 150081, China
- State Key Laboratory-Province Key Laboratories of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin 150081, China
- Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin 150081, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin 150081, China
| | - Xue Bai
- College of Pharmacy, Hainan University, Haikou 570228, China
| | - Huimin Li
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin 150081, China
- State Key Laboratory-Province Key Laboratories of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin 150081, China
- Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin 150081, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin 150081, China
| | - Jin Gao
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin 150081, China
- State Key Laboratory-Province Key Laboratories of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin 150081, China
- Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin 150081, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin 150081, China
| | - Yutong Hao
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin 150081, China
- State Key Laboratory-Province Key Laboratories of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin 150081, China
- Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin 150081, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin 150081, China
| | - Yiyang Li
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin 150081, China
- State Key Laboratory-Province Key Laboratories of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin 150081, China
- Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin 150081, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin 150081, China
| | - Yanli Xie
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin 150081, China
- State Key Laboratory-Province Key Laboratories of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin 150081, China
- Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin 150081, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin 150081, China
| | - Ange Hu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin 150081, China
- State Key Laboratory-Province Key Laboratories of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin 150081, China
- Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin 150081, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin 150081, China
| | - Qiang Huang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin 150081, China
- State Key Laboratory-Province Key Laboratories of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin 150081, China
- Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin 150081, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin 150081, China
| | - Xin Liu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin 150081, China
- State Key Laboratory-Province Key Laboratories of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin 150081, China
- Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin 150081, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin 150081, China
| | - Yong Zhang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin 150081, China
- State Key Laboratory-Province Key Laboratories of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin 150081, China
- Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin 150081, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin 150081, China
| |
Collapse
|
3
|
Yang H, Luo X, Wang X, Peng Y, Li Z, He Y, Cong J, Xie T, Zhang W. The PP2A inhibitor LB-100 mitigates lupus nephritis by suppressing tertiary lymphoid structure formation. Eur J Pharmacol 2024; 977:176703. [PMID: 38839028 DOI: 10.1016/j.ejphar.2024.176703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 05/29/2024] [Accepted: 05/30/2024] [Indexed: 06/07/2024]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by multi-organ involvement and autoantibody production. Patients with SLE face a substantial risk of developing lupus nephritis (LN), which imposes a substantial burden on both patients and their families. Protein phosphatase 2A (PP2A) is a widely distributed serine/threonine phosphatase that participates in regulating multiple signaling pathways. Inhibition of PP2A has been implicated in the treatment of various diseases. LB-100, a small molecule inhibitor of PP2A, has demonstrated anti-tumor therapeutic effects and high safety profile in preclinical experiments. However, the role of PP2A and its inhibitor has been insufficiently studied in LN. In this study, we assessed the potential effects of LB-100 in both MRL/lpr mice and R848-induced BALB/c mice. Our findings indicated that LB-100 administration led to reduced spleen enlargement, decreased deposition of immune complexes, ameliorated renal damage, and improved kidney function in both spontaneous and R848-induced lupus mouse models. Importantly, we observed the formation of tertiary lymphoid structures (TLSs) in the kidneys of two distinct lupus mouse models. The levels of signature genes of TLS were elevated in the kidneys of lupus mice, whereas LB-100 mitigated chemokine production and inhibited TLS formation. In addition, we confirmed that inhibition or knockdown of PP2A reduced the production of T cell-related chemokines by renal tubular epithelial cells (RTEC). In summary, our study highlighted the renal protective potential of the PP2A inhibitor LB-100 in two distinct lupus mouse models, suggesting its potential as a novel strategy for treating LN and other autoimmune diseases.
Collapse
Affiliation(s)
- Hongyu Yang
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Xiaoyu Luo
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Xuan Wang
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China; Department of General Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China; National Medical Metabolomics International Collaborative Research Center, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yi Peng
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Zhilan Li
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yifei He
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Jing Cong
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Tingting Xie
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China; Department of General Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China; National Medical Metabolomics International Collaborative Research Center, Xiangya Hospital, Central South University, Changsha, 410008, China.
| | - Weiru Zhang
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China; Department of General Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China; National Medical Metabolomics International Collaborative Research Center, Xiangya Hospital, Central South University, Changsha, 410008, China.
| |
Collapse
|
4
|
Müller-Calleja N, Ruf W, Lackner KJ. Lipid-binding antiphospholipid antibodies: significance for pathophysiology and diagnosis of the antiphospholipid syndrome. Crit Rev Clin Lab Sci 2024; 61:370-387. [PMID: 38293818 DOI: 10.1080/10408363.2024.2305121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 12/13/2023] [Accepted: 01/10/2024] [Indexed: 02/01/2024]
Abstract
The antiphospholipid syndrome (APS) is an autoimmune disease characterized by the presence of pathogenic antiphospholipid antibodies (aPL). Since approximately 30 years ago, lipid-binding aPL, which do not require a protein cofactor, have been regarded as irrelevant for APS pathogenesis even though anticardiolipin are a diagnostic criterion of APS. In this review, we will summarize the available evidence from in vitro studies, animal models, and epidemiologic studies, which suggest that this concept is no longer tenable. Accordingly, we will only briefly touch on the role of other aPL in APS. This topic has been amply reviewed in detail elsewhere. We will discuss the consequences for laboratory diagnostics and future research required to resolve open questions related to the pathogenic role of different aPL specificities.
Collapse
Affiliation(s)
- Nadine Müller-Calleja
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Wolfram Ruf
- Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA, USA
| | - Karl J Lackner
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| |
Collapse
|
5
|
Feng W, Qiao J, Tan Y, Liu Q, Wang Q, Yang B, Yang S, Cui L. Interaction of antiphospholipid antibodies with endothelial cells in antiphospholipid syndrome. Front Immunol 2024; 15:1361519. [PMID: 39044818 PMCID: PMC11263079 DOI: 10.3389/fimmu.2024.1361519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 06/19/2024] [Indexed: 07/25/2024] Open
Abstract
Antiphospholipid syndrome (APS) is an autoimmune disease with arteriovenous thrombosis and recurrent miscarriages as the main clinical manifestations. Due to the complexity of its mechanisms and the diversity of its manifestations, its diagnosis and treatment remain challenging issues. Antiphospholipid antibodies (aPL) not only serve as crucial "biomarkers" in diagnosing APS but also act as the "culprits" of the disease. Endothelial cells (ECs), as one of the core target cells of aPL, bridge the gap between the molecular level of these antibodies and the tissue and organ level of pathological changes. A more in-depth exploration of the relationship between ECs and the pathogenesis of APS holds the potential for significant advancements in the precise diagnosis, classification, and therapy of APS. Many researchers have highlighted the vital involvement of ECs in APS and the underlying mechanisms governing their functionality. Through extensive in vitro and in vivo experiments, they have identified multiple aPL receptors on the EC membrane and various intracellular pathways. This article furnishes a comprehensive overview and summary of these receptors and signaling pathways, offering prospective targets for APS therapy.
Collapse
Affiliation(s)
- Weimin Feng
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
- Institute of Medical Technology, Health Science Centre, Peking University, Beijing, China
| | - Jiao Qiao
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
- Institute of Medical Technology, Health Science Centre, Peking University, Beijing, China
| | - Yuan Tan
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
- Institute of Medical Technology, Health Science Centre, Peking University, Beijing, China
| | - Qi Liu
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
- Institute of Medical Technology, Health Science Centre, Peking University, Beijing, China
| | - Qingchen Wang
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
| | - Boxin Yang
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
| | - Shuo Yang
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
| | - Liyan Cui
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
- Institute of Medical Technology, Health Science Centre, Peking University, Beijing, China
| |
Collapse
|
6
|
Bitsadze V, Yakubova F, Khizroeva J, Lazarchuk A, Salnikova P, Vorobev A, Tretyakova M, Degtyareva N, Grigoreva K, Gashimova N, Kvaratskheliia M, Makatsariya N, Kudryavtseva E, Tomlenova A, Gris JC, Elalamy I, Ay C, Makatsariya A. Catastrophic Antiphospholipid Syndrome. Int J Mol Sci 2024; 25:668. [PMID: 38203837 PMCID: PMC10779422 DOI: 10.3390/ijms25010668] [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: 11/24/2023] [Revised: 12/26/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024] Open
Abstract
Unlike classic APS, CAPS causes multiple microthrombosis due to an increased inflammatory response, known as a "thrombotic storm". CAPS typically develops after infection, trauma, or surgery and begins with the following symptoms: fever, thrombocytopenia, muscle weakness, visual and cognitive disturbances, abdominal pain, renal failure, and disseminated intravascular coagulation. Although the presence of antiphospholipid antibodies in the blood is one of the diagnostic criteria, the level of these antibodies can fluctuate significantly, which complicates the diagnostic process and can lead to erroneous interpretation of rapidly developing symptoms. Triple therapy is often used to treat CAPS, which includes the use of anticoagulants, plasmapheresis, and high doses of glucocorticosteroids and, in some cases, additional intravenous immunoglobulins. The use of LMWH is recommended as the drug of choice due to its anti-inflammatory and anticoagulant properties. CAPS is a multifactorial disease that requires not only an interdisciplinary approach but also highly qualified medical care, adequate and timely diagnosis, and appropriate prevention in the context of relapse or occurrence of the disease. Improved new clinical protocols and education of medical personnel regarding CAPS can significantly improve the therapeutic approach and reduce mortality rates.
Collapse
Affiliation(s)
- Victoria Bitsadze
- Department of Obstetrics, Gynecology and Perinatal Medicine, N. F. Filatov Clinical Institute of Children’s Health, I. M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya Str. 8-2, 119991 Moscow, Russia; (F.Y.); (J.K.); (A.L.); (P.S.); (A.V.); (M.T.); (N.D.); (K.G.); (N.G.); (M.K.); (N.M.); (E.K.); (A.T.); (J.-C.G.); (I.E.); (C.A.)
| | - Fidan Yakubova
- Department of Obstetrics, Gynecology and Perinatal Medicine, N. F. Filatov Clinical Institute of Children’s Health, I. M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya Str. 8-2, 119991 Moscow, Russia; (F.Y.); (J.K.); (A.L.); (P.S.); (A.V.); (M.T.); (N.D.); (K.G.); (N.G.); (M.K.); (N.M.); (E.K.); (A.T.); (J.-C.G.); (I.E.); (C.A.)
| | - Jamilya Khizroeva
- Department of Obstetrics, Gynecology and Perinatal Medicine, N. F. Filatov Clinical Institute of Children’s Health, I. M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya Str. 8-2, 119991 Moscow, Russia; (F.Y.); (J.K.); (A.L.); (P.S.); (A.V.); (M.T.); (N.D.); (K.G.); (N.G.); (M.K.); (N.M.); (E.K.); (A.T.); (J.-C.G.); (I.E.); (C.A.)
| | - Arina Lazarchuk
- Department of Obstetrics, Gynecology and Perinatal Medicine, N. F. Filatov Clinical Institute of Children’s Health, I. M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya Str. 8-2, 119991 Moscow, Russia; (F.Y.); (J.K.); (A.L.); (P.S.); (A.V.); (M.T.); (N.D.); (K.G.); (N.G.); (M.K.); (N.M.); (E.K.); (A.T.); (J.-C.G.); (I.E.); (C.A.)
| | - Polina Salnikova
- Department of Obstetrics, Gynecology and Perinatal Medicine, N. F. Filatov Clinical Institute of Children’s Health, I. M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya Str. 8-2, 119991 Moscow, Russia; (F.Y.); (J.K.); (A.L.); (P.S.); (A.V.); (M.T.); (N.D.); (K.G.); (N.G.); (M.K.); (N.M.); (E.K.); (A.T.); (J.-C.G.); (I.E.); (C.A.)
| | - Alexander Vorobev
- Department of Obstetrics, Gynecology and Perinatal Medicine, N. F. Filatov Clinical Institute of Children’s Health, I. M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya Str. 8-2, 119991 Moscow, Russia; (F.Y.); (J.K.); (A.L.); (P.S.); (A.V.); (M.T.); (N.D.); (K.G.); (N.G.); (M.K.); (N.M.); (E.K.); (A.T.); (J.-C.G.); (I.E.); (C.A.)
| | - Maria Tretyakova
- Department of Obstetrics, Gynecology and Perinatal Medicine, N. F. Filatov Clinical Institute of Children’s Health, I. M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya Str. 8-2, 119991 Moscow, Russia; (F.Y.); (J.K.); (A.L.); (P.S.); (A.V.); (M.T.); (N.D.); (K.G.); (N.G.); (M.K.); (N.M.); (E.K.); (A.T.); (J.-C.G.); (I.E.); (C.A.)
| | - Natalia Degtyareva
- Department of Obstetrics, Gynecology and Perinatal Medicine, N. F. Filatov Clinical Institute of Children’s Health, I. M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya Str. 8-2, 119991 Moscow, Russia; (F.Y.); (J.K.); (A.L.); (P.S.); (A.V.); (M.T.); (N.D.); (K.G.); (N.G.); (M.K.); (N.M.); (E.K.); (A.T.); (J.-C.G.); (I.E.); (C.A.)
| | - Kristina Grigoreva
- Department of Obstetrics, Gynecology and Perinatal Medicine, N. F. Filatov Clinical Institute of Children’s Health, I. M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya Str. 8-2, 119991 Moscow, Russia; (F.Y.); (J.K.); (A.L.); (P.S.); (A.V.); (M.T.); (N.D.); (K.G.); (N.G.); (M.K.); (N.M.); (E.K.); (A.T.); (J.-C.G.); (I.E.); (C.A.)
| | - Nilufar Gashimova
- Department of Obstetrics, Gynecology and Perinatal Medicine, N. F. Filatov Clinical Institute of Children’s Health, I. M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya Str. 8-2, 119991 Moscow, Russia; (F.Y.); (J.K.); (A.L.); (P.S.); (A.V.); (M.T.); (N.D.); (K.G.); (N.G.); (M.K.); (N.M.); (E.K.); (A.T.); (J.-C.G.); (I.E.); (C.A.)
