1
|
Moon S, Ito Y. A simplified in vitro disease-mimicking culture system can determine the angiogenic effect of medicines on vascular diseases. Cytotechnology 2025; 77:75. [PMID: 40062227 PMCID: PMC11889311 DOI: 10.1007/s10616-025-00736-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 02/27/2025] [Indexed: 03/21/2025] Open
Abstract
Many patients undergoing clinical regenerative treatments experience severe conditions arising from endothelial disruption. In chronic cardiac and perivascular diseases, deficiencies in vascular endothelial growth factor (VEGF), insulin-like growth factor (IGF), and heparin, which are essential for maintaining and activating endothelial cells, can lead to angiogenic dysregulation. Endothelial disruption caused by ischemic hypoxia and a deficiency in these factors is associated with many vascular diseases. However, their pathogenic processes remain unclear at the cellular level. Therefore, the present study aimed to develop a culture system that mimics the disease environment to test the effectiveness of drug candidates in restoring damaged blood vessels in chronic vascular diseases, including coronary artery disease and peripheral vascular disease. This study focused on VEGF, IGF, and heparin and developed a pseudo-disease culture system by pre-treating human umbilical vein endothelial cells (HUVECs) with a starvation medium (EGM-2™ medium lacking VEGF, IGF, and heparin) to examine the ability of HUVECs to form a traditional 2D vascular network. The results indicated that a deficiency in these proteins results in disruptions in tube morphogenesis. Moreover, the results suggested that dysregulation of the PI3K/AKT pathway plays a key role for in vascular disruption in HUVECs. The proposed pseudo-disease starvation system provides a simple way to visualize pathological disruptions to blood vessels and assess the efficacy of drugs for vascular regeneration. Supplementary Information The online version contains supplementary material available at 10.1007/s10616-025-00736-4.
Collapse
Affiliation(s)
- SongHo Moon
- Institute of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki Japan
| | - Yuzuru Ito
- Institute of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki Japan
- Life Science Development Department, CHIYODA Corporation, Yokohama, Kanagawa Japan
- National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki Japan
| |
Collapse
|
2
|
Wang Y, Yang J, Luo Y, Zhao Z, Yuan Y, Li J, Liu Y, Yi Y, Xu X, Lan Y, Zou J, Li Q, Wang L, Pan Y, Yang Y, Xiong M, Wu M, Li J, Li W, Zhang Y, Cao Y, Zhu Y, Xiao ZXJ. Targeting IGF1-Induced Cellular Senescence to Rejuvenate Hair Follicle Aging. Aging Cell 2025:e70053. [PMID: 40159808 DOI: 10.1111/acel.70053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/28/2025] [Accepted: 03/13/2025] [Indexed: 04/02/2025] Open
Abstract
The insulin-like growth factor-1 (IGF-1) signaling pathway is known as a potent aging modifier, disruption of which consistently associates with lifespan extension across diverse species. Despite this established association, the mechanisms by which IGF-1 signaling modulates organ aging remain poorly understood. In this study, we assessed age-related changes in IGF-1 expression across multiple organs in mice and identified a more prominent increase in skin IGF-1 levels with aging-a phenomenon also observed in human skin. To explore the consequences of elevated IGF-1, we developed transgenic mice ectopically expressing human IGF-1 in the epidermis, driven by the bovine keratin 5 promoter (IGF-1 Tg). These mice exhibited premature aging of hair follicles, as evidenced by accelerated hair graying and loss. Single-cell RNA sequencing analyses of dorsal skin highlighted an upsurge in cellular senescence markers and the senescence-associated secretory phenotype (SASP) in hair follicle stem cells (HFSCs), alongside a decline in hair growth and HFSC exhaustion. Our findings indicate that excessive IGF-1 triggers HFSC senescence, thereby disrupting hair follicle homeostasis. Remarkably, interventions in IGF-1 signaling via downstream mechanisms-specifically blocking Ac-p53 activation via SIRT1 overexpression or senolytic treatment for senescent cell clearance, or reducing IGF-1 through dietary restriction-significantly reduced senescence markers, mitigated premature hair follicle aging phenotypes, and restored the stem cell pool. Our findings provide fundamental insights into the biological processes of hair aging and highlight the therapeutic promise of targeted interventions to rejuvenate aged HFSCs and promote hair follicle health.
Collapse
Affiliation(s)
- Yang Wang
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Jian Yang
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Yue Luo
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Zhiqiang Zhao
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Yawen Yuan
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Juan Li
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yang Liu
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Yong Yi
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Xiaoke Xu
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Yuankunyu Lan
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Juan Zou
- Department of Pathology, West China Second University Hospital, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| | - Qintong Li
- Departments of Obstetrics & Gynecology and Pediatrics, West China Second University Hospital, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Sichuan University, Chengdu, Sichuan, China
| | - Liang Wang
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Yang Pan
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Yuanhan Yang
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Muzhao Xiong
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Min Wu
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Jinsong Li
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Weiyuxin Li
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Yujun Zhang
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Yang Cao
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Yi Zhu
- Department of Physiology and Biomedical Engineering, Robert & Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota, USA
| | - Zhi-Xiong Jim Xiao
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
3
|
Fasoulopoulos A, Varras M, Varra FN, Philippou A, Myoteri D, Varra VK, Kouroglou E, Gryparis A, Papadopetraki A, Vlachos I, Papadopoulos K, Koutsilieris M, Konstantinidou AE. Expression of the IGF‑1Ea isoform in human placentas from third trimester normal and idiopathic intrauterine growth restriction singleton pregnancies: Correlations with clinical and histopathological parameters. Mol Med Rep 2025; 31:69. [PMID: 39791214 PMCID: PMC11751665 DOI: 10.3892/mmr.2025.13434] [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: 05/21/2024] [Accepted: 10/17/2024] [Indexed: 01/12/2025] Open
Abstract
Intrauterine growth restriction (IUGR) is the second most common obstetric complication after preterm labor. Appropriate trophoblast differentiation and placental structure, growth and function are key for the maintenance of pregnancy and normal fetal growth, development and survival. Extravillous trophoblast cell proliferation, migration and invasion are regulated by molecules produced by the fetomaternal interface, including autocrine factors produced by the trophoblast, such as insulin‑like growth factor (IGF)‑1. The aim of the present study was to investigate expression patterns of IGF‑1Ea isoform in IUGR placenta compared with appropriate for gestational age (AGA) pregnancies. Placental frozen tissues were collected from 13 AGA and 15 IUGR third trimester pregnancies for detection of IGF‑1Ea mRNA expression using reverse transcription‑quantitative PCR. Formalin‑fixed paraffin‑embedded samples from 15 AGA and 47 IUGR pregnancies were analyzed immunohistochemically for the identification and localization of the IGF‑1Ea peptide and comparison of clinical and histopathological parameters. To the best of our knowledge, the present study is the first to show IGF‑1Ea expression in third trimester human placenta. The results indicated that similar IGF‑1Ea mRNA expression levels were present in placental specimens from both groups. Cytoplasmic IGF‑1Ea expression was localized in the perivillous syncytiotrophoblast, extravillous trophoblast and endothelium of the villous and decidual vessels in both groups. No significant difference in the scores and intensity of IGF‑1Ea expression in perivillous syncytiotrophoblasts were noted in the IUGR vs. AGA pregnancies. Most IUGR cases showed negative IGF‑1Ea expression in the extravillous trophoblast, whereas AGA pregnancies showed predominantly positive immunostaining. A sex‑specific expression pattern was noted in the extravillous trophoblast, with negative IGF‑1Ea expression in the placentas of female IUGR cases. Additionally, positive immunostaining for IGF‑1Ea peptide in fetal villous and maternal decidual vessels, was more frequently observed in the IUGR group compared with AGA. In conclusion, no difference in total IGF‑1Ea mRNA placental expression was observed between IUGR and AGA pregnancies, likely due to heterogeneity of histological structures expressing this isoform. Negative IGF‑1Ea immunohistological expression in the extravillous trophoblast from IUGR placentas, associated with histological changes of maternal malperfusion, may reflect the involvement of this isoform in defective placentation. The presence of IGF‑1Ea peptide in the endothelium of the villous vessels in IUGR placentas may indicate a reactive autocrine regulation to compensate for malperfused villi in IUGR pregnancy by regulating angiogenesis and vasodilation. The observed sex differences in IGF‑1Ea expression between IUGR and AGA placentas may indicate interactions between sex hormones and selective IGF‑1 binding proteins in regulating IGF‑1Ea synthesis; however, this requires further elucidation.
Collapse
Affiliation(s)
- Apostolos Fasoulopoulos
- Fourth Obstetrics and Gynecology Department, ‘Elena Venizelou’ General Hospital of Athens, 11521 Athens, Greece
| | - Michail Varras
- Fourth Obstetrics and Gynecology Department, ‘Elena Venizelou’ General Hospital of Athens, 11521 Athens, Greece
| | - Fani-Niki Varra
- Medical School, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Anastasios Philippou
- Department of Physiology, Medical School, National Kapodistrian University of Athens, 11527 Athens, Greece
| | - Despina Myoteri
- Department of Pathology, Aretaieion University Hospital, Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | | | - Evgenia Kouroglou
- Fourth Obstetrics and Gynecology Department, ‘Elena Venizelou’ General Hospital of Athens, 11521 Athens, Greece
| | - Alexandros Gryparis
- Department of Speech and Language Therapy, University of Ioannina, 45500 Ioannina, Greece
| | - Argyro Papadopetraki
- Department of Physiology, Medical School, National Kapodistrian University of Athens, 11527 Athens, Greece
| | - Iakovos Vlachos
- Department of Pathology, Aretaieion University Hospital, Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | - Konstantinos Papadopoulos
- Department of Pathology, Aretaieion University Hospital, Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | - Michael Koutsilieris
- Department of Physiology, Medical School, National Kapodistrian University of Athens, 11527 Athens, Greece
| | - Anastasia Evangelia Konstantinidou
- Department of Pathology, Aretaieion University Hospital, Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece
- First Department of Pathology, Unit of Pediatric-Perinatal Pathology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
4
|
Silva-Reis A, Brill B, Brandao-Rangel MAR, Moraes-Ferreira R, Melamed D, Aquino-Santos HC, Frison CR, Albertini R, Lopes-Martins RÁB, de Oliveira LVF, Paixao-Santos G, Oliveira CR, Abbasi A, Vieira RP. Association Between Visceral Fat and Lung Function Impairment in Overweight and Grade I Obese Women: A Cross-Sectional Study. Adv Respir Med 2024; 92:548-558. [PMID: 39727499 PMCID: PMC11673606 DOI: 10.3390/arm92060048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 12/04/2024] [Accepted: 12/11/2024] [Indexed: 12/28/2024]
Abstract
Beyond the common comorbidities related to obesity, such as type 2 diabetes and cardiovascular diseases, impaired lung function is already known, but whether the fat distribution (sub-cutaneous, visceral) affects the lung function and pulmonary immune response are poorly known. Few evidence has shown that visceral fat is associated with insulin resistance, low-grade inflammation, and reduced lung function. In the present study, the body composition and fat distribution were evaluated by multi-frequency octopolar bioimpedance. This study demonstrated a possible association of increased visceral fat with impaired lung function in obesity grade I (n = 28; 45.46 ± 10.38 years old) women that was not observed in normal weight (n = 20; 43.20 ± 10.78 years old) and in overweight women (n = 30; 47.27 ± 10.25 years old). We also identified a negative correlation in FVC% (R2 = 0.9129; p < 0.0236), FEV1% (R2 = 0.1079; p < 0.0134), PEF% (R2 = 0.1673; p < 0.0018), and VC IN% (R2 = 0.1330; p < 0.0057) in the obesity grade I group, clearly demonstrating that higher levels of visceral fat correlate with reduced lung function, but not with sub-cutaneous fat. In addition, for the first time, a negative correlation among anti-fibrotic protein klotho (R2 = 0.09298; p < 0.0897) and anti-inflammatory IL-10 (R2 = 0.1653; p < 0.0487) in plasma was observed, in contrast to increased visceral fat. On the contrary, in breath condensate, a positive correlation for adiponectin (R2 = 0.5665; p < 0.0120), IL1-Ra (R2 = 0.2121; p < 0.0544), and IL1-Beta (R2 = 0.3270; p < 0.0084) was found. Thus, increased visceral fat directly influences the impairment of lung function and the systemic and pulmonary immune response of women with obesity grade I.
Collapse
Affiliation(s)
- Anamei Silva-Reis
- Laboratory of Pulmonary and Exercise Immunology (LABPEI), Evangelical University of Goiás (UniEvangélica), Avenida Universitária Km 3,5, Anápolis 75083-515, GO, Brazil; (A.S.-R.); (R.Á.B.L.-M.); (L.V.F.d.O.); (G.P.-S.)
- Post-Graduation Program in Sciences of Human Movement and Rehabilitation, Federal University of Sao Paulo, Avenida Ana Costa 95, Santos 11060-001, SP, Brazil; (M.A.R.B.-R.); (R.M.-F.); (H.C.A.-S.); (C.R.F.); (R.A.)
| | - Boris Brill
- Leniado Medical Center, Divrei Khayim St. 16, Nethanya 4244916, Israel;
| | - Maysa Alves Rodrigues Brandao-Rangel
- Post-Graduation Program in Sciences of Human Movement and Rehabilitation, Federal University of Sao Paulo, Avenida Ana Costa 95, Santos 11060-001, SP, Brazil; (M.A.R.B.-R.); (R.M.-F.); (H.C.A.-S.); (C.R.F.); (R.A.)
| | - Renilson Moraes-Ferreira
- Post-Graduation Program in Sciences of Human Movement and Rehabilitation, Federal University of Sao Paulo, Avenida Ana Costa 95, Santos 11060-001, SP, Brazil; (M.A.R.B.-R.); (R.M.-F.); (H.C.A.-S.); (C.R.F.); (R.A.)
| | - Dobroslav Melamed
- Department of Research and Development, Libi Pharm, Ben Gurion 70, Rechovot 7639461, Israel;
| | - Helida Cristina Aquino-Santos
- Post-Graduation Program in Sciences of Human Movement and Rehabilitation, Federal University of Sao Paulo, Avenida Ana Costa 95, Santos 11060-001, SP, Brazil; (M.A.R.B.-R.); (R.M.-F.); (H.C.A.-S.); (C.R.F.); (R.A.)
| | - Claudio Ricardo Frison
- Post-Graduation Program in Sciences of Human Movement and Rehabilitation, Federal University of Sao Paulo, Avenida Ana Costa 95, Santos 11060-001, SP, Brazil; (M.A.R.B.-R.); (R.M.-F.); (H.C.A.-S.); (C.R.F.); (R.A.)
| | - Regiane Albertini
- Post-Graduation Program in Sciences of Human Movement and Rehabilitation, Federal University of Sao Paulo, Avenida Ana Costa 95, Santos 11060-001, SP, Brazil; (M.A.R.B.-R.); (R.M.-F.); (H.C.A.-S.); (C.R.F.); (R.A.)
| | - Rodrigo Álvaro Brandao Lopes-Martins
- Laboratory of Pulmonary and Exercise Immunology (LABPEI), Evangelical University of Goiás (UniEvangélica), Avenida Universitária Km 3,5, Anápolis 75083-515, GO, Brazil; (A.S.-R.); (R.Á.B.L.-M.); (L.V.F.d.O.); (G.P.-S.)
| | - Luís Vicente Franco de Oliveira
- Laboratory of Pulmonary and Exercise Immunology (LABPEI), Evangelical University of Goiás (UniEvangélica), Avenida Universitária Km 3,5, Anápolis 75083-515, GO, Brazil; (A.S.-R.); (R.Á.B.L.-M.); (L.V.F.d.O.); (G.P.-S.)
| | - Gustavo Paixao-Santos
- Laboratory of Pulmonary and Exercise Immunology (LABPEI), Evangelical University of Goiás (UniEvangélica), Avenida Universitária Km 3,5, Anápolis 75083-515, GO, Brazil; (A.S.-R.); (R.Á.B.L.-M.); (L.V.F.d.O.); (G.P.-S.)
| | - Carlos Rocha Oliveira
- GAP Biotech Laboratory of Biotechnology and Bioinformatics, Rua Comendador Remo Cesaroni 223, São José dos Campos 12243-020, SP, Brazil;
| | - Asghar Abbasi
- Division of Respiratory and Critical Care Physiology and Medicine, Department of Medicine, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, 1124 W Carson St, CDCRC Building, Torrance, CA 90502, USA;
| | - Rodolfo P. Vieira
- Laboratory of Pulmonary and Exercise Immunology (LABPEI), Evangelical University of Goiás (UniEvangélica), Avenida Universitária Km 3,5, Anápolis 75083-515, GO, Brazil; (A.S.-R.); (R.Á.B.L.-M.); (L.V.F.d.O.); (G.P.-S.)
- Post-Graduation Program in Sciences of Human Movement and Rehabilitation, Federal University of Sao Paulo, Avenida Ana Costa 95, Santos 11060-001, SP, Brazil; (M.A.R.B.-R.); (R.M.-F.); (H.C.A.-S.); (C.R.F.); (R.A.)
- Brazilian Institute of Teaching and Research in Pulmonary and Exercise Immunology (IBEPIPE), Rua Pedro Ernesto 240, São José dos Campos 12245-520, SP, Brazil
| |
Collapse
|
5
|
Li L, Mang XY, Jiang KW, Zhao Y, Chen YR. Swimming training promotes angiogenesis of endothelial progenitor cells by upregulating IGF1 expression and activating the PI3K/AKT pathway in type 2 diabetic rats. Mol Med Rep 2024; 30:237. [PMID: 39422032 PMCID: PMC11529167 DOI: 10.3892/mmr.2024.13361] [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: 04/01/2024] [Accepted: 08/15/2024] [Indexed: 10/19/2024] Open
Abstract
The present study aimed to investigate the effect of swimming training on the angiogenesis of endothelial progenitor cells (EPCs) in type 2 diabetes mellitus (T2DM) rats by upregulating the insulin‑like growth factor 1 (IGF1) expression and to reveal its potential mechanism of action. Male Sprague‑Dawley rats were divided into the Control, Model, Model train, Model train + short interfering (si)‑NC and Model train + si‑IGF1 groups. Serum glucose levels were measured using the oral glucose tolerance test. EPCs were isolated from the bone marrow cavity and identified through morphological observation and immunofluorescence staining. The expression of IGF‑1 mRNA in rat serum and EPCs was analyzed by reverse transcription‑quantitative PCR. The fasting insulin levels in serum were assessed by ELISA. Cell Counting Kit‑8, scratch assay and tube formation assay were used to determine the cell viability, migration and tube formation of rat EPCs, and western blotting was employed to measure the expression levels of IGF1, phosphoinositide 3‑kinase (PI3K), phosphorylated‑PI3K, protein kinase B (AKT) and phosphorylated‑AKT. The present study demonstrated that swimming training significantly decreased the glucose levels and homeostatic model assessment of insulin resistance scores, but increased the fasting insulin levels and IGF1 mRNA expression. Microscopic observation and immunofluorescence identification suggested that EPCs were successfully isolated. In addition, swimming training markedly elevated the levels of IGF1 and promoted cell viability, migration and tube formation in rat EPCs. Furthermore, IGF1 knockdown experiments indicated that swimming training might play a regulatory role by elevating the IGF1 expression to activate the PI3K/AKT pathway. Overall, swimming training promoted the angiogenesis of EPCs in T2DM rats and its potential mechanism may be related to the upregulation of IGF1 expression and the activation of the PI3K/AKT pathway.
