1
|
Hong G, Zhou Y, Yang S, Yan S, Lu J, Xu B, Zhan Z, Jiang H, Wei B, Wang J. Metformin acts on miR-181a-5p/PAI-1 axis in stem cells providing new strategies for improving age-related osteogenic differentiation decline. Stem Cells 2024; 42:1055-1069. [PMID: 39283761 DOI: 10.1093/stmcls/sxae057] [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: 04/02/2024] [Accepted: 08/26/2024] [Indexed: 12/12/2024]
Abstract
A general decline in the osteogenic differentiation capacity of human bone marrow mesenchymal stem cells (hBMSCs) in the elderly is a clinical consensus, with diverse opinions on the mechanisms. Many studies have demonstrated that metformin (MF) significantly protects against osteoporosis and reduces fracture risk. However, the exact mechanism of this effect remains unclear. In this study, we found that the decreased miR-181a-5p expression triggered by MF treatment plays a critical role in recovering the osteogenic ability of aging hBMSCs (derived from elderly individuals). Notably, the miR-181a-5p expression in hBMSCs was significantly decreased with prolonged MF (1000 μM) treatment. Further investigation revealed that miR-181a-5p overexpression markedly impairs the osteogenic ability of hBMSCs, while miR-181a-5p inhibition reveals the opposite result. We also found that miR-181a-5p could suppress the protein translation process of plasminogen activator inhibitor-1 (PAI-1), as evidenced by luciferase assays and Western blots. Additionally, low PAI-1 levels were associated with diminished osteogenic ability, whereas high levels promoted it. These findings were further validated in human umbilical cord mesenchymal stem cells (hUCMSCs). Finally, our in vivo experiment with a bone defects rat model confirmed that the agomiR-181a-5p (long-lasting miR-181a-5p mimic) undermined bone defects recovery, while the antagomiR-181a-5p (long-lasting miR-181a-5p inhibitor) significantly promoted the bone defects recovery. In conclusion, we found that MF promotes bone tissue regeneration through the miR-181a-5p/PAI-1 axis by affecting MSC osteogenic ability, providing new strategies for the treatment of age-related bone regeneration disorders.
Collapse
Affiliation(s)
- Guanhao Hong
- Stem Cell Research and Cellular Therapy Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, People's Republic of China
| | - Yulan Zhou
- Reproductive Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, People's Republic of China
| | - Shukai Yang
- Orthopedic Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, People's Republic of China
| | - Shouquan Yan
- Stem Cell Research and Cellular Therapy Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, People's Republic of China
| | - Jiaxu Lu
- Stem Cell Research and Cellular Therapy Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, People's Republic of China
| | - Bo Xu
- Stem Cell Research and Cellular Therapy Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, People's Republic of China
| | - Zeyu Zhan
- Orthopedic Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, People's Republic of China
| | - Huasheng Jiang
- Orthopedic Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, People's Republic of China
| | - Bo Wei
- Orthopedic Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, People's Republic of China
| | - Jiafeng Wang
- Stem Cell Research and Cellular Therapy Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, People's Republic of China
| |
Collapse
|
2
|
Hong NE, Chaplin A, Di L, Ravodina A, Bevan GH, Gao H, Asase C, Gangwar RS, Cameron MJ, Mignery M, Cherepanova O, Finn AV, Nayak L, Pieper AA, Maiseyeu A. Nanoparticle-based itaconate treatment recapitulates low-cholesterol/low-fat diet-induced atherosclerotic plaque resolution. Cell Rep 2024; 43:114911. [PMID: 39466775 DOI: 10.1016/j.celrep.2024.114911] [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: 04/26/2024] [Revised: 08/22/2024] [Accepted: 10/09/2024] [Indexed: 10/30/2024] Open
Abstract
Current pharmacologic treatments for atherosclerosis do not completely protect patients; additional protection can be achieved by dietary modifications, such as a low-cholesterol/low-fat diet (LCLFD), that mediate plaque stabilization and inflammation reduction. However, this lifestyle modification can be challenging for patients. Unfortunately, incomplete understanding of the underlying mechanisms has thwarted efforts to mimic the protective effects of a LCLFD. Here, we report that the tricarboxylic acid cycle intermediate itaconate (ITA), produced by plaque macrophages, is key to diet-induced plaque resolution. ITA is produced by immunoresponsive gene 1 (IRG1), which we observe is highly elevated in myeloid cells of vulnerable plaques and absent from early or stable plaques in mice and humans. We additionally report development of an ITA-conjugated lipid nanoparticle that accumulates in plaque and bone marrow myeloid cells, epigenetically reduces inflammation via H3K27ac deacetylation, and reproduces the therapeutic effects of LCLFD-induced plaque resolution in multiple atherosclerosis models.
Collapse
Affiliation(s)
- Natalie E Hong
- Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cleveland, OH, USA; Department of Biomedical Engineering, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Alice Chaplin
- Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Lin Di
- Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cleveland, OH, USA; Department of Biomedical Engineering, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Anastasia Ravodina
- Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Graham H Bevan
- Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Huiyun Gao
- Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Courteney Asase
- Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Roopesh Singh Gangwar
- Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cleveland, OH, USA; Division of Allergy and Immunology, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Mark J Cameron
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Matthew Mignery
- Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Olga Cherepanova
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Aloke V Finn
- Department of Internal Medicine, Cardiovascular Division, University of Maryland School of Medicine, Baltimore, MD, USA; CVPath Institute, Inc., Gaithersburg, MD, USA
| | - Lalitha Nayak
- Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cleveland, OH, USA; Department of Hematology & Oncology, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Andrew A Pieper
- Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA; Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes VA Medical Center, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA; Department of Pathology, Case Western Reserve University, Cleveland, OH, USA; Department of Neurosciences, Case Western Reserve University, Cleveland, OH, USA
| | - Andrei Maiseyeu
- Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
3
|
dos Santos TW, Pereira QC, Fortunato IM, Oliveira FDS, Alvarez MC, Ribeiro ML. Body Composition and Senescence: Impact of Polyphenols on Aging-Associated Events. Nutrients 2024; 16:3621. [PMID: 39519454 PMCID: PMC11547493 DOI: 10.3390/nu16213621] [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: 09/09/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
Aging is a dynamic and progressive process characterized by the gradual accumulation of cellular damage. The continuous functional decline in the intrinsic capacity of living organisms to precisely regulate homeostasis leads to an increased susceptibility and vulnerability to diseases. Among the factors contributing to these changes, body composition-comprised of fat mass and lean mass deposits-plays a crucial role in the trajectory of a disability. Particularly, visceral and intermuscular fat deposits increase with aging and are associated with adverse health outcomes, having been linked to the pathogenesis of sarcopenia. Adipose tissue is involved in the secretion of bioactive factors that can ultimately mediate inter-organ pathology, including skeletal muscle pathology, through the induction of a pro-inflammatory profile such as a SASP, cellular senescence, and immunosenescence, among other events. Extensive research has shown that natural compounds have the ability to modulate the mechanisms associated with cellular senescence, in addition to exhibiting anti-inflammatory, antioxidant, and immunomodulatory potential, making them interesting strategies for promoting healthy aging. In this review, we will discuss how factors such as cellular senescence and the presence of a pro-inflammatory phenotype can negatively impact body composition and lead to the development of age-related diseases, as well as how the use of polyphenols can be a functional measure for restoring balance, maintaining tissue quality and composition, and promoting health.
Collapse
Affiliation(s)
- Tanila Wood dos Santos
- Laboratory of Immunopharmacology and Molecular Biology, Sao Francisco University, Av. Sao Francisco de Assis, 218, Braganca Paulista 12916-900, SP, Brazil; (T.W.d.S.); (Q.C.P.); (I.M.F.); (F.d.S.O.); (M.C.A.)
| | - Quélita Cristina Pereira
- Laboratory of Immunopharmacology and Molecular Biology, Sao Francisco University, Av. Sao Francisco de Assis, 218, Braganca Paulista 12916-900, SP, Brazil; (T.W.d.S.); (Q.C.P.); (I.M.F.); (F.d.S.O.); (M.C.A.)
| | - Isabela Monique Fortunato
- Laboratory of Immunopharmacology and Molecular Biology, Sao Francisco University, Av. Sao Francisco de Assis, 218, Braganca Paulista 12916-900, SP, Brazil; (T.W.d.S.); (Q.C.P.); (I.M.F.); (F.d.S.O.); (M.C.A.)
| | - Fabrício de Sousa Oliveira
- Laboratory of Immunopharmacology and Molecular Biology, Sao Francisco University, Av. Sao Francisco de Assis, 218, Braganca Paulista 12916-900, SP, Brazil; (T.W.d.S.); (Q.C.P.); (I.M.F.); (F.d.S.O.); (M.C.A.)
| | - Marisa Claudia Alvarez
- Laboratory of Immunopharmacology and Molecular Biology, Sao Francisco University, Av. Sao Francisco de Assis, 218, Braganca Paulista 12916-900, SP, Brazil; (T.W.d.S.); (Q.C.P.); (I.M.F.); (F.d.S.O.); (M.C.A.)
- Hematology and Transfusion Medicine Center, University of Campinas/Hemocentro, UNICAMP, Rua Carlos Chagas 480, Campinas 13083-878, SP, Brazil
| | - Marcelo Lima Ribeiro
- Laboratory of Immunopharmacology and Molecular Biology, Sao Francisco University, Av. Sao Francisco de Assis, 218, Braganca Paulista 12916-900, SP, Brazil; (T.W.d.S.); (Q.C.P.); (I.M.F.); (F.d.S.O.); (M.C.A.)
| |
Collapse
|
4
|
Khoukaz HB, Vadali M, Schoenherr A, Ramirez-Perez FI, Morales-Quinones M, Sun Z, Fujie S, Foote CA, Lyu Z, Zeng S, Augenreich MA, Cai D, Chen SY, Joshi T, Ji Y, Hill MA, Martinez-Lemus LA, Fay WP. PAI-1 Regulates the Cytoskeleton and Intrinsic Stiffness of Vascular Smooth Muscle Cells. Arterioscler Thromb Vasc Biol 2024; 44:2191-2203. [PMID: 38868940 PMCID: PMC11424258 DOI: 10.1161/atvbaha.124.320938] [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: 04/09/2024] [Accepted: 06/01/2024] [Indexed: 06/14/2024]
Abstract
BACKGROUND Plasma concentration of PAI-1 (plasminogen activator inhibitor-1) correlates with arterial stiffness. Vascular smooth muscle cells (SMCs) express PAI-1, and the intrinsic stiffness of SMCs is a major determinant of total arterial stiffness. We hypothesized that PAI-1 promotes SMC stiffness by regulating the cytoskeleton and that pharmacological inhibition of PAI-1 decreases SMC and aortic stiffness. METHODS PAI-039, a specific inhibitor of PAI-1, and small interfering RNA were used to inhibit PAI-1 expression in cultured human SMCs. Effects of PAI-1 inhibition on SMC stiffness, F-actin (filamentous actin) content, and cytoskeleton-modulating enzymes were assessed. WT (wild-type) and PAI-1-deficient murine SMCs were used to determine PAI-039 specificity. RNA sequencing was performed to determine the effects of PAI-039 on SMC gene expression. In vivo effects of PAI-039 were assessed by aortic pulse wave velocity. RESULTS PAI-039 significantly reduced intrinsic stiffness of human SMCs, which was accompanied by a significant decrease in cytoplasmic F-actin content. PAI-1 gene knockdown also decreased cytoplasmic F-actin. PAI-1 inhibition significantly increased the activity of cofilin, an F-actin depolymerase, in WT murine SMCs, but not in PAI-1-deficient SMCs. RNA-sequencing analysis suggested that PAI-039 upregulates AMPK (AMP-activated protein kinase) signaling in SMCs, which was confirmed by Western blotting. Inhibition of AMPK prevented activation of cofilin by PAI-039. In mice, PAI-039 significantly decreased aortic stiffness and tunica media F-actin content without altering the elastin or collagen content. CONCLUSIONS PAI-039 decreases intrinsic SMC stiffness and cytoplasmic stress fiber content. These effects are mediated by AMPK-dependent activation of cofilin. PAI-039 also decreases aortic stiffness in vivo. These findings suggest that PAI-1 is an important regulator of the SMC cytoskeleton and that pharmacological inhibition of PAI-1 has the potential to prevent and treat cardiovascular diseases involving arterial stiffening.