| | - Margaret Kvaratskheliia
- Department of Obstetrics, Gynecology and Perinatal Medicine, N. F. Filatov Clinical Institute of Children’s Health, I. M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya Str. 8-2, 119991 Moscow, Russia; (F.Y.); (J.K.); (A.L.); (P.S.); (A.V.); (M.T.); (N.D.); (K.G.); (N.G.); (M.K.); (N.M.); (E.K.); (A.T.); (J.-C.G.); (I.E.); (C.A.)
| | - Nataliya Makatsariya
- Department of Obstetrics, Gynecology and Perinatal Medicine, N. F. Filatov Clinical Institute of Children’s Health, I. M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya Str. 8-2, 119991 Moscow, Russia; (F.Y.); (J.K.); (A.L.); (P.S.); (A.V.); (M.T.); (N.D.); (K.G.); (N.G.); (M.K.); (N.M.); (E.K.); (A.T.); (J.-C.G.); (I.E.); (C.A.)
| | - Ekaterina Kudryavtseva
- Department of Obstetrics, Gynecology and Perinatal Medicine, N. F. Filatov Clinical Institute of Children’s Health, I. M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya Str. 8-2, 119991 Moscow, Russia; (F.Y.); (J.K.); (A.L.); (P.S.); (A.V.); (M.T.); (N.D.); (K.G.); (N.G.); (M.K.); (N.M.); (E.K.); (A.T.); (J.-C.G.); (I.E.); (C.A.)
| | - Anna Tomlenova
- Department of Obstetrics, Gynecology and Perinatal Medicine, N. F. Filatov Clinical Institute of Children’s Health, I. M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya Str. 8-2, 119991 Moscow, Russia; (F.Y.); (J.K.); (A.L.); (P.S.); (A.V.); (M.T.); (N.D.); (K.G.); (N.G.); (M.K.); (N.M.); (E.K.); (A.T.); (J.-C.G.); (I.E.); (C.A.)
| | - Jean-Christophe Gris
- Department of Obstetrics, Gynecology and Perinatal Medicine, N. F. Filatov Clinical Institute of Children’s Health, I. M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya Str. 8-2, 119991 Moscow, Russia; (F.Y.); (J.K.); (A.L.); (P.S.); (A.V.); (M.T.); (N.D.); (K.G.); (N.G.); (M.K.); (N.M.); (E.K.); (A.T.); (J.-C.G.); (I.E.); (C.A.)
- Faculty of Pharmaceutical and Biological Sciences, Montpellier University, 34093 Montpellier, France
| | - Ismail Elalamy
- Department of Obstetrics, Gynecology and Perinatal Medicine, N. F. Filatov Clinical Institute of Children’s Health, I. M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya Str. 8-2, 119991 Moscow, Russia; (F.Y.); (J.K.); (A.L.); (P.S.); (A.V.); (M.T.); (N.D.); (K.G.); (N.G.); (M.K.); (N.M.); (E.K.); (A.T.); (J.-C.G.); (I.E.); (C.A.)
- Department Hematology and Thrombosis Center, Medicine Sorbonne University, 75012 Paris, France
- Hospital Tenon, 4 Rue de la Chine, 75020 Paris, France
| | - Cihan Ay
- Department of Obstetrics, Gynecology and Perinatal Medicine, N. F. Filatov Clinical Institute of Children’s Health, I. M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya Str. 8-2, 119991 Moscow, Russia; (F.Y.); (J.K.); (A.L.); (P.S.); (A.V.); (M.T.); (N.D.); (K.G.); (N.G.); (M.K.); (N.M.); (E.K.); (A.T.); (J.-C.G.); (I.E.); (C.A.)
- Department of Medicine I, Clinical Division of Hematology and Hemostaseology, Medical University of Vienna, 1080 Vienna, Austria
| | - Alexander Makatsariya
- Department of Obstetrics, Gynecology and Perinatal Medicine, N. F. Filatov Clinical Institute of Children’s Health, I. M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya Str. 8-2, 119991 Moscow, Russia; (F.Y.); (J.K.); (A.L.); (P.S.); (A.V.); (M.T.); (N.D.); (K.G.); (N.G.); (M.K.); (N.M.); (E.K.); (A.T.); (J.-C.G.); (I.E.); (C.A.)
| |
Collapse
|
7
|
Riitano G, Capozzi A, Recalchi S, Augusto M, Conti F, Misasi R, Garofalo T, Sorice M, Manganelli V. Role of Lipid Rafts on LRP8 Signaling Triggered by Anti-β2-GPI Antibodies in Endothelial Cells. Biomedicines 2023; 11:3135. [PMID: 38137358 PMCID: PMC10740635 DOI: 10.3390/biomedicines11123135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 11/16/2023] [Accepted: 11/23/2023] [Indexed: 12/24/2023] Open
Abstract
Antiphospholipid antibody syndrome is an autoimmune disease characterized by thrombosis and/or pregnancy morbidity in association with circulating antiphospholipid antibodies, mainly anti-β2 glycoprotein 1 antibodies (anti-β2-GPI antibodies). Previous studies demonstrated that the signaling pathway may involve lipid rafts, plasma membrane microdomains enriched in glycosphingolipid and cholesterol. In this study, we analyzed the signaling pathway of LRP8/ApoER2, a putative receptor of anti-β2-GPI antibodies, through lipid rafts in human endothelial cells. LRP8, Dab2 and endothelial nitric oxide synthase (e-NOS) phosphorylation were evaluated using Western blot, Nitric Oxide (NO) production with cytofluorimetric analysis, LRP8 enrichment in lipid rafts via sucrose gradient fractionation, and scanning confocal microscopy analysis of its association with ganglioside GM1 was also conducted. The analyses demonstrated that affinity-purified anti-β2-GPI antibodies induced LRP8 and Dab-2 phosphorylation, together with a significant decrease in e-NOS phosphorylation, with consequent decrease in NO intracellular production. These effects were almost completely prevented by Methyl-β-cyclodextrin (MβCD), indicating the involvement of lipid rafts. It was supported with the observation of LRP8 enrichment in lipid raft fractions and its association with ganglioside GM1, detected with scanning confocal microscopy. These findings demonstrate that LRP8 signaling triggered by anti-β2-GPI antibodies in endothelial cells occurs through lipid rafts. It represents a new task for valuable therapeutic approaches, such as raft-targeted therapy, including cyclodextrins and statins.
Collapse
Affiliation(s)
- Gloria Riitano
- Department of Experimental Medicine, “Sapienza” University of Rome, 00161 Rome, Italy; (G.R.); (A.C.); (S.R.); (R.M.); (T.G.); (V.M.)
| | - Antonella Capozzi
- Department of Experimental Medicine, “Sapienza” University of Rome, 00161 Rome, Italy; (G.R.); (A.C.); (S.R.); (R.M.); (T.G.); (V.M.)
| | - Serena Recalchi
- Department of Experimental Medicine, “Sapienza” University of Rome, 00161 Rome, Italy; (G.R.); (A.C.); (S.R.); (R.M.); (T.G.); (V.M.)
| | | | - Fabrizio Conti
- Rheumatology Unit, Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, “Sapienza” University of Rome, 00161 Rome, Italy;
| | - Roberta Misasi
- Department of Experimental Medicine, “Sapienza” University of Rome, 00161 Rome, Italy; (G.R.); (A.C.); (S.R.); (R.M.); (T.G.); (V.M.)
| | - Tina Garofalo
- Department of Experimental Medicine, “Sapienza” University of Rome, 00161 Rome, Italy; (G.R.); (A.C.); (S.R.); (R.M.); (T.G.); (V.M.)
| | - Maurizio Sorice
- Department of Experimental Medicine, “Sapienza” University of Rome, 00161 Rome, Italy; (G.R.); (A.C.); (S.R.); (R.M.); (T.G.); (V.M.)
| | - Valeria Manganelli
- Department of Experimental Medicine, “Sapienza” University of Rome, 00161 Rome, Italy; (G.R.); (A.C.); (S.R.); (R.M.); (T.G.); (V.M.)
| |
Collapse
|
8
|
Qin R, Wu H, Guan H, Tang C, Zheng Z, Deng C, Chen C, Zou Q, Lu L, Ma K. Anti-phospholipid autoantibodies in human diseases. Clin Immunol 2023; 256:109803. [PMID: 37821073 DOI: 10.1016/j.clim.2023.109803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 09/24/2023] [Accepted: 10/05/2023] [Indexed: 10/13/2023]
Abstract
Anti-phospholipid autoantibodies are a group of antibodies that can specifically bind to anionic phospholipids and phospholipid protein complexes. Recent studies have reported elevated serum anti-phospholipid autoantibody levels in patients with antiphospholipid syndrome, systemic lupus erythematosus, rheumatoid arthritis, metabolic disorders, malaria, SARS-CoV-2 infection, obstetric diseases and cardiovascular diseases. However, the underlying mechanisms of anti-phospholipid autoantibodies in disease pathogenesis remain largely unclear. Emerging evidence indicate that anti-phospholipid autoantibodies modulate NETs formation, monocyte activation, blockade of apoptotic cell phagocytosis in macrophages, complement activation, dendritic cell activation and vascular endothelial cell activation. Herein, we provide an update on recent advances in elucidating the effector mechanisms of anti-phospholipid autoantibodies in the pathogenesis of various diseases, which may facilitate the development of potential therapeutic targets for the treatment of anti-phospholipid autoantibody-related disorders.
Collapse
Affiliation(s)
- Rencai Qin
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, Centre for Infection and Immunity Studies (CIIS), School of Medicine, The Seventh Affiliated Hospital, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, Guangdong 518107, China
| | - Haiqi Wu
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, Centre for Infection and Immunity Studies (CIIS), School of Medicine, The Seventh Affiliated Hospital, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, Guangdong 518107, China
| | - Hui Guan
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, Centre for Infection and Immunity Studies (CIIS), School of Medicine, The Seventh Affiliated Hospital, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, Guangdong 518107, China
| | - Chun Tang
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, Centre for Infection and Immunity Studies (CIIS), School of Medicine, The Seventh Affiliated Hospital, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, Guangdong 518107, China
| | - Zhihua Zheng
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, Centre for Infection and Immunity Studies (CIIS), School of Medicine, The Seventh Affiliated Hospital, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, Guangdong 518107, China
| | - Chong Deng
- Department of Pathology and Shenzhen Institute of Research and Innovation, The University of Hong Kong, Hong Kong 999077, China
| | - Chengshun Chen
- Department of Rheumatology and Immunology, First Affiliated Hospital of Army Medical University, Chongqing, China
| | - Qinghua Zou
- Department of Rheumatology and Immunology, First Affiliated Hospital of Army Medical University, Chongqing, China.
| | - Liwei Lu
- Department of Pathology and Shenzhen Institute of Research and Innovation, The University of Hong Kong, Hong Kong 999077, China.
| | - Kongyang Ma
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, Centre for Infection and Immunity Studies (CIIS), School of Medicine, The Seventh Affiliated Hospital, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, Guangdong 518107, China.
| |
Collapse
|
9
|
Mineo C, Shaul PW, Bermas BL. The pathogenesis of obstetric APS: a 2023 update. Clin Immunol 2023; 255:109745. [PMID: 37625670 PMCID: PMC11366079 DOI: 10.1016/j.clim.2023.109745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/16/2023] [Accepted: 08/18/2023] [Indexed: 08/27/2023]
Abstract
The antiphospholipid syndrome (APS) is an autoimmune disease characterized by the persistent presence of antibodies directed against phospholipids and phospholipid-binding proteins that are associated with thrombosis and pregnancy-related morbidity. The latter includes fetal deaths, premature birth and maternal complications. In the early 1990s, a distinct set of autoantibodies, termed collectively antiphospholipid antibodies (aPL), were identified as the causative agents of this disorder. Subsequently histological analyses of the placenta from APS pregnancies revealed various abnormalities, including inflammation at maternal-fetal interface and poor placentation manifested by reduced trophoblast invasion and limited uterine spiral artery remodeling. Further preclinical investigations identified the molecular targets of aPL and the downstream intracellular pathways of key placental cell types. While these discoveries suggest potential therapeutics for this disorder, definitive clinical trials have not been completed. This concise review focuses on the recent developments in the field of basic and translational research pursuing novel mechanisms underlying obstetric APS.
Collapse
Affiliation(s)
- Chieko Mineo
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, UT Southwestern Medical Center, Dallas, TX, United States.
| | - Philip W Shaul
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, UT Southwestern Medical Center, Dallas, TX, United States
| | - Bonnie L Bermas
- Division of Rheumatic Diseases, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
10
|
Yu H, Zaveri S, Sattar Z, Schaible M, Perez Gandara B, Uddin A, McGarvey LR, Ohlmeyer M, Geraghty P. Protein Phosphatase 2A as a Therapeutic Target in Pulmonary Diseases. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1552. [PMID: 37763671 PMCID: PMC10535831 DOI: 10.3390/medicina59091552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 08/22/2023] [Accepted: 08/24/2023] [Indexed: 09/29/2023]
Abstract
New disease targets and medicinal chemistry approaches are urgently needed to develop novel therapeutic strategies for treating pulmonary diseases. Emerging evidence suggests that reduced activity of protein phosphatase 2A (PP2A), a complex heterotrimeric enzyme that regulates dephosphorylation of serine and threonine residues from many proteins, is observed in multiple pulmonary diseases, including lung cancer, smoke-induced chronic obstructive pulmonary disease, alpha-1 antitrypsin deficiency, asthma, and idiopathic pulmonary fibrosis. Loss of PP2A responses is linked to many mechanisms associated with disease progressions, such as senescence, proliferation, inflammation, corticosteroid resistance, enhanced protease responses, and mRNA stability. Therefore, chemical restoration of PP2A may represent a novel treatment for these diseases. This review outlines the potential impact of reduced PP2A activity in pulmonary diseases, endogenous and exogenous inhibitors of PP2A, details the possible PP2A-dependent mechanisms observed in these conditions, and outlines potential therapeutic strategies for treatment. Substantial medicinal chemistry efforts are underway to develop therapeutics targeting PP2A activity. The development of specific activators of PP2A that selectively target PP2A holoenzymes could improve our understanding of the function of PP2A in pulmonary diseases. This may lead to the development of therapeutics for restoring normal PP2A responses within the lung.