Collapse
Affiliation(s)
- Lan Li
- Department of Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong 518020, P.R. China
- Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, Shenzhen, Guangdong 518020, P.R. China
| | - Xiao-Ying Mang
- Department of Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong 518020, P.R. China
- Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, Shenzhen, Guangdong 518020, P.R. China
| | - Ke-Wei Jiang
- Department of Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong 518020, P.R. China
- Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, Shenzhen, Guangdong 518020, P.R. China
| | - Ying Zhao
- Department of Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong 518020, P.R. China
- Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, Shenzhen, Guangdong 518020, P.R. China
| | - Yu-Rong Chen
- Department of Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong 518020, P.R. China
- Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, Shenzhen, Guangdong 518020, P.R. China
| |
Collapse
|
6
|
Nyúl-Tóth Á, Patai R, Csiszar A, Ungvari A, Gulej R, Mukli P, Yabluchanskiy A, Benyo Z, Sotonyi P, Prodan CI, Liotta EM, Toth P, Elahi F, Barsi P, Maurovich-Horvat P, Sorond FA, Tarantini S, Ungvari Z. Linking peripheral atherosclerosis to blood-brain barrier disruption: elucidating its role as a manifestation of cerebral small vessel disease in vascular cognitive impairment. GeroScience 2024; 46:6511-6536. [PMID: 38831182 PMCID: PMC11494622 DOI: 10.1007/s11357-024-01194-0] [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: 04/10/2024] [Accepted: 05/06/2024] [Indexed: 06/05/2024] Open
Abstract
Aging plays a pivotal role in the pathogenesis of cerebral small vessel disease (CSVD), contributing to the onset and progression of vascular cognitive impairment and dementia (VCID). In older adults, CSVD often leads to significant pathological outcomes, including blood-brain barrier (BBB) disruption, which in turn triggers neuroinflammation and white matter damage. This damage is frequently observed as white matter hyperintensities (WMHs) in neuroimaging studies. There is mounting evidence that older adults with atherosclerotic vascular diseases, such as peripheral artery disease, ischemic heart disease, and carotid artery stenosis, face a heightened risk of developing CSVD and VCID. This review explores the complex relationship between peripheral atherosclerosis, the pathogenesis of CSVD, and BBB disruption. It explores the continuum of vascular aging, emphasizing the shared pathomechanisms that underlie atherosclerosis in large arteries and BBB disruption in the cerebral microcirculation, exacerbating both CSVD and VCID. By reviewing current evidence, this paper discusses the impact of endothelial dysfunction, cellular senescence, inflammation, and oxidative stress on vascular and neurovascular health. This review aims to enhance understanding of these complex interactions and advocate for integrated approaches to manage vascular health, thereby mitigating the risk and progression of CSVD and VCID.
Collapse
Affiliation(s)
- Ádám Nyúl-Tóth
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Public Health, Semmelweis University, Semmelweis University, Budapest, Hungary
| | - Roland Patai
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anna Csiszar
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
| | - Anna Ungvari
- Department of Public Health, Semmelweis University, Semmelweis University, Budapest, Hungary.
| | - Rafal Gulej
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Peter Mukli
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Public Health, Semmelweis University, Semmelweis University, Budapest, Hungary
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Doctoral College/Department of Public Health, International Training Program in Geroscience, Semmelweis University, Budapest, Hungary
| | - Zoltan Benyo
- Institute of Translational Medicine, Semmelweis University, 1094, Budapest, Hungary
- Cerebrovascular and Neurocognitive Disorders Research Group, HUN-REN, Semmelweis University, 1094, Budapest, Hungary
| | - Peter Sotonyi
- Department of Vascular and Endovascular Surgery, Heart and Vascular Centre, Semmelweis University, 1122, Budapest, Hungary
| | - Calin I Prodan
- Veterans Affairs Medical Center, Oklahoma City, OK, USA
- Department of Neurology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Eric M Liotta
- Doctoral College/Department of Public Health, International Training Program in Geroscience, Semmelweis University, Budapest, Hungary
- Department of Neurology, Division of Stroke and Neurocritical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Peter Toth
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Public Health, Semmelweis University, Semmelweis University, Budapest, Hungary
- Department of Neurosurgery, Medical School, University of Pecs, Pecs, Hungary
- Neurotrauma Research Group, Szentagothai Research Centre, University of Pecs, Pecs, Hungary
- ELKH-PTE Clinical Neuroscience MR Research Group, University of Pecs, Pecs, Hungary
| | - Fanny Elahi
- Departments of Neurology and Neuroscience Ronald M. Loeb Center for Alzheimer's Disease Friedman Brain Institute Icahn School of Medicine at Mount Sinai, New York, NY, USA
- James J. Peters VA Medical Center, Bronx, NY, USA
| | - Péter Barsi
- ELKH-SE Cardiovascular Imaging Research Group, Department of Radiology, Medical Imaging Centre, Semmelweis University, Budapest, Hungary
| | - Pál Maurovich-Horvat
- ELKH-SE Cardiovascular Imaging Research Group, Department of Radiology, Medical Imaging Centre, Semmelweis University, Budapest, Hungary
| | - Farzaneh A Sorond
- Department of Neurology, Division of Stroke and Neurocritical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Stefano Tarantini
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Doctoral College/Department of Public Health, International Training Program in Geroscience, Semmelweis University, Budapest, Hungary
| | - Zoltan Ungvari
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Doctoral College/Department of Public Health, International Training Program in Geroscience, Semmelweis University, Budapest, Hungary
| |
Collapse
|
7
|
Deng S, Lao M, Zheng H, Hao J. Forkhead box P1 transcriptionally activates IGF-1 to lighten ox-LDL-induced endothelial cellular senescence by inactivating NLRP3 inflammasome. Biogerontology 2024; 26:15. [PMID: 39585426 DOI: 10.1007/s10522-024-10151-5] [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: 05/07/2024] [Accepted: 10/30/2024] [Indexed: 11/26/2024]
Abstract
Endothelial cell (EC) senescence is a major contributor in atherosclerosis (AS) development. Herein, the role of forkhead box P transcription factor 1 (FOXP1) and insulin-like growth factor (IGF)-1 in regulating EC senescence during AS progression was investigated. The mRNA and protein expressions were assessed using qRT-PCR and western blot. IL-1β and IL-18 secretion levels were analyzed by ELISA. Cell viability and pyroptosis were determined by MTT assay and flow cytometry, respectively. SA β-Gal staining was used to measure cell senescence. Tube formation assay was adopted to detect the angiogenesis ability. Dual-luciferase reporter and ChIP assays were used to investigate the relationship between FOXP1 and IGF‑1. ox-LDL stimulation significantly reduced FOXP1 and IGF-1 expression levels in human aortic endothelial cells (HAECs). FOXP1 or IGF-1 overexpression both mitigated ox-LDL-induced cellular senescence and NLRP3 activation in HAECs. It was subsequently revealed that FOXB1 transcriptionally activated IGF-1 expression in HAECs by binding to IGF-1 promoter. Rescue experiments demonstrated that IGF-1 silencing abolished the inhibitory impact of FOXP1 overexpression on ox-LDL-induced cellular senescence and NLRP3 activation in HAECs. FOXP1 transcriptionally activated IGF-1 to lighten ox-LDL-induced endothelial cellular senescence by inactivating NLRP3 inflammasome.
Collapse
Affiliation(s)
- Siqi Deng
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Key Laboratory of Brain Science Research Transformation in Tropical Environment of Hainan Province, School of Basic Medicine and Life Sciences, Hainan Medical University, No. 3, Xueyuan Road, Longhua District, Haikou, 571199, Hainan Province, China
| | - Meili Lao
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Key Laboratory of Brain Science Research Transformation in Tropical Environment of Hainan Province, School of Basic Medicine and Life Sciences, Hainan Medical University, No. 3, Xueyuan Road, Longhua District, Haikou, 571199, Hainan Province, China
| | - Huihui Zheng
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Key Laboratory of Brain Science Research Transformation in Tropical Environment of Hainan Province, School of Basic Medicine and Life Sciences, Hainan Medical University, No. 3, Xueyuan Road, Longhua District, Haikou, 571199, Hainan Province, China
| | - Jingwen Hao
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Key Laboratory of Brain Science Research Transformation in Tropical Environment of Hainan Province, School of Basic Medicine and Life Sciences, Hainan Medical University, No. 3, Xueyuan Road, Longhua District, Haikou, 571199, Hainan Province, China.
| |
Collapse
|
8
|
Lewitt MS, Boyd GW. Insulin-like Growth Factor-Binding Protein-1 (IGFBP-1) as a Biomarker of Cardiovascular Disease. Biomolecules 2024; 14:1475. [PMID: 39595651 PMCID: PMC11592324 DOI: 10.3390/biom14111475] [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: 10/02/2024] [Revised: 10/29/2024] [Accepted: 11/17/2024] [Indexed: 11/28/2024] Open
Abstract
Insulin-like growth factor-binding protein-1 (IGFBP-1) contributes to the regulation of IGFs for metabolism and growth and has IGF-independent actions. IGFBP-1 in the circulation is derived from the liver, where it is inhibited by insulin and stimulated by multiple factors, including proinflammatory cytokines. IGFBP-1 levels are influenced by sex and age, which also determine cardiometabolic risk and patterns of disease presentation. While lower circulating IGFBP-1 concentrations are associated with an unfavorable cardiometabolic risk profile, higher IGFBP-1 predicts worse cardiovascular disease outcomes. This review explores these associations and the possible roles of IGFBP-1 in the pathophysiology of atherosclerosis. We recommend the evaluation of dynamic approaches, such as simultaneous measurements of fasting IGFBP-1 and proinsulin level in response to an oral glucose challenge, as well as multi-marker approaches incorporating markers of inflammation.
Collapse
Affiliation(s)
- Moira S. Lewitt
- School of Health and Life Sciences, University of the West of Scotland, Paisley PA1 2BE, UK
| | - Gary W. Boyd
- School of Health and Life Sciences, University of the West of Scotland, Hamilton G72 0LH, UK;
| |
Collapse
|
9
|
Csiszar A, Ungvari A, Patai R, Gulej R, Yabluchanskiy A, Benyo Z, Kovacs I, Sotonyi P, Kirkpartrick AC, Prodan CI, Liotta EM, Zhang XA, Toth P, Tarantini S, Sorond FA, Ungvari Z. Atherosclerotic burden and cerebral small vessel disease: exploring the link through microvascular aging and cerebral microhemorrhages. GeroScience 2024; 46:5103-5132. [PMID: 38639833 PMCID: PMC11336042 DOI: 10.1007/s11357-024-01139-7] [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: 02/20/2024] [Accepted: 03/14/2024] [Indexed: 04/20/2024] Open
Abstract
Cerebral microhemorrhages (CMHs, also known as cerebral microbleeds) are a critical but frequently underestimated aspect of cerebral small vessel disease (CSVD), bearing substantial clinical consequences. Detectable through sensitive neuroimaging techniques, CMHs reveal an extensive pathological landscape. They are prevalent in the aging population, with multiple CMHs often being observed in a given individual. CMHs are closely associated with accelerated cognitive decline and are increasingly recognized as key contributors to the pathogenesis of vascular cognitive impairment and dementia (VCID) and Alzheimer's disease (AD). This review paper delves into the hypothesis that atherosclerosis, a prevalent age-related large vessel disease, extends its pathological influence into the cerebral microcirculation, thereby contributing to the development and progression of CSVD, with a specific focus on CMHs. We explore the concept of vascular aging as a continuum, bridging macrovascular pathologies like atherosclerosis with microvascular abnormalities characteristic of CSVD. We posit that the same risk factors precipitating accelerated aging in large vessels (i.e., atherogenesis), primarily through oxidative stress and inflammatory pathways, similarly instigate accelerated microvascular aging. Accelerated microvascular aging leads to increased microvascular fragility, which in turn predisposes to the formation of CMHs. The presence of hypertension and amyloid pathology further intensifies this process. We comprehensively overview the current body of evidence supporting this interconnected vascular hypothesis. Our review includes an examination of epidemiological data, which provides insights into the prevalence and impact of CMHs in the context of atherosclerosis and CSVD. Furthermore, we explore the shared mechanisms between large vessel aging, atherogenesis, microvascular aging, and CSVD, particularly focusing on how these intertwined processes contribute to the genesis of CMHs. By highlighting the role of vascular aging in the pathophysiology of CMHs, this review seeks to enhance the understanding of CSVD and its links to systemic vascular disorders. Our aim is to provide insights that could inform future therapeutic approaches and research directions in the realm of neurovascular health.
Collapse
Affiliation(s)
- Anna Csiszar
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anna Ungvari
- Department of Public Health, Semmelweis University, Semmelweis University, Budapest, Hungary.
| | - Roland Patai
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Rafal Gulej
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Zoltan Benyo
- Institute of Translational Medicine, Semmelweis University, 1094, Budapest, Hungary
- Cerebrovascular and Neurocognitive Disorders Research Group, HUN-REN, Semmelweis University, 1094, Budapest, Hungary
| | - Illes Kovacs
- Department of Ophthalmology, Semmelweis University, 1085, Budapest, Hungary
- Department of Ophthalmology, Weill Cornell Medical College, New York, NY, 10021, USA
| | - Peter Sotonyi
- Department of Vascular and Endovascular Surgery, Heart and Vascular Centre, Semmelweis University, 1122, Budapest, Hungary
| | - Angelia C Kirkpartrick
- Veterans Affairs Medical Center, Oklahoma City, OK, USA
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Calin I Prodan
- Veterans Affairs Medical Center, Oklahoma City, OK, USA
- Department of Neurology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Eric M Liotta
- International Training Program in Geroscience, Doctoral College/Department of Public Health, Semmelweis University, Budapest, Hungary
- Department of Neurology, Division of Stroke and Neurocritical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Xin A Zhang
- Department of Physiology, University of Oklahoma Health Science Center, Oklahoma City, OK, USA
| | - Peter Toth
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Public Health, Semmelweis University, Semmelweis University, Budapest, Hungary
- Department of Neurosurgery, Medical School, University of Pecs, Pecs, Hungary
- Neurotrauma Research Group, Szentagothai Research Centre, University of Pecs, Pecs, Hungary
- ELKH-PTE Clinical Neuroscience MR Research Group, University of Pecs, Pecs, Hungary
| | - Stefano Tarantini
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Farzaneh A Sorond
- Department of Neurology, Division of Stroke and Neurocritical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Zoltan Ungvari
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College/Department of Public Health, Semmelweis University, Budapest, Hungary
| |
Collapse
|
10
|
Butt TH, Tobiume M, Re DB, Kariya S. Physical Exercise Counteracts Aging-Associated White Matter Demyelination Causing Cognitive Decline. Aging Dis 2024; 15:2136-2148. [PMID: 38377028 PMCID: PMC11346408 DOI: 10.14336/ad.2024.0216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 02/16/2024] [Indexed: 02/22/2024] Open
Abstract
In the central nervous system, oligodendrocytes wrap around neuronal axons to form myelin, an insulating layer or sheath that allows for the efficient conductance of action potentials. In addition to structural insulation, myelin provides encased axons with nutrient, metabolic and defensive support. Demyelination, or myelin loss, can therefore cause axonal dysfunction, leading to neurological impairment and disease. In Alzheimer's disease (AD), progressive white matter demyelination is acknowledged as one of the earliest pathologies preceding symptom onset. Unfortunately, current pharmacotherapy for slowing demyelination or promoting remyelination in AD is nonexistent. Exercise is recognized for its wide-ranging benefits to human health, including improved mental health and the prevention of lifestyle-related diseases. Mounting evidence suggests the contribution of physical activity in delaying the progression of dementia in elderly populations. Recent mechanistic studies have shown that exercise facilitates myelination in the brain through the vitalization of intrinsic pro-myelination cues, such as increased neurotrophic factors and electrical activity. In this review, we summarize and discuss the potential of physical exercise on counteracting aging-associated white matter demyelination, which causes cognitive decline in AD. We highlight the need of further basic and clinical research investigations on this topic to establish novel approaches for healthy and improved brain aging.
Collapse
Affiliation(s)
- Tanya H Butt
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Makoto Tobiume
- Unit for Respiratory System & Dementia in the Division of Internal Medicine, Katsuren Hospital, Itoman, Okinawa, Japan
| | - Diane B Re
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, USA
- NIEHS Center for Environmental Health Sciences in Northern Manhattan, Columbia University, New York, NY, USA
- Center for Motor Neuron Biology and Disease, Columbia University, New York, NY, USA
| | - Shingo Kariya
- Unit for Nervous System & Dementia in the Division of Internal Medicine, Katsuren Hospital, Itoman, Okinawa, Japan
| |
Collapse
|
11
|
Hemat Jouy S, Mohan S, Scichilone G, Mostafa A, Mahmoud AM. Adipokines in the Crosstalk between Adipose Tissues and Other Organs: Implications in Cardiometabolic Diseases. Biomedicines 2024; 12:2129. [PMID: 39335642 PMCID: PMC11428859 DOI: 10.3390/biomedicines12092129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 09/14/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024] Open
Abstract
Adipose tissue was previously regarded as a dormant organ for lipid storage until the identification of adiponectin and leptin in the early 1990s. This revelation unveiled the dynamic endocrine function of adipose tissue, which has expanded further. Adipose tissue has emerged in recent decades as a multifunctional organ that plays a significant role in energy metabolism and homeostasis. Currently, it is evident that adipose tissue primarily performs its function by secreting a diverse array of signaling molecules known as adipokines. Apart from their pivotal function in energy expenditure and metabolism regulation, these adipokines exert significant influence over a multitude of biological processes, including but not limited to inflammation, thermoregulation, immune response, vascular function, and insulin sensitivity. Adipokines are pivotal in regulating numerous biological processes within adipose tissue and facilitating communication between adipose tissue and various organs, including the brain, gut, pancreas, endothelial cells, liver, muscle, and more. Dysregulated adipokines have been implicated in several metabolic diseases, like obesity and diabetes, as well as cardiovascular diseases. In this article, we attempted to describe the significance of adipokines in developing metabolic and cardiovascular diseases and highlight their role in the crosstalk between adipose tissues and other tissues and organs.