Collapse
Affiliation(s)
- Hekmat B Khoukaz
- Departments of Medicine (H.B.K., M.V., F.I.R.-P., M.M.-Q., Y.J., L.A.M.-L., W.P.F.), University of Missouri, Columbia
| | - Manisha Vadali
- Departments of Medicine (H.B.K., M.V., F.I.R.-P., M.M.-Q., Y.J., L.A.M.-L., W.P.F.), University of Missouri, Columbia
| | - Alex Schoenherr
- Medical Pharmacology and Physiology (A.S., C.A.F., S.-Y.C., M.A.H., L.A.M.-L., W.P.F.), University of Missouri, Columbia
| | - Francisco I Ramirez-Perez
- Departments of Medicine (H.B.K., M.V., F.I.R.-P., M.M.-Q., Y.J., L.A.M.-L., W.P.F.), University of Missouri, Columbia
| | - Mariana Morales-Quinones
- Departments of Medicine (H.B.K., M.V., F.I.R.-P., M.M.-Q., Y.J., L.A.M.-L., W.P.F.), University of Missouri, Columbia
| | - Zhe Sun
- Dalton Cardiovascular Research Center (Z.S., M.A.H., L.A.M.-L., W.P.F.), University of Missouri, Columbia
| | - Shumpei Fujie
- Faculty of Sport and Health Science, Ritsumeikan University, Kusatsu, Shiga, Japan (S.F.)
| | - Christopher A Foote
- Medical Pharmacology and Physiology (A.S., C.A.F., S.-Y.C., M.A.H., L.A.M.-L., W.P.F.), University of Missouri, Columbia
| | - Zhen Lyu
- Electrical Engineering and Computer Science (Z.L., S.Z.), University of Missouri, Columbia
| | - Shuai Zeng
- Electrical Engineering and Computer Science (Z.L., S.Z.), University of Missouri, Columbia
| | - Marc A Augenreich
- Nutrition and Exercise Physiology (M.A.A.), University of Missouri, Columbia
| | - Dunpeng Cai
- Surgery (D.C., S.-Y.C.), University of Missouri, Columbia
| | - Shi-You Chen
- Medical Pharmacology and Physiology (A.S., C.A.F., S.-Y.C., M.A.H., L.A.M.-L., W.P.F.), University of Missouri, Columbia
- Surgery (D.C., S.-Y.C.), University of Missouri, Columbia
- Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, MO (S.-Y.C., W.P.F.)
| | - Trupti Joshi
- Health Management and Informatics (T.J.), University of Missouri, Columbia
| | - Yan Ji
- Departments of Medicine (H.B.K., M.V., F.I.R.-P., M.M.-Q., Y.J., L.A.M.-L., W.P.F.), University of Missouri, Columbia
| | - Michael A Hill
- Medical Pharmacology and Physiology (A.S., C.A.F., S.-Y.C., M.A.H., L.A.M.-L., W.P.F.), University of Missouri, Columbia
- Dalton Cardiovascular Research Center (Z.S., M.A.H., L.A.M.-L., W.P.F.), University of Missouri, Columbia
| | - Luis A Martinez-Lemus
- Departments of Medicine (H.B.K., M.V., F.I.R.-P., M.M.-Q., Y.J., L.A.M.-L., W.P.F.), University of Missouri, Columbia
- Medical Pharmacology and Physiology (A.S., C.A.F., S.-Y.C., M.A.H., L.A.M.-L., W.P.F.), University of Missouri, Columbia
- Dalton Cardiovascular Research Center (Z.S., M.A.H., L.A.M.-L., W.P.F.), University of Missouri, Columbia
| | - William P Fay
- Departments of Medicine (H.B.K., M.V., F.I.R.-P., M.M.-Q., Y.J., L.A.M.-L., W.P.F.), University of Missouri, Columbia
- Medical Pharmacology and Physiology (A.S., C.A.F., S.-Y.C., M.A.H., L.A.M.-L., W.P.F.), University of Missouri, Columbia
- Dalton Cardiovascular Research Center (Z.S., M.A.H., L.A.M.-L., W.P.F.), University of Missouri, Columbia
- Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, MO (S.-Y.C., W.P.F.)
| |
Collapse
|
5
|
Castro-Leyva V, Manuel-Apolinar L, Basurto-Acevedo NE, Basurto L, González-Chávez A, Ruiz-Gastelum E, Martínez-Murillo C. Metabolic Changes Induced by Bariatric Surgery May be Mediated by PAI-1 and PCSK9 Crosstalk. Arch Med Res 2024; 55:103032. [PMID: 38971127 DOI: 10.1016/j.arcmed.2024.103032] [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: 01/29/2024] [Revised: 06/01/2024] [Accepted: 06/12/2024] [Indexed: 07/08/2024]
Abstract
BACKGROUND Adiposity favors several metabolic disorders with an exacerbated chronic pro-inflammatory status and tissue damage, with high levels of plasminogen activator inhibitor type 1 (PAI-1) and proprotein convertase subtilisin/kexin type 9 (PCSK9). OBJECTIVE To demonstrate the influence of bariatric surgery on the crosstalk between PAI-1 and PCSK9 to regulate metabolic markers. METHODS Observational and longitudinal study of 190 patients with obesity and obesity-related comorbidities who underwent bariatric surgery. We measured, before and after bariatric surgery, the anthropometric variables and we performed biochemical analysis by standard methods (glucose, insulin, triglycerides [TG], total cholesterol, high-density lipoprotein cholesterol [HDL-C], low-density lipoprotein cholesterol [LDL-C] and TG/HDL-C ratio, PAI-1 and PCSK9 were measured by ELISA). RESULTS PAI-1 levels decreased significantly after bariatric surgery, and were positively correlated with lipids, glucose, and TG, with significance on PCSK9 and TG/HDL-C alleviating the insulin resistance (IR) and inducing a state reversal of type 2 diabetes (T2D) with a significant decrease in body weight and BMI (p <0.0001). Multivariate regression analysis predicted a functional model in which PAI-1 acts as a regulator of PCSK9 (p <0.002), TG (p <0.05), and BMI; at the same time, PCSK9 modulates LDL-C HDL-C and PAI-1. CONCLUSIONS After bariatric surgery, we found a positive association and crosstalk between PAI-1 and PCSK9, which modulates the delicate balance of cholesterol, favoring the decrease of circulating lipids, TG, and PAI-1, which influences the glucose levels with amelioration of IR and T2D, demonstrating the crosstalk between fibrinolysis and lipid metabolism, the two main factors involved in atherosclerosis and cardiovascular disease in human obesity.
Collapse
Affiliation(s)
- Violeta Castro-Leyva
- Unidad de Investigación Médica en Enfermedades Endocrinas, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Leticia Manuel-Apolinar
- Unidad de Investigación Médica en Enfermedades Endocrinas, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Norma Eleane Basurto-Acevedo
- Servicio de Cirugía General, Clínica de Tracto Digestivo Superior, Cirugía Bariátrica y Metabólica, Hospital General de México Dr. Eduardo Liceaga, Mexico City, Mexico
| | - Lourdes Basurto
- Unidad de Investigación Médica en Enfermedades Endocrinas, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico.
| | - Antonio González-Chávez
- Clínica para la Atención Integral del Paciente con Diabetes y Obesidad, Hospital General de México Dr. Eduardo Liceaga, Mexico City, Mexico
| | - Edith Ruiz-Gastelum
- Servicio de Cardiología, Instituto de Seguridad y Servicios Sociales de los Trabajadores del Estado, Sonora, Mexico
| | - Carlos Martínez-Murillo
- Departamento de Hematología. Hospital General de México Dr. Eduardo Liceaga, Mexico City, Mexico
| |
Collapse
|
6
|
Fujita M, Sasada M, Iyoda T, Fukai F. Involvement of Matricellular Proteins in Cellular Senescence: Potential Therapeutic Targets for Age-Related Diseases. Int J Mol Sci 2024; 25:6591. [PMID: 38928297 PMCID: PMC11204155 DOI: 10.3390/ijms25126591] [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: 05/29/2024] [Revised: 06/10/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
Senescence is a physiological and pathological cellular program triggered by various types of cellular stress. Senescent cells exhibit multiple characteristic changes. Among them, the characteristic flattened and enlarged morphology exhibited in senescent cells is observed regardless of the stimuli causing the senescence. Several studies have provided important insights into pro-adhesive properties of cellular senescence, suggesting that cell adhesion to the extracellular matrix (ECM), which is involved in characteristic morphological changes, may play pivotal roles in cellular senescence. Matricellular proteins, a group of structurally unrelated ECM molecules that are secreted into the extracellular environment, have the unique ability to control cell adhesion to the ECM by binding to cell adhesion receptors, including integrins. Recent reports have certified that matricellular proteins are closely involved in cellular senescence. Through this biological function, matricellular proteins are thought to play important roles in the pathogenesis of age-related diseases, including fibrosis, osteoarthritis, intervertebral disc degeneration, atherosclerosis, and cancer. This review outlines recent studies on the role of matricellular proteins in inducing cellular senescence. We highlight the role of integrin-mediated signaling in inducing cellular senescence and provide new therapeutic options for age-related diseases targeting matricellular proteins and integrins.
Collapse
Affiliation(s)
- Motomichi Fujita
- Department of Molecular Patho-Physiology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda 278-8510, Chiba, Japan
| | - Manabu Sasada
- Clinical Research Center in Hiroshima, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-Ku, Hiroshima 734-8551, Japan
| | - Takuya Iyoda
- Department of Pharmacy, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, 1-1-1 Daigaku-Doori, Sanyo-Onoda 756-0884, Yamaguchi, Japan
| | - Fumio Fukai
- Department of Molecular Patho-Physiology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda 278-8510, Chiba, Japan
| |
Collapse
|
7
|
Varra FN, Varras M, Varra VK, Theodosis-Nobelos P. Molecular and pathophysiological relationship between obesity and chronic inflammation in the manifestation of metabolic dysfunctions and their inflammation‑mediating treatment options (Review). Mol Med Rep 2024; 29:95. [PMID: 38606791 PMCID: PMC11025031 DOI: 10.3892/mmr.2024.13219] [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] [Accepted: 01/17/2024] [Indexed: 04/13/2024] Open
Abstract
Obesity reaches up to epidemic proportions globally and increases the risk for a wide spectrum of co‑morbidities, including type‑2 diabetes mellitus (T2DM), hypertension, dyslipidemia, cardiovascular diseases, non‑alcoholic fatty liver disease, kidney diseases, respiratory disorders, sleep apnea, musculoskeletal disorders and osteoarthritis, subfertility, psychosocial problems and certain types of cancers. The underlying inflammatory mechanisms interconnecting obesity with metabolic dysfunction are not completely understood. Increased adiposity promotes pro‑inflammatory polarization of macrophages toward the M1 phenotype, in adipose tissue (AT), with subsequent increased production of pro‑inflammatory cytokines and adipokines, inducing therefore an overall, systemic, low‑grade inflammation, which contributes to metabolic syndrome (MetS), insulin resistance (IR) and T2DM. Targeting inflammatory mediators could be alternative therapies to treat obesity, but their safety and efficacy remains to be studied further and confirmed in future clinical trials. The present review highlights the molecular and pathophysiological mechanisms by which the chronic low‑grade inflammation in AT and the production of reactive oxygen species lead to MetS, IR and T2DM. In addition, focus is given on the role of anti‑inflammatory agents, in the resolution of chronic inflammation, through the blockade of chemotactic factors, such as monocytes chemotractant protein‑1, and/or the blockade of pro‑inflammatory mediators, such as IL‑1β, TNF‑α, visfatin, and plasminogen activator inhibitor‑1, and/or the increased synthesis of adipokines, such as adiponectin and apelin, in obesity‑associated metabolic dysfunction.
Collapse
Affiliation(s)
- Fani-Niki Varra
- Department of Pharmacy, School of Health Sciences, Frederick University, Nicosia 1036, Cyprus
- Medical School, Dimocritus University of Thrace, Alexandroupolis 68100, Greece
| | - Michail Varras
- Fourth Department of Obstetrics and Gynecology, ‘Elena Venizelou’ General Hospital, Athens 11521, Greece
| | | | | |
Collapse
|
8
|
Jin R, Forbes CM, Miller NL, Lafin J, Strand DW, Case T, Cates JM, Liu Q, Ramirez-Solano M, Mohler JL, Matusik RJ. Transcriptomic analysis of benign prostatic hyperplasia identifies critical pathways in prostatic overgrowth and 5-alpha reductase inhibitor resistance. Prostate 2024; 84:441-459. [PMID: 38168866 DOI: 10.1002/pros.24661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 11/06/2023] [Accepted: 12/05/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND The medical therapy of prostatic symptoms (MTOPS) trial randomized men with symptoms of benign prostatic hyperplasia (BPH) and followed response of treatment with a 5α-reductase inhibitor (5ARI), an alpha-adrenergic receptor antagonist (α-blocker), the combination of 5ARI and α-blocker or no medical therapy (none). Medical therapy reduced risk of clinical progression by 66% but the reasons for nonresponse or loss of therapeutic response in some patients remains unresolved. Our previous work showed that prostatic glucocorticoid levels are increased in 5ARI-treated patients and that glucocorticoids can increased branching of prostate epithelia in vitro. To understand the transcriptomic changes associated with 5ARI treatment, we performed bulk RNA sequencing of BPH and control samples from patients who received 5ARI versus those that did not. Deconvolution analysis was performed to estimate cellular composition. Bulk RNA sequencing was also performed on control versus glucocorticoid-treated prostate epithelia in 3D culture to determine underlying transcriptomic changes associated with branching morphogenesis. METHOD Surgical BPH (S-BPH) tissue was defined as benign prostatic tissue collected from the transition zone (TZ) of patients who failed medical therapy while control tissue termed Incidental BPH (I-BPH) was obtained from the TZ of men undergoing radical prostatectomy for low-volume/grade prostatic adenocarcinoma confined to the peripheral zone. S-BPH patients were divided into four subgroups: men on no medical therapy (none: n = 7), α-blocker alone (n = 10), 5ARI alone (n = 6) or combination therapy (α-blocker and 5ARI: n = 7). Control I-BPH tissue was from men on no medical therapy (none: n = 8) or on α-blocker (n = 6). A human prostatic cell line in 3D culture that buds and branches was used to identify genes involved in early prostatic growth. Snap-frozen prostatic tissue taken at the time of surgery and 3D organoids were used for RNA-seq analysis. Bulk RNAseq data were deconvoluted using CIBERSORTx. Differentially expressed genes (DEG) that were statistically significant among S-BPH, I-BPH, and during budding and branching of organoids were used for pathway analysis. RESULTS Transcriptomic analysis between S-BPH (n = 30) and I-BPH (n = 14) using a twofold cutoff (p < 0.05) identified 377 DEG (termed BPH377) and a cutoff < 0.05 identified 3377 DEG (termed BPH3377). Within the S-BPH, the subgroups none and α-blocker were compared to patients on 5ARI to reveal 361 DEG (termed 5ARI361) that were significantly changed. Deconvolution analysis of bulk RNA seq data with a human prostate single cell data set demonstrated increased levels of mast cells, NK cells, interstitial fibroblasts, and prostate luminal cells in S-BPH versus I-BPH. Glucocorticoid (GC)-induced budding and branching of benign prostatic cells in 3D culture was compared to control organoids to identify early events in prostatic morphogenesis. GC induced 369 DEG (termed GC359) in 3D culture. STRING analysis divided the large datasets into 20-80 genes centered around a hub. In general, biological processes induced in BPH supported growth and differentiation such as chromatin modification and DNA repair, transcription, cytoskeleton, mitochondrial electron transport, ubiquitination, protein folding, and cholesterol synthesis. Identified signaling pathways were pooled to create a list of DEG that fell into seven hubs/clusters. The hub gene centrality was used to name the network including AP-1, interleukin (IL)-6, NOTCH1 and NOTCH3, NEO1, IL-13, and HDAC/KDM. All hubs showed connections to inflammation, chromatin structure, and development. The same approach was applied to 5ARI361 giving multiple networks, but the EGF and sonic hedgehog (SHH) hub was of particular interest as a developmental pathway. The BPH3377, 5ARI363, and GC359 lists were compared and 67 significantly changed DEG were identified. Common genes to the 3D culture included an IL-6 hub that connected to genes identified in BPH hubs that defined AP1, IL-6, NOTCH, NEO1, IL-13, and HDAC/KDM. CONCLUSIONS Reduction analysis of BPH and 3D organoid culture uncovered networks previously identified in prostatic development as being reinitiated in BPH. Identification of these pathways provides insight into the failure of medical therapy for BPH and new therapeutic targets for BPH/LUTS.