Collapse
Affiliation(s)
- Howard Yu
- Department of Medicine, State University of New York Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203, USA; (H.Y.); (S.Z.); (Z.S.); (M.S.); (B.P.G.); (A.U.); (L.R.M.)
| | - Sahil Zaveri
- Department of Medicine, State University of New York Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203, USA; (H.Y.); (S.Z.); (Z.S.); (M.S.); (B.P.G.); (A.U.); (L.R.M.)
| | - Zeeshan Sattar
- Department of Medicine, State University of New York Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203, USA; (H.Y.); (S.Z.); (Z.S.); (M.S.); (B.P.G.); (A.U.); (L.R.M.)
| | - Michael Schaible
- Department of Medicine, State University of New York Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203, USA; (H.Y.); (S.Z.); (Z.S.); (M.S.); (B.P.G.); (A.U.); (L.R.M.)
| | - Brais Perez Gandara
- Department of Medicine, State University of New York Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203, USA; (H.Y.); (S.Z.); (Z.S.); (M.S.); (B.P.G.); (A.U.); (L.R.M.)
| | - Anwar Uddin
- Department of Medicine, State University of New York Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203, USA; (H.Y.); (S.Z.); (Z.S.); (M.S.); (B.P.G.); (A.U.); (L.R.M.)
| | - Lucas R. McGarvey
- Department of Medicine, State University of New York Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203, USA; (H.Y.); (S.Z.); (Z.S.); (M.S.); (B.P.G.); (A.U.); (L.R.M.)
| | | | - Patrick Geraghty
- Department of Medicine, State University of New York Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203, USA; (H.Y.); (S.Z.); (Z.S.); (M.S.); (B.P.G.); (A.U.); (L.R.M.)
| |
Collapse
|
11
|
Sacharidou A, Chambliss K, Peng J, Barrera J, Tanigaki K, Luby-Phelps K, Özdemir İ, Khan S, Sirsi SR, Kim SH, Katzenellenbogen BS, Katzenellenbogen JA, Kanchwala M, Sathe AA, Lemoff A, Xing C, Hoyt K, Mineo C, Shaul PW. Endothelial ERα promotes glucose tolerance by enhancing endothelial insulin transport to skeletal muscle. Nat Commun 2023; 14:4989. [PMID: 37591837 PMCID: PMC10435471 DOI: 10.1038/s41467-023-40562-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 08/01/2023] [Indexed: 08/19/2023] Open
Abstract
The estrogen receptor (ER) designated ERα has actions in many cell and tissue types that impact glucose homeostasis. It is unknown if these include mechanisms in endothelial cells, which have the potential to influence relative obesity, and processes in adipose tissue and skeletal muscle that impact glucose control. Here we show that independent of impact on events in adipose tissue, endothelial ERα promotes glucose tolerance by enhancing endothelial insulin transport to skeletal muscle. Endothelial ERα-deficient male mice are glucose intolerant and insulin resistant, and in females the antidiabetogenic actions of estradiol (E2) are absent. The glucose dysregulation is due to impaired skeletal muscle glucose disposal that results from attenuated muscle insulin delivery. Endothelial ERα activation stimulates insulin transcytosis by skeletal muscle microvascular endothelial cells. Mechanistically this involves nuclear ERα-dependent upregulation of vesicular trafficking regulator sorting nexin 5 (SNX5) expression, and PI3 kinase activation that drives plasma membrane recruitment of SNX5. Thus, coupled nuclear and non-nuclear actions of ERα promote endothelial insulin transport to skeletal muscle to foster normal glucose homeostasis.
Collapse
Affiliation(s)
- Anastasia Sacharidou
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Ken Chambliss
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Jun Peng
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Jose Barrera
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Keiji Tanigaki
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Katherine Luby-Phelps
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - İpek Özdemir
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Sohaib Khan
- University of Cincinnati Cancer Institute, Department of Cancer and Cell Biology, University of Cincinnati College of Medicine, Cincinnati, OH, 45219, USA
| | - Shashank R Sirsi
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Sung Hoon Kim
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Benita S Katzenellenbogen
- Departments of Physiology and Cell Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | | | - Mohammed Kanchwala
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Adwait A Sathe
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Andrew Lemoff
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Chao Xing
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Kenneth Hoyt
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Chieko Mineo
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| | - Philip W Shaul
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
12
|
Granada-Gómez M, Velásquez-Berrío M, Molina CR, Martín SS, Escudero C, Alvarez AM, Cadavid AP. Modulation of the activation of endothelial nitric oxide synthase and nitrosative stress biomarkers by aspirin triggered lipoxins: A possible mechanism of action of aspirin in the antiphospholipid syndrome. Am J Reprod Immunol 2023; 90:e13753. [PMID: 37491919 DOI: 10.1111/aji.13753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 06/20/2023] [Accepted: 07/04/2023] [Indexed: 07/27/2023] Open
Abstract
PROBLEM Antiphospholipid syndrome (APS) is characterized by the clinical manifestation of vascular thrombosis (VT) or pregnancy morbidity (PM) and antiphospholipid antibodies (aPL) that can modify the nitric oxide production. Low-dose aspirin is used in the prevention and treatment of diverse alterations of pregnancy. One of the mechanisms of action of aspirin is to induce the production of aspirin-triggered-lipoxins (ATL). The aim of this study was to evaluate the modulatory effect of ATL over the activation of endothelial nitric oxide synthase (eNOS) and nitrosative stress biomarkers induced by aPL. METHODS We used polyclonal IgG and sera from women with aPL and PM/VT or VT only, and from women with PM only and positive for non-criteria aPL (SN-OAPS). In these sera, biomarkers of nitrosative stress (nitrites and nitrotyrosine) were measured. The protein expression of nitrotyrosine and the phosphorylation of eNOS (at Ser1177) were estimated in human umbilical vein endothelial cells (HUVECs) stimulated with polyclonal IgG with or without ATL. RESULTS Women with SN-OAPS showed increased circulating levels of nitrites and nitrotyrosine. Likewise, polyclonal IgG from either SN-OAPS or VT patients stimulated nitrotyrosine expression in HUVECs. ATL decreased the nitrotyrosine expression induced by polyclonal IgG from the SN-OAPS group. ATL also recovered the reduced eNOS phosphorylation at Ser1177 in HUVECs stimulated with polyclonal IgG from women with PM/VT or SN-OAPS. CONCLUSIONS Increased nitrosative stress present in serum of women with SN-OAPS is associated with IgG-mediated impaired endothelial NO synthesis in endothelial cells. ATL prevent these cellular changes.
Collapse
Affiliation(s)
- Manuel Granada-Gómez
- Grupo Reproducción, Facultad de Medicina, Dpto. Microbiología y Parasitología, Universidad de Antioquia UdeA, Medellín, Colombia
| | - Manuela Velásquez-Berrío
- Grupo Reproducción, Facultad de Medicina, Dpto. Microbiología y Parasitología, Universidad de Antioquia UdeA, Medellín, Colombia
| | - Carolina Rúa Molina
- Grupo de Investigación en Trombosis, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, Colombia
| | - Sebastián San Martín
- Biomedical Research Center School of Medicine, Universidad de Valparaiso, Valparaiso, Chile
- Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillan, Chile
| | - Carlos Escudero
- Vascular Physiology Laboratory, Basic Sciences Department, Faculty of Sciences, Universidad del Bio-Bio, Chillán, Chile
- Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillan, Chile
- Red Iberoamericana de Alteraciones Vasculares Asociadas a TRanstornos del EMbarazo (RIVATREM)
| | - Angela M Alvarez
- Grupo Reproducción, Facultad de Medicina, Dpto. Microbiología y Parasitología, Universidad de Antioquia UdeA, Medellín, Colombia
| | - Angela P Cadavid
- Grupo Reproducción, Facultad de Medicina, Dpto. Microbiología y Parasitología, Universidad de Antioquia UdeA, Medellín, Colombia
- Grupo de Investigación en Trombosis, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, Colombia
- Red Iberoamericana de Alteraciones Vasculares Asociadas a TRanstornos del EMbarazo (RIVATREM)
| |
Collapse
|
13
|
Patsouras M, Alexopoulou E, Foutadakis S, Tsiki E, Karagianni P, Agelopoulos M, Vlachoyiannopoulos PG. Antiphospholipid antibodies induce proinflammatory and procoagulant pathways in endothelial cells. J Transl Autoimmun 2023; 6:100202. [PMID: 37216142 PMCID: PMC10197110 DOI: 10.1016/j.jtauto.2023.100202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 03/01/2023] [Accepted: 04/30/2023] [Indexed: 05/24/2023] Open
Abstract
Antiphospholipid syndrome (APS) is an autoimmune thrombophilia characterized by recurrent thrombotic events and/or pregnancy morbidity in the presence of antiphospholipid antibodies detected either as anti-cardiolipin, anti-β2 Glycoprotein I (anti-β2GPI) or Lupus anticoagulant (LA). Endothelial deregulation characterizes the syndrome. To address gene expression changes accompanying the development of autoimmune phenotype in endothelial cells in the context of APS, we performed transcriptomics analysis in Human Umbilical Vein Endothelial Cells (HUVECs) stimulated with IgG from APS patients and β2GPI, followed by intersection of RNA-seq data with published microarray and ChIP-seq results (Chromatin Immunoprecipitation). Our strategy revealed that during HUVEC activation diverse signaling pathways such as TNF-α, TGF-β, MAPK38, and Hippo are triggered as indicated by Gene Ontology (GO) classification and pathway analysis. Finally, cell biology approaches performed side-by-side in naïve and stimulated cultured HUVECs, as well as, in placenta specimens derived from Healthy donors (HDs) and APS-patients verified the evolution of an APS-characteristic gene expression program in endothelial cells during the initial stages of the disease's development.
Collapse
Affiliation(s)
- Markos Patsouras
- Department of Pathophysiology, Medical School, National and Kapodistrian University of Athens, Greece
| | - Eirini Alexopoulou
- Department of Pathophysiology, Medical School, National and Kapodistrian University of Athens, Greece
| | - Spyros Foutadakis
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, 4 Soranou Ephessiou Street, Athens, 11527, Greece
| | - Eirini Tsiki
- Department of Pathophysiology, Medical School, National and Kapodistrian University of Athens, Greece
| | - Panagiota Karagianni
- Department of Pathophysiology, Medical School, National and Kapodistrian University of Athens, Greece
| | - Marios Agelopoulos
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, 4 Soranou Ephessiou Street, Athens, 11527, Greece
| | | |
Collapse
|
14
|
El Hasbani G, Saliba AN, Uthman I, Taher AT. Hematological manifestations of antiphospholipid syndrome: Going beyond thrombosis. Blood Rev 2023; 58:101015. [PMID: 36175215 DOI: 10.1016/j.blre.2022.101015] [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: 08/17/2022] [Revised: 09/09/2022] [Accepted: 09/12/2022] [Indexed: 11/30/2022]
Abstract
Thrombotic complications are a hallmark of antiphospholipid syndrome (APS). These vascular - arterial, venous, and/or small vessel - complications are well described and known to hematologists and healthcare providers caring for patients with this disease. In this review, we shed light on other hematological manifestations of the disease, including bleeding, thrombocytopenia, autoimmune hemolytic anemia, and thrombotic microangiopathy syndromes. While these manifestations are not bona fide clinical criteria for the diagnosis of APS, they frequently interact and contribute to the complexity of clinical management of APS.
Collapse
Affiliation(s)
- Georges El Hasbani
- Department of Internal Medicine, Hartford Healthcare, St. Vincent's Medical Center, Bridgeport, CT 06606, USA
| | - Antoine N Saliba
- Division of Hematology, Department of Medicine, Mayo Clinic, 200 First Street Southwest, Rochester, MN 55905, USA
| | - Imad Uthman
- Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Ali T Taher
- Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon..
| |
Collapse
|
15
|
Abstract
Antiphospholipid syndrome (APS) is a thrombo-inflammatory disease propelled by circulating autoantibodies that recognize cell surface phospholipids and phospholipid binding proteins. The result is an increased risk of thrombotic events, pregnancy morbidity, and various other autoimmune and inflammatory complications. Although antiphospholipid syndrome was first recognized in patients with lupus, the stand alone presentation of antiphospholipid syndrome is at least equally common. Overall, the diagnosis appears to affect at least one in 2000 people. Studies of antiphospholipid syndrome pathogenesis have long focused on logical candidates such as coagulation factors, endothelial cells, and platelets. Recent work has shed light on additional potential therapeutic targets within the innate immune system, including the complement system and neutrophil extracellular traps. Vitamin K antagonists remain the mainstay of treatment for most patients with thrombotic antiphospholipid syndrome and, based on current data, appear superior to the more targeted direct oral anticoagulants. The potential role of immunomodulatory treatments in antiphospholipid syndrome management is receiving increased attention. As for many systemic autoimmune diseases, the most important future direction is to more precisely identify mechanistic drivers of disease heterogeneity in pursuit of unlocking personalized and proactive treatments for patients.