Collapse
Affiliation(s)
- Shaghayegh Hemat Jouy
- Department of Exercise Physiology, Faculty of Physical Education and Sport Sciences, Central Tehran Branch, Islamic Azad University, Tehran 14778-93855, Iran;
| | - Sukrutha Mohan
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (S.M.); (G.S.)
| | - Giorgia Scichilone
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (S.M.); (G.S.)
| | - Amro Mostafa
- Department of Pharmacology, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA;
| | - Abeer M. Mahmoud
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (S.M.); (G.S.)
- Department of Kinesiology and Nutrition, College of Applied Health Sciences, University of Illinois Chicago, Chicago, IL 60612, USA
| |
Collapse
|
12
|
Liu T, Li F, Fei Y, Sun F, Chen M, Tian X, Zheng W, Zhu Z, Wang W. Serum insulin-like growth factor-1 as a potential prognostic biomarker for heart failure with reduced ejection fraction: a meta-analysis. Front Cardiovasc Med 2024; 11:1415238. [PMID: 39355348 PMCID: PMC11442213 DOI: 10.3389/fcvm.2024.1415238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 09/06/2024] [Indexed: 10/03/2024] Open
Abstract
Background Most studies have indicated that peripheral insulin-like growth levels factor-1 (IGF-1) is valuable in diagnosing heart failure, although the results have been inconsistent. To help solve the debate, we performed a meta-analysis to explore the relationship between IGF-1 and heart failure (HF). Methods We conducted an extensive search across various databases such as Embase, Cochrane Library, Pubmed, Medline, and Web of Science on May 30, 2023. From the extensive pool of studies, we selected 16 relevant articles, encompassing a total of 1,380 cases and 1,153 controls, to conduct a rigorous meta-analysis. Results The total results indicated that there is an association between lower IGF-1 level and HF. The random-effects model yielded a pooled standardized mean difference (SMD) of -0.598 (95% CI: -1.081 to -0.116, P = 0.015). Further subgroup analysis also showed that IGF-1 levels were associated with HF in the age difference ≥5 years subgroup and body mass index difference >1 subgroup. Additionally, significant association between IGF-1 levels and HF were detected in the "serum" samples and "Europe" subgroups. Importantly, we observed IGF-1 showed significant lower levels in patients with reduced ejection fraction (HFrEF) compared to the controls, not in patients with preserved ejection fraction (HFpEF). The Begg's and Egger's tests revealed no indication of publication bias. Conclusions Our meta-analysis has provided evidence suggesting a substantial correlation between reduced levels of IGF-1 and the occurrence of HF. Further prospective studies are necessary to ascertain the use of IGF-1 as a reliable biomarker for diagnosing HF, especially for HFrEF. But the diagnosis of HFpEF should be cautious.
Collapse
Affiliation(s)
- Tingting Liu
- Department of Experimental Animal Laboratory, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Fangyu Li
- Innovation Center for Neurological Disorders and Department of Neurology, National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Yihuan Fei
- School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, Hebei, China
| | - Fangling Sun
- Department of Experimental Animal Laboratory, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Mengqi Chen
- Department of Experimental Animal Laboratory, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Xin Tian
- Department of Experimental Animal Laboratory, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Wenrong Zheng
- Department of Experimental Animal Laboratory, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Zixin Zhu
- Department of Experimental Animal Laboratory, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Wen Wang
- Department of Experimental Animal Laboratory, Xuanwu Hospital of Capital Medical University, Beijing, China
- Beijing Institute for Brain Disorders, Beijing, China
| |
Collapse
|
13
|
Salah A, Bouzid F, Dhouib W, Benmarzoug R, Triki N, Rebai A, Kharrat N. Integrative Bioinformatics Approaches to Uncover Hub Genes and Pathways Involved in Cardiovascular Diseases. Cell Biochem Biophys 2024; 82:2107-2127. [PMID: 38809349 DOI: 10.1007/s12013-024-01319-4] [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] [Accepted: 05/15/2024] [Indexed: 05/30/2024]
Abstract
Cardiovascular diseases (CVD) represent a significant global health challenge resulting from a complex interplay of genetic, environmental, and lifestyle factors. However, the molecular pathways and genetic factors involved in the onset and progression of CVDs remain incompletely understood. Here, we performed an integrative bioinformatic analysis to highlight specific genes and signaling pathways implicated in the pathogenesis of 80 CVDs. Differentially expressed genes (DEGs) were identified through the integrated analysis of microarray and GWAS datasets. Then, hub genes were identified after gene ontology functional annotation analysis and protein-protein internet (PPI) analysis. In addition, pathways were identified through KEGG and gene ontology enrichment analyses. A total of 821 hub genes related to 80 CVDs were identified, including 135 common and frequent CVD-associated genes. TNF, IL6, VEGFA, and TGFB.1 genes were the central core genes expressed in 50% or more of CVDs, confirming that the inflammation is a key pathological feature of CVDs. Analysis of hub genes by KEGG enrichment revealed predominant enrichment in 201 KEGG pathways, of which the AGE-RAGE signaling pathway in diabetic complications was identified as the common key KEGG implicated in 62 CVDs. In addition, the outcomes showed an overrepresentation in pathways categorized under human diseases, particularly in the subcategories of infectious diseases and cancers, which may be common risk factors for CVDs. In conclusion, this powerful approach for in silico fine-mapping of genes and pathways allowed the identification of determinant hubs genes and pathways implicated in the pathogenesis of CVDs which could be employed in developing more targeted and effective interventions for preventing, diagnosing, and treating CVDs. The function of these hub genes in CVDs needs further exploration to elucidate their biological characteristics.
Collapse
Affiliation(s)
- Awatef Salah
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia.
| | - Fériel Bouzid
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | - Wala Dhouib
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | - Riadh Benmarzoug
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | - Nesrine Triki
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | - Ahmed Rebai
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | - Najla Kharrat
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| |
Collapse
|
14
|
Lee WS, Abel ED, Kim J. New Insights into IGF-1 Signaling in the Heart. Physiology (Bethesda) 2024; 39:0. [PMID: 38713091 DOI: 10.1152/physiol.00003.2024] [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: 01/10/2024] [Revised: 04/24/2024] [Accepted: 05/04/2024] [Indexed: 05/08/2024] Open
Abstract
Insulin-like growth factor-1 (IGF-1) signaling has multiple physiological roles in cellular growth, metabolism, and aging. Myocardial hypertrophy, cell death, senescence, fibrosis, and electrical remodeling are hallmarks of various heart diseases and contribute to the progression of heart failure. This review highlights the critical role of IGF-1 and its cognate receptor in cardiac hypertrophy, aging, and remodeling.
Collapse
Affiliation(s)
- Wang-Soo Lee
- Division of Cardiology, Department of Internal Medicine, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - E Dale Abel
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States
| | - Jaetaek Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| |
Collapse
|
15
|
Thapa R, Moglad E, Goyal A, Bhat AA, Almalki WH, Kazmi I, Alzarea SI, Ali H, Oliver BG, MacLoughlin R, Dureja H, Singh SK, Dua K, Gupta G. Deciphering NF-kappaB pathways in smoking-related lung carcinogenesis. EXCLI JOURNAL 2024; 23:991-1017. [PMID: 39253534 PMCID: PMC11382301 DOI: 10.17179/excli2024-7475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Accepted: 07/01/2024] [Indexed: 09/11/2024]
Abstract
One of the main causes of death worldwide is lung cancer, which is largely caused by cigarette smoking. The crucial transcription factor NF-κB, which controls inflammatory responses and various cellular processes, is a constitutively present cytoplasmic protein strictly regulated by inhibitors like IκB proteins. Upon activation by external stimuli, it undergoes phosphorylation, translocates into the nucleus, and modulates the expression of specific genes. The incontrovertible association between pulmonary malignancy and tobacco consumption underscores and highlights a public health concern. Polycyclic aromatic hydrocarbons and nitrosamines, potent carcinogenic compounds present in the aerosol emitted from combusted tobacco, elicit profound deleterious effects upon inhalation, resulting in severe perturbation of pulmonary tissue integrity. The pathogenesis of smoking-induced lung cancer encompasses an intricate process wherein NF-κB activation plays a pivotal role, triggered by exposure to cigarette smoke through diverse signaling pathways, including those associated with oxidative stress and pro-inflammatory cytokines. Unraveling the participation of NF-κB in smoking-induced lung cancer provides pivotal insights into molecular processes, wherein intricate crosstalk between NF-κB and pathways such as MAPK and PI3K-Akt amplifies the inflammatory response, fostering an environment conducive to the formation of lung cancer. This study reviews the critical function of NF-κB in the complex molecular pathways linked to the initiation and advancement of lung carcinogenesis as well as potential treatment targets. See also the graphical abstract(Fig. 1).
Collapse
Affiliation(s)
- Riya Thapa
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Ehssan Moglad
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj 11942, Saudi Arabia
| | - Ahsas Goyal
- Institute of Pharmaceutical Research, GLA University, Mathura, U.P., India
| | - Asif Ahmad Bhat
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, 21589, Jeddah, Saudi Arabia
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, 72341, Sakaka, Al-Jouf, Saudi Arabia
| | - Haider Ali
- Center for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India
- Department of Pharmacology, Kyrgyz State Medical College, Bishkek, Kyrgyzstan
| | - Brian Gregory Oliver
- Woolcock Institute of Medical Research, Macquarie University, Sydney, NSW 2137 Australia
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007 Australia
| | - Ronan MacLoughlin
- Research and Development, Aerogen Limited, IDA Business Park, Galway, Connacht, H91 HE94 Ireland
- School of Pharmacy & Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Leinster, D02 YN77 Ireland
- School of Pharmacy & Pharmaceutical Sciences, Trinity College, Dublin, Leinster, D02 PN40 Ireland
| | - Harish Dureja
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, 124001, Haryana, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia
- School of Medical and Life Sciences, Sunway University, Sunway City, 47500, Malaysia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW 2007, Australia
| | - Gaurav Gupta
- Center for Research Impact & Outcome-Chitkara College of Pharmacy, Chitkara University, Punjab
- Center of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| |
Collapse
|
16
|
Dubin RF, Deo R, Ren Y, Wang J, Pico AR, Mychaleckyj JC, Kozlitina J, Arthur V, Lee H, Shah A, Feldman H, Bansal N, Zelnick L, Rao P, Sukul N, Raj DS, Mehta R, Rosas SE, Bhat Z, Weir MR, He J, Chen J, Kansal M, Kimmel PL, Ramachandran VS, Waikar SS, Segal MR, Ganz P. Incident heart failure in chronic kidney disease: proteomics informs biology and risk stratification. Eur Heart J 2024; 45:2752-2767. [PMID: 38757788 PMCID: PMC11313584 DOI: 10.1093/eurheartj/ehae288] [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: 07/30/2023] [Revised: 04/09/2024] [Accepted: 04/25/2024] [Indexed: 05/18/2024] Open
Abstract
BACKGROUND AND AIMS Incident heart failure (HF) among individuals with chronic kidney disease (CKD) incurs hospitalizations that burden patients and health care systems. There are few preventative therapies, and the Pooled Cohort equations to Prevent Heart Failure (PCP-HF) perform poorly in the setting of CKD. New drug targets and better risk stratification are urgently needed. METHODS In this analysis of incident HF, SomaScan V4.0 (4638 proteins) was analysed in 2906 participants of the Chronic Renal Insufficiency Cohort (CRIC) with validation in the Atherosclerosis Risk in Communities (ARIC) study. The primary outcome was 14-year incident HF (390 events); secondary outcomes included 4-year HF (183 events), HF with reduced ejection fraction (137 events), and HF with preserved ejection fraction (165 events). Mendelian randomization and Gene Ontology were applied to examine causality and pathways. The performance of novel multi-protein risk models was compared to the PCP-HF risk score. RESULTS Over 200 proteins were associated with incident HF after adjustment for estimated glomerular filtration rate at P < 1 × 10-5. After adjustment for covariates including N-terminal pro-B-type natriuretic peptide, 17 proteins remained associated at P < 1 × 10-5. Mendelian randomization associations were found for six proteins, of which four are druggable targets: FCG2B, IGFBP3, CAH6, and ASGR1. For the primary outcome, the C-statistic (95% confidence interval [CI]) for the 48-protein model in CRIC was 0.790 (0.735, 0.844) vs. 0.703 (0.644, 0.762) for the PCP-HF model (P = .001). C-statistic (95% CI) for the protein model in ARIC was 0.747 (0.707, 0.787). CONCLUSIONS Large-scale proteomics reveal novel circulating protein biomarkers and potential mediators of HF in CKD. Proteomic risk models improve upon the PCP-HF risk score in this population.
Collapse
Affiliation(s)
- Ruth F Dubin
- Division of Nephrology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, H5.122E, Dallas, TX 75390, USA
| | - Rajat Deo
- Division of Cardiovascular Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Yue Ren
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jianqiao Wang
- Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Alexander R Pico
- Institute of Data Science and Biotechnology, Gladstone Institutes, San Francisco, CA, USA
| | - Josyf C Mychaleckyj
- Center for Public Health Genomics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Julia Kozlitina
- McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Victoria Arthur
- Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Hongzhe Lee
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Amil Shah
- Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Harold Feldman
- Patient-Centered Outcomes Research Institute, Washington, DC, USA
| | - Nisha Bansal
- Division of Nephrology, University of Washington Medical Center, Seattle, WA, USA
| | - Leila Zelnick
- Division of Nephrology, University of Washington Medical Center, Seattle, WA, USA
| | - Panduranga Rao
- Division of Nephrology, University of Michigan, Ann Arbor, MI, USA
| | - Nidhi Sukul
- Division of Nephrology, University of Michigan, Ann Arbor, MI, USA
| | - Dominic S Raj
- Division of Kidney Diseases and Hypertension, George Washington University School of Medicine, Washington, DC, USA
| | - Rupal Mehta
- Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, USA
| | - Sylvia E Rosas
- Joslin Diabetes Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Zeenat Bhat
- Division of Nephrology, University of Michigan, Ann Arbor, MI, USA
| | - Matthew R Weir
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jiang He
- Department of Epidemiology, Tulane University, New Orleans, LA, USA
| | - Jing Chen
- Department of Epidemiology, Tulane University, New Orleans, LA, USA
| | - Mayank Kansal
- Division of Cardiology, University of Illinois College of Medicine, Chicago, IL, USA
| | - Paul L Kimmel
- Division of Kidney, Urologic, and Hematologic Diseases, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Vasan S Ramachandran
- University of Texas School of Public Health San Antonio and the University of Texas Health Sciences Center in San Antonio, Section of Preventive Medicine and Epidemiology, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Sushrut S Waikar
- Section of Nephrology, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Mark R Segal
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Peter Ganz
- Division of Cardiology, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
17
|
Adasheva DA, Serebryanaya DV. IGF Signaling in the Heart in Health and Disease. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:1402-1428. [PMID: 39245453 DOI: 10.1134/s0006297924080042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 06/11/2024] [Accepted: 06/22/2024] [Indexed: 09/10/2024]
Abstract
One of the most vital processes of the body is the cardiovascular system's proper operation. Physiological processes in the heart are regulated by the balance of cardioprotective and pathological mechanisms. The insulin-like growth factor system (IGF system, IGF signaling pathway) plays a pivotal role in regulating growth and development of various cells and tissues. In myocardium, the IGF system provides cardioprotective effects as well as participates in pathological processes. This review summarizes recent data on the role of IGF signaling in cardioprotection and pathogenesis of various cardiovascular diseases, as well as analyzes severity of these effects in various scenarios.
Collapse
Affiliation(s)
- Daria A Adasheva
- Department of Biochemistry, Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
| | - Daria V Serebryanaya
- Department of Biochemistry, Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia.
- Pirogov Russian National Research Medical University, Moscow, 117997, Russia
| |
Collapse
|
18
|
Chun-peng ZHANG, Tian CAO, Xue YANG. Pharmacological mechanisms of Taohe Chengqi decoction in diabetic cardiovascular complications: A systematic review, network pharmacology and molecular docking. Heliyon 2024; 10:e33308. [PMID: 39044965 PMCID: PMC11263673 DOI: 10.1016/j.heliyon.2024.e33308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 06/18/2024] [Accepted: 06/19/2024] [Indexed: 07/25/2024] Open
Abstract
Background Diabetic cardiovascular complications are the leading cause of diabetes-related deaths. These complications place an enormous and growing burden on global health systems and economies. The objective of this study was to conduct a systematic review on the therapeutic mechanisms of Taohe Chengqi Decoction (THCQD) in the treatment of diabetic cardiovascular complications. To predict the potential mechanisms of action of THCQD on diabetic cardiovascular complications using network pharmacology, and to validate these predictions through molecular docking analysis. Methods To collect relevant animal experiments, we searched a total of 6 databases. Eligibility for the study was determined based on inclusion and exclusion criteria. Data extraction was then performed on the literature. Methodological quality of animal studies was assessed using SYRCLE criteria. Based on network pharmacology, intersecting genes for THCQD and diabetic cardiovascular complications were obtained using Venny, PPI analysis and topology analysis of intersecting genes were performed; GO and KEGG were used for enrichment analysis and prediction of new targets of action. Molecular docking techniques were employed to model the interactions between drug components and target genes, thereby validating the results of network pharmacology predictions. Results A total of 16 studies were finally identified that fit the direction of this review. Included 6 studies of the myocardium, 1 study of the aortic arch, 5 studies of the femoral artery, 4 studies of the thoracic aorta. THCQD exhibited anti-inflammatory, anti-fibrotic and anti-atherosclerotic effects on cardiovascular complications in diabetic rats. Network pharmacology results showed that C0363 (Resveratrol), C0041 (Emodin), and C1114 (Baicalein) were the key components in the treatment of diabetic cardiovascular complications by THCQD. PPI results showed that INS, AKT1, TNF, ALB, IL6, IL1B as the genes that interact with the top 6. KEGG enrichment analysis identified the AGE-RAGE signaling pathway in diabetic complications as the most prominent pathway enriched by THCQD for diabetic cardiovascular complications genes. The results of molecular docking showed that the key active components demonstrated favorable interactions with their corresponding target genes. Conclusion In conclusion, the results of both basic and web-based pharmacological studies support the beneficial effects of the natural herbal formulation THCQD on diabetic cardiovascular complications. This decoction has anti-inflammatory and antifibrotic properties and is effective in ameliorating diabetic cardiovascular disease. The network pharmacology results further support these ideas and identify the AGE-RAGE signaling pathway in diabetic complications as possibly the most relevant pathway for THCQD in the treatment of diabetic cardiovascular complications. The extent of the therapeutic potential of all-natural herbal components in the treatment of diabetic cardiovascular disease merits further investigation.