Collapse
Affiliation(s)
- Renjie Jin
- Department of Urology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Connor M Forbes
- Department of Urology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Urology Department, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Nicole L Miller
- Department of Urology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - John Lafin
- Department of Urology, University of Texas, Southwestern, Dallas, Texas, USA
- Department of Urology, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Douglas W Strand
- Department of Urology, University of Texas, Southwestern, Dallas, Texas, USA
- Department of Urology, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Thomas Case
- Department of Urology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Justin M Cates
- Department of Pathology Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Qi Liu
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Marisol Ramirez-Solano
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - James L Mohler
- Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Robert J Matusik
- Department of Urology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
9
|
Xu Y, Hu Y, Wu G, Niu L, Fang C, Li Y, Jiang L, Yuan C, Huang M. Specific inhibition on PAI-1 reduces the dose of Alteplase for ischemic stroke treatment. Int J Biol Macromol 2024; 257:128618. [PMID: 38070813 DOI: 10.1016/j.ijbiomac.2023.128618] [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: 09/08/2023] [Revised: 11/30/2023] [Accepted: 12/02/2023] [Indexed: 12/21/2023]
Abstract
Administration of recombinant tPA (rtPA, or trade name Alteplase®) is an FDA-approved therapy for acute ischemic stroke (AIS), but poses the risk of hemorrhagic complications. Recombinant tPA can be rapidly inactivated by the endogenous inhibitor, plasminogen activator inhibitor 1 (PAI-1). In this work, we study a novel treatment approach that combines a PAI-1 inhibitor, PAItrap4, with a reduced dose of rtPA to address the hemorrhagic concern of rtPA. PAItrap4 is a highly specific and very potent protein-based inhibitor of PAI-1, comprising of a variant of uPA serine protease domain, human serum albumin, and a cyclic RGD peptide. PAItrap4 efficiently targets and inhibits PAI-1 on activated platelets, and also possesses a long half-life in vivo. Our results demonstrate that PAItrap4 effectively counteracts the inhibitory effects of PAI-1 on rtPA, preserving rtPA activity based on amidolytic and clot lysis assays. In an in vivo murine stroke model, PAItrap4, together with low-dose rtPA, enhances the blood perfusion in the stroke-affected areas, reduces infarct size, and promotes neurological recovery in mice. Importantly, such treatment does not increase the amount of cerebral hemorrhage, thus reducing the risk of cerebral hemorrhage. In addition, PAItrap4 does not compromise the normal blood coagulation function in mice, demonstrating its safety as a therapeutic agent. These findings highlight this combination therapy as a promising alternative for the treatment of ischemic stroke, offering improved safety and efficacy.
Collapse
Affiliation(s)
- Yanyan Xu
- College of Chemical Engineering, Fuzhou University, Fujian 350108, China; National Joint Research Center on Biomedical Photodynamic Technology, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Yinping Hu
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, China; National Joint Research Center on Biomedical Photodynamic Technology, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Guangqian Wu
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, China; National Joint Research Center on Biomedical Photodynamic Technology, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Lili Niu
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, China; National Joint Research Center on Biomedical Photodynamic Technology, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Chao Fang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Yongkun Li
- Department of Neurology, Fujian Provincial Hospital, Shengli Clinical College of Fujian Medical University, No. 134 Dong Street, Fuzhou, Fujian 350001, China
| | - Longguang Jiang
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, China; National Joint Research Center on Biomedical Photodynamic Technology, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Cai Yuan
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian, 350108, China; National Joint Research Center on Biomedical Photodynamic Technology, Fuzhou University, Fuzhou, Fujian 350108, China.
| | - Mingdong Huang
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, China; National Joint Research Center on Biomedical Photodynamic Technology, Fuzhou University, Fuzhou, Fujian 350108, China.
| |
Collapse
|
10
|
Zhao Q, Cheng N, Sun X, Yan L, Li W. The application of nanomedicine in clinical settings. Front Bioeng Biotechnol 2023; 11:1219054. [PMID: 37441195 PMCID: PMC10335748 DOI: 10.3389/fbioe.2023.1219054] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 06/05/2023] [Indexed: 07/15/2023] Open
Abstract
As nanotechnology develops in the fields of mechanical engineering, electrical engineering, information and communication, and medical care, it has shown great promises. In recent years, medical nanorobots have made significant progress in terms of the selection of materials, fabrication methods, driving force sources, and clinical applications, such as nanomedicine. It involves bypassing biological tissues and delivering drugs directly to lesions and target cells using nanorobots, thus increasing concentration. It has also proved useful for monitoring disease progression, complementary diagnosis, and minimally invasive surgery. Also, we examine the development of nanomedicine and its applications in medicine, focusing on the use of nanomedicine in the treatment of various major diseases, including how they are generalized and how they are modified. The purpose of this review is to provide a summary and discussion of current research for the future development in nanomedicine.
Collapse
Affiliation(s)
- Qingsong Zhao
- Postdoctoral Programme of Meteria Medica Institute of Harbin University of Commerce, Harbin, China
| | - Nuo Cheng
- Department of Endocrinology, The Fourth Hospital of Harbin Medical University, Harbin, China
| | - Xuyan Sun
- Department of Endocrinology, The Fourth Hospital of Harbin Medical University, Harbin, China
| | - Lijun Yan
- Postdoctoral Programme of Meteria Medica Institute of Harbin University of Commerce, Harbin, China
| | - Wenlan Li
- Postdoctoral Programme of Meteria Medica Institute of Harbin University of Commerce, Harbin, China
| |
Collapse
|
11
|
Kopytek M, Ząbczyk M, Mazur P, Undas A, Natorska J. PAI-1 Overexpression in Valvular Interstitial Cells Contributes to Hypofibrinolysis in Aortic Stenosis. Cells 2023; 12:1402. [PMID: 37408236 PMCID: PMC10216522 DOI: 10.3390/cells12101402] [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: 03/23/2023] [Revised: 05/11/2023] [Accepted: 05/15/2023] [Indexed: 07/07/2023] Open
Abstract
Aortic stenosis (AS) is associated with hypofibrinolysis, but its mechanism is poorly understood. We investigated whether LDL cholesterol affects plasminogen activator inhibitor 1 (PAI-1) expression, which may contribute to hypofibrinolysis in AS. Stenotic valves were obtained from 75 severe AS patients during valve replacement to assess lipids accumulation, together with PAI-1 and nuclear factor-κB (NF-κB) expression. Five control valves from autopsy healthy individuals served as controls. The expression of PAI-1 in valve interstitial cells (VICs) after LDL stimulation was assessed at protein and mRNA levels. PAI-1 activity inhibitor (TM5275) and NF-κB inhibitor (BAY 11-7082) were used to suppress PAI-1 activity or NF-κB pathway. Clot lysis time (CLT) was performed to assess fibrinolytic capacity in VICs cultures. Solely AS valves showed PAI-1 expression, the amount of which was correlated with lipid accumulation and AS severity and co-expressed with NF-κB. In vitro VICs showed abundant PAI-1 expression. LDL stimulation increased PAI-1 levels in VICs supernatants and prolonged CLT. PAI-1 activity inhibition shortened CLT, while NF-κB inhibition decreased PAI-1 and SERPINE1 expression in VICs, its level in supernatants and shortened CLT. In severe AS, valvular PAI-1 overexpression driven by lipids accumulation contributes to hypofibrinolysis and AS severity.
Collapse
Affiliation(s)
- Magdalena Kopytek
- Thromboembolic Disorders Department, Institute of Cardiology, Jagiellonian University Medical College, 80 Pradnicka St., 31-202 Krakow, Poland; (M.K.); (M.Z.); (A.U.)
- Krakow Centre for Medical Research and Technologies, John Paul II Hospital, 80 Pradnicka St., 31-202 Krakow, Poland
| | - Michał Ząbczyk
- Thromboembolic Disorders Department, Institute of Cardiology, Jagiellonian University Medical College, 80 Pradnicka St., 31-202 Krakow, Poland; (M.K.); (M.Z.); (A.U.)
- Krakow Centre for Medical Research and Technologies, John Paul II Hospital, 80 Pradnicka St., 31-202 Krakow, Poland
| | - Piotr Mazur
- Department of Cardiovascular Surgery, Mayo Clinic, 200 First St. SW, Rochester, MN 55905, USA
- Department of Cardiovascular Surgery and Transplantology, Institute of Cardiology, Jagiellonian University Medical College, 80 Pradnicka St., 31-202 Krakow, Poland
| | - Anetta Undas
- Thromboembolic Disorders Department, Institute of Cardiology, Jagiellonian University Medical College, 80 Pradnicka St., 31-202 Krakow, Poland; (M.K.); (M.Z.); (A.U.)
- Krakow Centre for Medical Research and Technologies, John Paul II Hospital, 80 Pradnicka St., 31-202 Krakow, Poland
| | - Joanna Natorska
- Thromboembolic Disorders Department, Institute of Cardiology, Jagiellonian University Medical College, 80 Pradnicka St., 31-202 Krakow, Poland; (M.K.); (M.Z.); (A.U.)
- Krakow Centre for Medical Research and Technologies, John Paul II Hospital, 80 Pradnicka St., 31-202 Krakow, Poland
| |
Collapse
|
12
|
Gunner CB, Azmoon P, Mantuano E, Das L, Zampieri C, Pizzo SV, Gonias SL. An antibody that targets cell-surface glucose-regulated protein-78 inhibits expression of inflammatory cytokines and plasminogen activator inhibitors by macrophages. J Cell Biochem 2023; 124:743-752. [PMID: 36947703 PMCID: PMC10200756 DOI: 10.1002/jcb.30401] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 03/02/2023] [Accepted: 03/12/2023] [Indexed: 03/24/2023]
Abstract
Glucose-regulated protein-78 (Grp78) is an endoplasmic reticulum chaperone, which is secreted by cells and associates with cell surfaces, where it functions as a receptor for activated α2 -macroglobulin (α2 M) and tissue-type plasminogen activator (tPA). In macrophages, α2 M and tPA also bind to the transmembrane receptor, LDL receptor-related protein-1 (LRP1), activating a cell-signaling receptor assembly that includes the NMDA receptor (NMDA-R) to suppress innate immunity. Herein, we demonstrate that an antibody targeting Grp78 (N88) inhibits NFκB activation and expression of proinflammatory cytokines in bone marrow-derived macrophages (BMDMs) treated with the toll-like receptor-4 (TLR4) ligand, lipopolysaccharide, or with agonists that activate TLR2, TLR7, or TLR9. Pharmacologic inhibition of the NMDA-R or deletion of the gene encoding LRP1 (Lrp1) in BMDMs neutralizes the activity of N88. The fibrinolysis protease inhibitor, plasminogen activator inhibitor-1 (PAI1), has been implicated in diverse diseases including metabolic syndrome, cardiovascular disease, and type 2 diabetes. Deletion of Lrp1 independently increased expression of PAI1 and PAI2 in BMDMs, as did treatment of wild-type BMDMs with TLR agonists. tPA, α2 M, and N88 inhibited expression of PAI1 and PAI2 in BMDMs treated with TLR-activating agents. Inhibiting Src family kinases blocked the ability of both N88 and tPA to function as anti-inflammatory agents, suggesting that the cell-signaling pathway activated by tPA and N88, downstream of LRP1 and the NMDA-R, may be equivalent. We conclude that targeting cell-surface Grp78 may be effective in suppressing innate immunity by a mechanism that requires LRP1 and the NMDA-R.