Collapse
Affiliation(s)
- Jason S Knight
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - D Ware Branch
- James R. and Jo Scott Research Chair, Department of Obstetrics and Gynecology, University of Utah Health and Intermountain Healthcare, Salt Lake City, Utah, USA
| | - Thomas L Ortel
- Division of Hematology, Departments of Medicine and Pathology, Duke University, Durham, North Carolina, USA
| |
Collapse
|
16
|
Foret T, Dufrost V, Heymonet M, Risse J, Faure GC, Louis H, Lagrange J, Lacolley P, Devreese K, Gibot S, Regnault V, Zuily S, Wahl D. Circulating Endothelial Cells are Associated with Thromboembolic Events in Patients with Antiphospholipid Antibodies. Thromb Haemost 2023; 123:76-84. [PMID: 35977699 DOI: 10.1055/a-1926-0453] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
BACKGROUND Endothelial damage has been described in antiphospholipid antibody (aPL)-positive patients. However, it is uncertain whether circulating endothelial cells (CECs)-which are released when endothelial injury occurs-can be a marker of patients at high risk for thrombosis. METHODS Ninety-seven patients with aPL and/or systemic lupus erythematosus (SLE) were included. CECs were determined by an automated CellSearch system. We also assayed plasma levels of tissue factor-bearing extracellular vesicles (TF+/EVs) and soluble triggering receptor expressed on myeloid cells 1 (sTREM-1) as markers of endothelial dysfunction/damage. RESULTS Patients' mean age was 46.1 ± 13.9 years, 77 were women. Thirty-seven had SLE and 75 patients were suffering from antiphospholipid syndrome. Thirty-seven percent of patients presented a medical history of arterial thrombosis and 46% a history of venous thromboembolism (VTE). Thirteen patients had increased levels of CECs (>20/mL), with a mean CEC level of 48.3 ± 21.3 per mL. In univariate analysis, patients with obesity or medical history of myocardial infarction (MI), VTE, or nephropathy had a significant increased CEC level. In multivariate analysis, obesity (odds ratio [OR] = 6.07, 95% confidence interval [CI]: 1.42-25.94), VTE (OR = 7.59 [95% CI: 1.38-41.66]), and MI (OR = 5.5 [95% CI: 1.1-26.6)] were independently and significantly associated with elevated CECs. We also identified significant correlations between CECs and other markers of endothelial dysfunction: sTREM-1 and TF+/EVs. CONCLUSION This study demonstrated that endothelial injury assessed by the levels of CECs was associated with thromboembolic events in patients with aPL and/or autoimmune diseases.
Collapse
Affiliation(s)
- Thomas Foret
- INSERM, DCAC, Université de Lorraine, Nancy, France.,Division of Vascular Medicine, CHRU-Nancy, Regional Competence Center for Rare Vascular and Systemic Autoimmune Diseases, Nancy, France
| | - Virginie Dufrost
- INSERM, DCAC, Université de Lorraine, Nancy, France.,Division of Vascular Medicine, CHRU-Nancy, Regional Competence Center for Rare Vascular and Systemic Autoimmune Diseases, Nancy, France
| | | | - Jessie Risse
- INSERM, DCAC, Université de Lorraine, Nancy, France.,Division of Vascular Medicine, CHRU-Nancy, Regional Competence Center for Rare Vascular and Systemic Autoimmune Diseases, Nancy, France.,CH de Sarreguemines, Sarreguemines, France
| | - Gilbert C Faure
- Laboratory of Immunology, CHRU-Nancy, Nancytomique, Pôle Laboratoire.,CRAN UMR CNRS 7039, Nancy, France
| | | | - Jeremy Lagrange
- INSERM, DCAC, Université de Lorraine, Nancy, France.,Délégation à la Recherche Clinique et à l'Innovation, CHRU-Nancy, Nancy, France
| | - Patrick Lacolley
- INSERM, DCAC, Université de Lorraine, Nancy, France.,Délégation à la Recherche Clinique et à l'Innovation, CHRU-Nancy, Nancy, France
| | - Katrien Devreese
- Department of Diagnostic Sciences, Coagulation Laboratory, Ghent University Hospital, Ghent University, Ghent, Belgium
| | | | - Veronique Regnault
- INSERM, DCAC, Université de Lorraine, Nancy, France.,Délégation à la Recherche Clinique et à l'Innovation, CHRU-Nancy, Nancy, France
| | - Stéphane Zuily
- INSERM, DCAC, Université de Lorraine, Nancy, France.,Division of Vascular Medicine, CHRU-Nancy, Regional Competence Center for Rare Vascular and Systemic Autoimmune Diseases, Nancy, France
| | - Denis Wahl
- INSERM, DCAC, Université de Lorraine, Nancy, France.,Division of Vascular Medicine, CHRU-Nancy, Regional Competence Center for Rare Vascular and Systemic Autoimmune Diseases, Nancy, France
| |
Collapse
|
17
|
Butt A, Erkan D, Lee AI. COVID-19 and antiphospholipid antibodies. Best Pract Res Clin Haematol 2022; 35:101402. [PMID: 36494152 PMCID: PMC9568270 DOI: 10.1016/j.beha.2022.101402] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 10/07/2022] [Accepted: 10/11/2022] [Indexed: 12/14/2022]
Abstract
Antiphospholipid syndrome and the coagulopathy of COVID-19 share many pathophysiologic features, including endotheliopathy, hypercoagulability, and activation of platelets, complement pathways, and neutrophil extracellular traps, all acting in concert via a model of immunothrombosis. Antiphospholipid antibody production in COVID-19 is common, with 50% of COVID-19 patients being positive for lupus anticoagulant in some studies, and with non-Sapporo criteria antiphospholipid antibodies being prevalent as well. The biological significance of antiphospholipid antibodies in COVID-19 is uncertain, as such antibodies are usually transient, and studies examining clinical outcomes in COVID-19 patients with and without antiphospholipid antibodies have yielded conflicting results. In this review, we explore the biology of antiphospholipid antibodies in COVID-19 and other infections and discuss mechanisms of thrombogenesis in antiphospholipid syndrome and parallels with COVID-19 coagulopathy. In addition, we review the existing literature on safety of COVID-19 vaccination in patients with antiphospholipid antibodies and antiphospholipid syndrome.
Collapse
Affiliation(s)
- Ayesha Butt
- Section of Hematology, Department of Medicine, Yale School of Medicine, 333 Cedar St., New Haven, CT, 06520, USA.
| | - Doruk Erkan
- Barbara Volcker Center for Women and Rheumatic Diseases, Hospital for Special Surgery and Weill Cornell Medicine, 535 E. 70th St., 6th floor, New York, NY, 10021, USA.
| | - Alfred Ian Lee
- Section of Hematology, Department of Medicine, Yale School of Medicine, 333 Cedar St., New Haven, CT, 06520, USA.
| |
Collapse
|
18
|
Leonardi GR, Lescano CH, Costa JL, Mazetto B, Orsi FA, Monica FZ. Adenosine diphosphate-induced aggregation is enhanced in platelets obtained from patients with thrombotic primary antiphospholipid syndrome (t-PAPS): Role of P2Y 12 -cAMP signaling pathway. J Thromb Haemost 2022; 20:1699-1711. [PMID: 35395698 DOI: 10.1111/jth.15724] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 03/22/2022] [Accepted: 04/04/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND Thrombotic antiphospholipid syndrome (t-PAPS) is characterized by arterial, venous, or microvascular occlusions, which are explained, in part, by the presence of antiphospholipid (aPL) antibodies. Although there is much evidence indicating that isolated aPL antibodies increase the activity of platelets obtained from healthy volunteers, platelet function in t-PAPS has not been as widely studied. OBJECTIVE To evaluate platelet reactivity in t-PAPS patients. METHODS Platelet aggregation, protein expression, and cyclic nucleotide levels were carried out in platelet rich plasma (PRP) or washed platelets (WPs) obtained from t-PAPS or healthy volunteers. RESULTS ADP-induced aggregation was significantly higher in PRP obtained from t-PAPS than obtained from the control. The protein expression of P2Y12 receptor and Gs alpha was significantly higher and lower, respectively in WPs from t-PAPS patients. In PRP incubated with iloprost or sodium nitroprusside, the residual platelet reactivity induced by ADP was still higher in PRP from t-PAPS than from the control. Lower intracellular levels of cyclic guanosine monophosphate (cGMP) and cyclic adenosine monophosphate (cAMP) were observed in unstimulated PRP from t-PAPS patients. The protein expression of soluble guanylate cyclase subunits and phosphodiesterases types 3 and 5 did not differ. The antiplatelet activity of ticagrelor was similar between the groups and cilostazol significantly potentiated this response. Isolated aPL antibodies obtained from t-PAPS patients potentiated ADP-induced aggregation in healthy platelets but did not affect the inhibitory responses induced by iloprost or sodium nitroprusside. CONCLUSIONS The overexpression of P2Y12 receptor, accompanied by lower levels of cAMP and cGMP levels produced greater amplitude of ADP aggregation in platelets from t-PAPS patients.
Collapse
Affiliation(s)
- Guilherme Ruiz Leonardi
- Department of Translation Medicine (Pharmacology), Faculty of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Caroline Honaiser Lescano
- Department of Translation Medicine (Pharmacology), Faculty of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Jose Luiz Costa
- Campinas Poison Control Center, University of Campinas, Campinas, Brazil
| | - Bruna Mazetto
- Hematology and Hemotherapy Center, University of Campinas, Campinas, Brazil
| | - Fernanda Andrade Orsi
- Hematology and Hemotherapy Center, University of Campinas, Campinas, Brazil
- Department of Clinical Pathology, Faculty of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Fabiola Zakia Monica
- Department of Translation Medicine (Pharmacology), Faculty of Medical Sciences, University of Campinas, Campinas, Brazil
| |
Collapse
|
19
|
Fang Z, Zhong M, Zhou L, Le Y, Wang H, Fang Z. Low-density lipoprotein receptor-related protein 8 facilitates the proliferation and invasion of non-small cell lung cancer cells by regulating the Wnt/β-catenin signaling pathway. Bioengineered 2022; 13:6807-6818. [PMID: 35246020 PMCID: PMC8974054 DOI: 10.1080/21655979.2022.2036917] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Low-density lipoprotein receptor-related protein 8 (LRP8) is involved in the development of multiple tumors, including lung cancer. However, the exact mechanism by which LRP8 exerts its oncogenic role in non-small cell lung cancer (NSCLC) remains elusive. Hence, in this study, we aimed to unravel the expression and role of LRP8 in the progression of NSCLC. We used online bioinformatics databases to identify the expression of LRP8 in multiple types of lung cancer. We validated LRP8 expression in NSCLC cell lines and tissues by Western blotting and immunohistochemistry. The functions of LRP8 in NSCLC carcinogenesis and progression were determined using in vitro and in vivo systems. The Wnt pathway activator LiCl was further used to validate the regulatory role of LRP8 in Wnt/β-catenin signaling. We demonstrated that LRP8 was markedly overexpressed in NSCLC tissues and cell lines, and its overexpression significantly correlated with poor clinicopathological characteristics and prognosis. Moreover, LRP8 depletion suppressed cell proliferation, migration, invasion, and epithelial-mesenchymal transition in vitro and impeded tumor growth in vivo. Mechanistically, LPR8 knockdown elicited tumor-suppressive functions by suppressing the Wnt/β-catenin pathway, which was partially reversed by LiCl. Hence, our study revealed that LRP8 facilitates NSCLC cell proliferation and invasion via the Wnt/β-catenin pathway, and thus LRP8 could be a novel therapeutic target for NSCLC.
Collapse
Affiliation(s)
- Zhi Fang
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China.,Department of Abdominal Oncology, Jiangxi Key Laboratory for Individualized Cancer Therapy, Nanchang, People's Republic of China
| | - Min Zhong
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China.,Department of Abdominal Oncology, Jiangxi Key Laboratory for Individualized Cancer Therapy, Nanchang, People's Republic of China
| | - Ling Zhou
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China.,Department of Abdominal Oncology, Jiangxi Key Laboratory for Individualized Cancer Therapy, Nanchang, People's Republic of China
| | - Yi Le
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China.,Department of Abdominal Oncology, Jiangxi Key Laboratory for Individualized Cancer Therapy, Nanchang, People's Republic of China
| | - Heng Wang
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| | - Ziling Fang
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China.,Department of Abdominal Oncology, Jiangxi Key Laboratory for Individualized Cancer Therapy, Nanchang, People's Republic of China
| |
Collapse
|
20
|
Tektonidou MG. Cardiovascular disease risk in antiphospholipid syndrome: Thrombo-inflammation and atherothrombosis. J Autoimmun 2022; 128:102813. [PMID: 35247655 DOI: 10.1016/j.jaut.2022.102813] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 02/23/2022] [Accepted: 02/27/2022] [Indexed: 12/11/2022]
Abstract
Antiphospholipid syndrome (APS) is an autoimmune disorder characterized by the presence of antiphospholipid antibodies (aPL) (lupus anticoagulant, anticardiolipin antibodies and anti-beta2glycoprotein I (anti-β2GPI) antibodies) and a plethora of macro- and micro-vascular manifestations, affecting predominantly young adults. Cardiovascular events are the leading causes of morbidity and mortality in APS. APL-mediated thrombo-inflammation and atherothrombosis are emerging pathogenetic mechanisms of cardiovascular disease (CVD) in APS, involving endothelial cell and monocyte activation, cytokines and adhesion molecules expression, complement and neutrophils activation, neutrophil extracellular traps formation, platelet cell activation and aggregation, and subsequent thrombin generation, in parallel with an oxidized low-density lipoprotein (oxLDL)-β2GPI complex induced macrophage differentiation to foam cells. High risk aPL profile, especially the presence of lupus anticoagulant and triple aPL positivity (all three aPL subtypes), co-existence with Systemic Lupus Erythematosus (SLE), as well as traditional risk factors such as smoking, hypertension, hyperlipemia and obesity are associated with both subclinical atherosclerosis and cardiovascular events in APS. Increased awareness of CVD risk by the physicians and patients, regular assessment and strict control of traditional risk factors, and lifestyle modifications are recommended. Use of low-dose aspirin should be considered for cardiovascular prevention in asymptomatic aPL carriers or SLE patients with high-risk aPL profile. The role of older agents such as hydroxychloroquine and statins or new potential targeted treatments against immuno- and athero-thrombosis has been demonstrated by experimental and some clinical studies and needs to be further evaluated by randomized controlled studies. This review summarizes the available evidence about the pathogenetic mechanisms and prevalence of cardiovascular events and subclinical atherosclerosis, the interrelationship between traditional and disease-related CVD risk factors, and the cardiovascular risk assessment and management in APS.