Collapse
Affiliation(s)
- ZHANG Chun-peng
- Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - CAO Tian
- Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - YANG Xue
- Department of Traditional Chinese Medicine, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, 200090, China
| |
Collapse
|
19
|
Trastulla L, Dolgalev G, Moser S, Jiménez-Barrón LT, Andlauer TFM, von Scheidt M, Budde M, Heilbronner U, Papiol S, Teumer A, Homuth G, Völzke H, Dörr M, Falkai P, Schulze TG, Gagneur J, Iorio F, Müller-Myhsok B, Schunkert H, Ziller MJ. Distinct genetic liability profiles define clinically relevant patient strata across common diseases. Nat Commun 2024; 15:5534. [PMID: 38951512 PMCID: PMC11217418 DOI: 10.1038/s41467-024-49338-2] [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: 02/20/2024] [Accepted: 05/31/2024] [Indexed: 07/03/2024] Open
Abstract
Stratified medicine holds great promise to tailor treatment to the needs of individual patients. While genetics holds great potential to aid patient stratification, it remains a major challenge to operationalize complex genetic risk factor profiles to deconstruct clinical heterogeneity. Contemporary approaches to this problem rely on polygenic risk scores (PRS), which provide only limited clinical utility and lack a clear biological foundation. To overcome these limitations, we develop the CASTom-iGEx approach to stratify individuals based on the aggregated impact of their genetic risk factor profiles on tissue specific gene expression levels. The paradigmatic application of this approach to coronary artery disease or schizophrenia patient cohorts identified diverse strata or biotypes. These biotypes are characterized by distinct endophenotype profiles as well as clinical parameters and are fundamentally distinct from PRS based groupings. In stark contrast to the latter, the CASTom-iGEx strategy discovers biologically meaningful and clinically actionable patient subgroups, where complex genetic liabilities are not randomly distributed across individuals but rather converge onto distinct disease relevant biological processes. These results support the notion of different patient biotypes characterized by partially distinct pathomechanisms. Thus, the universally applicable approach presented here has the potential to constitute an important component of future personalized medicine paradigms.
Collapse
Affiliation(s)
- Lucia Trastulla
- Max Planck Institute of Psychiatry, Munich, Germany
- Technische Universität München Medical Graduate Center Experimental Medicine, Munich, Germany
- Human Technopole, Milan, Italy
- Department of Psychiatry, University of Münster, Münster, Germany
| | - Georgii Dolgalev
- Department of Psychiatry, University of Münster, Münster, Germany
| | - Sylvain Moser
- Max Planck Institute of Psychiatry, Munich, Germany
- Technische Universität München Medical Graduate Center Experimental Medicine, Munich, Germany
- International Max Planck Research School for Translational Psychiatry (IMPRS-TP), Munich, Germany
| | - Laura T Jiménez-Barrón
- Max Planck Institute of Psychiatry, Munich, Germany
- International Max Planck Research School for Translational Psychiatry (IMPRS-TP), Munich, Germany
| | - Till F M Andlauer
- Max Planck Institute of Psychiatry, Munich, Germany
- Department of Neurology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Moritz von Scheidt
- Klinik für Herz-und Kreislauferkrankungen, Deutsches Herzzentrum München, Technical University Munich, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Monika Budde
- Institute of Psychiatric Phenomics and Genomics (IPPG), LMU University Hospital, LMU Munich, Munich, 80336, Germany
| | - Urs Heilbronner
- Institute of Psychiatric Phenomics and Genomics (IPPG), LMU University Hospital, LMU Munich, Munich, 80336, Germany
| | - Sergi Papiol
- Max Planck Institute of Psychiatry, Munich, Germany
- Institute of Psychiatric Phenomics and Genomics (IPPG), LMU University Hospital, LMU Munich, Munich, 80336, Germany
| | - Alexander Teumer
- German Center for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, Germany
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | - Georg Homuth
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Henry Völzke
- German Center for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, Germany
- Institute of Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Marcus Dörr
- German Center for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, Germany
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
| | - Peter Falkai
- Max Planck Institute of Psychiatry, Munich, Germany
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, 80336, Germany
| | - Thomas G Schulze
- Institute of Psychiatric Phenomics and Genomics (IPPG), LMU University Hospital, LMU Munich, Munich, 80336, Germany
- Department of Psychiatry and Behavioral Sciences, SUNY Upstate Medical University, Syracuse, NY, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Julien Gagneur
- School of Computation, Information and Technology, Technical University of Munich, Garching, Germany
- Institute of Human Genetics, School of Medicine and Health, Technical University of Munich, Munich, Germany
- Computational Health Center, Helmholtz Center Munich, Neuherberg, Germany
| | | | - Bertram Müller-Myhsok
- Max Planck Institute of Psychiatry, Munich, Germany
- Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Heribert Schunkert
- Klinik für Herz-und Kreislauferkrankungen, Deutsches Herzzentrum München, Technical University Munich, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Michael J Ziller
- Max Planck Institute of Psychiatry, Munich, Germany.
- Department of Psychiatry, University of Münster, Münster, Germany.
- Center for Soft Nanoscience, University of Münster, Münster, Germany.
| |
Collapse
|
20
|
Cheng B, Yang R, Xu H, Wang L, Jiang N, Song T, Dong C. Peripheral Blood miRNA Expression in Patients with Essential Hypertension in the Han Chinese Population in Hefei, China. Biochem Genet 2024:10.1007/s10528-024-10867-6. [PMID: 38907084 DOI: 10.1007/s10528-024-10867-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 06/10/2024] [Indexed: 06/23/2024]
Abstract
Primary hypertension is a significant risk factor for cardiovascular diseases. However, the pathogenesis of primary hypertension involves multiple biological processes, including the nervous system, circulatory system, endocrine system, and more. Despite extensive research, there is no clear understanding of the regulatory mechanism underlying its pathogenesis. In recent years, miRNAs have gained attention as a regulatory factor capable of modulating the expression of related molecules through gene silencing. Therefore, exploring differentially expressed miRNAs in patients with essential hypertension (EH) may offer a novel approach for future diagnosis and treatment of EH. This study included a total of twenty Han Chinese population samples from Hefei, China. The samples consisted of 10 healthy individuals and 10 patients with EH. Statistical analysis was conducted to analyze the general information of the two-sample groups. High-throughput sequencing and base identification were performed to obtain the original sequencing sequences. These sequences were then annotated using various databases including Rfam, cDNA sequences, species repetitive sequences library, and miRBase database. The number of miRNA species contained in the samples was measured. Next, TPM values were calculated to determine the expression level of each miRNA. The bioinformatics of the differentiated miRNAs were analyzed using the OECloud tool, and RPM values were calculated. Furthermore, the reliability of the expression was analyzed by calculating the area under the Roc curve using the OECloud tools. Statistical analysis revealed no significant differences between the two samples in terms of age distribution, gender composition, smoking history, and alcohol consumption history (P > 0.05). However, there was a notable presence of family genetic history and high BMI in the EH population (P < 0.05). The sequencing results identified a total of 245 miRNAs, out of which 16 miRNAs exhibited differential expression. Among the highly expressed miRNAs were let-7d-5p, miR-101-3p, miR-122-5p, miR-122b-3p, miR-192-5p, and miR-6722-3p. On the other hand, the lowly expressed miRNAs included miR-103a-3p, miR-16-5p, miR-181a-2-3p, miR-200a-3p, miR-200b-3p, miR-200c-3p, miR-221-3p, miR-30d-5p, miR-342-5p, and miR-543. This study initially identified 16 miRNAs that are aberrantly expressed and function in various processes associated with the onset and progression of essential hypertension. These miRNAs have the potential to be targeted for future diagnosis and treatment of EH. However, further samples are required to provide additional support for this study.
Collapse
Affiliation(s)
- Bin Cheng
- Department of Anhui University of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Ronglu Yang
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Hui Xu
- College of Traditional Chinese Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Li Wang
- College of Traditional Chinese Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Nan Jiang
- College of Traditional Chinese Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Tingting Song
- The First Clinical Medical School, Anhui University of Chinese Medicine, Hefei, China.
| | - Changwu Dong
- The Second Clinical Medical School, Anhui University of Chinese Medicine, Hefei, China.
| |
Collapse
|
21
|
Miller LR, Bickel MA, Vance ML, Vaden H, Nagykaldi D, Nyul-Toth A, Bullen EC, Gautam T, Tarantini S, Yabluchanskiy A, Kiss T, Ungvari Z, Conley SM. Vascular smooth muscle cell-specific Igf1r deficiency exacerbates the development of hypertension-induced cerebral microhemorrhages and gait defects. GeroScience 2024; 46:3481-3501. [PMID: 38388918 PMCID: PMC11009188 DOI: 10.1007/s11357-024-01090-7] [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: 12/05/2023] [Accepted: 02/03/2024] [Indexed: 02/24/2024] Open
Abstract
Cerebrovascular fragility and cerebral microhemorrhages (CMH) contribute to age-related cognitive impairment, mobility defects, and vascular cognitive impairment and dementia, impairing healthspan and reducing quality of life in the elderly. Insulin-like growth factor 1 (IGF-1) is a key vasoprotective growth factor that is reduced during aging. Circulating IGF-1 deficiency leads to the development of CMH and other signs of cerebrovascular dysfunction. Here our goal was to understand the contribution of IGF-1 signaling on vascular smooth muscle cells (VSMCs) to the development of CMH and associated gait defects. We used an inducible VSMC-specific promoter and an IGF-1 receptor (Igf1r) floxed mouse line (Myh11-CreERT2 Igf1rf/f) to knockdown Igf1r. Angiotensin II in combination with L-NAME-induced hypertension was used to elicit CMH. We observed that VSMC-specific Igf1r knockdown mice had accelerated development of CMH, and subsequent associated gait irregularities. These phenotypes were accompanied by upregulation of a cluster of pro-inflammatory genes associated with VSMC maladaptation. Collectively our findings support an essential role for VSMCs as a target for the vasoprotective effects of IGF-1, and suggest that VSMC dysfunction in aging may contribute to the development of CMH.
Collapse
Affiliation(s)
- Lauren R Miller
- Department of Cell Biology, University of Oklahoma Health Sciences Center, 940 Stanton L. Young Blvd, BMSB 553, Oklahoma City, OK, 73104, USA
- Currently at: Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Marisa A Bickel
- Department of Cell Biology, University of Oklahoma Health Sciences Center, 940 Stanton L. Young Blvd, BMSB 553, Oklahoma City, OK, 73104, USA
| | - Michaela L Vance
- Department of Cell Biology, University of Oklahoma Health Sciences Center, 940 Stanton L. Young Blvd, BMSB 553, Oklahoma City, OK, 73104, USA
| | - Hannah Vaden
- Department of Cell Biology, University of Oklahoma Health Sciences Center, 940 Stanton L. Young Blvd, BMSB 553, Oklahoma City, OK, 73104, USA
| | - Domonkos Nagykaldi
- Department of Cell Biology, University of Oklahoma Health Sciences Center, 940 Stanton L. Young Blvd, BMSB 553, Oklahoma City, OK, 73104, USA
| | - Adam Nyul-Toth
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Elizabeth C Bullen
- Department of Cell Biology, University of Oklahoma Health Sciences Center, 940 Stanton L. Young Blvd, BMSB 553, Oklahoma City, OK, 73104, USA
| | - Tripti Gautam
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Stefano Tarantini
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Tamas Kiss
- Pediatric Center, Semmelweis University, Budapest, Hungary
- Eötvös Loránd Research Network and Semmelweis University Cerebrovascular and Neurocognitive Disorders Research Group, Budapest, Hungary
| | - Zoltan Ungvari
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Shannon M Conley
- Department of Cell Biology, University of Oklahoma Health Sciences Center, 940 Stanton L. Young Blvd, BMSB 553, Oklahoma City, OK, 73104, USA.
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
22
|
Banjac K, Obradovic M, Zafirovic S, Essack M, Gluvic Z, Sunderic M, Nedic O, Isenovic ER. The involvement of Akt, mTOR, and S6K in the in vivo effect of IGF-1 on the regulation of rat cardiac Na +/K +-ATPase. Mol Biol Rep 2024; 51:517. [PMID: 38622478 DOI: 10.1007/s11033-024-09451-3] [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: 12/01/2023] [Accepted: 03/15/2024] [Indexed: 04/17/2024]
Abstract
BACKGROUND We previously demonstrated that insulin-like growth factor-1 (IGF-1) regulates sodium/potassium adenosine triphosphatase (Na+/K+-ATPase) in vascular smooth muscle cells (VSMC) via phosphatidylinositol-3 kinase (PI3K). Taking into account that others' work show that IGF-1 activates the PI3K/protein kinase B (Akt) signaling pathway in many different cells, we here further questioned if the Akt/mammalian target of rapamycin (mTOR)/ribosomal protein p70 S6 kinase (S6K) pathway stimulates Na+/K+-ATPase, an essential protein for maintaining normal heart function. METHODS AND RESULTS There were 14 adult male Wistar rats, half of whom received bolus injections of IGF-1 (50 μg/kg) for 24 h. We evaluated cardiac Na+/K+-ATPase expression, activity, and serum IGF-1 levels. Additionally, we examined the phosphorylated forms of the following proteins: insulin receptor substrate (IRS), phosphoinositide-dependent kinase-1 (PDK-1), Akt, mTOR, S6K, and α subunit of Na+/K+-ATPase. Additionally, the mRNA expression of the Na+/K+-ATPase α1 subunit was evaluated. Treatment with IGF-1 increases levels of serum IGF-1 and stimulates Na+/K+-ATPase activity, phosphorylation of α subunit of Na+/K+-ATPase on Ser23, and protein expression of α2 subunit. Furthermore, IGF-1 treatment increased phosphorylation of IRS-1 on Tyr1222, Akt on Ser473, PDK-1 on Ser241, mTOR on Ser2481 and Ser2448, and S6K on Thr421/Ser424. The concentration of IGF-1 in serum positively correlates with Na+/K+-ATPase activity and the phosphorylated form of mTOR (Ser2448), while Na+/K+-ATPase activity positively correlates with the phosphorylated form of IRS-1 (Tyr1222) and mTOR (Ser2448). CONCLUSION These results indicate that the Akt/mTOR/S6K signalling pathway may be involved in the IGF-1 regulating cardiac Na+/K+-ATPase expression and activity.
Collapse
Affiliation(s)
- Katarina Banjac
- Department of Radiobiology and Molecular Genetics, "VINCA" Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, P.O.Box 522, Belgrade, 11000, Serbia
| | - Milan Obradovic
- Department of Radiobiology and Molecular Genetics, "VINCA" Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, P.O.Box 522, Belgrade, 11000, Serbia.
| | - Sonja Zafirovic
- Department of Radiobiology and Molecular Genetics, "VINCA" Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, P.O.Box 522, Belgrade, 11000, Serbia
| | - Magbubah Essack
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Zoran Gluvic
- Clinic of Internal Medicine, School of Medicine, University Clinical-Hospital Centre Zemun-Belgrade, University of Belgrade, Vukova 9, Belgrade, 11080, Serbia
| | - Milos Sunderic
- Institute for the Application of Nuclear Energy, Department for Metabolism, University of Belgrade, Banatska 31b, Belgrade, Serbia
| | - Olgica Nedic
- Institute for the Application of Nuclear Energy, Department for Metabolism, University of Belgrade, Banatska 31b, Belgrade, Serbia
| | - Esma R Isenovic
- Department of Radiobiology and Molecular Genetics, "VINCA" Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, P.O.Box 522, Belgrade, 11000, Serbia
| |
Collapse
|
23
|
Szydlowska-Gladysz J, Gorecka AE, Stepien J, Rysz I, Ben-Skowronek I. IGF-1 and IGF-2 as Molecules Linked to Causes and Consequences of Obesity from Fetal Life to Adulthood: A Systematic Review. Int J Mol Sci 2024; 25:3966. [PMID: 38612776 PMCID: PMC11012406 DOI: 10.3390/ijms25073966] [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: 02/20/2024] [Revised: 03/28/2024] [Accepted: 03/29/2024] [Indexed: 04/14/2024] Open
Abstract
This study examines the impact of insulin-like growth factor 1 (IGF-1) and insulin-like growth factor 2 (IGF-2) on various aspects of children's health-from the realms of growth and puberty to the nuanced characteristics of metabolic syndrome, diabetes, liver pathology, carcinogenic potential, and cardiovascular disorders. A comprehensive literature review was conducted using PubMed, with a Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) method employing specific keywords related to child health, obesity, and insulin-like growth factors. This study reveals associations between insulin-like growth factor 1 and birth weight, early growth, and adiposity. Moreover, insulin-like growth factors play a pivotal role in regulating bone development and height during childhood, with potential implications for puberty onset. This research uncovers insulin-like growth factor 1 and insulin-like growth factor 2 as potential biomarkers and therapeutic targets for metabolic dysfunction-associated liver disease and hepatocellular carcinoma, and it also highlights the association between insulin-like growth factors (IGFs) and cancer. Additionally, this research explores the impact of insulin-like growth factors on cardiovascular health, noting their role in cardiomyocyte hypertrophy. Insulin-like growth factors play vital roles in human physiology, influencing growth and development from fetal stages to adulthood. The impact of maternal obesity on children's IGF levels is complex, influencing growth and carrying potential metabolic consequences. Imbalances in IGF levels are linked to a range of health conditions (e.g., insulin resistance, glucose intolerance, metabolic syndrome, and diabetes), prompting researchers to seek novel therapies and preventive strategies, offering challenges and opportunities in healthcare.
Collapse
Affiliation(s)
- Justyna Szydlowska-Gladysz
- Department of Pediatric Endocrinology and Diabetology with Endocrine-Metabolic Laboratory, Medical University in Lublin, 20-093 Lublin, Poland
| | | | | | | | - Iwona Ben-Skowronek
- Department of Pediatric Endocrinology and Diabetology with Endocrine-Metabolic Laboratory, Medical University in Lublin, 20-093 Lublin, Poland
| |
Collapse
|
24
|
Miller LR, Bickel MA, Tarantini S, Runion ME, Matacchiera Z, Vance ML, Hibbs C, Vaden H, Nagykaldi D, Martin T, Bullen EC, Pinckard J, Kiss T, Howard EW, Yabluchanskiy A, Conley SM. IGF1R deficiency in vascular smooth muscle cells impairs myogenic autoregulation and cognition in mice. Front Aging Neurosci 2024; 16:1320808. [PMID: 38425784 PMCID: PMC10902040 DOI: 10.3389/fnagi.2024.1320808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 01/22/2024] [Indexed: 03/02/2024] Open
Abstract
Introduction Cerebrovascular pathologies contribute to cognitive decline during aging, leading to vascular cognitive impairment and dementia (VCID). Levels of circulating insulin-like growth factor 1 (IGF-1), a vasoprotective hormone, decrease during aging. Decreased circulating IGF-1 in animal models leads to the development of VCID-like symptoms, but the cellular mechanisms underlying IGF-1-deficiency associated pathologies in the aged cerebrovasculature remain poorly understood. Here, we test the hypothesis that vascular smooth muscle cells (VSMCs) play an integral part in mediating the vasoprotective effects of IGF-1. Methods We used a hypertension-based model of cerebrovascular dysfunction in mice with VSMC-specific IGF-1 receptor (Igf1r) deficiency and evaluated the development of cerebrovascular pathologies and cognitive dysfunction. Results VSMC-specific Igf1r deficiency led to impaired cerebral myogenic autoregulation, independent of blood pressure changes, which was also associated with impaired spatial learning and memory function as measured by radial arm water maze and impaired motor learning measured by rotarod. In contrast, VSMC-specific IGF-1 receptor knockdown did not lead to cerebral microvascular rarefaction. Discussion These studies suggest that VSMCs are key targets for IGF-1 in the context of cerebrovascular health, playing a role in vessel stability alongside other cells in the neurovascular unit, and that VSMC dysfunction in aging likely contributes to VCID.