Collapse
Affiliation(s)
- Cory B. Gunner
- Department of Pathology, University of San Diego California School of Medicine, La Jolla, CA, USA
| | - Pardis Azmoon
- Department of Pathology, University of San Diego California School of Medicine, La Jolla, CA, USA
| | - Elisabetta Mantuano
- Department of Pathology, University of San Diego California School of Medicine, La Jolla, CA, USA
| | - Lipsa Das
- Department of Pathology, University of San Diego California School of Medicine, La Jolla, CA, USA
| | - Carlotta Zampieri
- Department of Pathology, University of San Diego California School of Medicine, La Jolla, CA, USA
| | - Salvatore V. Pizzo
- Department of Pathology, Duke University Medical Center, Durham, NC, USA
| | - Steven L. Gonias
- Department of Pathology, University of San Diego California School of Medicine, La Jolla, CA, USA
| |
Collapse
|
13
|
Ghosh AK, Kalousdian AA, Shang M, Lux E, Eren M, Keating A, Wilsbacher LD, Vaughan DE. Cardiomyocyte PAI-1 influences the cardiac transcriptome and limits the extent of cardiac fibrosis in response to left ventricular pressure overload. Cell Signal 2023; 104:110555. [PMID: 36584735 DOI: 10.1016/j.cellsig.2022.110555] [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: 09/25/2022] [Revised: 11/26/2022] [Accepted: 12/02/2022] [Indexed: 12/29/2022]
Abstract
Plasminogen activator inhibitor-1 (PAI-1) is a specific and rapid-acting inhibitor of endogenous plasminogen activators (uPA and tPA). The global PAI-1 knockout mice (PAI-1KO) develop age-dependent cardiac-selective fibrosis, and young global PAI-1KO mice exhibit augmented susceptibility to developing cardiac fibrosis in response to hypertension. Here, we tested the hypothesis that cardiomyocyte PAI-1 is necessary to provide cardioprotective effects in a left ventricular pressure overload-induced murine model of cardiac hypertrophy and fibrosis using cardiomyocyte-specific PAI-1 knockout (cmPAI-1KO) mice. The results revealed that cmPAI-1KO mice display significantly worse cardiac fibrosis than controls. To investigate the molecular mechanisms responsible for these effects, genome-wide cardiac transcriptome analysis was performed. Loss of cardiomyocyte PAI-1 led to differential expression of 978 genes compared to controls in response to left ventricular pressure overload. Pathway enrichment analysis identified the inflammatory response, cell substrate adhesion, regulation of cytokine production, leukocyte migration, extracellular matrix organization, and cytokine-mediated signaling pathways as being significantly upregulated in cmPAI-1KO hearts. Conversely, specific epigenetic repressors, cation transmembrane transport, muscle system processes, and nitric oxide signaling were significantly downregulated in cmPAI-1KO hearts compared to control hearts in response to left ventricular pressure overload. Collectively, the present study provides strong evidence of the impact of cardiomyocyte PAI-1 in regulation of the transcriptome network involved in the cardiac stress response. In response to stress, the deregulatory impact of cardiomyocyte PAI-1 loss on the cardiac transcriptome may be the underlying cause of cardiac-selective accelerated fibrogenesis in global PAI-1-deficient mice.
Collapse
Affiliation(s)
- Asish K Ghosh
- Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| | - Anthony A Kalousdian
- Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Meng Shang
- Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Elizabeth Lux
- Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Mesut Eren
- Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Anna Keating
- Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Lisa D Wilsbacher
- Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Douglas E Vaughan
- Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
14
|
Phair IR, Nisr RB, Howden AJM, Sovakova M, Alqurashi N, Foretz M, Lamont D, Viollet B, Rena G. AMPK integrates metabolite and kinase-based immunometabolic control in macrophages. Mol Metab 2023; 68:101661. [PMID: 36586434 PMCID: PMC9842865 DOI: 10.1016/j.molmet.2022.101661] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 11/25/2022] [Accepted: 12/16/2022] [Indexed: 12/29/2022] Open
Abstract
OBJECTIVE Previous mechanistic studies on immunometabolism have focused on metabolite-based paradigms of regulation, such as itaconate. Here, we, demonstrate integration of metabolite and kinase-based immunometabolic control by AMP kinase. METHODS We combined whole cell quantitative proteomics with gene knockout of AMPKα1. RESULTS Comparing macrophages with AMPKα1 catalytic subunit deletion with wild-type, inflammatory markers are largely unchanged in unstimulated cells, but with an LPS stimulus, AMPKα1 knockout leads to a striking M1 hyperpolarisation. Deletion of AMPKα1 also resulted in increased expression of rate-limiting enzymes involved in itaconate synthesis, metabolism of glucose, arginine, prostaglandins and cholesterol. Consistent with this, we observed functional changes in prostaglandin synthesis and arginine metabolism. Selective AMPKα1 activation also unlocks additional regulation of IL-6 and IL-12 in M1 macrophages. CONCLUSIONS Together, our results validate AMPK as a pivotal immunometabolic regulator in macrophages.
Collapse
Affiliation(s)
- Iain R Phair
- Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee, DD1 9SY, UK.
| | - Raid B Nisr
- Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee, DD1 9SY, UK.
| | - Andrew J M Howden
- Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK.
| | - Magdalena Sovakova
- Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee, DD1 9SY, UK.
| | - Noor Alqurashi
- Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee, DD1 9SY, UK.
| | - Marc Foretz
- Université Paris Cité, Institut Cochin, CNRS, INSERM, F-75014 Paris, France.
| | - Douglas Lamont
- Centre for Advanced Scientific Technologies, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK.
| | - Benoit Viollet
- Université Paris Cité, Institut Cochin, CNRS, INSERM, F-75014 Paris, France.
| | - Graham Rena
- Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee, DD1 9SY, UK.
| |
Collapse
|
15
|
Tanaka K, Harada H, Kamuro H, Sakai H, Yamamoto A, Tomimatsu M, Ikeda A, Chosokabe R, Tanaka S, Okada Y, Fujio Y, Obana M. Arid5a/IL-6/PAI-1 Signaling Is Involved in the Pathogenesis of Lipopolysaccharide-Induced Kidney Injury. Biol Pharm Bull 2023; 46:1753-1760. [PMID: 38044094 DOI: 10.1248/bpb.b23-00482] [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] [Indexed: 12/05/2023]
Abstract
A systemic inflammatory response leads to widespread organ dysfunction, such as kidney dysfunction. Plasminogen activator inhibitor-1 (PAI-1) is involved in the pathogenesis of inflammatory kidney injury; however, the regulatory mechanism of PAI-1 in injured kidneys remains unclear. PAI-1 is induced by interleukin (IL)-6 in patients with sepsis. In addition, the stabilization of IL-6 is regulated by the adenine-thymine-rich interactive domain-containing protein 5a (Arid5a). Therefore, the aim of the present study was to examine the involvement of Arid5a/IL-6/PAI-1 signaling in lipopolysaccharide (LPS)-induced inflammatory kidney injury. LPS treatment to C57BL/6J mice upregulated Pai-1 mRNA in the kidneys. Enzyme-linked immunosorbent assay (ELISA) revealed that PAI-1 expression was induced in the culture supernatants of LPS-treated human umbilical vein endothelial cells, but not in those of LPS-treated human kidney 2 (HK-2) cells, a tubular cell line. Combined with single-cell analysis, endothelial cells were found to be responsible for PAI-1 elevation in LPS-treated kidneys. Administration of TM5441, a PAI-1 inhibitor, reduced the urinary albumin/creatinine ratio, concomitant with downregulation of Il-6 and Arid5a mRNA expressions. IL-6 treatment in LPS model mice further upregulated Pai-1 mRNA expression compared with LPS alone, accompanied by renal impairment. Furthermore, the expression of Il-6 and Pai-1 mRNA was lower in Arid5a knockout mice than in wild-type mice after LPS treatment. Taken together, the vicious cycle of Arid5a/IL-6/PAI-1 signaling is involved in LPS-induced kidney injury.
Collapse
Affiliation(s)
- Koki Tanaka
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University
| | - Hiroki Harada
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University
| | - Hiroyasu Kamuro
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University
| | - Hibiki Sakai
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University
| | - Ayaha Yamamoto
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University
| | - Masashi Tomimatsu
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University
| | - Akari Ikeda
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University
| | - Renya Chosokabe
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University
| | - Shota Tanaka
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University
| | - Yoshiaki Okada
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University
- Center for Infectious Disease Education and Research (CiDER), Osaka University
| | - Yasushi Fujio
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University
- Center for Infectious Disease Education and Research (CiDER), Osaka University
- Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University
| | - Masanori Obana
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University
- Center for Infectious Disease Education and Research (CiDER), Osaka University
- Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University
- Global Center for Medical Engineering and Informatics (MEI), Osaka University
- Radioisotope Research Center, Institute for Radiation Sciences, Osaka University
| |
Collapse
|
16
|
Lee S, Affandi J, Waters S, Price P. Human Cytomegalovirus Infection and Cardiovascular Disease: Current Perspectives. Viral Immunol 2023; 36:13-24. [PMID: 36622943 DOI: 10.1089/vim.2022.0139] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Infections with human cytomegalovirus (HCMV) are often asymptomatic in healthy adults but can be severe in people with a compromised immune system. While several studies have demonstrated associations between cardiovascular disease in older adults and HCMV seropositivity, the underlying mechanisms are unclear. We review evidence published within the last 5 years establishing how HCMV can contribute directly and indirectly to the development and progression of atherosclerotic plaques. We also discuss associations between HCMV infection and cardiovascular outcomes in populations with a high or very high burden of HCMV, including patients with renal or autoimmune disease, transplant recipients, and people living with HIV.
Collapse
Affiliation(s)
- Silvia Lee
- Department of Microbiology, Pathwest Laboratory Medicine, Perth, Western Australia, Australia.,Department of Advanced Clinical and Translational Cardiovascular Imaging, Harry Perkins Institute of Medical Research, Murdoch, Western Australia, Australia.,Curtin Medical School and the Curtin Health Innovation Research Institute (CHIRI); Bentley, Western Australia, Australia
| | - Jacquita Affandi
- Curtin School of Population Health; Curtin University, Bentley, Western Australia, Australia
| | - Shelley Waters
- Curtin Medical School and the Curtin Health Innovation Research Institute (CHIRI); Bentley, Western Australia, Australia
| | - Patricia Price
- Curtin Medical School and the Curtin Health Innovation Research Institute (CHIRI); Bentley, Western Australia, Australia
| |
Collapse
|
17
|
Mokry M, Boltjes A, Slenders L, Bel-Bordes G, Cui K, Brouwer E, Mekke JM, Depuydt MA, Timmerman N, Waissi F, Verwer MC, Turner AW, Khan MD, Hodonsky CJ, Benavente ED, Hartman RJ, van den Dungen NAM, Lansu N, Nagyova E, Prange KH, Kovacic JC, Björkegren JL, Pavlos E, Andreakos E, Schunkert H, Owens GK, Monaco C, Finn AV, Virmani R, Leeper NJ, de Winther MP, Kuiper J, de Borst GJ, Stroes ES, Civelek M, de Kleijn DP, den Ruijter HM, Asselbergs FW, van der Laan SW, Miller CL, Pasterkamp G. Transcriptomic-based clustering of human atherosclerotic plaques identifies subgroups with different underlying biology and clinical presentation. NATURE CARDIOVASCULAR RESEARCH 2022; 1:1140-1155. [PMID: 37920851 PMCID: PMC10621615 DOI: 10.1038/s44161-022-00171-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 10/20/2022] [Indexed: 11/04/2023]
Abstract
Histopathological studies have revealed key processes of atherosclerotic plaque thrombosis. However, the diversity and complexity of lesion types highlight the need for improved sub-phenotyping. Here we analyze the gene expression profiles of 654 advanced human carotid plaques. The unsupervised, transcriptome-driven clustering revealed five dominant plaque types. These plaque phenotypes were associated with clinical presentation and showed differences in cellular compositions. Validation in coronary segments showed that the molecular signature of these plaques was linked to coronary ischemia. One of the plaque types with the most severe clinical symptoms pointed to both inflammatory and fibrotic cell lineages. Further, we did a preliminary analysis of potential circulating biomarkers that mark the different plaques phenotypes. In conclusion, the definition of the plaque at risk for a thrombotic event can be fine-tuned by in-depth transcriptomic-based phenotyping. These differential plaque phenotypes prove clinically relevant for both carotid and coronary artery plaques and point to distinct underlying biology of symptomatic lesions.