Collapse
Affiliation(s)
- Maria G Tektonidou
- First Department of Propaedeutic Internal Medicine, Joint Academic Rheumatology Program, National and Kapodistrian University of Athens, Medical School, Athens, Greece.
| |
Collapse
|
21
|
Knight JS, Kanthi Y. Mechanisms of immunothrombosis and vasculopathy in antiphospholipid syndrome. Semin Immunopathol 2022; 44:347-362. [PMID: 35122116 PMCID: PMC8816310 DOI: 10.1007/s00281-022-00916-w] [Citation(s) in RCA: 62] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 01/19/2022] [Indexed: 12/14/2022]
Abstract
Antiphospholipid syndrome (APS) is an autoimmune thrombophilia propelled by circulating antiphospholipid antibodies that herald vascular thrombosis and obstetrical complications. Antiphospholipid antibodies recognize phospholipids and phospholipid-binding proteins and are not only markers of disease but also key drivers of APS pathophysiology. Thrombotic events in APS can be attributed to various conspirators including activated endothelial cells, platelets, and myeloid-lineage cells, as well as derangements in coagulation and fibrinolytic systems. Furthermore, recent work has especially highlighted the role of neutrophil extracellular traps (NETs) and the complement system in APS thrombosis. Beyond acute thrombosis, patients with APS can also develop an occlusive vasculopathy, a long-term consequence of APS characterized by cell proliferation and infiltration that progressively expands the intima and leads to organ damage. This review will highlight known pathogenic factors in APS and will also briefly discuss similarities between APS and the thrombophilic coagulopathy of COVID-19.
Collapse
Affiliation(s)
- Jason S Knight
- Division of Rheumatology, University of Michigan, 1150 West Medical Center Drive, Ann Arbor, MI, 48109, USA.
| | - Yogendra Kanthi
- Division of Intramural Research National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| |
Collapse
|
22
|
Li R, Zhang C, Xie F, Zhou X, Hu X, Shi J, Du X, Lin Z, Dong N. Protein Phosphatase 2A Deficiency in Macrophages Increases Foam Cell Formation and Accelerates Atherosclerotic Lesion Development. Front Cardiovasc Med 2022; 8:745009. [PMID: 35118139 PMCID: PMC8803755 DOI: 10.3389/fcvm.2021.745009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 12/27/2021] [Indexed: 11/13/2022] Open
Abstract
Protein phosphatase 2A (PP2A), a crucial serine/threonine phosphatase, has recently been reported to play an important role in cardiovascular disease. Previous studies have hinted that PP2A is involved in atherosclerosis formation, but the associated mechanisms remain poorly understood. In this study, we investigate the role of PP2A in the pathogenesis of atherosclerosis. In human atherosclerotic coronary arteries, we found that the expression and activity of PP2A decreased significantly when compared to non-atherosclerotic arteries. Additional experiments demonstrated that pharmacological inhibition of PP2A aggravated atherosclerosis of ApoE−/− mice. Considering the central role of macrophages in atherosclerosis, mice with conditional knockout of the PP2A-Cα subunit in myeloid cells were produced to investigate the function of PP2A in macrophages. Results showed that PP2A deficiency in myeloid cells aggravated atherosclerotic lesions in mice. in vitro experiments indicated that PP2A-deficient macrophages had an enhanced ability of lipid uptake and foam cell formation. Mechanistically, the deficiency of the PP2A in macrophages led to an increase in the phosphorylation level of p38, which contributed to the elevated expression of scavenger receptor CD36, a key factor involved in lipoprotein uptake. Our data suggest that PP2A participates in the pathophysiological process of atherosclerosis. The decrease of PP2A expression and activity in macrophages is a crucial determinant for foam cell formation and the initiation of atherosclerosis. Our study may provide a potential novel approach for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Rui Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chao Zhang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fei Xie
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xianming Zhou
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xingjian Hu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiawei Shi
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinling Du
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Xinling Du
| | - Zhiyong Lin
- Cardiology Division, Emory University School of Medicine, Atlanta, GA, United States
- Zhiyong Lin
| | - Nianguo Dong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Nianguo Dong
| |
Collapse
|
23
|
Islabão AG, Trindade VC, da Mota LMH, Andrade DCO, Silva CA. Managing Antiphospholipid Syndrome in Children and Adolescents: Current and Future Prospects. Paediatr Drugs 2022; 24:13-27. [PMID: 34904182 PMCID: PMC8667978 DOI: 10.1007/s40272-021-00484-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/03/2021] [Indexed: 11/28/2022]
Abstract
Pediatric antiphospholipid syndrome (APS) is a rare acquired multisystem autoimmune thromboinflammatory condition characterized by thrombotic and non-thrombotic clinical manifestations. APS in children and adolescents typically presents with large-vessel thrombosis, thrombotic microangiopathy, and, rarely, obstetric morbidity. Non-thrombotic clinical manifestations are frequently seen in pediatric APS and may be present even before the vascular thrombotic events occur. We review insights into the pathogenesis of APS and discuss potential targets for therapy. The identification of multiple immunologic abnormalities in patients with APS reveals molecular targets for current or future treatment. Management strategies, especially for APS in adolescents, require screening for additional prothrombotic risk factors and consideration of counseling regarding contraceptive strategies, lifestyle recommendations, treatment adherence, and mental health issues associated with this autoimmune thrombophilia. The main goal of therapy in pediatric APS is the prevention of thrombosis. The management of acute thrombosis events in children and adolescents is the same as for primary APS, which involves isolated occurrences, and secondary APS, which is seen in association with another autoimmune disease, e.g., systemic lupus erythematosus. A pediatric hematologist should be consulted so other differential thrombophilic conditions can be eliminated. Therapy includes unfractionated heparin or low-molecular-weight heparin followed by vitamin K antagonists. Treatment of catastrophic APS involves triple therapy (anticoagulation, intravenous corticosteroid pulse therapy, and plasma exchange) and may include intravenous immunoglobulin for children and adolescents with this condition. New drugs such as eculizumab and sirolimus seem to be promising drugs for APS.
Collapse
Affiliation(s)
- Aline Garcia Islabão
- Pediatric Rheumatology Unit, Hospital da Criança de Brasília Jose Alencar, Brasília, DF Brazil ,Programa de Pós-graduação em Ciências Médicas, Faculdade de Medicina, Universidade de Brasília, Brasília, DF Brazil
| | - Vitor Cavalcanti Trindade
- Faculdade de Medicina, Children and Adolescent Institute, Hospital das Clinicas HCFMUSP, Universidade de São Paulo, Av. Dr. Enéas Carvalho de Aguiar, 647-Cerqueira César, São Paulo, SP 05403-000 Brazil
| | - Licia Maria Henrique da Mota
- Programa de Pós-graduação em Ciências Médicas, Faculdade de Medicina, Universidade de Brasília, Brasília, DF Brazil ,Rheumatology Unit, Hospital Universitário de Brasília, Universidade de Brasília, Brasília, Brazil
| | | | - Clovis Artur Silva
- Faculdade de Medicina, Children and Adolescent Institute, Hospital das Clinicas HCFMUSP, Universidade de São Paulo, Av. Dr. Enéas Carvalho de Aguiar, 647-Cerqueira César, São Paulo, SP, 05403-000, Brazil. .,Rheumatology Division, Faculdade de Medicina, Hospital das Clinicas HCFMUSP, Universidade de São Paulo, São Paulo, Brazil.
| |
Collapse
|
24
|
Velásquez M, Peláez LF, Rojas M, Narváez-Sánchez R, Velásquez JA, Escudero C, San Martín S, Cadavid ÁP. Differences in Endothelial Activation and Dysfunction Induced by Antiphospholipid Antibodies Among Groups of Patients With Thrombotic, Refractory, and Non-refractory Antiphospholipid Syndrome. Front Physiol 2021; 12:764702. [PMID: 34925061 PMCID: PMC8675389 DOI: 10.3389/fphys.2021.764702] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 11/02/2021] [Indexed: 01/20/2023] Open
Abstract
Antiphospholipid syndrome (APS) is an autoimmune disorder characterized by pregnancy morbidity or thrombosis and persistent antiphospholipid antibodies (aPL) that bind to the endothelium and induce endothelial activation, which is evidenced by the expression of adhesion molecules and the production of reactive oxygen species (ROS) and subsequent endothelial dysfunction marked by a decrease in the synthesis and release of nitric oxide (NO). These endothelial alterations are the key components for the development of severe pathological processes in APS. Patients with APS can be grouped according to the presence of other autoimmune diseases (secondary APS), thrombosis alone (thrombotic APS), pregnancy morbidity (obstetric APS), and refractoriness to conventional treatment regimens (refractory APS). Typically, patients with severe and refractory obstetric APS exhibit thrombosis and are classified as those having primary or secondary APS. The elucidation of the mechanisms underlying these alterations according to the different groups of patients with APS could help establish new therapies, particularly necessary for severe and refractory cases. Therefore, this study aimed to evaluate the differences in endothelial activation and dysfunction induced by aPL between patients with refractory obstetric APS and other APS clinical manifestations. Human umbilical vein endothelial cells (HUVECs) were stimulated with polyclonal immunoglobulin-G (IgG) from different groups of patients n = 21), including those with primary (VTI) and secondary thrombotic APS (VTII) and refractory primary (RI+), refractory secondary (RII+), and non-refractory primary (NR+) obstetric APS. All of them with thrombosis. The expression of adhesion molecules; the production of ROS, NO, vascular endothelial growth factor (VEGF), and endothelin-1; and the generation of microparticles were used to evaluate endothelial activation and dysfunction. VTI IgG induced the expression of adhesion molecules and the generation of microparticles and VEGF. RI+ IgG induced the expression of adhesion molecules and decreased NO production. RII+ IgG increased the production of microparticles, ROS, and endothelin-1 and reduced NO release. NR+ IgG increased the production of microparticles and endothelin-1 and decreased the production of VEGF and NO. These findings reveal differences in endothelial activation and dysfunction among groups of patients with APS, which should be considered in future studies to evaluate new therapies, especially in refractory cases.
Collapse
Affiliation(s)
- Manuela Velásquez
- Grupo Reproducción, Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, Colombia
| | - Luisa F Peláez
- Grupo Reproducción, Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, Colombia
| | - Mauricio Rojas
- Grupo de Inmunología Celular e Inmunogenética, Facultad de Medicina, Universidad de Antioquia UdeA, Unidad de Citometría de Flujo, Sede de Investigación Universitaria, Medellín, Colombia
| | - Raúl Narváez-Sánchez
- Physiology and Biochemistry Research Group-PHYSIS, Faculty of Medicine, University of Antioquia UdeA, Medellín, Colombia
| | | | - Carlos Escudero
- Red Iberoamericana de Alteraciones Vasculares Asociadas a TRanstornos del EMbarazo (RIVATREM), Chillán, Chile.,Vascular Physiology Laboratory, Basic Sciences Department, Faculty of Sciences, Universidad del Bio-Bio, Chillán, Chile.,Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán, Chile
| | - Sebastián San Martín
- Red Iberoamericana de Alteraciones Vasculares Asociadas a TRanstornos del EMbarazo (RIVATREM), Chillán, Chile.,Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán, Chile.,Biomedical Research Center School of Medicine, Universidad de Valparaíso, Valparaíso, Chile
| | - Ángela P Cadavid
- Grupo Reproducción, Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, Colombia.,Red Iberoamericana de Alteraciones Vasculares Asociadas a TRanstornos del EMbarazo (RIVATREM), Chillán, Chile.,Grupo de Investigación en Trombosis, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, Colombia
| |
Collapse
|
25
|
Green D. Pathophysiology of the Antiphospholipid Antibody Syndrome. Thromb Haemost 2021; 122:1085-1095. [PMID: 34794200 PMCID: PMC9391091 DOI: 10.1055/a-1701-2809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The antiphospholipid syndrome is characterized by antibodies directed against phospholipid-binding proteins and phospholipids attached to cell membrane receptors, mitochondria, oxidized lipoproteins, and activated complement components. When antibodies bind to these complex antigens, cells are activated and the coagulation and complement cascades are triggered, culminating in thrombotic events and pregnancy morbidity that further define the syndrome. The phospholipid-binding proteins most often involved are annexins II and V, β2-glycoprotein I, prothrombin, and cardiolipin. A distinguishing feature of the antiphospholipid syndrome is the "lupus anticoagulant". This is not a single entity but rather a family of antibodies directed against complex antigens consisting of β2-glycoprotein I and/or prothrombin bound to an anionic phospholipid. Although these antibodies prolong in vitro clotting times by competing with clotting factors for phospholipid binding sites, they are not associated with clinical bleeding. Rather, they are thrombogenic because they augment thrombin production in vivo by concentrating prothrombin on phospholipid surfaces. Other antiphospholipid antibodies decrease the clot-inhibitory properties of the endothelium and enhance platelet adherence and aggregation. Some are atherogenic because they increase lipid peroxidation by reducing paraoxonase activity, and others impair fetal nutrition by diminishing placental antithrombotic and fibrinolytic activity. This plethora of destructive autoantibodies is currently managed with immunomodulatory agents, but new approaches to treatment might include vaccines against specific autoantigens, blocking the antibodies generated by exposure to cytoplasmic DNA, and selective targeting of aberrant B-cells to reduce or eliminate autoantibody production.