Collapse
Affiliation(s)
- Lauren R. Miller
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Marisa A. Bickel
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Stefano Tarantini
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Megan E. Runion
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Zoe Matacchiera
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Michaela L. Vance
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Clara Hibbs
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Hannah Vaden
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Domonkos Nagykaldi
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Teryn Martin
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Elizabeth C. Bullen
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Jessica Pinckard
- Division of Comparative Medicine, Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Tamas Kiss
- Pediatric Center, Semmelweis University, Budapest, Hungary
- Eötvös Loránd Research Network and Semmelweis University Cerebrovascular and Neurocognitive Disorders Research Group, Budapest, Hungary
| | - Eric W. Howard
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Shannon M. Conley
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| |
Collapse
|
25
|
Gulej R, Csik B, Faakye J, Tarantini S, Shanmugarama S, Chandragiri SS, Mukli P, Conley S, Csiszar A, Ungvari Z, Yabluchanskiy A, Nyúl-Tóth Á. Endothelial deficiency of insulin-like growth factor-1 receptor leads to blood-brain barrier disruption and accelerated endothelial senescence in mice, mimicking aspects of the brain aging phenotype. Microcirculation 2024; 31:e12840. [PMID: 38082450 PMCID: PMC10922445 DOI: 10.1111/micc.12840] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 11/07/2023] [Accepted: 11/22/2023] [Indexed: 01/11/2024]
Abstract
INTRODUCTION Age-related blood-brain barrier (BBB) disruption, cerebromicrovascular senescence, and microvascular rarefaction substantially contribute to the pathogenesis of vascular cognitive impairment (VCI) and Alzheimer's disease (AD). Previous studies established a causal link between age-related decline in circulating levels of insulin-like growth factor-1 (IGF-1), cerebromicrovascular dysfunction, and cognitive decline. The aim of our study was to determine the effect of IGF-1 signaling on senescence, BBB permeability, and vascular density in middle-age and old brains. METHODS Accelerated endothelial senescence was assessed in senescence reporter mice (VE-Cadherin-CreERT2 /Igf1rfl/fl × p16-3MR) using flow cytometry. To determine the functional consequences of impaired IGF-1 input to cerebromicrovascular endothelial cells, BBB integrity and capillary density were studied in mice with endothelium-specific knockout of IGF1R (VE-Cadherin-CreERT2 /Igf1rfl/fl ) using intravital two-photon microscopy. RESULTS In VE-Cadherin-CreERT2 /Igf1rfl/fl mice: (1) there was an increased presence of senescent endothelial cells; (2) cumulative permeability of the microvessels to fluorescent tracers of different molecular weights (0.3-40 kDa) is significantly increased, as compared to that of control mice, whereas decline in cortical capillary density does not reach statistical significance. CONCLUSIONS These findings support the notion that IGF-1 signaling plays a crucial role in preserving a youthful cerebromicrovascular endothelial phenotype and maintaining the integrity of the BBB.
Collapse
Affiliation(s)
- Rafal Gulej
- Vascular Cognitive Impairment, Neurodegeneration, and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Boglarka Csik
- Vascular Cognitive Impairment, Neurodegeneration, and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Janet Faakye
- Vascular Cognitive Impairment, Neurodegeneration, and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Stefano Tarantini
- Vascular Cognitive Impairment, Neurodegeneration, and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
| | - Santny Shanmugarama
- Vascular Cognitive Impairment, Neurodegeneration, and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Siva Sai Chandragiri
- Vascular Cognitive Impairment, Neurodegeneration, and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Peter Mukli
- Vascular Cognitive Impairment, Neurodegeneration, and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Shannon Conley
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anna Csiszar
- Vascular Cognitive Impairment, Neurodegeneration, and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Zoltan Ungvari
- Vascular Cognitive Impairment, Neurodegeneration, and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment, Neurodegeneration, and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Ádám Nyúl-Tóth
- Vascular Cognitive Impairment, Neurodegeneration, and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| |
Collapse
|
26
|
Herman R, Janez A, Mikhailidis DP, Poredos P, Blinc A, Sabovic M, Studen KB, Schernthaner GH, Anagnostis P, Antignani PL, Jensterle M. Growth Hormone, Atherosclerosis and Peripheral Arterial Disease: Exploring the Spectrum from Acromegaly to Growth Hormone Deficiency. Curr Vasc Pharmacol 2024; 22:28-35. [PMID: 37962050 DOI: 10.2174/0115701611269162231106042956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 09/19/2023] [Accepted: 10/03/2023] [Indexed: 11/15/2023]
Abstract
Growth hormone (GH) and insulin-like growth factor 1 (IGF-1) are increasingly recognised for their role in cardiovascular (CV) physiology. The GH-IGF-1 axis plays an essential role in the development of the CV system as well as in the complex molecular network that regulates cardiac and endothelial structure and function. A considerable correlation between GH levels and CV mortality exists even among individuals in the general population without a notable deviation in the GHIGF- 1 axis functioning. In addition, over the last decades, evidence has demonstrated that pathologic conditions involving the GH-IGF-1 axis, as seen in GH excess to GH deficiency, are associated with an increased risk for CV morbidity and mortality. A significant part of that risk can be attributed to several accompanying comorbidities. In both conditions, disease control is associated with a consistent improvement of CV risk factors, reduction of CV mortality, and achievement of standardised mortality ratio similar to that of the general population. Data on the prevalence of peripheral arterial disease in patients with acromegaly or growth hormone deficiency and the effects of GH and IGF-1 levels on the disease progression is limited. In this review, we will consider the pivotal role of the GH-IGF-1 axis on CV system function, as well as the far-reaching consequences that arise when disorders within this axis occur, particularly in relation to the atherosclerosis process.
Collapse
Affiliation(s)
- R Herman
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Department of Internal Medicine, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - A Janez
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Department of Internal Medicine, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - D P Mikhailidis
- Department of Surgical Biotechnology, Division of Surgery and Interventional Science, University College London Medical School, University College London (UCL) and Department of Clinical Biochemistry, Royal Free Hospital Campus (UCL), London, UK
| | - P Poredos
- Department of Vascular Diseases, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - A Blinc
- Department of Internal Medicine, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Department of Vascular Diseases, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - M Sabovic
- Department of Internal Medicine, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Department of Vascular Diseases, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - K Bajuk Studen
- Department of Internal Medicine, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Department of Nuclear Medicine, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - G H Schernthaner
- Department of Medicine 2, Division of Angiology, Medical University of Vienna, Vienna, Austria
| | - P Anagnostis
- Unit of Reproductive Endocrinology, 1st Department of Obstetrics and Gynaecology, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | - M Jensterle
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Department of Internal Medicine, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
27
|
Wu Z. Compression Promotes the Osteogenic Differentiation of Human Periodontal Ligament Stem Cells by Regulating METTL14-mediated IGF1. Curr Stem Cell Res Ther 2024; 19:1120-1128. [PMID: 38279741 DOI: 10.2174/011574888x244047231012103752] [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: 01/30/2023] [Revised: 07/17/2023] [Accepted: 07/26/2023] [Indexed: 01/28/2024]
Abstract
BACKGROUND AND OBJECTIVES Orthodontic treatment involves the application of mechanical force to induce periodontal tissue remodeling and ultimately promote tooth movement. It is essential to study the response mechanisms of human periodontal ligament stem cells (hPDLSCs) to improve orthodontic treatment. METHODS In this study, hPDLSCs treated with compressive force were used to simulate orthodontic treatment. Cell viability and cell death were assessed using the CCK-8 assay and TUNEL staining. Alkaline phosphatase (ALP) and alizarin red staining were performed to evaluate osteogenic differentiation. The binding relationship between IGF1 and METTL14 was assessed using RIP and dual-luciferase reporter assays. RESULTS The compressive force treatment promoted the viability and osteogenic differentiation of hPDLSCs. Additionally, m6A and METTL14 levels in hPDLSCs increased after compressive force treatment, whereas METTL14 knockdown decreased cell viability and inhibited the osteogenic differentiation of hPDLSCs treated with compressive force. Furthermore, the upregulation of METTL14 increased m6A levels, mRNA stability, and IGF1 expression. RIP and dual-luciferase reporter assays confirmed the interaction between METTL14 and IGF1. Furthermore, rescue experiments demonstrated that IGF1 overexpression reversed the effects of METTL14 knockdown in hPDLSCs treated with compressive force. CONCLUSIONS In conclusion, this study demonstrated that compressive force promotes cell viability and osteogenic differentiation of hPDLSCs by regulating IGF1 levels mediated by METTL14.
Collapse
Affiliation(s)
- Zengbo Wu
- North Sichuan Medical College, Xinglin Community, Sihai Street, Shunqing District, Nanchong, Sichuan, 637000, China
| |
Collapse
|
28
|
Ren Y, Ruan P, Segal M, Dobre M, Schelling JR, Banerjee U, Shafi T, Ganz P, Dubin RF. Evaluation of a large-scale aptamer proteomics platform among patients with kidney failure on dialysis. PLoS One 2023; 18:e0293945. [PMID: 38079395 PMCID: PMC10712847 DOI: 10.1371/journal.pone.0293945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 10/22/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Patients with kidney failure suffer high mortality, and we currently lack markers for risk stratification for these patients. We carried out a quality control study of a modified aptamer assay (SomaScan v.4.0) that measures ~ 5000 proteins, in preparation for a larger study using this platform in cohorts with kidney failure. METHODS Forty participants from the Cardiac, Endothelial Function and Arterial Stiffness in End-Stage Renal Disease (CERES study) were selected to analyze technical and short-term biological variability, orthogonal correlations and differential protein expression in plasma from patients who died during 2.5 year follow-up. Long-term (one year) variability was studied in 421 participants in the Chronic Renal Insufficiency Cohort. We evaluated 4849 aptamers (4607 unique proteins) using data formats including raw data and data formatted using Adaptive Normalization by Maximum Likelihood (ANML), an algorithm developed for SomaScan data in individuals with normal kidney function. RESULTS In ANML format, median[IQR] intra-assay coefficient of variation (CV) was 2.38%[1.76, 3.40] and inter-assay CV was 7.38%[4.61, 13.12]. Short-term within-subject CV was 5.76% [3.35, 9.72]; long-term CV was 8.71%[5.91, 13.37]. Spearman correlations between aptamer and traditional assays for PTH, NT-proBNP, FGF-23 and CRP were all > 0.7. Fold-change (FC) in protein levels among non-survivors, significant after Bonferroni correction, included SVEP1 (FC[95% CI] 2.14 [1.62, 2.82]), keratocan (1.74 [1.40, 2.15]) and LanC-like protein 1 (0.56 [0.45, 0.70]). Compared to raw aptamer data, technical and short-term biological variability in paired samples was lower in ANML-formatted data. ANML formatting had minimal impact on orthogonal correlations with traditional assays or the associations of proteins with the phenotype of mortality. CONCLUSIONS SomaScan had excellent technical variability and low within-subject short-term variability. ANML formatting could facilitate comparison of biomarker results with other studies that utilize this format. We expect SomaScan to provide novel and reproducible information in patients with kidney failure on dialysis.
Collapse
Affiliation(s)
- Yue Ren
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Peifeng Ruan
- Peter O’Donnell Jr School of Public Health, UT Southwestern, Dallas, Texas, United States of America
| | - Mark Segal
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California, United States of America
| | - Mirela Dobre
- Division of Nephrology and Hypertension, University Hospitals Cleveland Medical Center, Cleveland, Ohio, United States of America
| | - Jeffrey R. Schelling
- Department of Physiology & Biophysics, Case Western Reserve University of School of Medicine, Cleveland, Ohio, United States of America
| | - Upasana Banerjee
- Department of Internal Medicine, Hurley Medical Center/Michigan State University, Flint, Michigan, United States of America
| | - Tariq Shafi
- Division of Kidney Diseases, Hypertension and Transplantation, Houston Methodist Hospital, Houston, Texas, United States of America
| | - Peter Ganz
- Division of Cardiology, University of California, San Francisco, San Francisco, California, United States of America
| | - Ruth F. Dubin
- Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | | |
Collapse
|
29
|
Adam LN, Al-Habib OAM, Shekha MS. Exploring the role of Sirtuin 3 gene polymorphisms and oxidative stress markers in the susceptibility to coronary artery disease. Mol Biol Rep 2023; 50:9221-9228. [PMID: 37801276 DOI: 10.1007/s11033-023-08825-3] [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: 07/06/2023] [Accepted: 09/12/2023] [Indexed: 10/07/2023]
Abstract
OBJECTIVE Coronary artery disease (CAD) is a complex disorder influenced by genetic and environmental factors. This case-control study investigated the association between Sirtuin SIRT3 gene polymorphisms, serum malondialdehyde (MDA) levels, and CAD susceptibility. METHODS Blood samples were collected from 70 CAD cases and 30 controls at the Cardiac Center, Azadi Teaching Hospital, Duhok, Iraq. Genomic DNA was extracted, and PCR-based allele genotyping determined SIRT3 rs11246029 T/C polymorphisms. Serum MDA levels were measured using ELISA. Statistical analysis included t-tests, Mann-Whitney tests, and Spearman correlations. Odds ratios (OR) with 95% confidence intervals (CI) assessed genotypes/alleles and CAD associations. The accuracy of serum MDA in predicting the severity of CAD was evaluated using receiver operating characteristic (ROC) curve analysis. RESULTS There were no significant variations in serum MDA levels between controls and CAD patients in the study. The diagnostic accuracy of serum MDA for CAD severity prediction was modest (Area Under Curve (AUC) = 0.56). Correlations revealed associations between MDA and total bilirubin (negative) and Troponin (positive). CRP correlated positively with LDH, glucose, cholesterol, LDL, CKmB, and Troponin. CKmB and Troponin are positively associated with clinical characteristics. Genotype analysis identified a significantly higher CAD risk with the CC genotype compared to controls. CONCLUSION These findings shed light on the potential role of SIRT3 gene polymorphisms and serum MDA levels in CAD susceptibility. Further research is needed to understand underlying mechanisms and therapeutic implications based on these markers. TRIAL REGISTRATION 15092021-9-12. Registered 15 September 2021.
Collapse
Affiliation(s)
- Lina N Adam
- Department of Biology, Faculty of Science, University of Zakho, Duhok, Kurdistan Region, Iraq.
| | - Omar A M Al-Habib
- Department of Biology, College of Science, University of Nawroz, Duhok, Kurdistan Region, Iraq
| | - Mudhir S Shekha
- Department of medical cell biology, Upsala University, Upsala, Sweden
- Department of Biology, College of Science, Salahaddin University -Erbil, Erbil, Kurdistan Region, Iraq
| |
Collapse
|
30
|
Backeljauw PF, Andrews M, Bang P, Dalle Molle L, Deal CL, Harvey J, Langham S, Petriczko E, Polak M, Storr HL, Dattani MT. Challenges in the care of individuals with severe primary insulin-like growth factor-I deficiency (SPIGFD): an international, multi-stakeholder perspective. Orphanet J Rare Dis 2023; 18:312. [PMID: 37805563 PMCID: PMC10559630 DOI: 10.1186/s13023-023-02928-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 09/24/2023] [Indexed: 10/09/2023] Open
Abstract
BACKGROUND Severe primary insulin-like growth factor-I (IGF-I) deficiency (SPIGFD) is a rare growth disorder characterized by short stature (standard deviation score [SDS] ≤ 3.0), low circulating concentrations of IGF-I (SDS ≤ 3.0), and normal or elevated concentrations of growth hormone (GH). Laron syndrome is the best characterized form of SPIGFD, caused by a defect in the GH receptor (GHR) gene. However, awareness of SPIGFD remains low, and individuals living with SPIGFD continue to face challenges associated with diagnosis, treatment and care. OBJECTIVE To gather perspectives on the key challenges for individuals and families living with SPIGFD through a multi-stakeholder approach. By highlighting critical gaps in the awareness, diagnosis, and management of SPIGFD, this report aims to provide recommendations to improve care for people affected by SPIGFD globally. METHODS An international group of clinical experts, researchers, and patient and caregiver representatives from the SPIGFD community participated in a virtual, half-day meeting to discuss key unmet needs and opportunities to improve the care of people living with SPIGFD. RESULTS As a rare disorder, limited awareness and understanding of SPIGFD amongst healthcare professionals (HCPs) poses significant challenges in the diagnosis and treatment of those affected. Patients often face difficulties associated with receiving a formal diagnosis, delayed treatment initiation and limited access to appropriate therapy. This has a considerable impact on the physical health and quality of life for patients, highlighting a need for more education and clearer guidance for HCPs. Support from patient advocacy groups is valuable in helping patients and their families to find appropriate care. However, there remains a need to better understand the burden that SPIGFD has on individuals beyond height, including the impact on physical, emotional, and social wellbeing. CONCLUSIONS To address the challenges faced by individuals and families affected by SPIGFD, greater awareness of SPIGFD is needed within the healthcare community, and a consensus on best practice in the care of individuals affected by this condition. Continued efforts are also needed at a global level to challenge existing perceptions around SPIGFD, and identify solutions that promote equitable access to appropriate care. Medical writing support was industry-sponsored.
Collapse
Affiliation(s)
- Philippe F Backeljauw
- Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| | - Mary Andrews
- The Major Aspects of Growth in Children (MAGIC) Foundation, Warrenville, IL, USA
- The MAGIC Foundation International Coalition for Organizations Supporting Endocrine Patients (MAGIC-ICOSEP), Atlanta, GA, USA
| | - Peter Bang
- Division of Children's and Women's Health, Department of Biomedical and Clinical Sciences (BKV), Faculty of Health Sciences, Linköping University, Linköping, Sweden
| | | | - Cheri L Deal
- Université de Montréal, Montréal, QC, Canada
- Centre Hospitalier Universitaire (CHU) Sainte-Justine, Montréal, QC, Canada
| | - Jamie Harvey
- The Major Aspects of Growth in Children (MAGIC) Foundation, Warrenville, IL, USA
- The MAGIC Foundation International Coalition for Organizations Supporting Endocrine Patients (MAGIC-ICOSEP), Atlanta, GA, USA
| | - Shirley Langham
- Paediatric Endocrinology, Great Ormond Street Hospital UCL Hospitals, London, UK
| | - Elżbieta Petriczko
- Department of Paediatrics, Endocrinology, Diabetology, Metabolic Disorders, and Cardiology of Developmental Age, Pomeranian Medical University, Szczecin, Poland
| | - Michel Polak
- Department of Pediatric Endocrinology, Gynecology and Diabetology, Hôpital Universitaire Necker Enfants Malades, Assistance Publique Hôpitaux de Paris, Université Paris Cité, Paris, France
| | - Helen L Storr
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Mehul T Dattani
- Paediatric Endocrinology, Great Ormond Street Hospital UCL Hospitals, London, UK.