Collapse
Affiliation(s)
- Michal Mokry
- Central Diagnostics Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
- Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Arjan Boltjes
- Central Diagnostics Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Lotte Slenders
- Central Diagnostics Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Gemma Bel-Bordes
- Central Diagnostics Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Kai Cui
- Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Eli Brouwer
- Central Diagnostics Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Joost M. Mekke
- Department of Vascular Surgery, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Marie A.C. Depuydt
- Leiden Academic Centre for Drug Research, Division of Biotherapeutics, Leiden University, Leiden, The Netherlands
| | - Nathalie Timmerman
- Department of Vascular Surgery, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Farahnaz Waissi
- Department of Vascular Surgery, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Maarten C Verwer
- Department of Vascular Surgery, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Adam W. Turner
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Mohammad Daud Khan
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Chani J. Hodonsky
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Ernest Diez Benavente
- Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Robin J.G. Hartman
- Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Noortje A M van den Dungen
- Central Diagnostics Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Nico Lansu
- Central Diagnostics Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Emilia Nagyova
- Central Diagnostics Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Koen H.M. Prange
- Amsterdam University Medical Centers – location AMC, University of Amsterdam, Experimental Vascular Biology, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam Infection and Immunity, Amsterdam, The Netherlands
| | - Jason C. Kovacic
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Victor Chang Cardiac Research Institute, Darlinghurst, Australia; and St Vincent’s Clinical School, University of New South Wales, Australia
| | - Johan L.M. Björkegren
- Department of Medicine, Karolinska Institutet, Karolinska Universitetssjukhuset, Huddinge, Sweden
- Department of Genetics & Genomic Sciences, Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029-6574, USA
| | - Eleftherios Pavlos
- Laboratory of Immunobiology, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Evangelos Andreakos
- Laboratory of Immunobiology, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Heribert Schunkert
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK e.V.), Partner Site Munich Heart Alliance, Munich, Germany
| | - Gary K. Owens
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA
| | - Claudia Monaco
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford
| | | | | | - Nicholas J. Leeper
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, CA
| | - Menno P.J. de Winther
- Amsterdam University Medical Centers – location AMC, University of Amsterdam, Experimental Vascular Biology, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam Infection and Immunity, Amsterdam, The Netherlands
| | - Johan Kuiper
- Leiden Academic Centre for Drug Research, Division of Biotherapeutics, Leiden University, Leiden, The Netherlands
| | - Gert J. de Borst
- Department of Vascular Surgery, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Erik S.G. Stroes
- Department of Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, the Netherlands
| | - Mete Civelek
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | | | - Hester M. den Ruijter
- Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Folkert W. Asselbergs
- Department of Cardiology, Division Heart & Lungs, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
- Health Data Research UK and Institute of Health Informatics, University College London, London, United Kingdom
| | - Sander W. van der Laan
- Central Diagnostics Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Clint L. Miller
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
- Department of Biomedical Engineering, Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, USA
- Department of Public Health Sciences, University of Virginia, Charlottesville, VA, USA
| | - Gerard Pasterkamp
- Central Diagnostics Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| |
Collapse
|
18
|
Granulocyte Colony-Stimulating Factor Ameliorates Endothelial Activation and Thrombotic Diathesis Biomarkers in a Murine Model of Hind Limb Ischemia. Biomedicines 2022; 10:biomedicines10092303. [PMID: 36140404 PMCID: PMC9496113 DOI: 10.3390/biomedicines10092303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 08/29/2022] [Accepted: 09/15/2022] [Indexed: 12/24/2022] Open
Abstract
Novel therapies in peripheral arterial disease, such as granulocyte colony-stimulating factor (GCSF) administration, might result in anti-atherosclerotic effects. In this study, we used 10-week-old male ApoE−/− mice, which were fed an atherosclerosis-inducing diet for four weeks. At the end of the four weeks, hind limb ischemia was induced through left femoral artery ligation, the atherosclerosis-inducing diet was discontinued, and a normal diet was initiated. Mice were then randomized into a control group (intramuscular 0.4 mL normal saline 0.9% for 7 days) and a group in which GCSF was administrated intramuscularly in the left hind limb for 7 days (100 mg/kg). In the GCSF group, but not in the control group, we observed significant reductions in the soluble adhesion molecules (vascular cell adhesion molecule-1 (sVCAM-1) and intercellular adhesion molecule-1 (sICAM-1)), sE-Selectin, and plasminogen activator inhibitor (PAI)-1 when they were measured through ELISA on the 1st and the 28th days after hind limb ischemia induction. Therefore, GCSF administration in an atherosclerotic mouse model of hind limb ischemia led to decreases in the biomarkers associated with endothelial activation and thrombosis. These findings warrant further validation in future preclinical studies.
Collapse
|
19
|
Jung RG, Duchez AC, Simard T, Dhaliwal S, Gillmore T, Di Santo P, Labinaz A, Ramirez FD, Rasheed A, Robichaud S, Ouimet M, Short S, Clifford C, Xiao F, Lordkipanidzé M, Burger D, Gadde S, Rayner KJ, Hibbert B. Plasminogen Activator Inhibitor-1–Positive Platelet-Derived Extracellular Vesicles Predicts MACE and the Proinflammatory SMC Phenotype. JACC Basic Transl Sci 2022; 7:985-997. [PMID: 36337926 PMCID: PMC9626902 DOI: 10.1016/j.jacbts.2022.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 05/02/2022] [Accepted: 05/02/2022] [Indexed: 12/01/2022]
Abstract
This study shows the existence of PAI-1+ PEVs. Approximately 20% of plasma PAI-1 is composed of PAI-1+ PEVs. Elevated PAI-1+ PEV levels were predictive of 1-year major adverse cardiac events in both the discovery and the validation cohort, with larger effect sizes than other clinical biomarkers. High PAI-1+ PEV levels did not affect thrombogenicity. Increasing doses of PAI-1+ PEVs promoted the proinflammatory VSMC state by enhancing proliferation and migration. Inhibition of the PAI-1:low-density lipoprotein–related receptor-1 pathway dampened the proinflammatory VSMC changes. PAI-1+ PEV is a promising biomarker for major adverse cardiac events, and targeting the PAI-1+ PEV–VSMC interaction may offer a novel target to modulate cardiac events in patients with coronary artery disease.
Patients with established coronary artery disease remain at elevated risk of major adverse cardiac events. The goal of this study was to evaluate the utility of plasminogen activator inhibitor-1–positive platelet-derived extracellular vesicles as a biomarker for major adverse cardiac events and to explore potential underlying mechanisms. Our study suggests these extracellular vesicles as a potential biomarker to identify and a therapeutic target to ameliorate neointimal formation of high-risk patients.
Collapse
Affiliation(s)
- Richard G. Jung
- CAPITAL Research Group, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
- Vascular Biology and Experimental Medicine Laboratory, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Anne-Claire Duchez
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Trevor Simard
- CAPITAL Research Group, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
- Vascular Biology and Experimental Medicine Laboratory, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Division of Cardiology, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Shan Dhaliwal
- CAPITAL Research Group, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
- Vascular Biology and Experimental Medicine Laboratory, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Ontario, Canada
| | - Taylor Gillmore
- Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Pietro Di Santo
- CAPITAL Research Group, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
- Vascular Biology and Experimental Medicine Laboratory, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Ontario, Canada
| | - Alisha Labinaz
- CAPITAL Research Group, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
- Vascular Biology and Experimental Medicine Laboratory, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - F. Daniel Ramirez
- CAPITAL Research Group, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
- Vascular Biology and Experimental Medicine Laboratory, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
- Division of Cardiology, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Adil Rasheed
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Sabrina Robichaud
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Mireille Ouimet
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Spencer Short
- Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Cole Clifford
- Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Fengxia Xiao
- Kidney Research Centre, Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Marie Lordkipanidzé
- Faculté de Pharmacie, Université de Montréal, Montréal, Québec, Canada
- Research Center, Montreal Heart Institute, Montréal, Québec, Canada
| | - Dylan Burger
- Kidney Research Centre, Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Suresh Gadde
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Katey J. Rayner
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Benjamin Hibbert
- CAPITAL Research Group, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
- Vascular Biology and Experimental Medicine Laboratory, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Division of Cardiology, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
- Address for correspondence: Dr Benjamin Hibbert, University of Ottawa Heart Institute, 40 Ruskin Street, H-4238, Ottawa, Ontario K1Y 4W7, Canada.
| |
Collapse
|
20
|
Badran M, Gozal D. PAI-1: A Major Player in the Vascular Dysfunction in Obstructive Sleep Apnea? Int J Mol Sci 2022; 23:5516. [PMID: 35628326 PMCID: PMC9141273 DOI: 10.3390/ijms23105516] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/05/2022] [Accepted: 05/12/2022] [Indexed: 02/04/2023] Open
Abstract
Obstructive sleep apnea is a chronic and prevalent condition that is associated with endothelial dysfunction, atherosclerosis, and imposes excess overall cardiovascular risk and mortality. Despite its high prevalence and the susceptibility of CVD patients to OSA-mediated stressors, OSA is still under-recognized and untreated in cardiovascular practice. Moreover, conventional OSA treatments have yielded either controversial or disappointing results in terms of protection against CVD, prompting the need for the identification of additional mechanisms and associated adjuvant therapies. Plasminogen activator inhibitor-1 (PAI-1), the primary inhibitor of tissue-type plasminogen activator (tPA) and urinary-type plasminogen activator (uPA), is a key regulator of fibrinolysis and cell migration. Indeed, elevated PAI-1 expression is associated with major cardiovascular adverse events that have been attributed to its antifibrinolytic activity. However, extensive evidence indicates that PAI-1 can induce endothelial dysfunction and atherosclerosis through complex interactions within the vasculature in an antifibrinolytic-independent matter. Elevated PAI-1 levels have been reported in OSA patients. However, the impact of PAI-1 on OSA-induced CVD has not been addressed to date. Here, we provide a comprehensive review on the mechanisms by which OSA and its most detrimental perturbation, intermittent hypoxia (IH), can enhance the transcription of PAI-1. We also propose causal pathways by which PAI-1 can promote atherosclerosis in OSA, thereby identifying PAI-1 as a potential therapeutic target in OSA-induced CVD.
Collapse
Affiliation(s)
- Mohammad Badran
- Department of Child Health and Child Health Research Institute, School of Medicine, University of Missouri, 400 N Keene St, Suite 010, Columbia, MO 65201, USA;
| | - David Gozal
- Department of Child Health and Child Health Research Institute, School of Medicine, University of Missouri, 400 N Keene St, Suite 010, Columbia, MO 65201, USA;
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO 65201, USA
| |
Collapse
|
21
|
Xiang Q, Tian F, Xu J, Du X, Zhang S, Liu L. New insight into dyslipidemia‐induced cellular senescence in atherosclerosis. Biol Rev Camb Philos Soc 2022; 97:1844-1867. [PMID: 35569818 PMCID: PMC9541442 DOI: 10.1111/brv.12866] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 04/18/2022] [Accepted: 04/28/2022] [Indexed: 11/28/2022]
Abstract
Atherosclerosis, characterized by lipid‐rich plaques in the arterial wall, is an age‐related disorder and a leading cause of mortality worldwide. However, the specific mechanisms remain complex. Recently, emerging evidence has demonstrated that senescence of various types of cells, such as endothelial cells (ECs), vascular smooth muscle cells (VSMCs), macrophages, endothelial progenitor cells (EPCs), and adipose‐derived mesenchymal stem cells (AMSCs) contributes to atherosclerosis. Cellular senescence and atherosclerosis share various causative stimuli, in which dyslipidemia has attracted much attention. Dyslipidemia, mainly referred to elevated plasma levels of atherogenic lipids or lipoproteins, or functional impairment of anti‐atherogenic lipids or lipoproteins, plays a pivotal role both in cellular senescence and atherosclerosis. In this review, we summarize the current evidence for dyslipidemia‐induced cellular senescence during atherosclerosis, with a focus on low‐density lipoprotein (LDL) and its modifications, hydrolysate of triglyceride‐rich lipoproteins (TRLs), and high‐density lipoprotein (HDL), respectively. Furthermore, we describe the underlying mechanisms linking dyslipidemia‐induced cellular senescence and atherosclerosis. Finally, we discuss the senescence‐related therapeutic strategies for atherosclerosis, with special attention given to the anti‐atherosclerotic effects of promising geroprotectors as well as anti‐senescence effects of current lipid‐lowering drugs.
Collapse
Affiliation(s)
- Qunyan Xiang
- Department of Geriatrics, The Second Xiangya Hospital Central South University Changsha Hunan 410011 PR China
- Institute of Aging and Age‐related Disease Research Central South University Changsha Hunan 410011 PR China
| | - Feng Tian
- Department of Geriatric Cardiology The First Affiliated Hospital of Zhengzhou University Zhengzhou Henan 450000 PR China
| | - Jin Xu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital Central South University Changsha Hunan 410011 PR China
- Research Institute of Blood Lipid and Atherosclerosis Central South University Changsha Hunan 410011 PR China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province Changsha Hunan 410011 PR China
- Cardiovascular Disease Research Center of Hunan Province Changsha Hunan 410011 PR China
| | - Xiao Du
- Department of Cardiovascular Medicine, The Second Xiangya Hospital Central South University Changsha Hunan 410011 PR China
- Research Institute of Blood Lipid and Atherosclerosis Central South University Changsha Hunan 410011 PR China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province Changsha Hunan 410011 PR China
- Cardiovascular Disease Research Center of Hunan Province Changsha Hunan 410011 PR China
| | - Shilan Zhang
- Department of Gastroenterology, The Second Xiangya Hospital Central South University Changsha Hunan 410011 PR China
| | - Ling Liu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital Central South University Changsha Hunan 410011 PR China
- Research Institute of Blood Lipid and Atherosclerosis Central South University Changsha Hunan 410011 PR China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province Changsha Hunan 410011 PR China
- Cardiovascular Disease Research Center of Hunan Province Changsha Hunan 410011 PR China
| |
Collapse
|
22
|
Liu L, Shi Z, Ji X, Zhang W, Luan J, Zahr T, Qiang L. Adipokines, adiposity, and atherosclerosis. Cell Mol Life Sci 2022; 79:272. [PMID: 35503385 PMCID: PMC11073100 DOI: 10.1007/s00018-022-04286-2] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 03/11/2022] [Accepted: 04/03/2022] [Indexed: 12/12/2022]
Abstract
Characterized by a surplus of whole-body adiposity, obesity is strongly associated with the prognosis of atherosclerosis, a hallmark of coronary artery disease (CAD) and the major contributor to cardiovascular disease (CVD) mortality. Adipose tissue serves a primary role as a lipid-storage organ, secreting cytokines known as adipokines that affect whole-body metabolism, inflammation, and endocrine functions. Emerging evidence suggests that adipokines can play important roles in atherosclerosis development, progression, as well as regression. Here, we review the versatile functions of various adipokines in atherosclerosis and divide these respective functions into three major groups: protective, deteriorative, and undefined. The protective adipokines represented here are adiponectin, fibroblast growth factor 21 (FGF-21), C1q tumor necrosis factor-related protein 9 (CTRP9), and progranulin, while the deteriorative adipokines listed include leptin, chemerin, resistin, Interleukin- 6 (IL-6), and more, with additional adipokines that have unclear roles denoted as undefined adipokines. Comprehensively categorizing adipokines in the context of atherosclerosis can help elucidate the various pathways involved and potentially pave novel therapeutic approaches to treat CVDs.