Collapse
Affiliation(s)
- David Green
- Medicine/Hematology/Oncology, Northwestern University Feinberg School of Medicine, Chicago, United States
| |
Collapse
|
26
|
Chu H, Sacharidou A, Nguyen A, Li C, Chambliss KL, Salmon JE, Shen YM, Lo J, Leone GW, Herz J, Hui DY, Marciano DK, Abrahams VM, Natale BV, Montalbano AP, Xiao X, Xu L, Natale DR, Shaul PW, Mineo C. Protein Phosphatase 2A Activation Via ApoER2 in Trophoblasts Drives Preeclampsia in a Mouse Model of the Antiphospholipid Syndrome. Circ Res 2021; 129:735-750. [PMID: 34404233 DOI: 10.1161/circresaha.120.318941] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Haiyan Chu
- Center for Pulmonary and Vascular Biology, Pediatrics (H.C., A.S., A.N., C.L., K.L.C., P.W.S., C.M.)
| | - Anastasia Sacharidou
- Center for Pulmonary and Vascular Biology, Pediatrics (H.C., A.S., A.N., C.L., K.L.C., P.W.S., C.M.)
| | - An Nguyen
- Center for Pulmonary and Vascular Biology, Pediatrics (H.C., A.S., A.N., C.L., K.L.C., P.W.S., C.M.)
| | - Chun Li
- Center for Pulmonary and Vascular Biology, Pediatrics (H.C., A.S., A.N., C.L., K.L.C., P.W.S., C.M.)
| | - Ken L Chambliss
- Center for Pulmonary and Vascular Biology, Pediatrics (H.C., A.S., A.N., C.L., K.L.C., P.W.S., C.M.)
| | - Jane E Salmon
- Medicine, Hospital for Special Surgery, Weill Cornell Medical College, New York (J.E.S.)
| | - Yu-Min Shen
- Internal Medicine (Y.-M.S., D.K.M.), University of Texas Southwestern Medical Center, Dallas
| | - Julie Lo
- Obstetrics and Gynecology (J.L.), University of Texas Southwestern Medical Center, Dallas
| | - Gustavo W Leone
- Froedtert-Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee (G.W.L.)
| | - Joachim Herz
- Molecular Genetics (J.H.), University of Texas Southwestern Medical Center, Dallas
| | - David Y Hui
- Pathology, University of Cincinnati College of Medicine (D.Y.H.)
| | - Denise K Marciano
- Internal Medicine (Y.-M.S., D.K.M.), University of Texas Southwestern Medical Center, Dallas.,Cell Biology (D.K.M., C.M.), University of Texas Southwestern Medical Center, Dallas
| | - Vikki M Abrahams
- Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, New Haven, CT (V.M.A.)
| | - Bryony V Natale
- Obstetrics, Gynecology & Reproductive Science, University of California San Diego, La Jolla (B.V.N., D.R.N.).,Obstetrics and Gynaecology, School of Medicine, Queen's University, Ontario, Canada (B.V.N., D.R.N.)
| | - Alina P Montalbano
- Biochemistry and Obstetrics and Gynecology (A.P.M.), University of Texas Southwestern Medical Center, Dallas
| | - Xue Xiao
- Population and Data Sciences and Pediatrics (X.X., L.X.), University of Texas Southwestern Medical Center, Dallas
| | - Lin Xu
- Population and Data Sciences and Pediatrics (X.X., L.X.), University of Texas Southwestern Medical Center, Dallas
| | - David R Natale
- Obstetrics, Gynecology & Reproductive Science, University of California San Diego, La Jolla (B.V.N., D.R.N.).,Obstetrics and Gynaecology, School of Medicine, Queen's University, Ontario, Canada (B.V.N., D.R.N.)
| | - Philip W Shaul
- Center for Pulmonary and Vascular Biology, Pediatrics (H.C., A.S., A.N., C.L., K.L.C., P.W.S., C.M.)
| | - Chieko Mineo
- Center for Pulmonary and Vascular Biology, Pediatrics (H.C., A.S., A.N., C.L., K.L.C., P.W.S., C.M.).,Cell Biology (D.K.M., C.M.), University of Texas Southwestern Medical Center, Dallas
| |
Collapse
|
27
|
Calvier L, Manouchehri N, Sacharidou A, Mineo C, Shaul PW, Hui DY, Kounnas MZ, Stüve O, Herz J. Apolipoprotein E receptor 2 deficiency decreases endothelial adhesion of monocytes and protects against autoimmune encephalomyelitis. Sci Immunol 2021; 6:eabd0931. [PMID: 34452924 PMCID: PMC8627794 DOI: 10.1126/sciimmunol.abd0931] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 07/29/2021] [Indexed: 01/10/2023]
Abstract
Under normal conditions, the blood-brain barrier effectively regulates the passage of immune cells into the central nervous system (CNS). However, under pathological conditions such as multiple sclerosis (MS), leukocytes, especially monocytes, infiltrate the CNS where they promote inflammatory demyelination, resulting in paralysis. Therapies targeting the immune cells directly and preventing leukocyte infiltration exist for MS but may compromise the immune system. Here, we explore how apolipoprotein E receptor 2 (ApoER2) regulates vascular adhesion and infiltration of monocytes during inflammation. We induced experimental autoimmune encephalitis in ApoER2 knockout mice and in mice carrying a loss-of-function mutation in the ApoER2 cytoplasmic domain. In both models, paralysis and neuroinflammation were largely abolished as a result of greatly diminished monocyte adherence due to reduced expression of adhesion molecules on the endothelial surface. Our findings expand our mechanistic understanding of the vascular barrier, the regulation of inflammation and vascular permeability, and the therapeutic potential of ApoER2-targeted therapies.
Collapse
Affiliation(s)
- Laurent Calvier
- Department of Molecular Genetics, University of Texas (UT) Southwestern Medical Center, Dallas, TX, USA.
- Center for Translational Neurodegeneration Research, UT Southwestern Medical Center, Dallas, TX, USA
| | - Navid Manouchehri
- Department of Neurology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Anastasia Sacharidou
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, UT Southwestern Medical Center, Dallas, TX, USA
| | - Chieko Mineo
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, UT Southwestern Medical Center, Dallas, TX, USA
| | - Philip W Shaul
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, UT Southwestern Medical Center, Dallas, TX, USA
| | - David Y Hui
- Department of Pathology, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | | | - Olaf Stüve
- Department of Neurology, UT Southwestern Medical Center, Dallas, TX, USA
- Department of Neurology, VA North Texas Health Care System, Medical Service, Dallas, TX, USA
| | - Joachim Herz
- Department of Molecular Genetics, University of Texas (UT) Southwestern Medical Center, Dallas, TX, USA.
- Center for Translational Neurodegeneration Research, UT Southwestern Medical Center, Dallas, TX, USA
- Department of Neurology, UT Southwestern Medical Center, Dallas, TX, USA
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
28
|
Liu J, Li Y. Thrombosis associated with mycoplasma pneumoniae infection (Review). Exp Ther Med 2021; 22:967. [PMID: 34335909 PMCID: PMC8290426 DOI: 10.3892/etm.2021.10399] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 06/18/2021] [Indexed: 12/12/2022] Open
Abstract
Mycoplasma pneumoniae is a common pathogen causing respiratory infections in children and adults. In addition to respiratory diseases, Mycoplasma pneumoniae is also involved in numerous extrapulmonary diseases. Thrombosis is an extrapulmonary manifestation associated with Mycoplasma pneumoniae infection. In recent years, an increasing number of case reports have been published identifying thrombosis secondary to Mycoplasma pneumoniae infection. In the present study, the available relevant literature in English available on PubMed, Medline and Web of Science was consulted. The results of the present study demonstrated that in patients with thrombosis caused by Mycoplasma pneumoniae infection, some of the factors causing thrombosis are transient and some are due to hereditary thrombophilia. Following timely treatment, the majority of patients recovered completely but some patients had a poor prognosis. The present review focuses on the pathogenesis, clinical features, treatment and prognosis of this crucial issue, which contributes toward the understanding of the disease.
Collapse
Affiliation(s)
- Jingwei Liu
- Department of Pediatrics Intensive Care Unit, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yumei Li
- Department of Pediatrics Intensive Care Unit, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
29
|
Calvier L, Xian X, Lee R, Sacharidou A, Mineo C, Shaul PW, Kounnas MZ, Tsai S, Herz J. Reelin Depletion Protects Against Atherosclerosis by Decreasing Vascular Adhesion of Leukocytes. Arterioscler Thromb Vasc Biol 2021; 41:1309-1318. [PMID: 33626909 PMCID: PMC7990715 DOI: 10.1161/atvbaha.121.316000] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
MESH Headings
- Animals
- Antibodies, Neutralizing/pharmacology
- Atherosclerosis/genetics
- Atherosclerosis/immunology
- Atherosclerosis/metabolism
- Atherosclerosis/prevention & control
- CX3C Chemokine Receptor 1/genetics
- Cell Adhesion/drug effects
- Cell Adhesion Molecules, Neuronal/antagonists & inhibitors
- Cell Adhesion Molecules, Neuronal/deficiency
- Cell Adhesion Molecules, Neuronal/genetics
- Coculture Techniques
- Disease Models, Animal
- Endothelial Cells/drug effects
- Endothelial Cells/immunology
- Endothelial Cells/metabolism
- Extracellular Matrix Proteins/antagonists & inhibitors
- Extracellular Matrix Proteins/deficiency
- Extracellular Matrix Proteins/genetics
- Female
- Green Fluorescent Proteins/genetics
- Green Fluorescent Proteins/metabolism
- Humans
- LDL-Receptor Related Proteins/metabolism
- Leukocyte Rolling/drug effects
- Leukocytes/drug effects
- Leukocytes/immunology
- Leukocytes/metabolism
- Male
- Mice, Transgenic
- Nerve Tissue Proteins/antagonists & inhibitors
- Nerve Tissue Proteins/deficiency
- Nerve Tissue Proteins/genetics
- Oligonucleotides, Antisense/pharmacology
- Plaque, Atherosclerotic
- Receptors, LDL/deficiency
- Receptors, LDL/genetics
- Reelin Protein
- Serine Endopeptidases/deficiency
- Serine Endopeptidases/genetics
- Signal Transduction
- U937 Cells
Collapse
Affiliation(s)
- Laurent Calvier
- Department of Molecular Genetics, University of Texas (UT) Southwestern Medical Center, Dallas TX, USA
- Center for Translational Neurodegeneration Research, UT Southwestern Medical Center, Dallas TX, USA
| | - Xunde Xian
- Department of Molecular Genetics, University of Texas (UT) Southwestern Medical Center, Dallas TX, USA
- Center for Translational Neurodegeneration Research, UT Southwestern Medical Center, Dallas TX, USA
| | - Richard Lee
- Ionis Pharmaceuticals, Inc. San Diego CA, USA
| | - Anastasia Sacharidou
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas TX, USA
| | - Chieko Mineo
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas TX, USA
| | - Philip W. Shaul
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas TX, USA
| | | | - Shirling Tsai
- Department of Surgery, UT Southwestern Medical Center, Dallas TX, USA
- Dallas VA Medical Center, Dallas TX, USA
| | - Joachim Herz
- Department of Molecular Genetics, University of Texas (UT) Southwestern Medical Center, Dallas TX, USA
- Center for Translational Neurodegeneration Research, UT Southwestern Medical Center, Dallas TX, USA
- Department of Neuroscience, UT Southwestern Medical Center, Dallas TX, USA
- Department of Neurology and Neurotherapeutics, UT Southwestern Medical Center, Dallas TX, USA
| |
Collapse
|
30
|
More about complement in the antiphospholipid syndrome. Blood 2021; 136:1456-1459. [PMID: 32462204 DOI: 10.1182/blood.2020005171] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 04/24/2020] [Indexed: 01/27/2023] Open
|
31
|
Gu SX, Tyagi T, Jain K, Gu VW, Lee SH, Hwa JM, Kwan JM, Krause DS, Lee AI, Halene S, Martin KA, Chun HJ, Hwa J. Thrombocytopathy and endotheliopathy: crucial contributors to COVID-19 thromboinflammation. Nat Rev Cardiol 2021; 18:194-209. [PMID: 33214651 PMCID: PMC7675396 DOI: 10.1038/s41569-020-00469-1] [Citation(s) in RCA: 255] [Impact Index Per Article: 85.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/19/2020] [Indexed: 02/06/2023]
Abstract
The core pathology of coronavirus disease 2019 (COVID-19) is infection of airway cells by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that results in excessive inflammation and respiratory disease, with cytokine storm and acute respiratory distress syndrome implicated in the most severe cases. Thrombotic complications are a major cause of morbidity and mortality in patients with COVID-19. Patients with pre-existing cardiovascular disease and/or traditional cardiovascular risk factors, including obesity, diabetes mellitus, hypertension and advanced age, are at the highest risk of death from COVID-19. In this Review, we summarize new lines of evidence that point to both platelet and endothelial dysfunction as essential components of COVID-19 pathology and describe the mechanisms that might account for the contribution of cardiovascular risk factors to the most severe outcomes in COVID-19. We highlight the distinct contributions of coagulopathy, thrombocytopathy and endotheliopathy to the pathogenesis of COVID-19 and discuss potential therapeutic strategies in the management of patients with COVD-19. Harnessing the expertise of the biomedical and clinical communities is imperative to expand the available therapeutics beyond anticoagulants and to target both thrombocytopathy and endotheliopathy. Only with such collaborative efforts can we better prepare for further waves and for future coronavirus-related pandemics.