- UCL Great Ormond Street Institute of Child Health, London, UK.
- Adolescent Endocrinology, UCL Hospitals, London, UK.
| |
Collapse
|
31
|
Zare S, Hasani M, Estêvão MD, Tahmasebi R, Azadbakht L, Shidfar F, Heshmati J, Ziaei S. Muscle Strength and Biochemical Markers as Predictors of Depression in Hemodialysis Patients: A Cross-Sectional Study. Clin Nutr Res 2023; 12:293-303. [PMID: 37969939 PMCID: PMC10641328 DOI: 10.7762/cnr.2023.12.4.293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 09/29/2023] [Accepted: 10/06/2023] [Indexed: 11/17/2023] Open
Abstract
Patients with chronic renal failure, many of which treated with hemodialysis, present a high prevalence of impaired muscle strength which suggest that muscle mass parameters may be used as markers for changes in muscle in these patients. Measurement of handgrip strength (HGS) is a common, simple, and quick measure of muscle function an indicator of overall muscle strength which has been associated with physical activity and several anthropometric traits. Intercellular adhesion molecule-1 (ICAM-1) and insulin-like growth factor-1 (IGF-1) are biochemical markers associated with inflammatory processes which are a common consequence of dialysis. Additionally, hemodialysis patients frequently present signs of malnutrition and depression. This cross-sectional study aimed to evaluate if muscle and biochemical markers could be used to predict the risk of depression in hemodialysis patients. Several anthropometric parameters, nutrient intake, depression state and the serum levels of ICAM-1 and IGF-1 were determined and Pearson's correlation coefficient and/or Spearman's correlation coefficient were used to test the correlation between them. Our results do not show a correlation between HGF, IGF-1 and ICAM-1 with the depression status of the patients, but mid-arm muscle circumference (MAMC) was statistically and positively correlated with depression. Additionally, ICAM-1 levels were negatively correlated with HGS, MAMC, and IGF-1. Overall, the results of the present study suggest that HGS may be used as an indicator of cardiovascular diseases and MAMC may be a good predictor of the level of depression in hemodialysis patients, although further studies are required.
Collapse
Affiliation(s)
- Soudabeh Zare
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Motahareh Hasani
- Department of Nutritional Sciences, School of Health, Golestan University of Medical Sciences, Gorgan 49341-74515, Iran
| | - M. Dulce Estêvão
- Universidade do Algarve, Escola Superior de Saúde, Campus de Gambelas, Faro 8005-139, Portugal
| | - Rahim Tahmasebi
- Department of Epidemiology & Biostatistics, School of Health, Bushehr University of Medical Sciences, Bushehr 7514633341, Iran
| | - Leila Azadbakht
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran 141556117, Iran
| | - Farzad Shidfar
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Javad Heshmati
- ICU Department, Imam Reza Hospital, Kermanshah University of Medical Sciences, Kermanshah 6714415333, Iran
| | - Somayeh Ziaei
- ICU Department, Imam Reza Hospital, Kermanshah University of Medical Sciences, Kermanshah 6714415333, Iran
| |
Collapse
|
32
|
Xiong Y, Wang Y, Yang T, Luo Y, Xu S, Li L. Receptor Tyrosine Kinase: Still an Interesting Target to Inhibit the Proliferation of Vascular Smooth Muscle Cells. Am J Cardiovasc Drugs 2023; 23:497-518. [PMID: 37524956 DOI: 10.1007/s40256-023-00596-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/05/2023] [Indexed: 08/02/2023]
Abstract
Vascular smooth muscle cells (VSMCs) proliferation is a critical event that contributes to the pathogenesis of vascular remodeling such as hypertension, restenosis, and pulmonary hypertension. Increasing evidences have revealed that VSMCs proliferation is associated with the activation of receptor tyrosine kinases (RTKs) by their ligands, including the insulin-like growth factor receptor (IGFR), fibroblast growth factor receptor (FGFR), epidermal growth factor receptor (EGFR), vascular endothelial growth factor receptor (VEGFR), and platelet-derived growth factor receptor (PDGFR). Moreover, some receptor tyrosinase inhibitors (TKIs) have been found and can prevent VSMCs proliferation to attenuate vascular remodeling. Therefore, this review will describe recent research progress on the role of RTKs and their inhibitors in controlling VSMCs proliferation, which helps to better understand the function of VSMCs proliferation in cardiovascular events and is beneficial for the prevention and treatment of vascular disease.
Collapse
Affiliation(s)
- Yilin Xiong
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563000, China
- Department of Pharmacology, Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, No. 6 Xuefu West Road, Zunyi, 563000, Guizhou, China
| | - Yan Wang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563000, China
- Department of Pharmacology, Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, No. 6 Xuefu West Road, Zunyi, 563000, Guizhou, China
| | - Tao Yang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563000, China
- Department of Pharmacology, Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, No. 6 Xuefu West Road, Zunyi, 563000, Guizhou, China
| | - Yunmei Luo
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563000, China
- Department of Pharmacology, Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, No. 6 Xuefu West Road, Zunyi, 563000, Guizhou, China
| | - Shangfu Xu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563000, China
- Department of Pharmacology, Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, No. 6 Xuefu West Road, Zunyi, 563000, Guizhou, China
| | - Lisheng Li
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563000, China.
- Department of Pharmacology, Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, No. 6 Xuefu West Road, Zunyi, 563000, Guizhou, China.
| |
Collapse
|
33
|
Salisbury DL, Li D, Todd M, Ng TKS, Yu F. Aerobic Exercise, Training Dose, and Cardiorespiratory Fitness: Effects and Relationships with Resting Plasma Neurotrophic Factors in Alzheimer's Dementia. JOURNAL OF VASCULAR DISEASES 2023; 2:351-366. [PMID: 39328309 PMCID: PMC11426414 DOI: 10.3390/jvd2030027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/28/2024]
Abstract
Background Vascular health is increasingly recognized for its roles in the pathogenesis and progression of Alzheimer's disease (AD). The objective of this study was to investigate effects of exercise training, dose, and cardiorespiratory fitness (CRF) on neurotrophic factors in community-dwelling, older adults with mild-to-moderate AD dementia. Methods This was a pilot blood ancillary study of the FIT-AD trial. Participants in the parent study were randomized to 6-month aerobic exercise (AEx) or stretching control. For this ancillary study, resting plasma brain-derived neurotrophic factor (BDNF), irisin, fibroblast growth factor-21 (FGF-21), and insulin-like growth factor-1 (IGF-1) biomarkers were assessed at baseline, 3, and 6 months. Estimates of within- and between-group effect sizes were calculated (Cohen's d). Relationships of biomarker change with dose and CRF change were explored with multivariable linear regression and repeated measures correlations. Results The sample (n = 26, 18 AEx/8 stretching) averaged 77.6 ± 6.9 years old, with the majority being male (65.4%), and non-Hispanic White (92.3%); between-group effect sizes were generally small except for irisin (d = -0.44)), AEx group relative to stretching group. Associations of dose and changes in CRF with changes in neurotrophic biomarker were weak (r2 ≤ 0.025). Conclusions The effects of exercise on BDNF, irisin, IGF-1, and FGF-21 were heterogeneous in AD. Our findings need validation in future, adequately powered exercise studies in AD.
Collapse
Affiliation(s)
| | - Danni Li
- School of Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| | - Michael Todd
- Edison College of Nursing and Health Innovation, Arizona State University, Tempe, AZ 85281, USA
| | - Ted K. S. Ng
- Department of Internal Medicine & Rush Institute of Healthy Aging, Rush University Medical Center, Chicago, IL 60612, USA
| | - Fang Yu
- Edison College of Nursing and Health Innovation, Arizona State University, Tempe, AZ 85281, USA
| |
Collapse
|
34
|
Lin J, Yang L, Huang J, Liu Y, Lei X, Chen R, Xu B, Huang C, Dou W, Wei X, Liu D, Zhang P, Huang Y, Ma Z, Zhang H. Insulin-Like Growth Factor 1 and Risk of Cardiovascular Disease: Results From the UK Biobank Cohort Study. J Clin Endocrinol Metab 2023; 108:e850-e860. [PMID: 36810801 DOI: 10.1210/clinem/dgad105] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 02/10/2023] [Accepted: 02/16/2023] [Indexed: 02/24/2023]
Abstract
CONTEXT Relationships between insulin-like growth factor 1 (IGF-1) levels and cardiovascular disease (CVD) in the general population remain unclear. OBJECTIVE This study aims to investigate the association of circulating IGF-1 concentrations with CVD from a population-based cohort study. METHODS A total of 394 082 participants without CVD and cancer at baseline from UK Biobank were included with measurements of serum IGF-1 at baseline. Main outcomes were incidence of CVD, including CVD mortality, coronary heart disease (CHD), myocardial infarction (MI), heart failure (HF), and stroke. RESULTS Over a median 11.6 years of follow-up, UK Biobank documented 35 803 incident CVD cases, including 4231 from CVD-related death, 27 051 from CHD, 10 014 from MI, 7661 from HF, and 6802 from stroke. Dose-response analysis showed a U-shaped relationship between IGF-1 levels and cardiovascular events. Compared with the third quintile of IGF-1, the lowest category of IGF-1 was associated with increased risk of CVD (hazard ratio 1.128; 95% CI, 1.093 to 1.164), CVD mortality (1.294; 1.181 to 1.418), CHD (1.118; 1.078 to 1.159), MI (1.071; 1.008 to 1.139), HF (1.185; 1.107 to 1.268), and stroke (1.149, 1.070 to 1.235); also, the highest category was associated with increased risk of CVD (1.056; 1.020 to 1.094), CVD mortality (1.111; 1.000 to 1.236), CHD (1.070; 1.028 to 1.114), MI (1.111; 1.041 to 1.187) and HF (1.098; 1.015 to 1.188) after multivariable adjustment. CONCLUSION This study indicates that both low and high levels of circulating IGF-1 are associated with increased risk of CVD in general population. These results highlight the importance of monitoring IGF-1 status on cardiovascular health.
Collapse
Affiliation(s)
- Jiayang Lin
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Linjie Yang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Junlin Huang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yating Liu
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xuzhen Lei
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Ruxin Chen
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Bingyan Xu
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Chensihan Huang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Weijuan Dou
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xueyun Wei
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Deying Liu
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Peizhen Zhang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yan Huang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Zhimin Ma
- Department of Endocrinology, The Affiliated Suzhou Science & Technology Town Hospital, Nanjing Medical University, Jiangsu 215153, China
| | - Huijie Zhang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
35
|
Wu TT, Zheng YY, Ma X, Xiu WJ, Yang HT, Hou XG, Yang Y, Chen Y, Ma YT, Xie X. Mutated CYP17A1 promotes atherosclerosis and early-onset coronary artery disease. Cell Commun Signal 2023; 21:155. [PMID: 37370070 PMCID: PMC10294473 DOI: 10.1186/s12964-023-01061-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 01/29/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND Coronary artery disease (CAD) is a multi-factor complex trait and is heritable, especially in early-onset families. However, the genetic factors affecting the susceptibility of early-onset CAD are not fully characterized. METHODS In the present study, we identified a rare nonsense variant in the CYP17A1 gene from a Chinese Han family with CAD. To validate the effect of this variation on atherosclerosis and early-onset coronary artery disease, we conducted studies on population, cells, and mice. RESULTS The mutation precisely congregated with the clinical syndrome in all the affected family members and was absent in unaffected family members and unrelated controls. Similar to the human phenotype, the CYP17A1-deficient mice present the phenotype of metabolic syndrome with hypertension, increased serum glucose concentration, and presentation of central obesity and fatty liver. Furthermore, CYP17A1 knockout mice or CYP17A1 + ApoE double knockout mice developed more atherosclerotic lesions than wild type (WT) with high fat diary. In cell models, CYP17A1 was found to be involved in glucose metabolism by increasing glucose intake and utilization, through activating IGF1/mTOR/HIF1-α signaling way, which was consistent in CYP17A1 knockout mice with impaired glucose tolerance and insulin resistance. CONCLUSIONS Through our study of cells, mice and humans, we identified CYP17A1 as a key protein participating in the pathophysiology of the atherosclerotic process and the possible mechanism of CYP17A1 C987X mutation induced atherosclerosis and early-onset CAD involving glucose homeostasis regulation was revealed. Video Abstract.
Collapse
Affiliation(s)
- Ting-Ting Wu
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, No. 137, Liyushan Road, Urumqi, 830011, People's Republic of China
| | - Ying-Ying Zheng
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, No. 137, Liyushan Road, Urumqi, 830011, People's Republic of China
| | - Xiang Ma
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, No. 137, Liyushan Road, Urumqi, 830011, People's Republic of China
| | - Wen-Juan Xiu
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, No. 137, Liyushan Road, Urumqi, 830011, People's Republic of China
| | - Hai-Tao Yang
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, No. 137, Liyushan Road, Urumqi, 830011, People's Republic of China
| | - Xian-Geng Hou
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, No. 137, Liyushan Road, Urumqi, 830011, People's Republic of China
| | - Yi Yang
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, No. 137, Liyushan Road, Urumqi, 830011, People's Republic of China
| | - You Chen
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, No. 137, Liyushan Road, Urumqi, 830011, People's Republic of China
| | - Yi-Tong Ma
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, No. 137, Liyushan Road, Urumqi, 830011, People's Republic of China.
| | - Xiang Xie
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, No. 137, Liyushan Road, Urumqi, 830011, People's Republic of China.
| |
Collapse
|
36
|
Deo R, Dubin RF, Ren Y, Murthy AC, Wang J, Zheng H, Zheng Z, Feldman H, Shou H, Coresh J, Grams M, Surapaneni AL, Bhat Z, Cohen JB, Rahman M, He J, Saraf SL, Go AS, Kimmel PL, Vasan RS, Segal MR, Li H, Ganz P. Proteomic cardiovascular risk assessment in chronic kidney disease. Eur Heart J 2023; 44:2095-2110. [PMID: 37014015 PMCID: PMC10281556 DOI: 10.1093/eurheartj/ehad115] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 01/21/2023] [Accepted: 02/16/2023] [Indexed: 04/05/2023] Open
Abstract
AIMS Chronic kidney disease (CKD) is widely prevalent and independently increases cardiovascular risk. Cardiovascular risk prediction tools derived in the general population perform poorly in CKD. Through large-scale proteomics discovery, this study aimed to create more accurate cardiovascular risk models. METHODS AND RESULTS Elastic net regression was used to derive a proteomic risk model for incident cardiovascular risk in 2182 participants from the Chronic Renal Insufficiency Cohort. The model was then validated in 485 participants from the Atherosclerosis Risk in Communities cohort. All participants had CKD and no history of cardiovascular disease at study baseline when ∼5000 proteins were measured. The proteomic risk model, which consisted of 32 proteins, was superior to both the 2013 ACC/AHA Pooled Cohort Equation and a modified Pooled Cohort Equation that included estimated glomerular filtrate rate. The Chronic Renal Insufficiency Cohort internal validation set demonstrated annualized receiver operating characteristic area under the curve values from 1 to 10 years ranging between 0.84 and 0.89 for the protein and 0.70 and 0.73 for the clinical models. Similar findings were observed in the Atherosclerosis Risk in Communities validation cohort. For nearly half of the individual proteins independently associated with cardiovascular risk, Mendelian randomization suggested a causal link to cardiovascular events or risk factors. Pathway analyses revealed enrichment of proteins involved in immunologic function, vascular and neuronal development, and hepatic fibrosis. CONCLUSION In two sizeable populations with CKD, a proteomic risk model for incident cardiovascular disease surpassed clinical risk models recommended in clinical practice, even after including estimated glomerular filtration rate. New biological insights may prioritize the development of therapeutic strategies for cardiovascular risk reduction in the CKD population.