Collapse
Affiliation(s)
- Longhua Liu
- School of Kinesiology, Shanghai University of Sport, Shanghai, People's Republic of China.
| | - Zunhan Shi
- School of Kinesiology, Shanghai University of Sport, Shanghai, People's Republic of China
| | - Xiaohui Ji
- School of Kinesiology, Shanghai University of Sport, Shanghai, People's Republic of China
| | - Wenqian Zhang
- School of Kinesiology, Shanghai University of Sport, Shanghai, People's Republic of China
| | - Jinwen Luan
- School of Kinesiology, Shanghai University of Sport, Shanghai, People's Republic of China
| | - Tarik Zahr
- Department of Pharmacology, Columbia University, New York, NY, USA
| | - Li Qiang
- Department of Pathology and Cellular Biology and Naomi Berrie Diabetes Center, Columbia University, New York, NY, USA.
| |
Collapse
|
23
|
Ramírez-Moreno E, Arias-Rico J, Jiménez-Sánchez RC, Estrada-Luna D, Jiménez-Osorio AS, Zafra-Rojas QY, Ariza-Ortega JA, Flores-Chávez OR, Morales-Castillejos L, Sandoval-Gallegos EM. Role of Bioactive Compounds in Obesity: Metabolic Mechanism Focused on Inflammation. Foods 2022; 11:foods11091232. [PMID: 35563955 PMCID: PMC9101148 DOI: 10.3390/foods11091232] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/17/2022] [Accepted: 04/18/2022] [Indexed: 02/01/2023] Open
Abstract
Obesity is a disease characterized by an inflammatory process in the adipose tissue due to diverse infiltrated immune cells, an increased secretion of proinflammatory molecules, and a decreased secretion of anti-inflammatory molecules. On the other hand, obesity increases the risk of several diseases, such as cardiovascular diseases, diabetes, and cancer. Their treatment is based on nutritional and pharmacological strategies. However, natural products are currently implemented as complementary and alternative medicine (CAM). Polyphenols and fiber are naturally compounds with potential action to reduce inflammation through several pathways and play an important role in the prevention and treatment of obesity, as well as in other non-communicable diseases. Hence, this review focuses on the recent evidence of the molecular mechanisms of polyphenols and dietary fiber, from Scopus, Science Direct, and PubMed, among others, by using key words and based on recent in vitro and in vivo studies.
Collapse
Affiliation(s)
- Esther Ramírez-Moreno
- Academic Area of Nutrition, Interdisciplinary Research Center, Institute of Health Sciences, Circuit Actopan Tilcuautla s/n, Ex hacienda La Concepción, San Agustin Tlaxiaca, Pachuca 42160, Mexico; (E.R.-M.); (Q.Y.Z.-R.); (J.A.A.-O.)
| | - José Arias-Rico
- Academic Area of Nursing; Institute of Health Sciences, Circuit Actopan Tilcuautla s/n, Ex hacienda La Concepción, San Agustin Tlaxiaca, Pachuca 42160, Mexico; (J.A.-R.); (R.C.J.-S.); (D.E.-L.); (A.S.J.-O.); (O.R.F.-C.); (L.M.-C.)
| | - Reyna Cristina Jiménez-Sánchez
- Academic Area of Nursing; Institute of Health Sciences, Circuit Actopan Tilcuautla s/n, Ex hacienda La Concepción, San Agustin Tlaxiaca, Pachuca 42160, Mexico; (J.A.-R.); (R.C.J.-S.); (D.E.-L.); (A.S.J.-O.); (O.R.F.-C.); (L.M.-C.)
| | - Diego Estrada-Luna
- Academic Area of Nursing; Institute of Health Sciences, Circuit Actopan Tilcuautla s/n, Ex hacienda La Concepción, San Agustin Tlaxiaca, Pachuca 42160, Mexico; (J.A.-R.); (R.C.J.-S.); (D.E.-L.); (A.S.J.-O.); (O.R.F.-C.); (L.M.-C.)
| | - Angélica Saraí Jiménez-Osorio
- Academic Area of Nursing; Institute of Health Sciences, Circuit Actopan Tilcuautla s/n, Ex hacienda La Concepción, San Agustin Tlaxiaca, Pachuca 42160, Mexico; (J.A.-R.); (R.C.J.-S.); (D.E.-L.); (A.S.J.-O.); (O.R.F.-C.); (L.M.-C.)
| | - Quinatzin Yadira Zafra-Rojas
- Academic Area of Nutrition, Interdisciplinary Research Center, Institute of Health Sciences, Circuit Actopan Tilcuautla s/n, Ex hacienda La Concepción, San Agustin Tlaxiaca, Pachuca 42160, Mexico; (E.R.-M.); (Q.Y.Z.-R.); (J.A.A.-O.)
| | - José Alberto Ariza-Ortega
- Academic Area of Nutrition, Interdisciplinary Research Center, Institute of Health Sciences, Circuit Actopan Tilcuautla s/n, Ex hacienda La Concepción, San Agustin Tlaxiaca, Pachuca 42160, Mexico; (E.R.-M.); (Q.Y.Z.-R.); (J.A.A.-O.)
| | - Olga Rocío Flores-Chávez
- Academic Area of Nursing; Institute of Health Sciences, Circuit Actopan Tilcuautla s/n, Ex hacienda La Concepción, San Agustin Tlaxiaca, Pachuca 42160, Mexico; (J.A.-R.); (R.C.J.-S.); (D.E.-L.); (A.S.J.-O.); (O.R.F.-C.); (L.M.-C.)
| | - Lizbeth Morales-Castillejos
- Academic Area of Nursing; Institute of Health Sciences, Circuit Actopan Tilcuautla s/n, Ex hacienda La Concepción, San Agustin Tlaxiaca, Pachuca 42160, Mexico; (J.A.-R.); (R.C.J.-S.); (D.E.-L.); (A.S.J.-O.); (O.R.F.-C.); (L.M.-C.)
| | - Eli Mireya Sandoval-Gallegos
- Academic Area of Nutrition, Interdisciplinary Research Center, Institute of Health Sciences, Circuit Actopan Tilcuautla s/n, Ex hacienda La Concepción, San Agustin Tlaxiaca, Pachuca 42160, Mexico; (E.R.-M.); (Q.Y.Z.-R.); (J.A.A.-O.)
- Correspondence:
| |
Collapse
|
24
|
Cansby E, Kumari S, Caputo M, Xia Y, Porosk R, Robinson J, Wang H, Olsson BM, Vallin J, Grantham J, Soomets U, Svensson LT, Sihlbom C, Marschall HU, Edsfeldt A, Goncalves I, Mahlapuu M. Silencing of STE20-type kinase STK25 in human aortic endothelial and smooth muscle cells is atheroprotective. Commun Biol 2022; 5:379. [PMID: 35440683 PMCID: PMC9018782 DOI: 10.1038/s42003-022-03309-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 03/22/2022] [Indexed: 02/07/2023] Open
Abstract
Recent studies highlight the importance of lipotoxic damage in aortic cells as the major pathogenetic contributor to atherosclerotic disease. Since the STE20-type kinase STK25 has been shown to exacerbate ectopic lipid storage and associated cell injury in several metabolic organs, we here investigate its role in the main cell types of vasculature. We depleted STK25 by small interfering RNA in human aortic endothelial and smooth muscle cells exposed to oleic acid and oxidized LDL. In both cell types, the silencing of STK25 reduces lipid accumulation and suppresses activation of inflammatory and fibrotic pathways as well as lowering oxidative and endoplasmic reticulum stress. Notably, in smooth muscle cells, STK25 inactivation hinders the shift from a contractile to a synthetic phenotype. Together, we provide several lines of evidence that antagonizing STK25 signaling in human aortic endothelial and smooth muscle cells is atheroprotective, highlighting this kinase as a new potential therapeutic target for atherosclerotic disease. Silencing of STK25, an STE20-type kinase, in human aortic endothelial and smooth muscle cells reduces lipid accumulation and suppresses inflammation and fibrotic pathways, ultimately exerting atheroprotective effects.
Collapse
Affiliation(s)
- Emmelie Cansby
- Department of Chemistry and Molecular Biology, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Sima Kumari
- Department of Chemistry and Molecular Biology, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Mara Caputo
- Department of Chemistry and Molecular Biology, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Ying Xia
- Department of Chemistry and Molecular Biology, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Rando Porosk
- Department of Biochemistry, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Jonathan Robinson
- Department of Biology and Biological Engineering, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Chalmers University of Technology, Gothenburg, Sweden
| | - Hao Wang
- Department of Biology and Biological Engineering, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Chalmers University of Technology, Gothenburg, Sweden
| | | | - Josefine Vallin
- Department of Chemistry and Molecular Biology, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Julie Grantham
- Department of Chemistry and Molecular Biology, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Ursel Soomets
- Department of Biochemistry, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - L Thomas Svensson
- Department of Biology and Biological Engineering, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Chalmers University of Technology, Gothenburg, Sweden
| | - Carina Sihlbom
- Proteomics Core Facility, University of Gothenburg, Gothenburg, Sweden
| | - Hanns-Ulrich Marschall
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Andreas Edsfeldt
- Department of Clinical Sciences Malmö, Clinical Research Center, Lund University, Malmö, Sweden.,Department of Cardiology, Skåne University Hospital, Lund/Malmö, Sweden.,Wallenberg Center for Molecular Medicine, Lund University, Malmö, Sweden
| | - Isabel Goncalves
- Department of Clinical Sciences Malmö, Clinical Research Center, Lund University, Malmö, Sweden.,Department of Cardiology, Skåne University Hospital, Lund/Malmö, Sweden
| | - Margit Mahlapuu
- Department of Chemistry and Molecular Biology, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden.
| |
Collapse
|
25
|
Vecchiola A, García K, González-Gómez LM, Tapia-Castillo A, Artigas R, Baudrand R, Kalergis AM, Carvajal CA, Fardella CE. Plasminogen Activator Inhibitor-1 and Adiponectin Are Associated With Metabolic Syndrome Components. Am J Hypertens 2022; 35:311-318. [PMID: 34525175 DOI: 10.1093/ajh/hpab138] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 08/01/2021] [Accepted: 09/14/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND We aimed to study the associations of adipocytokines, endothelial damage markers, and high-sensitivity C-reactive protein (hs-CRP) with metabolic syndrome (MetS) components. METHODS This cross-sectional study included 202 subjects categorized into MetS and No-MetS according to Harmonizing Adult Treatment Panel III. RESULTS Subjects with MetS showed higher levels of proinflammatory molecules but significantly lower adiponectin levels than subjects with No-MetS. Among the studied adipocytokines, plasminogen activator inhibitor-1 (PAI-1) and adiponectin showed the strongest associations with most MetS components. PAI-1 was associated with MetS (odds ratio (OR) 1.107 (1.065-1.151), P < 0.0001), whereas adiponectin was inversely associated with MetS (OR 0.710 (0.610-0.825), P < 0.0001). Following adjustment by sex, age, body mass index, and 24-hour urinary sodium excretion in a multivariate analysis, the association of PAI-1 (OR 1.090 (1.044-1.137), P < 0.0001) and adiponectin (OR 0.634 (0.519-0.775), P < 0.0001) with MetS remained significant. Multivariate analyses supported a model in which systolic blood pressure (BP) could be predicted by PAI-1, hs-CRP, and matrix metalloproteinase 2 (R2 = 0.125; P = 0.04); diastolic BP (R2 = 0.218; P = 0.0001) and glucose (R2 = 0.074; P = 0.0001) could be predicted by PAI-1; waist circumference could be predicted by PAI-1 and hs-CRP (R2 = 0.28; P = 0.016). Receiver operating characteristic curve analysis showed that a PAI-1 concentration had the best sensitivity and specificity for discriminating subjects with MetS. CONCLUSION PAI-1 and adiponectin rendered the most robust associations with MetS components in a general population, indicating that unfavorable adipose tissue performance is a key contributor to these metabolic anomalies. Further prospective analyses should allow establishing whether these adipocytokines can anticipate the progress of MetS and cardiovascular risk.