Collapse
Affiliation(s)
- Sean X Gu
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Tarun Tyagi
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Kanika Jain
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Vivian W Gu
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Seung Hee Lee
- Division of Cardiovascular Diseases, Center for Biomedical Sciences, National Institute of Health, Cheongju, Chungbuk, Korea
| | - Jonathan M Hwa
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Jennifer M Kwan
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Diane S Krause
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Alfred I Lee
- Section of Hematology, Department of Internal Medicine and Yale Comprehensive Cancer Center, Yale University School of Medicine, New Haven, CT, USA
| | - Stephanie Halene
- Section of Hematology, Department of Internal Medicine and Yale Comprehensive Cancer Center, Yale University School of Medicine, New Haven, CT, USA
| | - Kathleen A Martin
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Hyung J Chun
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - John Hwa
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
32
|
FcRn augments induction of tissue factor activity by IgG-containing immune complexes. Blood 2021; 135:2085-2093. [PMID: 32187355 DOI: 10.1182/blood.2019001133] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Accepted: 03/03/2020] [Indexed: 12/17/2022] Open
Abstract
Thromboembolism complicates disorders caused by immunoglobulin G (IgG)-containing immune complexes (ICs), but the underlying mechanisms are incompletely understood. Prior evidence indicates that induction of tissue factor (TF) on monocytes, a pivotal step in the initiation, localization, and propagation of coagulation by ICs, is mediated through Fcγ receptor IIa (FcγRIIa); however, the involvement of other receptors has not been investigated in detail. The neonatal Fc receptor (FcRn) that mediates IgG and albumin recycling also participates in cellular responses to IgG-containing ICs. Here we asked whether FcRn is also involved in the induction of TF-dependent factor Xa (FXa) activity by IgG-containing ICs by THP-1 monocytic cells and human monocytes. Induction of FXa activity by ICs containing IgG antibodies to platelet factor 4 (PF4) involved in heparin-induced thrombocytopenia (HIT), β-2-glycoprotein-1 implicated in antiphospholipid syndrome, or red blood cells coated with anti-(α)-Rh(D) antibodies that mediate hemolysis in vivo was inhibited by a humanized monoclonal antibody (mAb) that blocks IgG binding to human FcRn. IgG-containing ICs that bind to FcγR and FcRn induced FXa activity, whereas IgG-containing ICs with an Fc engineered to be unable to engage FcRn did not. Infusion of an α-FcRn mAb prevented fibrin deposition after microvascular injury in a murine model of HIT in which human FcγRIIa was expressed as a transgene. These data implicate FcRn in TF-dependent FXa activity induced by soluble and cell-associated IgG-containing ICs. Antibodies to FcRn, now in clinical trials in warm autoimmune hemolytic anemia to lower IgG antibodies and IgG containing ICs may also reduce the risk of venous thromboembolism.
Collapse
|
33
|
Cheng C, Cheng GY, Denas G, Pengo V. Arterial thrombosis in antiphospholipid syndrome (APS): Clinical approach and treatment. A systematic review. Blood Rev 2020; 48:100788. [PMID: 33341301 DOI: 10.1016/j.blre.2020.100788] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 11/17/2020] [Accepted: 12/08/2020] [Indexed: 12/17/2022]
Abstract
Thrombotic Antiphospholipid Syndrome (APS) is a condition affecting young individuals in whom a thromboembolic event occurs in the presence of circulating antiphospholipid antibodies (aPL). An extensive body of literature has covered the most common clinical presentation of the syndrome, venous thromboembolism. Arterial thrombosis in APS, a lesser clinical expression, is less studied. This review will concentrate on the body of literature concerning pathogenesis, clinical presentation and management of arterial thrombosis in APS.
Collapse
Affiliation(s)
- Chunyan Cheng
- Thrombosis Research Laboratory, Department of Cardiac-Thoracic-Vascular Sciences and Public Health, University of Padova, Padua, Italy
| | - Gang-Yi Cheng
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Gentian Denas
- Thrombosis Research Laboratory, Department of Cardiac-Thoracic-Vascular Sciences and Public Health, University of Padova, Padua, Italy
| | - Vittorio Pengo
- Thrombosis Research Laboratory, Department of Cardiac-Thoracic-Vascular Sciences and Public Health, University of Padova, Padua, Italy; Arianna Foundation on Anticoagulation, Bologna, Italy.
| |
Collapse
|
34
|
Abstract
PURPOSE OF REVIEW Antiphospholipid syndrome (APS) is a thrombo-inflammatory disease that is primarily treated with anticoagulation. Better understanding the inflammatory aspects of APS could lead to safer, more effective, and more personalized therapeutic options. To this end, we sought to understand recent literature related to the role of neutrophils and, in particular, neutrophil extracellular traps (NETs) in APS. RECENT FINDINGS Expression of genes associated with type I interferons, endothelial adhesion, and pregnancy regulation are increased in APS neutrophils. APS neutrophils have a reduced threshold for NET release, which likely potentiates thrombotic events and perhaps especially large-vein thrombosis. Neutrophil-derived reactive oxygen species also appear to play a role in APS pathogenesis. There are new approaches for preventing and disrupting NETs that could potentially be leveraged to reduce the risk of APS-associated thrombosis. Neutrophils and NETs contribute to APS pathophysiology. More precisely understanding their roles at a mechanistic level should help identify new therapeutic targets for inhibiting NET formation, enhancing NET dissolution, and altering neutrophil adhesion. Such approaches may ultimately lead to better clinical management of APS patients and thereby reduce the chronic burden of this disease.
Collapse
|
35
|
Wei A, Xiao H, Xu G, Yu X, Guo J, Jing Z, Shi S, Song Y. Hyperoside Protects Human Umbilical Vein Endothelial Cells Against Anticardiolipin Antibody-Induced Injury by Activating Autophagy. Front Pharmacol 2020; 11:762. [PMID: 32508661 PMCID: PMC7253676 DOI: 10.3389/fphar.2020.00762] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 05/07/2020] [Indexed: 12/28/2022] Open
Abstract
Anticardiolipin antibody (aCL), an important characterization of antiphospholipid syndrome, shows an intense association with vascular endothelial injury. Hyperoside is a flavonoid extracted from medicinal plants traditionally used in Chinese medicines, displaying anti-inflammatory, anti-cancer, and anti-oxidative properties in various diseases. Recent studies have shifted the focus on the protective effects of hyperoside on vascular endothelial injury. However, little is known about the mechanisms involved. In the present study, we investigated the effect of hyperoside on aCL-induced injury of human umbilical vein endothelial cells (HUVECs) in vitro. Our data illustrated that aCL induced HUVEC injury via inhibiting autophagy. Hyperoside reduced aCL-induced secretion of proinflammatory cytokines IL-1β and IL-8 and endothelial adhesion cytokines TF, ICAM1, and VCAM1 in HUVECs. Additionally, hyperoside activated autophagy and suppressed the mTOR/S6K and TLR4/Myd88/NF-κB signaling transduction pathways in aCL-induced HUVECs. To the best of our knowledge, this is the first study to investigate the effect of hyperoside on aCL-induced injury, as well as offer insights into the involved mechanisms, which is of great significance for the treatment of antiphospholipid syndrome.
Collapse
Affiliation(s)
- Aiwu Wei
- Department of Reproductive Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Huidongzi Xiao
- Department of Reproductive Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Guangli Xu
- Department of Reproductive Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Xile Yu
- Department of Reproductive Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Jingjing Guo
- Department of Reproductive Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Zhuqing Jing
- Department of Reproductive Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Shaoqi Shi
- Department of Reproductive Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Yanli Song
- Department of Reproductive Medicine, Henan Province Hospital of Traditional Chinese Medicine, Zhengzhou, China
| |
Collapse
|
36
|
McDonnell T, Wincup C, Buchholz I, Pericleous C, Giles I, Ripoll V, Cohen H, Delcea M, Rahman A. The role of beta-2-glycoprotein I in health and disease associating structure with function: More than just APS. Blood Rev 2020; 39:100610. [PMID: 31471128 PMCID: PMC7014586 DOI: 10.1016/j.blre.2019.100610] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 08/09/2019] [Accepted: 08/15/2019] [Indexed: 12/22/2022]
Abstract
Beta-2-Glycoprotein I (β2GPI) plays a number of essential roles throughout the body. β2GPI, C-reactive protein and thrombomodulin are the only three proteins that possess the dual capability to up and down regulate the complement and coagulation systems depending upon external stimulus. Clinically, β2GPI is the primary antigen in the autoimmune condition antiphospholipid syndrome (APS), which is typically characterised by pregnancy morbidity and vascular thrombosis. This protein is also capable of adopting at least two distinct structural forms, but it has been argued that several other intermediate forms may exist. Thus, β2GPI is a unique protein with a key role in haemostasis, homeostasis and immunity. In this review, we examine the genetics, structure and function of β2GPI in the body and how these factors may influence its contribution to disease pathogenesis. We also consider the clinical implications of β2GPI in the diagnosis of APS and as a potentially novel therapeutic target.
Collapse
Affiliation(s)
- Thomas McDonnell
- Rheumatology, Division of Medicine, Rayne Institute, University College London, UK.
| | - Chris Wincup
- Rheumatology, Division of Medicine, Rayne Institute, University College London, UK
| | - Ina Buchholz
- Nanostructure Group, Institute of Biochemistry, University of Greifswald, Germany
| | - Charis Pericleous
- Imperial College London, Imperial College Vascular Sciences, National Heart & Lung Institute, ICTEM, Hammersmith Campus, Du Cane Road, London, UK
| | - Ian Giles
- Rheumatology, Division of Medicine, Rayne Institute, University College London, UK
| | - Vera Ripoll
- Rheumatology, Division of Medicine, Rayne Institute, University College London, UK
| | - Hannah Cohen
- Department of Haematology, University College London Hospitals NHS Foundation Trust, London, UK
| | - Mihaela Delcea
- Nanostructure Group, Institute of Biochemistry, University of Greifswald, Germany
| | - Anisur Rahman
- Rheumatology, Division of Medicine, Rayne Institute, University College London, UK
| |
Collapse
|
37
|
Oxidative stress in endothelial cells induced by the serum of women with different clinical manifestations of the antiphospholipid syndrome. ACTA ACUST UNITED AC 2019; 39:673-688. [PMID: 31860179 PMCID: PMC7363350 DOI: 10.7705/biomedica.4701] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Indexed: 12/27/2022]
Abstract
Introducción. El síndrome antifosfolípido se caracteriza por la presencia persistente de anticuerpos antifosfolípidos y manifestaciones clínicas de trombosis o morbilidad gestacional, las cuales se asocian con estrés oxidativo y disfunción endotelial. Objetivo. Evaluar los marcadores de estrés oxidativo en células endoteliales, inducidos por el suero de mujeres con diferentes manifestaciones clínicas del síndrome antifosfolípido y analizar la capacidad antioxidante de los sueros. Materiales y métodos. Se incluyeron 48 mujeres que fueron clasificadas así: presencia de anticuerpos antifosfolípidos y criterios clínicos de morbilidad gestacional, trombosis vascular o ambas. Como grupos control se incluyeron mujeres negativas para anticuerpos antifosfolípidos. En un modelo in vitro de células endoteliales estimuladas con los sueros de las mujeres del estudio, se determinaron algunos marcadores de estrés oxidativo por citometría de flujo. También, se analizó la capacidad antioxidante de los sueros incluidos. Resultados. Los sueros de los grupos de mujeres con síndrome antifosfolípido que presentaban trombosis, con morbilidad gestacional o sin ella, generaron un incremento significativo (p<0,05 y p<0,001) en los marcadores de estrés oxidativo endotelial, en contraste con el control de suero humano normal. No se observaron diferencias en el efecto de los sueros de los diferentes grupos de estudio sobre la lipoperoxidación endotelial. Tampoco se encontró diferencia en la actividad antioxidante de los sueros. Conclusión. El estrés oxidativo mitocondrial en el endotelio se asocia con la presencia de trombosis. Sin embargo, cuando esta se asocia con morbilidad gestacional, también se genera estrés oxidativo intracelular.
Collapse
|
38
|
Komaravolu RK, Waltmann MD, Konaniah E, Jaeschke A, Hui DY. ApoER2 (Apolipoprotein E Receptor-2) Deficiency Accelerates Smooth Muscle Cell Senescence via Cytokinesis Impairment and Promotes Fibrotic Neointima After Vascular Injury. Arterioscler Thromb Vasc Biol 2019; 39:2132-2144. [PMID: 31412739 PMCID: PMC6761011 DOI: 10.1161/atvbaha.119.313194] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Genome-wide studies showed that mutation in apoER2 (apolipoprotein E receptor-2) is additive with ε4 polymorphism in the APOE gene on cardiovascular disease risk in humans. ApoE or apoER2 deficiency also accelerates atherosclerosis lesion necrosis in hypercholesterolemic mice and promotes neointima formation after vascular injury. This study tests the hypothesis that apoE and apoER2 modulate vascular occlusive diseases through distinct mechanisms. Approach and Results: Carotid endothelial denudation induced robust neointima formation in both apoE-/- and apoER2-deficient Lrp8-/- mice. The intima in apoE-/- mice was rich in smooth muscle cells, but the intima in Lrp8-/- mice was cell-poor and rich in extracellular matrix. Vascular smooth muscle cells isolated from apoE-/- mice were hyperplastic whereas Lrp8-/- smooth muscle cells showed reduced proliferation but responded robustly to TGF (transforming growth factor)-β-induced fibronectin synthesis indicative of a senescence-associated secretory phenotype, which was confirmed by increased β-galactosidase activity, p16INK4a immunofluorescence, and number of multinucleated cells. Western blot analysis of cell cycle-associated proteins showed that apoER2 deficiency promotes cell cycle arrest at the metaphase/anaphase. Coimmunoprecipitation experiments revealed that apoER2 interacts with the catalytic subunit of protein phosphatase 2A. In the absence of apoER2, PP2A-C (protein phosphatase 2A catalytic subunit) failed to interact with CDC20 (cell-division cycle protein 20) thus resulting in inactive anaphase-promoting complex and impaired cell cycle exit. CONCLUSIONS This study showed that apoER2 participates in APC (anaphase-promoting complex)/CDC20 complex formation during mitosis, and its absence impedes cytokinesis abscission thereby accelerating premature cell senescence and vascular disease. This mechanism is distinct from apoE deficiency, which causes smooth muscle cell hyperplasia to accelerate vascular disease.