Collapse
Affiliation(s)
- Rajat Deo
- Division of Cardiovascular Medicine, Electrophysiology Section, Perelman School of Medicine at the University of Pennsylvania, One Convention Avenue, Level 2 / City Side, Philadelphia, PA 19104, USA
| | - Ruth F Dubin
- Division of Nephrology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Yue Ren
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, 215 Blockley Hall, 423 Guardian Drive, Philadelphia, PA 19104, USA
| | - Ashwin C Murthy
- Division of Cardiovascular Medicine, Electrophysiology Section, Perelman School of Medicine at the University of Pennsylvania, One Convention Avenue, Level 2 / City Side, Philadelphia, PA 19104, USA
| | - Jianqiao Wang
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, 215 Blockley Hall, 423 Guardian Drive, Philadelphia, PA 19104, USA
| | - Haotian Zheng
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, 215 Blockley Hall, 423 Guardian Drive, Philadelphia, PA 19104, USA
| | - Zihe Zheng
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, 215 Blockley Hall, 423 Guardian Drive, Philadelphia, PA 19104, USA
| | - Harold Feldman
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, 215 Blockley Hall, 423 Guardian Drive, Philadelphia, PA 19104, USA
| | - Haochang Shou
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, 215 Blockley Hall, 423 Guardian Drive, Philadelphia, PA 19104, USA
| | - Josef Coresh
- Department of Epidemiology; Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University Bloomberg School of Public Health, 615 N Wolfe St, Baltimore, MD 21205, USA
- Department of Medicine, Johns Hopkins University, 2024 E. Monument Street, Room 2-635, Suite 2-600, Baltimore, MD 21287, USA
| | - Morgan Grams
- Department of Epidemiology; Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University Bloomberg School of Public Health, 615 N Wolfe St, Baltimore, MD 21205, USA
- Department of Medicine, Johns Hopkins University, 2024 E. Monument Street, Room 2-635, Suite 2-600, Baltimore, MD 21287, USA
| | - Aditya L Surapaneni
- Department of Epidemiology; Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University Bloomberg School of Public Health, 615 N Wolfe St, Baltimore, MD 21205, USA
| | - Zeenat Bhat
- Division of Nephrology, University of Michigan, 5100 Brehm Tower, 1000 Wall Street, Ann Arbor, MI 48105, USA
| | - Jordana B Cohen
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, 215 Blockley Hall, 423 Guardian Drive, Philadelphia, PA 19104, USA
- Renal, Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, 831 Blockley Hall, 423 Guardian Drive, Philadelphia, PA 19104, USA
| | - Mahboob Rahman
- Department of Medicine, Case Western Reserve University School of Medicine, 11100 Euclid Avenue, Wearn Bldg. 3 Floor. Rm 352, Cleveland, OH 44106, USA
| | - Jiang He
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, 1440 Canal Street, SL 18, New Orleans, LA 70112, USA
| | - Santosh L Saraf
- Division of Hematology and Oncology, University of Illinois at Chicago, 1740 West Taylor Street, Chicago, IL 60612, USA
| | - Alan S Go
- Division of Research, Kaiser Permanente Northern California, 2000 Broadway, Oakland, CA 94612, USA
- Departments of Epidemiology, Biostatistics and Medicine, University of California at San Francisco, San Francisco, CA, USA
| | - Paul L Kimmel
- Division of Kidney, Urologic, and Hematologic Diseases, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Ramachandran S Vasan
- Section of Preventive Medicine and Epidemiology, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
- Section of Cardiology, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
| | - Mark R Segal
- Department of Epidemiology and Biostatistics, University of California, 550 16th Street, 2nd Floor, Box #0560, San Francisco, CA 94143, USA
| | - Hongzhe Li
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, 215 Blockley Hall, 423 Guardian Drive, Philadelphia, PA 19104, USA
| | - Peter Ganz
- Division of Cardiology, Zuckerberg San Francisco General Hospital and Department of Medicine, University of California, San Francisco, 1001 Potrero Avenue, 5G1, San Francisco, CA 94110, USA
| |
Collapse
|
37
|
Coskun M, Altinova AE, Babayeva A, Sel AT, Yapar D, Karaca M, Yalcin MM, Akturk M, Toruner FB, Karakoc MA, Yetkin I. Leukocyte Telomere Length and Neuregulin-4 Levels in Female Patients with Acromegaly: The Relationship between Disease Activity and Body Fat Distribution. J Clin Med 2023; 12:4108. [PMID: 37373801 DOI: 10.3390/jcm12124108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 06/06/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
The study aimed to examine leukocyte telomere length (LTL) and serum neuregulin-4 levels and their relationship with disease activity, co-morbidities and body fat distribution in female acromegaly patients. Forty female patients with acromegaly and thirty-nine age and body mass index (BMI) similar healthy female volunteers were included in the study. Patients were classified into two groups: active acromegaly (AA) and controlled acromegaly (CA). The quantitative polymerase chain reaction (PCR) method was used to study LTL, and T/S ratio < 1 was accepted as shortened telomere length. Neuregulin-4 was studied by ELISA. There was no difference in median LTL between acromegaly and the control group (p = 0.530). The percentage of T/S < 1 in patients with acromegaly (60.0%) was similar to that of the control group (43.6%) (p = 0.144). However, serum neuregulin-4 was significantly higher in patients with acromegaly than those in the control group (p = 0.037). There were no significant differences concerning LTL, percentage of T/S < 1 and neuregulin-4 levels between active and controlled acromegaly groups (p > 0.05). Neuregulin-4 correlated positively with fasting glucose, triglyceride (TG), triglyceride/glucose (TyG) index, and lean body mass in the acromegaly group. A negative correlation was observed between LTL and neuregulin-4 in the control group (p = 0.039). When the factors affecting neuregulin-4 were evaluated by multivariate linear regression analysis with an enter method, TG (β: 0.316, p = 0.025) was independently and positively associated with neuregulin-4. Our findings indicate that acromegaly is associated with unchanged LTL and high neuregulin-4 levels in female patients. However, the relationship between acromegaly, the aging process, and neuregulin-4 involves complex mechanisms, and further studies are needed.
Collapse
Affiliation(s)
- Meric Coskun
- Department of Endocrinology and Metabolism, Faculty of Medicine, Gazi University, Ankara 06100, Turkey
| | - Alev Eroglu Altinova
- Department of Endocrinology and Metabolism, Faculty of Medicine, Gazi University, Ankara 06100, Turkey
| | - Afruz Babayeva
- Department of Endocrinology and Metabolism, Faculty of Medicine, Gazi University, Ankara 06100, Turkey
| | - Aydin Tuncer Sel
- Department of Endocrinology and Metabolism, Faculty of Medicine, Gazi University, Ankara 06100, Turkey
| | - Dilek Yapar
- Department of Public Health, Faculty of Medicine, Gazi University, Ankara 06100, Turkey
| | - Mine Karaca
- Department of Internal Medicine, Faculty of Medicine, Gazi University, Ankara 06100, Turkey
| | - Mehmet Muhittin Yalcin
- Department of Endocrinology and Metabolism, Faculty of Medicine, Gazi University, Ankara 06100, Turkey
| | - Mujde Akturk
- Department of Endocrinology and Metabolism, Faculty of Medicine, Gazi University, Ankara 06100, Turkey
| | - Fusun Balos Toruner
- Department of Endocrinology and Metabolism, Faculty of Medicine, Gazi University, Ankara 06100, Turkey
| | - Mehmet Ayhan Karakoc
- Department of Endocrinology and Metabolism, Faculty of Medicine, Gazi University, Ankara 06100, Turkey
| | - Ilhan Yetkin
- Department of Endocrinology and Metabolism, Faculty of Medicine, Gazi University, Ankara 06100, Turkey
| |
Collapse
|
38
|
Costa D, Pellicano C, Mercuri V, Arnone JM, Rizzo F, Leodori G, Gargiulo P, Rosato E. In acromegalic patients the serum levels of interleukin-33 and Resolvin D1 influence skin perfusion of hands: A pilot study. Microvasc Res 2023; 149:104571. [PMID: 37321455 DOI: 10.1016/j.mvr.2023.104571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/12/2023] [Accepted: 06/12/2023] [Indexed: 06/17/2023]
Abstract
AIM Acromegaly is a rare chronic disease, caused by the over-secretion of growth hormone (GH), that creates a pro-inflammatory state, but the exact mechanisms by which GH or insulin-like growth factor 1 (IGF-I) act on inflammatory cells are not fully understood. Aim of the study was to evaluate Interleukin-33 (IL33) and D-series resolvins 1 (RvD1) and the skin perfusion of hands in patients with acromegaly (AP) and healthy controls (HC). METHODS IL33 and RvD1 have been assessed in 20 AP and 20 HC. Nailfold videocapillaroscopy (NVC) was performed and skin perfusion of hands was assessed by laser speckle contrast analysis (LASCA) in both populations. RESULTS IL33 was significantly higher in AP compared to HC [73.08 pg/ml (IQR 47.11-100.80 pg/ml) vs 41.5 4 pg/ml (IQR 20.16-55.49 pg/ml), p < 0.05] and RvD1 was significantly lower in AP than HC [36.1 pg/ml (IQR 27.88-66.21 pg/ml) vs 60.01 pg/ml (IQR 46.88-74.69 pg/ml), p < 0.05]. At LASCA, peripheral blood perfusion (PBP) was significantly lower in AP compared to HC [56.66 pU (IQR 46.29-65.44 pU) vs 87 pU (IQR 80-98 pU), p < 0.001]. The median values of ROI1 and ROI3 were significantly lower in AP compared to HC [112.81 pU (IQR 83.36-121.69 pU) vs 131 pU (IQR 108-135 pU), p < 0.05] and [59.78 pU (IQR 46.84-79.75 pU) vs 85 pU (IQR 78-98 pU), p < 0.05], respectively. The proximal-distal gradient (PDG) was observed in 8 of 20 (40 %) AP. CONCLUSION Serum IL33 is higher in AP compared to HC; conversely, RvD1 is lower in AP compared to HC. Reduction of PBP of hands was present in AP compared to HC, probably due to endothelial dysfunction.
Collapse
Affiliation(s)
- Denise Costa
- Department of Experimental Medicine, Endocrinology-Pituitary Disease, Sapienza University of Rome, Rome, Italy
| | - Chiara Pellicano
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Valeria Mercuri
- Department of Experimental Medicine, Endocrinology-Pituitary Disease, Sapienza University of Rome, Rome, Italy
| | - Jacopo Maria Arnone
- Department of Experimental Medicine, Endocrinology-Pituitary Disease, Sapienza University of Rome, Rome, Italy
| | - Flavio Rizzo
- Laboratory of Seminology-Sperm Bank "Loredana Gandini", Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Giorgia Leodori
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Patrizia Gargiulo
- Department of Experimental Medicine, Endocrinology-Pituitary Disease, Sapienza University of Rome, Rome, Italy
| | - Edoardo Rosato
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy.
| |
Collapse
|
39
|
Rivera FB, Taliño MK, Ansay MF, Mangubat GF, Mahilum ML, Menghrajani RH, Placino S, Cha SW, Aparece JP, Yu MG, Co ML, Lerma E, Vijayaraghavan K, McCullough PA. Cardiovascular Effects of Excess Growth Hormone: How Real is the Threat? Rev Cardiovasc Med 2023; 24:95. [PMID: 39076279 PMCID: PMC11273026 DOI: 10.31083/j.rcm2404095] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 01/16/2023] [Accepted: 01/31/2023] [Indexed: 07/31/2024] Open
Abstract
Patients with acromegaly carry a high risk of developing cardiovascular diseases (CVD). In fact, CVD is the leading cause of mortality among this group of patients. The most frequent cardiovascular complications are heart failure (HF), valvular disease, hypertension, arrhythmias, and coronary artery disease (CAD). The pathophysiology centers on the family of growth hormone (GH). These hormones are involved in normal cardiac development and function; however, excess of insulin-like growth factor-1 (IGF-1), the principally active hormone, can also cause negative effects on the cardiovascular system. HF in acromegaly usually presents with biventricular enlargement and diastolic dysfunction and is strongly associated with the duration of GH excess rather than the degree of hormone elevation. There is a high prevalence of valvular disease affecting aortic and mitral valves among patients with longer disease duration. The development of hypertension in acromegaly may be attributed to the effects of chronic GH/IGF-1 excess on different organ systems, which act via several mechanisms. The aspect of arrhythmia and CAD complicating acromegaly are currently not fully understood.
Collapse
Affiliation(s)
| | | | | | | | - Mer Lorraine Mahilum
- Department of Medicine, Southern Philippines Medical Center, 8000 Davao City, Philippines
| | - Rajiv Hans Menghrajani
- St. Luke’s Medical Center College of Medicine - William H. Quasha Memorial, 1102 Manila, Philippines
| | - Siena Placino
- St. Luke’s Medical Center College of Medicine - William H. Quasha Memorial, 1102 Manila, Philippines
| | - Sung Whoy Cha
- Cebu Institute of Medicine, 6000 Cebu City, Philippines
| | | | - Marc Gregory Yu
- Section of Vascular Cell Biology, Joslin Diabetes Center and Harvard Medical School, Boston, MA 02215, USA
| | - Michael Lawrenz Co
- Section of Cardiology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Edgar Lerma
- Section of Nephrology, University of Illinois at Chicago College of Medicine/Advocate Christ Medical Center, Oak Lawn, IL 60612, USA
| | | | | |
Collapse
|
40
|
Wang W, Yu K, Zhao SY, Mo DG, Liu JH, Han LJ, Li T, Yao HC. The impact of circulating IGF-1 and IGFBP-2 on cardiovascular prognosis in patients with acute coronary syndrome. Front Cardiovasc Med 2023; 10:1126093. [PMID: 36970368 PMCID: PMC10036580 DOI: 10.3389/fcvm.2023.1126093] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 02/22/2023] [Indexed: 03/12/2023] Open
Abstract
BackgroundWhile insulin-like growth factor 1 (IGF-1) exerts a cardioprotective effect in the setting of atherosclerosis, insulin-like growth factor binding protein 2 (IGFBP-2) is involved in metabolic syndrome. Although IGF-1 and IGFBP-2 are known to be predictors for mortality in patients with heart failure, their use in clinic as prognostic biomarkers for acute coronary syndrome (ACS) requires investigation. We evaluated the relationship between IGF-1 and IGFBP-2 levels at admission and the risk of major adverse cardiovascular events (MACEs) in patients with ACS.MethodsA total of 277 ACS patients and 42 healthy controls were included in this prospective cohort study. Plasma samples were obtained and analyzed at admission. Patients were followed for MACEs after hospitalization.ResultsAmong patients who suffered acute myocardial infarction, plasma levels of IGF-1 and IGFBP-2 were lower and higher, respectively, as compared to healthy controls (both p < 0.05). The mean follow-up period was 5.22 (1.0–6.0) months and MACEs incidence was 22.4% (62 of 277 patients). Kaplan–Meier survival analysis revealed that patients with low IGFBP-2 levels had a greater event-free survival rate than patients with high IGFBP-2 levels (p < 0.001). Multivariate Cox proportional hazards analysis revealed IGFBP-2, but not IGF-1, to be a positive predictor of MACEs (hazard ratio 2.412, 95% CI 1.360–4.277; p = 0.003).ConclusionOur findings suggest that high IGFBP-2 levels are associated with the development of MACEs following ACS. Moreover, IGFBP-2 is likely an independent predictive marker of clinical outcomes in ACS.
Collapse
Affiliation(s)
- Wei Wang
- Department of Cardiology, Liaocheng People's Hospital, Shandong University, Jinan, China
- Department of Cardiology, Liaocheng People's Hospital Affiliated to Shandong First Medical University, Liaocheng, China
| | - Kang Yu
- Department of Laboratory Medicine, Liaocheng People's Hospital Affiliated to Shandong First Medical University, Liaocheng, China
| | - Shou-Yong Zhao
- Department of Laboratory Medicine, Liaocheng People's Hospital Affiliated to Shandong First Medical University, Liaocheng, China
| | - De-Gang Mo
- Department of Cardiology, Liaocheng People's Hospital Affiliated to Shandong First Medical University, Liaocheng, China
| | - Jia-Hui Liu
- Department of Cardiology, Liaocheng People's Hospital Affiliated to Shandong First Medical University, Liaocheng, China
| | - Li-Jinn Han
- Department of Cardiology, Liaocheng People's Hospital Affiliated to Shandong First Medical University, Liaocheng, China
| | - Tai Li
- Department of Nursing, Liaocheng Vocational & Technical College, Liaocheng, China
| | - Heng-Chen Yao
- Department of Cardiology, Liaocheng People's Hospital, Shandong University, Jinan, China
- Department of Cardiology, Liaocheng People's Hospital Affiliated to Shandong First Medical University, Liaocheng, China
- Correspondence: Heng-Chen Yao
| |
Collapse
|
41
|
Anabolic and Inflammatory Response to High- and Low-Load Resistance Training in Patients with Coronary Artery Disease: A Randomized Controlled Trial. J Cardiopulm Rehabil Prev 2023:01273116-990000000-00076. [PMID: 36867715 DOI: 10.1097/hcr.0000000000000783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/05/2023]
|
42
|
Sukhanov S, Higashi Y, Yoshida T, Danchuk S, Alfortish M, Goodchild T, Scarborough A, Sharp T, Jenkins JS, Garcia D, Ivey J, Tharp DL, Schumacher J, Rozenbaum Z, Kolls JK, Bowles D, Lefer D, Delafontaine P. Insulin-like growth factor 1 reduces coronary atherosclerosis in pigs with familial hypercholesterolemia. JCI Insight 2023; 8:e165713. [PMID: 36602878 PMCID: PMC9990768 DOI: 10.1172/jci.insight.165713] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 01/03/2023] [Indexed: 01/06/2023] Open
Abstract
Although murine models of coronary atherosclerotic disease have been used extensively to determine mechanisms, limited new therapeutic options have emerged. Pigs with familial hypercholesterolemia (FH pigs) develop complex coronary atheromas that are almost identical to human lesions. We reported previously that insulin-like growth factor 1 (IGF-1) reduced aortic atherosclerosis and promoted features of stable plaque in a murine model. We administered human recombinant IGF-1 or saline (control) in atherosclerotic FH pigs for 6 months. IGF-1 decreased relative coronary atheroma in vivo (intravascular ultrasound) and reduced lesion cross-sectional area (postmortem histology). IGF-1 increased plaque's fibrous cap thickness, and reduced necrotic core, macrophage content, and cell apoptosis, consistent with promotion of a stable plaque phenotype. IGF-1 reduced circulating triglycerides, markers of systemic oxidative stress, and CXCL12 chemokine levels. We used spatial transcriptomics (ST) to identify global transcriptome changes in advanced plaque compartments and to obtain mechanistic insights into IGF-1 effects. ST analysis showed that IGF-1 suppressed FOS/FOSB factors and gene expression of MMP9 and CXCL14 in plaque macrophages, suggesting possible involvement of these molecules in IGF-1's effect on atherosclerosis. Thus, IGF-1 reduced coronary plaque burden and promoted features of stable plaque in a pig model, providing support for consideration of clinical trials.
Collapse
Affiliation(s)
- Sergiy Sukhanov
- Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Yusuke Higashi
- Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Tadashi Yoshida
- Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Svitlana Danchuk
- Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Mitzi Alfortish
- Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Traci Goodchild
- Cardiovascular Center of Excellence, School of Medicine, Louisiana State University, New Orleans, Louisiana, USA
| | - Amy Scarborough
- Cardiovascular Center of Excellence, School of Medicine, Louisiana State University, New Orleans, Louisiana, USA
| | - Thomas Sharp
- Cardiovascular Center of Excellence, School of Medicine, Louisiana State University, New Orleans, Louisiana, USA
| | | | | | - Jan Ivey
- Ochsner Medical Center, New Orleans, Louisiana, USA
| | - Darla L. Tharp
- Department of Biomedical Sciences, University of Missouri-Columbia, Missouri, USA
| | - Jeffrey Schumacher
- Cardiovascular Center of Excellence, School of Medicine, Louisiana State University, New Orleans, Louisiana, USA
| | - Zach Rozenbaum
- Tulane University School of Medicine, New Orleans, Louisiana, USA
- Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Jay K. Kolls
- Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Douglas Bowles
- Department of Biomedical Sciences, University of Missouri-Columbia, Missouri, USA
| | - David Lefer
- Cardiovascular Center of Excellence, School of Medicine, Louisiana State University, New Orleans, Louisiana, USA
| | | |
Collapse
|
43
|
Macvanin M, Gluvic Z, Radovanovic J, Essack M, Gao X, Isenovic ER. New insights on the cardiovascular effects of IGF-1. Front Endocrinol (Lausanne) 2023; 14:1142644. [PMID: 36843588 PMCID: PMC9947133 DOI: 10.3389/fendo.2023.1142644] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 01/26/2023] [Indexed: 02/11/2023] Open
Abstract
INTRODUCTION Cardiovascular (CV) disorders are steadily increasing, making them the world's most prevalent health issue. New research highlights the importance of insulin-like growth factor 1 (IGF-1) for maintaining CV health. METHODS We searched PubMed and MEDLINE for English and non-English articles with English abstracts published between 1957 (when the first report on IGF-1 identification was published) and 2022. The top search terms were: IGF-1, cardiovascular disease, IGF-1 receptors, IGF-1 and microRNAs, therapeutic interventions with IGF-1, IGF-1 and diabetes, IGF-1 and cardiovascular disease. The search retrieved original peer-reviewed articles, which were further analyzed, focusing on the role of IGF-1 in pathophysiological conditions. We specifically focused on including the most recent findings published in the past five years. RESULTS IGF-1, an anabolic growth factor, regulates cell division, proliferation, and survival. In addition to its well-known growth-promoting and metabolic effects, there is mounting evidence that IGF-1 plays a specialized role in the complex activities that underpin CV function. IGF-1 promotes cardiac development and improves cardiac output, stroke volume, contractility, and ejection fraction. Furthermore, IGF-1 mediates many growth hormones (GH) actions. IGF-1 stimulates contractility and tissue remodeling in humans to improve heart function after myocardial infarction. IGF-1 also improves the lipid profile, lowers insulin levels, increases insulin sensitivity, and promotes glucose metabolism. These findings point to the intriguing medicinal potential of IGF-1. Human studies associate low serum levels of free or total IGF-1 with an increased risk of CV and cerebrovascular illness. Extensive human trials are being conducted to investigate the therapeutic efficacy and outcomes of IGF-1-related therapy. DISCUSSION We anticipate the development of novel IGF-1-related therapy with minimal side effects. This review discusses recent findings on the role of IGF-1 in the cardiovascular (CVD) system, including both normal and pathological conditions. We also discuss progress in therapeutic interventions aimed at targeting the IGF axis and provide insights into the epigenetic regulation of IGF-1 mediated by microRNAs.