Collapse
Affiliation(s)
- Andrea Vecchiola
- Departmento de Endocrinología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
- Millenium Institute on Immunology and Immunotherapy IMII, Santiago, Chile
- Translational Endocrinology (CETREN), Faculty of Medicine, Department of Endocrinology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Killén García
- Departmento de Endocrinología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Luis M González-Gómez
- Departmento de Endocrinología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
- Millenium Institute on Immunology and Immunotherapy IMII, Santiago, Chile
| | - Alejandra Tapia-Castillo
- Departmento de Endocrinología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
- Millenium Institute on Immunology and Immunotherapy IMII, Santiago, Chile
- Translational Endocrinology (CETREN), Faculty of Medicine, Department of Endocrinology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Rocío Artigas
- Advanced Center for Chronic Diseases (ACCDiS), Core Biodata, Santiago, Chile
| | - René Baudrand
- Departmento de Endocrinología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
- Translational Endocrinology (CETREN), Faculty of Medicine, Department of Endocrinology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M Kalergis
- Departmento de Endocrinología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
- Millenium Institute on Immunology and Immunotherapy IMII, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Cristian A Carvajal
- Departmento de Endocrinología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
- Millenium Institute on Immunology and Immunotherapy IMII, Santiago, Chile
- Translational Endocrinology (CETREN), Faculty of Medicine, Department of Endocrinology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Carlos E Fardella
- Departmento de Endocrinología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
- Millenium Institute on Immunology and Immunotherapy IMII, Santiago, Chile
- Translational Endocrinology (CETREN), Faculty of Medicine, Department of Endocrinology, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
26
|
Plasminogen activator inhibitor-1 mediate downregulation of adiponectin in type 2 diabetes patients with metabolic syndrome. Cytokine X 2022; 4:100064. [PMID: 35128381 PMCID: PMC8803603 DOI: 10.1016/j.cytox.2022.100064] [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: 12/10/2020] [Revised: 09/15/2021] [Accepted: 01/17/2022] [Indexed: 01/02/2023] Open
Abstract
Both Adiponectin and PAI-1 levels are associated with the Metabolic abnormalities. This study demonstrates that subjects with MetS have low adiponectin and higher PAI-1 levels compare to non-MetS. Higher PAI-1 levels are associated with higher odds of risk and prevalence of MetS. Pharmacological targeting of PAI-1 is necessary for MetS management.
Introduction Metabolic syndrome (MetS) is a multifactorial disease characterized by metabolic abnormalities. Plasminogen activator inhibitor-1(PAI-1) is a key factor of the fibrinolysis its expression is elevated in insulin resistance, obesity, and MetS. In addition, an adiponectin produced by adipocytes is also key factor in MetS. This study aimed to investigate the relationship between PAI-1, adiponectin levels in MetS. Patients and Methods A total of 379 subjects were analyse in this cross-sectional study. MetS was defined by NCEP ATP-III criteria. Anthropometric, fasting blood glucose, HbA1c, total cholesterol, high-density lipoprotein, low-density lipoprotein, triglycerides, PAI-1, and adiponectin were measured. Results PAI-1 levels were higher in MetS compared with non-MetS. In addition, adiponectin levels were significantly lower in MetS compared to non-MetS. Furthermore, increased level of PAI-1 corresponds with increase in prevalence of MetS. PAI-1 levels were significantly associated with MetS (OR = 2.51, CI = 1.23 – 5.14; p = 0.039). Conclusion PAI-1 increases the risk of MetS. PAI-1 and adiponectin regulation is useful in assesing the presence and severity of MetS. Further pharmacological targeting of PAI-1 studies are necessary for MetS management.
Collapse
|
27
|
Afroz R, Kumarapperuma H, Nguyen QVN, Mohamed R, Little PJ, Kamato D. Lipopolysaccharide acting via toll-like receptor 4 transactivates the TGF-β receptor in vascular smooth muscle cells. Cell Mol Life Sci 2022; 79:121. [PMID: 35122536 PMCID: PMC8817999 DOI: 10.1007/s00018-022-04159-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 01/06/2022] [Accepted: 01/20/2022] [Indexed: 12/11/2022]
Abstract
Toll-like receptors (TLRs) recognise pathogen‑associated molecular patterns, which allow the detection of microbial infection by host cells. Bacterial-derived toxin lipopolysaccharide activates TLR4 and leads to the activation of the Smad2 transcription factor. The phosphorylation of the Smad2 transcription factor is the result of the activation of the transforming growth factor-β receptor 1 (TGFBR1). Therefore, we sought to investigate LPS via TLR4-mediated Smad2 carboxy terminal phosphorylation dependent on the transactivation of the TGFBR1. The in vitro model used human aortic vascular smooth muscle cells to assess the implications of TLR4 transactivation of the TGFBR1 in vascular pathophysiology. We show that LPS-mediated Smad2 carboxy terminal phosphorylation is inhibited in the presence of TGFBR1 inhibitor, SB431542. Treatment with MyD88 and TRIF pathway antagonists does not affect LPS-mediated phosphorylation of Smad2 carboxy terminal; however, LPS-mediated Smad2 phosphorylation was inhibited in the presence of MMP inhibitor, GM6001, and unaffected in the presence of ROCK inhibitor Y27632 or ROS/NOX inhibitor DPI. LPS via transactivation of the TGFBR1 stimulates PAI-1 mRNA expression. TLRs are first in line to respond to exogenous invading substances and endogenous molecules; our findings characterise a novel signalling pathway in the context of cell biology. Identifying TLR transactivation of the TGFBR1 may provide future insight into the detrimental implications of pathogens in pathophysiology.
Collapse
Affiliation(s)
- Rizwana Afroz
- School of Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, QLD, 4102, Australia.,Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, QLD, 4111, Australia
| | - Hirushi Kumarapperuma
- School of Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, QLD, 4102, Australia
| | - Quang V N Nguyen
- School of Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, QLD, 4102, Australia
| | - Raafat Mohamed
- School of Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, QLD, 4102, Australia.,Department of Basic Sciences, College of Dentistry, University of Mosul, Mosul, Iraq
| | - Peter J Little
- School of Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, QLD, 4102, Australia.,Department of Pharmacy, Xinhua College of Sun Yat-Sen University, Tianhe District, Guangzhou, 510520, China.,Sunshine Coast Health Institute, University of the Sunshine Coast, Birtinya, QLD, 4575, Australia
| | - Danielle Kamato
- School of Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, QLD, 4102, Australia.
| |
Collapse
|
28
|
The Roles and Associated Mechanisms of Adipokines in Development of Metabolic Syndrome. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27020334. [PMID: 35056647 PMCID: PMC8781412 DOI: 10.3390/molecules27020334] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/30/2021] [Accepted: 01/03/2022] [Indexed: 12/12/2022]
Abstract
Metabolic syndrome is a cluster of metabolic indicators that increase the risk of diabetes and cardiovascular diseases. Visceral obesity and factors derived from altered adipose tissue, adipokines, play critical roles in the development of metabolic syndrome. Although the adipokines leptin and adiponectin improve insulin sensitivity, others contribute to the development of glucose intolerance, including visfatin, fetuin-A, resistin, and plasminogen activator inhibitor-1 (PAI-1). Leptin and adiponectin increase fatty acid oxidation, prevent foam cell formation, and improve lipid metabolism, while visfatin, fetuin-A, PAI-1, and resistin have pro-atherogenic properties. In this review, we briefly summarize the role of various adipokines in the development of metabolic syndrome, focusing on glucose homeostasis and lipid metabolism.
Collapse
|
29
|
Medcalf RL, Keragala CB. The Fibrinolytic System: Mysteries and Opportunities. Hemasphere 2021; 5:e570. [PMID: 34095754 PMCID: PMC8171360 DOI: 10.1097/hs9.0000000000000570] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 04/07/2021] [Indexed: 02/07/2023] Open
Abstract
The deposition and removal of fibrin has been the primary role of coagulation and fibrinolysis, respectively. There is also little doubt that these 2 enzyme cascades influence each other given they share the same serine protease family ancestry and changes to 1 arm of the hemostatic pathway would influence the other. The fibrinolytic system in particular has also been known for its capacity to clear various non-fibrin proteins and to activate other enzyme systems, including complement and the contact pathway. Furthermore, it can also convert a number of growth factors into their mature, active forms. More recent findings have extended the reach of this system even further. Here we will review some of these developments and also provide an account of the influence of individual players of the fibrinolytic (plasminogen activating) pathway in relation to physiological and pathophysiological events, including aging and metabolism.
Collapse
Affiliation(s)
- Robert L. Medcalf
- Molecular Neurotrauma and Haemostasis, Australian Centre for Blood Diseases, Central Clinical School, Monash University, Victoria, Australia
| | - Charithani B. Keragala
- Molecular Neurotrauma and Haemostasis, Australian Centre for Blood Diseases, Central Clinical School, Monash University, Victoria, Australia
| |
Collapse
|
30
|
Han M, Pandey D. ZMPSTE24 Regulates SARS-CoV-2 Spike Protein-enhanced Expression of Endothelial Plasminogen Activator Inhibitor-1. Am J Respir Cell Mol Biol 2021; 65:300-308. [PMID: 34003736 PMCID: PMC8485999 DOI: 10.1165/rcmb.2020-0544oc] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Endothelial dysfunction is implicated in the thrombotic events reported in COVID-19 patients, but underlying molecular mechanisms are unknown. Circulating levels of the coagulation cascade activator PAI-1 are substantially higher in COVID-19 patients with severe respiratory dysfunction than in patients with bacterial-sepsis and ARDS. Indeed, the elevation of PAI-1 is recognized as an early marker of endothelial dysfunction. Here, we report that recombinant SARS-CoV-2-S1 stimulated robust production of PAI-1 by HPMEC. We examined the role of protein degradation in this SARS-CoV-2-S1 induction of PAI-1 and found that the proteasomal degradation inhibitor bortezomib inhibited SARS-CoV-2-S1 mediated changes in PAI-1. Our data further show that bortezomib upregulated KLF2, a shear-stress-regulated transcription factor that suppresses PAI-1 expression. Aging and metabolic disorders are known to increase the mortality and morbidity in COVID-19 patients. We therefore examined the role of ZMPSTE24, a metalloprotease with a demonstrated role in host defense against RNA viruses that is decreased in the elderly and in metabolic syndrome, in the induction of PAI-1 in HPMEC by SARS-CoV-2-S1. Indeed, overexpression of ZMPSTE24 blunted enhancement of PAI-1 production in spike protein-exposed HPMEC. Additionally, we found that membrane expression of the SARS-CoV-2 entry receptor ACE2 was reduced by ZMPSTE24-mediated cleavage and shedding of the ACE2 ectodomain, leading to accumulation of ACE2 decoy fragments that may bind SARS-CoV-2. These data indicate that decreases in ZMPSTE24 with age and comorbidities may increase vulnerability to vascular endothelial injury by SARS-CoV-2 viruses and that enhanced production of endothelial PAI-1 might play role in prothrombotic events in COVID-19 patients. This article is open access and distributed under the terms of the Creative Commons Attribution Non-Commercial No Derivatives License 4.0 (http://creativecommons.org/licenses/by-nc-nd/4.0/).
Collapse
Affiliation(s)
- Mingming Han
- Johns Hopkins University, 1466, Baltimore, Maryland, United States.,University of Science and Technology of China, 12652, Hefei, China
| | - Deepesh Pandey
- Johns Hopkins University, 1466, Baltimore, Maryland, United States;
| |
Collapse
|
31
|
MH-76, a Novel Non-Quinazoline α 1-Adrenoceptor Antagonist, but Not Prazosin Reduces Inflammation and Improves Insulin Signaling in Adipose Tissue of Fructose-Fed Rats. Pharmaceuticals (Basel) 2021; 14:ph14050477. [PMID: 34069933 PMCID: PMC8157569 DOI: 10.3390/ph14050477] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/09/2021] [Accepted: 05/12/2021] [Indexed: 11/20/2022] Open
Abstract
Background: Quinazoline α1-adrenoceptors antagonists have been shown to exert moderately favorable effects on the metabolic profile in hypertensive patients. However, based on AntiHypertensive and Lipid-Lowering Treatment to Prevent Heart Attack Trial (ALLHAT) results, they are no longer recommended as a first line therapy of hypertension. Recent studies have shown that quinazoline-based α1-adrenoceptors antagonists (prazosin, doxazosin) induce the apoptosis and necrosis, which may be responsible for ALLHAT outcomes; however, these effects were proven to be independent of α1-adrenoceptor blockade and were associated with the presence of quinazoline moiety. MH-76 (1-[3-(2,6-dimethylphenoxy)propyl]-4-(2-methoxyphenyl)piperazine hydrochloride)) is a non-quinazoline α1-adrenoceptor antagonist which, in fructose-fed rats, exerted antihypertensive effect, and, contrary to prazosin, reduced insulin resistance and abdominal adiposity. In this study we aimed to further investigate and compare the effects of MH-76 and prazosin on inflammation in adipose tissue of fructose-fed rats. Methods: Abdominal adipose tissue was collected from four groups of fructose-fed rats (Control, Fructose, Fructose + MH-76 and Fructose + Prazosin) and subjected to biochemical, histopathological and immunohistochemical studies. Moreover, selected tissue distribution studies were performed. Results: Treatment with MH-76 but not with prazosin improved endothelial integrity, reduced adipose tissue inflammation and infiltration by immune cells, resulting in lowering leptin, MCP-1, IL-6, TNF-α and PAI-1 levels. In adipose tissue from Fructose + MH-76 animals, a higher amount of eosinophils accompanied with higher IL-4 concentration was observed. Treatment with MH-76 but not with prazosin markedly reduced phosphorylation of IRS-1 at Ser307. Conclusion: MH-76 may improve insulin signaling in adipose tissue by reducing the pro-inflammatory cytokine production and inhibiting the inflammatory cells recruitment. In contrast, in adipose tissue from animals treated with prazosin, the inflammatory effect was clearly enhanced.