Collapse
Affiliation(s)
- Ravi K. Komaravolu
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Research Center, University of Cincinnati College of Medicine, Cincinnati, OH 45237
| | - Meaghan D. Waltmann
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Research Center, University of Cincinnati College of Medicine, Cincinnati, OH 45237
| | - Eddy Konaniah
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Research Center, University of Cincinnati College of Medicine, Cincinnati, OH 45237
| | - Anja Jaeschke
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Research Center, University of Cincinnati College of Medicine, Cincinnati, OH 45237
| | - David Y. Hui
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Research Center, University of Cincinnati College of Medicine, Cincinnati, OH 45237
| |
Collapse
|
39
|
Tissue factor pathway inhibitor primes monocytes for antiphospholipid antibody-induced thrombosis. Blood 2019; 134:1119-1131. [PMID: 31434703 DOI: 10.1182/blood.2019001530] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 08/06/2019] [Indexed: 12/28/2022] Open
Abstract
Antiphospholipid antibodies (aPLs) with complex lipid and/or protein reactivities cause complement-dependent thrombosis and pregnancy complications. Although cross-reactivities with coagulation regulatory proteins contribute to the risk for developing thrombosis in patients with antiphospholipid syndrome, the majority of pathogenic aPLs retain reactivity with membrane lipid components and rapidly induce reactive oxygen species-dependent proinflammatory signaling and tissue factor (TF) procoagulant activation. Here, we show that lipid-reactive aPLs activate a common species-conserved TF signaling pathway. aPLs dissociate an inhibited TF coagulation initiation complex on the cell surface of monocytes, thereby liberating factor Xa for thrombin generation and protease activated receptor 1/2 heterodimer signaling. In addition to proteolytic signaling, aPLs promote complement- and protein disulfide isomerase-dependent TF-integrin β1 trafficking that translocates aPLs and NADPH oxidase to the endosome. Cell surface TF pathway inhibitor (TFPI) synthesized by monocytes is required for TF inhibition, and disabling TFPI prevents aPL signaling, indicating a paradoxical prothrombotic role for TFPI. Myeloid cell-specific TFPI inactivation has no effect on models of arterial or venous thrombus development, but remarkably prevents experimental aPL-induced thrombosis in mice. Thus, the physiological control of TF primes monocytes for rapid aPL pathogenic signaling and thrombosis amplification in an unexpected crosstalk between complement activation and coagulation signaling.
Collapse
|
40
|
Miranda S, Billoir P, Damian L, Thiebaut PA, Schapman D, Le Besnerais M, Jouen F, Galas L, Levesque H, Le Cam-Duchez V, Joannides R, Richard V, Benhamou Y. Hydroxychloroquine reverses the prothrombotic state in a mouse model of antiphospholipid syndrome: Role of reduced inflammation and endothelial dysfunction. PLoS One 2019; 14:e0212614. [PMID: 30870459 PMCID: PMC6417644 DOI: 10.1371/journal.pone.0212614] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 02/06/2019] [Indexed: 01/01/2023] Open
Abstract
Antiphospholipid antibodies (aPL) promote endothelial dysfunction, inflammation and procoagulant state. We investigated the effect of hydroxychloroquine (HCQ) on prothrombotic state and endothelial function in mice and in human aortic endothelial cells (HAEC). Human aPL were injected to C57BL/6 mice treated or not with HCQ. Vascular endothelial function and eNOS were assessed in isolated mesenteric arteries. Thrombosis was assessed both in vitro by measuring thrombin generation time (TGT) and tissue factor (TF) expression and in vivo by the measurement of the time to occlusion in carotid and the total thrombosis area in mesenteric arteries. TGT, TF, and VCAM1 expression were evaluated in HAEC. aPL increased VCAM-1 expression and reduced endothelium dependent relaxation to acetylcholine. In parallel, aPL shortened the time to occlusion and extended thrombus area in mice. This was associated with an overexpression of TF and an increased TGT in mice and in HAEC. HCQ reduced clot formation as well as TGT, and improved endothelial-dependent relaxations. Finally, HCQ increased the p-eNOS/eNOS ratio. This study provides new evidence that HCQ improves procoagulant status and vascular function in APS by modulating eNOS, leading to an improvement in the production of NO.
Collapse
Affiliation(s)
- Sébastien Miranda
- Rouen University Hospital, Department of Internal Medicine, Rouen, France
- Normandie Univ, UNIROUEN, INSERM U1096 EnVI, Rouen, France
- University of Rouen, Institute for Research and Innovation in Biomedicine, Rouen, France
| | - Paul Billoir
- Normandie Univ, UNIROUEN, INSERM U1096 EnVI, Rouen, France
- Rouen University Hospital, Department of Vascular Haemostasis, Rouen France
| | - Louise Damian
- Rouen University Hospital, Department of Internal Medicine, Rouen, France
- Normandie Univ, UNIROUEN, INSERM U1096 EnVI, Rouen, France
- University of Rouen, Institute for Research and Innovation in Biomedicine, Rouen, France
| | - Pierre Alain Thiebaut
- Rouen University Hospital, Department of Internal Medicine, Rouen, France
- Normandie Univ, UNIROUEN, INSERM U1096 EnVI, Rouen, France
- University of Rouen, Institute for Research and Innovation in Biomedicine, Rouen, France
| | - Damien Schapman
- University of Rouen, Institute for Research and Innovation in Biomedicine, Rouen, France
- Univ, Inserm, UNIROUEN, PRIMACEN, Cell Imaging Platform of Normandy, Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Maelle Le Besnerais
- Rouen University Hospital, Department of Internal Medicine, Rouen, France
- Normandie Univ, UNIROUEN, INSERM U1096 EnVI, Rouen, France
- University of Rouen, Institute for Research and Innovation in Biomedicine, Rouen, France
| | - Fabienne Jouen
- University of Rouen, Institute for Research and Innovation in Biomedicine, Rouen, France
- Inserm U1234, Rouen, France
| | - Ludovic Galas
- University of Rouen, Institute for Research and Innovation in Biomedicine, Rouen, France
- Univ, Inserm, UNIROUEN, PRIMACEN, Cell Imaging Platform of Normandy, Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Hervé Levesque
- Rouen University Hospital, Department of Internal Medicine, Rouen, France
- Normandie Univ, UNIROUEN, INSERM U1096 EnVI, Rouen, France
- University of Rouen, Institute for Research and Innovation in Biomedicine, Rouen, France
| | - Véronique Le Cam-Duchez
- Normandie Univ, UNIROUEN, INSERM U1096 EnVI, Rouen, France
- Rouen University Hospital, Department of Vascular Haemostasis, Rouen France
| | - Robinson Joannides
- Normandie Univ, UNIROUEN, INSERM U1096 EnVI, Rouen, France
- University of Rouen, Institute for Research and Innovation in Biomedicine, Rouen, France
| | - Vincent Richard
- Normandie Univ, UNIROUEN, INSERM U1096 EnVI, Rouen, France
- University of Rouen, Institute for Research and Innovation in Biomedicine, Rouen, France
| | - Ygal Benhamou
- Rouen University Hospital, Department of Internal Medicine, Rouen, France
- Normandie Univ, UNIROUEN, INSERM U1096 EnVI, Rouen, France
- University of Rouen, Institute for Research and Innovation in Biomedicine, Rouen, France
- * E-mail:
| |
Collapse
|
41
|
Velásquez M, Rojas M, Abrahams VM, Escudero C, Cadavid ÁP. Mechanisms of Endothelial Dysfunction in Antiphospholipid Syndrome: Association With Clinical Manifestations. Front Physiol 2018; 9:1840. [PMID: 30627104 PMCID: PMC6309735 DOI: 10.3389/fphys.2018.01840] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 12/06/2018] [Indexed: 12/13/2022] Open
Abstract
The endothelium is a monolayer of cells that covers the inner surface of blood vessels and its integrity is essential for the maintenance of vascular health. Endothelial dysfunction is a key pathological component of antiphospholipid syndrome (APS). Its systemic complications include thrombotic endocarditis, valvular dysfunction, cerebrovascular occlusions, proliferative nephritis, deep vein thrombosis, and pulmonary embolism. In women, APS is also associated with pregnancy complications (obstetric APS). The conventional treatment regimens for APS are ineffective when the clinical symptoms are severe. Therefore, a better understanding of alterations in the endothelium caused by antiphospholipid antibodies (aPL) may lead to more effective therapies in patients with elevated aPL titers and severe clinical symptoms. Currently, while in vivo analyses of endothelial dysfunction in patients with APS have been reported, most research has been performed using in vitro models with endothelial cells exposed to either patient serum/plasma, monoclonal aPL, or IgGs isolated from patients with APS. These studies have described a reduction in endothelial cell nitric oxide synthesis, the induction of inflammatory and procoagulant phenotypes, an increase in endothelial proliferation, and impairments in vascular remodeling and angiogenesis. Despite these lines of evidence, further research is required to better understand the pathophysiology of endothelial dysfunction in patients with APS. In this review, we have compared the current understanding about the mechanisms of endothelial dysfunction induced by patient-derived aPL under the two main clinical manifestations of APS: thrombosis and gestational complications, either alone or in combination. We also discuss gaps in our current knowledge regarding aPL-induced endothelial dysfunction.
Collapse
Affiliation(s)
- Manuela Velásquez
- Grupo Reproducción, Departamento de Microbiología y Parasitología, Escuela de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Mauricio Rojas
- Grupo de Inmunología Celular e Inmunogenética, Facultad de Medicina, Coordinador Unidad de Citometría de Flujo, Sede de Investigación Universitaria, Universidad de Antioquia, Medellín, Colombia
| | - Vikki M Abrahams
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, United States
| | - Carlos Escudero
- Vascular Physiology Laboratory, Group of Investigation in Tumor Angiogenesis (GIANT), Department of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile.,Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillan, Chile.,Red Iberoamericana de Alteraciones Vasculares Asociadas a Transtornos del Embarazo, Chillan, Chile
| | - Ángela P Cadavid
- Grupo Reproducción, Departamento de Microbiología y Parasitología, Escuela de Medicina, Universidad de Antioquia, Medellín, Colombia.,Red Iberoamericana de Alteraciones Vasculares Asociadas a Transtornos del Embarazo, Chillan, Chile
| |
Collapse
|
42
|
Lackner KJ, Müller-Calleja N. Pathogenesis of antiphospholipid syndrome: recent insights and emerging concepts. Expert Rev Clin Immunol 2018; 15:199-209. [PMID: 30412684 DOI: 10.1080/1744666x.2019.1546578] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Introduction: Even though our understanding of the antiphospholipid syndrome (APS) has improved tremendously over the last decades, we are still not in a position to replace symptomatic anticoagulation by pathogenesis based causal treatments. Areas covered: Recent years have provided further insights into pathogenetically relevant mechanisms. These include a differentiation of pathogenic subtypes of antiphospholipid antibodies (aPL), novel mechanisms modulating disease activity, for example, extracellular vesicles and microRNA, and novel players in pathogenesis, for example, neutrophils and neutrophil extracellular traps (NETs). Expert commentary: It is evident that aPL induce a proinflammatory and procoagulant state and recent data suggest that different aPL species activate different signaling pathways which sometimes converge into a common cellular response. This implies that presence of more than one aPL species may disproportionally increase the risk for the major manifestations of APS, that is, thrombosis and fetal loss. Further delineation of the pathogenic mechanisms will hopefully provide clues to causal rather than symptomatic treatments of APS.
Collapse
Affiliation(s)
- Karl J Lackner
- a Institute of Clinical Chemistry and Laboratory Medicine , University Medical Center of the Johannes Gutenberg University , Mainz , Germany.,b Center for Translational Vascular Biology , University Medical Center of the Johannes Gutenberg University , Mainz , Germany
| | - Nadine Müller-Calleja
- a Institute of Clinical Chemistry and Laboratory Medicine , University Medical Center of the Johannes Gutenberg University , Mainz , Germany.,b Center for Translational Vascular Biology , University Medical Center of the Johannes Gutenberg University , Mainz , Germany.,c Center for Thrombosis and Hemostasis , University Medical Center of the Johannes Gutenberg University , Mainz , Germany
| |
Collapse
|
43
|
Morrell CN, Pariser DN, Hilt ZT, Vega Ocasio D. The Platelet Napoleon Complex-Small Cells, but Big Immune Regulatory Functions. Annu Rev Immunol 2018; 37:125-144. [PMID: 30485751 DOI: 10.1146/annurev-immunol-042718-041607] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Platelets have dual physiologic roles as both cellular mediators of thrombosis and immune modulatory cells. Historically, the thrombotic function of platelets has received significant research and clinical attention, but emerging research indicates that the immune regulatory roles of platelets may be just as important. We now know that in addition to their role in the acute thrombotic event at the time of myocardial infarction, platelets initiate and accelerate inflammatory processes that are part of the pathogenesis of atherosclerosis and myocardial infarction expansion. Furthermore, it is increasingly apparent from recent studies that platelets impact the pathogenesis of many vascular inflammatory processes such as autoimmune diseases, sepsis, viral infections, and growth and metastasis of many types of tumors. Therefore, we must consider platelets as immune cells that affect all phases of immune responses.
Collapse
Affiliation(s)
- Craig N Morrell
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine, Rochester, New York 14642, USA;
| | - Daphne N Pariser
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine, Rochester, New York 14642, USA;
| | - Zachary T Hilt
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine, Rochester, New York 14642, USA;
| | - Denisse Vega Ocasio
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine, Rochester, New York 14642, USA;
| |
Collapse
|
44
|
The antiphospholipid syndrome finally fathomed? Blood 2018; 131:2091-2092. [PMID: 29748168 DOI: 10.1182/blood-2018-03-837880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|