Collapse
|
32
|
He Z, Wang G, Wu J, Tang Z, Luo M. The molecular mechanism of LRP1 in physiological vascular homeostasis and signal transduction pathways. Biomed Pharmacother 2021; 139:111667. [PMID: 34243608 DOI: 10.1016/j.biopha.2021.111667] [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: 01/21/2021] [Revised: 04/07/2021] [Accepted: 04/23/2021] [Indexed: 01/10/2023] Open
Abstract
Interactions between vascular smooth muscle cells (VSMCs), endothelial cells (ECs), pericytes (PCs) and macrophages (MФ), the major components of blood vessels, play a crucial role in maintaining vascular structural and functional homeostasis. Low-density lipoprotein (LDL) receptor-related protein-1 (LRP1), a transmembrane receptor protein belonging to the LDL receptor family, plays multifunctional roles in maintaining endocytosis, homeostasis, and signal transduction. Accumulating evidence suggests that LRP1 modulates vascular homeostasis mainly by regulating vasoactive substances and specific intracellular signaling pathways, including the plasminogen activator inhibitor 1 (PAI-1) signaling pathway, platelet-derived growth factor (PDGF) signaling pathway, transforming growth factor-β (TGF-β) signaling pathway and vascular endothelial growth factor (VEGF) signaling pathway. The aim of the present review is to focus on recent advances in the discovery and mechanism of vascular homeostasis regulated by LRP1-dependent signaling pathways. These recent discoveries expand our understanding of the mechanisms controlling LRP1 as a target for studies on vascular complications.
Collapse
Affiliation(s)
- Zhaohui He
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Drug Discovery Reseach Center, Southwest Medical University, 319 Zhongshan Road, Luzhou, Sichuan 646000, China; Department of Clinical Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Gang Wang
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Drug Discovery Reseach Center, Southwest Medical University, 319 Zhongshan Road, Luzhou, Sichuan 646000, China; Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, the School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Jianbo Wu
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Drug Discovery Reseach Center, Southwest Medical University, 319 Zhongshan Road, Luzhou, Sichuan 646000, China; Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, the School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China; Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States
| | - Zonghao Tang
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Drug Discovery Reseach Center, Southwest Medical University, 319 Zhongshan Road, Luzhou, Sichuan 646000, China; Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, the School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.
| | - Mao Luo
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Drug Discovery Reseach Center, Southwest Medical University, 319 Zhongshan Road, Luzhou, Sichuan 646000, China; Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, the School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.
| |
Collapse
|
33
|
Morrow GB, Whyte CS, Mutch NJ. A Serpin With a Finger in Many PAIs: PAI-1's Central Function in Thromboinflammation and Cardiovascular Disease. Front Cardiovasc Med 2021; 8:653655. [PMID: 33937363 PMCID: PMC8085275 DOI: 10.3389/fcvm.2021.653655] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 02/23/2021] [Indexed: 12/27/2022] Open
Abstract
Plasminogen activator inhibitor 1 (PAI-1) is a member of the serine protease inhibitor (serpin) superfamily. PAI-1 is the principal inhibitor of the plasminogen activators, tissue plasminogen activator (tPA), and urokinase-type plasminogen activator (uPA). Turbulence in the levels of PAI-1 tilts the balance of the hemostatic system resulting in bleeding or thrombotic complications. Not surprisingly, there is strong evidence that documents the role of PAI-1 in cardiovascular disease. The more recent uncovering of the coalition between the hemostatic and inflammatory pathways has exposed a distinct role for PAI-1. The storm of proinflammatory cytokines liberated during inflammation, including IL-6 and TNF-α, directly influence PAI-1 synthesis and increase circulating levels of this serpin. Consequently, elevated levels of PAI-1 are commonplace during infection and are frequently associated with a hypofibrinolytic state and thrombotic complications. Elevated PAI-1 levels are also a feature of metabolic syndrome, which is defined by a cluster of abnormalities including obesity, type 2 diabetes, hypertension, and elevated triglyceride. Metabolic syndrome is in itself defined as a proinflammatory state associated with elevated levels of cytokines. In addition, insulin has a direct impact on PAI-1 synthesis bridging these pathways. This review describes the key physiological functions of PAI-1 and how these become perturbed during disease processes. We focus on the direct relationship between PAI-1 and inflammation and the repercussion in terms of an ensuing hypofibrinolytic state and thromboembolic complications. Collectively, these observations strengthen the utility of PAI-1 as a viable drug target for the treatment of various diseases.
Collapse
Affiliation(s)
- Gael B Morrow
- Aberdeen Cardiovascular and Diabetes Centre, Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, United Kingdom.,Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Claire S Whyte
- Aberdeen Cardiovascular and Diabetes Centre, Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, United Kingdom
| | - Nicola J Mutch
- Aberdeen Cardiovascular and Diabetes Centre, Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, United Kingdom
| |
Collapse
|
34
|
Levine JA, Olivares S, Miyata T, Vaughan DE, Henkel AS. Inhibition of PAI-1 Promotes Lipolysis and Enhances Weight Loss in Obese Mice. Obesity (Silver Spring) 2021; 29:713-720. [PMID: 33594826 PMCID: PMC8842994 DOI: 10.1002/oby.23112] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 11/30/2020] [Accepted: 12/14/2020] [Indexed: 12/20/2022]
Abstract
OBJECTIVE This study investigates the therapeutic potential of a small molecule inhibitor of plasminogen activator inhibitor-1 (PAI-1), TM5441, in reversing diet-induced obesity in mice. METHODS Wild-type C57BL/6J mice were fed a high-fat high-sugar (HFHS) diet for 8 weeks to induce obesity. After the first 8 weeks, TM5441 was added to the diet for an additional 8 weeks. In order to determine the efficacy of PAI-1 inhibition in conjunction with dietary modification, mice were fed an HFHS diet for 8 weeks to induce obesity and were then switched to a low-fat diet with or without TM5441 for an additional 2 to 8 weeks. RESULTS Obese mice showed weight reduction and significant improvement in hepatic steatosis when TM5441 was added to the HFHS diet. Obese mice that were treated with TM5441 in conjunction with dietary modification showed enhanced weight loss and a more rapid reversal of hepatic steatosis compared with obese mice treated with dietary modification alone. The enhanced weight loss among mice treated with TM5441 was associated with increased adipose tissue expression of adipose triglyceride lipase, phosphorylated hormone-sensitive lipase, and phosphorylated perilipin-1 as well as induction of adipose tissue lipolysis. CONCLUSIONS Pharmacologic PAI-1 inhibition stimulates adipose tissue lipolysis and enhances weight loss in obese mice.
Collapse
Affiliation(s)
- Joshua A. Levine
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Shantel Olivares
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
- Department of Medicine, Jesse Brown VA Medical Center, Chicago, IL
| | - Toshio Miyata
- Department of Molecular Medicine and Therapy, United Centers for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Douglas E. Vaughan
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Anne S. Henkel
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
- Department of Medicine, Jesse Brown VA Medical Center, Chicago, IL
| |
Collapse
|
35
|
Sillen M, Declerck PJ. A Narrative Review on Plasminogen Activator Inhibitor-1 and Its (Patho)Physiological Role: To Target or Not to Target? Int J Mol Sci 2021; 22:ijms22052721. [PMID: 33800359 PMCID: PMC7962805 DOI: 10.3390/ijms22052721] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 02/28/2021] [Accepted: 03/03/2021] [Indexed: 02/06/2023] Open
Abstract
Plasminogen activator inhibitor-1 (PAI-1) is the main physiological inhibitor of plasminogen activators (PAs) and is therefore an important inhibitor of the plasminogen/plasmin system. Being the fast-acting inhibitor of tissue-type PA (tPA), PAI-1 primarily attenuates fibrinolysis. Through inhibition of urokinase-type PA (uPA) and interaction with biological ligands such as vitronectin and cell-surface receptors, the function of PAI-1 extends to pericellular proteolysis, tissue remodeling and other processes including cell migration. This review aims at providing a general overview of the properties of PAI-1 and the role it plays in many biological processes and touches upon the possible use of PAI-1 inhibitors as therapeutics.
Collapse
|
36
|
Qian LL, Ji JJ, Guo JQ, Wu YP, Ma GS, Yao YY. Protective role of serpina3c as a novel thrombin inhibitor against atherosclerosis in mice. Clin Sci (Lond) 2021; 135:447-463. [PMID: 33458764 DOI: 10.1042/cs20201235] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 01/13/2021] [Accepted: 01/18/2021] [Indexed: 12/26/2022]
Abstract
Abnormal vascular smooth muscle cell (VSMC) proliferation is a critical step in the development of atherosclerosis. Serpina3c is a serine protease inhibitor (serpin) that plays a key role in metabolic diseases. The present study aimed to investigate the role of serpina3c in atherosclerosis and regulation of VSMC proliferation and possible mechanisms. Serpina3c is down-regulated during high-fat diet (HFD)-induced atherosclerosis. An Apoe-/-/serpina3c-/--double-knockout mouse model was used to determine the role of serpina3c in atherosclerosis after HFD for 12 weeks. Compared with Apoe-/- mice, the Apoe-/-/serpina3c-/- mice developed more severe atherosclerosis, and the number of VSMCs and macrophages in aortic plaques was significantly increased. The present study revealed serpina3c as a novel thrombin inhibitor that suppressed thrombin activity. In circulating plasma, thrombin activity was high in the Apoe-/-/serpina3c-/- mice, compared with Apoe-/- mice. Immunofluorescence staining showed thrombin and serpina3c colocalization in the liver and aortic cusp. In addition, inhibition of thrombin by dabigatran in serpina3c-/- mice reduced neointima lesion formation due to partial carotid artery ligation. Moreover, an in vitro study confirmed that thrombin activity was also decreased by serpina3c protein, supernatant and cell lysate that overexpressed serpina3c. The results of experiments showed that serpina3c negatively regulated VSMC proliferation in culture. The possible mechanism may involve serpina3c inhibition of ERK1/2 and JNK signaling in thrombin/PAR-1 system-mediated VSMC proliferation. Our results highlight a protective role for serpina3c as a novel thrombin inhibitor in the development of atherosclerosis, with serpina3c conferring protection through the thrombin/PAR-1 system to negatively regulate VSMC proliferation through ERK1/2 and JNK signaling.
Collapse
Affiliation(s)
- Ling-Lin Qian
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao, Nanjing, Jiangsu 210009, China
| | - Jing-Jing Ji
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao, Nanjing, Jiangsu 210009, China
| | - Jia-Qi Guo
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao, Nanjing, Jiangsu 210009, China
| | - Yan-Ping Wu
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao, Nanjing, Jiangsu 210009, China
| | - Gen-Shan Ma
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao, Nanjing, Jiangsu 210009, China
| | - Yu-Yu Yao
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao, Nanjing, Jiangsu 210009, China
| |
Collapse
|
37
|
Nam DE, Seong HC, Hahn YS. Plasminogen Activator Inhibitor-1 and Oncogenesis in the Liver Disease. JOURNAL OF CELLULAR SIGNALING 2021; 2:221-227. [PMID: 34671766 PMCID: PMC8525887 DOI: 10.33696/signaling.2.054] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2023]
Abstract
Hepatocellular carcinoma (HCC) is a significant cause of cancer mortality worldwide. Chronic hepatic inflammation and fibrosis play a critical role in the development of HCC. Liver fibrosis develops as a result of response to injury such that a persistent and excessive wound healing response induces extracellular matrix (ECM) deposition leading to HCC. PAI-1 is a fibrinolysis inhibitor involved in regulating protein degradation and homeostasis while assisting wound healing. PAI-1 presents increased levels in various diseases such as fibrosis, cancer, obesity and metabolic syndrome. Moreover, PAI-1 has been extensively studied for developing potential therapies against fibrosis. In the present review, we summarize how PAI-1 affects oncogenesis during liver disease progression based on the recently published literatures. Although there are controversies regarding the role of PAI-1 and approaches to treatment, this review suggests that proper manipulation of PAI-1 activity could provide a novel therapeutic option on the development of chronic liver disease via modulation of cancer stem-like cells (CSCs) differentiation.
Collapse
Affiliation(s)
- Da-eun Nam
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, USA
| | - Hae Chang Seong
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, USA
| | - Young S. Hahn
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, USA
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, USA
- Correspondence should be addressed to Young S. Hahn;
| |
Collapse
|
38
|
PAI-1, the Plasminogen System, and Skeletal Muscle. Int J Mol Sci 2020; 21:ijms21197066. [PMID: 32993026 PMCID: PMC7582753 DOI: 10.3390/ijms21197066] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/21/2020] [Accepted: 09/23/2020] [Indexed: 12/14/2022] Open
Abstract
The plasminogen system is a critical proteolytic system responsible for the remodeling of the extracellular matrix (ECM). The master regulator of the plasminogen system, plasminogen activator inhibitor-1 (PAI-1), has been implicated for its role in exacerbating various disease states not only through the accumulation of ECM (i.e., fibrosis) but also its role in altering cell fate/behaviour. Examination of PAI-1 has extended through various tissues and cell-types with recent investigations showing its presence in skeletal muscle. In skeletal muscle, the role of this protein has been implicated throughout the regeneration process, and in skeletal muscle pathologies (muscular dystrophy, diabetes, and aging-driven pathology). Needless to say, the complete function of this protein in skeletal muscle has yet to be fully elucidated. Given the importance of skeletal muscle in maintaining overall health and quality of life, it is critical to understand the alterations—particularly in PAI-1—that occur to negatively impact this organ. Thus, we provide a comprehensive review of the importance of PAI-1 in skeletal muscle health and function. We aim to shed light on the relevance of this protein in skeletal muscle and propose potential therapeutic approaches to aid in the maintenance of skeletal muscle health.
Collapse
|