Collapse
Affiliation(s)
- Mirjana Macvanin
- Department of Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Zoran Gluvic
- Clinic for Internal Medicine, Department of Endocrinology and Diabetes, Zemun Clinical Hospital, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Jelena Radovanovic
- Department of Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Magbubah Essack
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
- Computer Science Program, Computer, Electrical and Mathematical Sciences and Engineering Division (CEMSE), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Xin Gao
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
- Computer Science Program, Computer, Electrical and Mathematical Sciences and Engineering Division (CEMSE), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Esma R. Isenovic
- Department of Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
44
|
Ferraguti G, Terracina S, Micangeli G, Lucarelli M, Tarani L, Ceccanti M, Spaziani M, D'Orazi V, Petrella C, Fiore M. NGF and BDNF in pediatrics syndromes. Neurosci Biobehav Rev 2023; 145:105015. [PMID: 36563920 DOI: 10.1016/j.neubiorev.2022.105015] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 11/02/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022]
Abstract
Neurotrophins (NTs) as nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF) play multiple roles in different settings including neuronal development, function and survival in both the peripheral and the central nervous systems from early stages. This report aims to provide a summary and subsequent review of evidences on the role of NTs in rare and non-common pediatric human diseases associated with changes in neurodevelopment. A variety of diseases has been analyzed and many have been linked to NTs neurobiological effects, including chronic granulomatous disease, hereditary sensory and autonomic neuropathy, Duchenne muscular dystrophy, Bardet-Biedl syndrome, Angelman syndrome, fragile X syndrome, trisomy 16, Williams-Beuren syndrome, Prader-Willi syndrome, WAGR syndrome, fetal alcohol spectrum disorders, Down syndrome and Klinefelter Syndrome. NTs alterations have been associated with numerous pathologic manifestations including cognitive defects, behavioral abnormalities, epilepsy, obesity, tumorigenesis as well as muscle-skeletal, immunity, bowel, pain sensibility and cilia diseases. In this report, we discuss that further studies are needed to clear a possible therapeutic role of NTs in these still often uncurable diseases.
Collapse
Affiliation(s)
- Giampiero Ferraguti
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Sergio Terracina
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Ginevra Micangeli
- Department of Maternal Infantile and Urological Sciences, Sapienza University of Rome, Italy
| | - Marco Lucarelli
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Luigi Tarani
- Department of Maternal Infantile and Urological Sciences, Sapienza University of Rome, Italy
| | - Mauro Ceccanti
- SITAC, Società Italiana per il Trattamento dell'Alcolismo e le sue Complicanze, Rome, Italy
| | - Matteo Spaziani
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Valerio D'Orazi
- Department of Surgical Sciences, Sapienza University of Rome, Rome, Italy
| | - Carla Petrella
- Institute of Biochemistry and Cell Biology, IBBC-CNR, Rome, Italy.
| | - Marco Fiore
- Institute of Biochemistry and Cell Biology, IBBC-CNR, Rome, Italy.
| |
Collapse
|
45
|
Bickel MA, Csik B, Gulej R, Ungvari A, Nyul-Toth A, Conley SM. Cell non-autonomous regulation of cerebrovascular aging processes by the somatotropic axis. Front Endocrinol (Lausanne) 2023; 14:1087053. [PMID: 36755922 PMCID: PMC9900125 DOI: 10.3389/fendo.2023.1087053] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 01/04/2023] [Indexed: 01/24/2023] Open
Abstract
Age-related cerebrovascular pathologies, ranging from cerebromicrovascular functional and structural alterations to large vessel atherosclerosis, promote the genesis of vascular cognitive impairment and dementia (VCID) and exacerbate Alzheimer's disease. Recent advances in geroscience, including results from studies on heterochronic parabiosis models, reinforce the hypothesis that cell non-autonomous mechanisms play a key role in regulating cerebrovascular aging processes. Growth hormone (GH) and insulin-like growth factor 1 (IGF-1) exert multifaceted vasoprotective effects and production of both hormones is significantly reduced in aging. This brief overview focuses on the role of age-related GH/IGF-1 deficiency in the development of cerebrovascular pathologies and VCID. It explores the mechanistic links among alterations in the somatotropic axis, specific macrovascular and microvascular pathologies (including capillary rarefaction, microhemorrhages, impaired endothelial regulation of cerebral blood flow, disruption of the blood brain barrier, decreased neurovascular coupling, and atherogenesis) and cognitive impairment. Improved understanding of cell non-autonomous mechanisms of vascular aging is crucial to identify targets for intervention to promote cerebrovascular and brain health in older adults.
Collapse
Affiliation(s)
- Marisa A. Bickel
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Boglarka Csik
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Rafal Gulej
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Anna Ungvari
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- International Training Program in Geroscience, Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Adam Nyul-Toth
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- International Training Program in Geroscience, Department of Public Health, Semmelweis University, Budapest, Hungary
- Institute of Biophysics, Biological Research Centre, Eötvös Lorand Research Network (ELKH), Szeged, Hungary
| | - Shannon M. Conley
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| |
Collapse
|
46
|
Guan J, Li F, Kang D, Anderson T, Pitcher T, Dalrymple-Alford J, Shorten P, Singh-Mallah G. Cyclic Glycine-Proline (cGP) Normalises Insulin-Like Growth Factor-1 (IGF-1) Function: Clinical Significance in the Ageing Brain and in Age-Related Neurological Conditions. Molecules 2023; 28:molecules28031021. [PMID: 36770687 PMCID: PMC9919809 DOI: 10.3390/molecules28031021] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 01/16/2023] [Accepted: 01/17/2023] [Indexed: 01/20/2023] Open
Abstract
Insulin-like growth factor-1 (IGF-1) function declines with age and is associated with brain ageing and the progression of age-related neurological conditions. The reversible binding of IGF-1 to IGF binding protein (IGFBP)-3 regulates the amount of bioavailable, functional IGF-1 in circulation. Cyclic glycine-proline (cGP), a metabolite from the binding site of IGF-1, retains its affinity for IGFBP-3 and competes against IGF-1 for IGFBP-3 binding. Thus, cGP and IGFBP-3 collectively regulate the bioavailability of IGF-1. The molar ratio of cGP/IGF-1 represents the amount of bioavailable and functional IGF-1 in circulation. The cGP/IGF-1 molar ratio is low in patients with age-related conditions, including hypertension, stroke, and neurological disorders with cognitive impairment. Stroke patients with a higher cGP/IGF-1 molar ratio have more favourable clinical outcomes. The elderly with more cGP have better memory retention. An increase in the cGP/IGF-1 molar ratio with age is associated with normal cognition, whereas a decrease in this ratio with age is associated with dementia in Parkinson disease. In addition, cGP administration reduces systolic blood pressure, improves memory, and aids in stroke recovery. These clinical and experimental observations demonstrate the role of cGP in regulating IGF-1 function and its potential clinical applications in age-related brain diseases as a plasma biomarker for-and an intervention to improve-IGF-1 function.
Collapse
Affiliation(s)
- Jian Guan
- Department of Pharmacology and Clinical Pharmacology, Faculty of Medicine and Health Sciences, School of Biomedical Sciences, The University of Auckland, Auckland 1142, New Zealand
- Centre for Brain Research, Faculty of Medicine and Health Sciences, School of Biomedical Sciences, The University of Auckland, Auckland 1142, New Zealand
- Brain Research New Zealand, The Centre for Research Excellent, Dunedin 9016, New Zealand
- The cGP Lab Limited New Zealand, Auckland 1021, New Zealand
- Correspondence: ; Tel.: +64-9-923-6134
| | - Fengxia Li
- Department of Pharmacology and Clinical Pharmacology, Faculty of Medicine and Health Sciences, School of Biomedical Sciences, The University of Auckland, Auckland 1142, New Zealand
- Centre for Brain Research, Faculty of Medicine and Health Sciences, School of Biomedical Sciences, The University of Auckland, Auckland 1142, New Zealand
- Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, Guangzhou 510075, China
- The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
| | - Dali Kang
- Department of Pharmacology and Clinical Pharmacology, Faculty of Medicine and Health Sciences, School of Biomedical Sciences, The University of Auckland, Auckland 1142, New Zealand
- Centre for Brain Research, Faculty of Medicine and Health Sciences, School of Biomedical Sciences, The University of Auckland, Auckland 1142, New Zealand
- Brain Research New Zealand, The Centre for Research Excellent, Dunedin 9016, New Zealand
- Shenyang Medical College, Shenyang 110034, China
| | - Tim Anderson
- New Zealand Brain Research Institute, Christchurch 4710, New Zealand
- Department of Medicine, University of Otago, Dunedin 9016, New Zealand
- Department of Neurology, Canterbury District Health Board, Christchurch 4710, New Zealand
| | - Toni Pitcher
- New Zealand Brain Research Institute, Christchurch 4710, New Zealand
- Department of Medicine, University of Otago, Dunedin 9016, New Zealand
- Department of Neurology, Canterbury District Health Board, Christchurch 4710, New Zealand
| | - John Dalrymple-Alford
- Department of Neurology, Canterbury District Health Board, Christchurch 4710, New Zealand
- Department of Psychology, University of Canterbury, Christchurch 4710, New Zealand
| | - Paul Shorten
- AgResearch Ltd., Ruakura Research Centre, Hamilton 3214, New Zealand
- Riddet Institute, Massey University, Palmerston North 4474, New Zealand
| | - Gagandeep Singh-Mallah
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden
| |
Collapse
|
47
|
Truong T, Silkiss RZ. The Role of Insulin-like Growth Factor-1 and Its Receptor in the Eye: A Review and Implications for IGF-1R Inhibition. Ophthalmic Plast Reconstr Surg 2023; 39:4-12. [PMID: 36598389 DOI: 10.1097/iop.0000000000002146] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
PURPOSE FDA approval of teprotumumab for thyroid eye disease in January 2020 reinforced interest in the pharmacologic potential of insulin-like growth factor-1 (IGF-1) and its receptor, IGF-1R. Despite recent approval and adaptation for ophthalmic use, IGF-1R inhibitors are not a new therapeutic class. In 1986, Yamashita described aIR3, a monoclonal antibody to IGF-1R (anti-IGF-1R), that inhibited the effect of IGF-1 on growth hormone release. Given the widespread presence of IGF-1R, interrupting this receptor can lead to systemic physiologic effects, some adverse. We aim to review what is known about IGF-1/IGF-1R in the eye and consider the possible local side effects, unintended consequences, and potential uses of this medication class. METHODS A PubMed database search utilizing the keywords "insulin-like growth factor-1, eye, inhibitor, antibody, side effect" was performed to identify publications discussing IGF-1 in the human eye from January 2011 to August 2021. Criteria for acceptance included studies discussing human subjects or human tissue specifically related to the eye. RESULTS Out of a total of 230 articles, 47 were organized in 3 subject groups for discussion: thyroid-associated orbitopathy, cornea and the ocular surface, and the retina and neovascularization. Review of the literature demonstrated that IGF-1 affects growth and development of the eye, epithelial proliferation, retinal angiogenesis, inflammation, and is associated with thyroid-associated orbitopathy. CONCLUSIONS IGF-1R exists throughout in the human body, including the cornea, retina, and orbit. Research regarding ocular effects of IGF-1/IGF-1R outside thyroid eye disease is limited. Carefully designed studies and clinical assessments of patients undergoing treatment with anti-IGF-1R may identify ocular side effects and foster consideration of the role of anti-IGF-1R in ocular therapeutics. Given the increasing use of anti-IGF-1R antibodies, understanding their ocular effects, side effects, and potential systemic implications for use in disease is critical.
Collapse
Affiliation(s)
- Timothy Truong
- Department of Ophthalmology, Division of Oculofacial Plastic Surgery, California Pacific Medical Center, San Francisco, California, U.S.A
| | - Rona Z Silkiss
- Department of Ophthalmology, Division of Oculofacial Plastic Surgery, California Pacific Medical Center, San Francisco, California, U.S.A
- Department of Ophthalmology, Division of Oculofacial Plastic Surgery, Silkiss Eye Surgery, San Francisco, California, U.S.A
| |
Collapse
|
48
|
Wang H, Wang J, Geng C, Wang C, Gu R, Zhu Z. A variant rs6214 within IGF-1 confers risk for ulcerative colitis in Chinese Han populations. Funct Integr Genomics 2022; 23:1. [PMID: 36520299 DOI: 10.1007/s10142-022-00921-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/15/2022] [Accepted: 11/22/2022] [Indexed: 12/16/2022]
Abstract
Insulin growth factor-1 (IGF-1) has been found to correlate with various diseases such as cancer and cardiovascular diseases including ulcerative colitis (UC). The present study aimed to investigate the plausible association of rs6214 (C > T) within IGF-1 and UC susceptibility in Chinese Han populations. A total of 977 UC patients and 1029 healthy controls were enrolled, and rs6214 was genotyped with PCR and direct sequencing on the ABI 3730XL DNA analyzer platform. Logistic regression analysis was applied for the correlation of rs6214 and UC susceptibility via calculation of odds ratio (OR) with a 95% confidence interval (95% CI) adjusted for age and sex under different genetic models. The difference of clinical parameters between genotypes was measured by one-way analysis of variance (ANOVA). Additional functional assays were conducted to establish the probable relationship. The results indicated that the T allele of rs6214 showed roughly 37% greater risk for UC risk in the additive model (OR = 1.37, 95% CI = 1.21-1.55, P < 0.000001) when compared with C allele carriers, and the pattern was similar in other three genetic models. Further stratified analysis suggested that the association was particularly noteworthy in UC patients with extensive colitis and severe condition. Moreover, the blood level of IGF-1 was downregulated in UC patients, and the mRNA level was lower in T allele carriers in rectal tissues of UC cases. Additional luciferase assay demonstrated that rs6214 regulates IGF-1 expression via promoting miR-2053. Collectively, rs6214 increased UC susceptibility and suppresses IGF-1 expression by enhancing miR-2053 binding. The current findings provided evidence that rs6214 is a promising biomarker for UC prediction and prognosis.
Collapse
Affiliation(s)
- Huiping Wang
- Xuzhou Engineering Research Center of Medical Genetics and Transformation, Laboratory of Genetic Foundation and Clinical Application, Department of Genetics, School of Life Sciences, Xuzhou Medical University, Xuzhou, 221004, China
| | - Jin Wang
- Laboratory of Experimental and Clinical Pathology, Departments of Pathology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, 221004, China
| | - Chunsong Geng
- Department of Laboratory, Suzhou Kowloon Hospital Shanghai Jiao Tong University School of Medicine, Suzhou, 215000, China
| | - Cong Wang
- Laboratory of Experimental and Clinical Pathology, Departments of Pathology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, 221004, China
| | - Rongrong Gu
- School of Life Sciences, Xuzhou Medical University, Xuzhou, 221004, China
| | - Zhansheng Zhu
- Laboratory of Experimental and Clinical Pathology, Departments of Pathology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, 221004, China.
| |
Collapse
|
49
|
Toth L, Czigler A, Hegedus E, Komaromy H, Amrein K, Czeiter E, Yabluchanskiy A, Koller A, Orsi G, Perlaki G, Schwarcz A, Buki A, Ungvari Z, Toth PJ. Age-related decline in circulating IGF-1 associates with impaired neurovascular coupling responses in older adults. GeroScience 2022; 44:2771-2783. [PMID: 35869380 PMCID: PMC9768079 DOI: 10.1007/s11357-022-00623-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 07/09/2022] [Indexed: 01/07/2023] Open
Abstract
Impairment of moment-to-moment adjustment of cerebral blood flow (CBF) to the increased oxygen and energy requirements of active brain regions via neurovascular coupling (NVC) contributes to the genesis of age-related cognitive impairment. Aging is associated with marked deficiency in the vasoprotective hormone insulin-like growth factor-1 (IGF-1). Preclinical studies on animal models of aging suggest that circulating IGF-1 deficiency is causally linked to impairment of NVC responses. The present study was designed to test the hypotheses that decreases in circulating IGF-1 levels in older adults also predict the magnitude of age-related decline of NVC responses. In a single-center cross-sectional study, we enrolled healthy young (n = 31, 11 female, 20 male, mean age: 28.4 + / - 4.2 years) and aged volunteers (n = 32, 18 female, 14 male, mean age: 67.9 + / - 4.1 years). Serum IGF-1 level, basal CBF (phase contrast magnetic resonance imaging (MRI)), and NVC responses during the trail making task (with transcranial Doppler sonography) were assessed. We found that circulating IGF-1 levels were significantly decreased with age and associated with decreased basal CBF. Age-related decline in IGF-1 levels predicted the magnitude of age-related decline in NVC responses. In conclusion, our study provides additional evidence in support of the concept that age-related circulating IGF-1 deficiency contributes to neurovascular aging, impairing CBF and functional hyperemia in older adults.
Collapse
Affiliation(s)
- Luca Toth
- Department of Neurosurgery, Medical School, University of Pecs, 2 Ret Street, Pecs, 7624, Hungary
- Institute for Translational Medicine, Medical School, University of Pecs, Pecs, Hungary
| | - Andras Czigler
- Department of Neurosurgery, Medical School, University of Pecs, 2 Ret Street, Pecs, 7624, Hungary
- Institute for Translational Medicine, Medical School, University of Pecs, Pecs, Hungary
| | - Emoke Hegedus
- Department of Neurosurgery, Medical School, University of Pecs, 2 Ret Street, Pecs, 7624, Hungary
| | - Hedvig Komaromy
- Department of Neurosurgery, Medical School, University of Pecs, 2 Ret Street, Pecs, 7624, Hungary
| | - Krisztina Amrein
- Department of Neurosurgery, Medical School, University of Pecs, 2 Ret Street, Pecs, 7624, Hungary
| | - Endre Czeiter
- Department of Neurosurgery, Medical School, University of Pecs, 2 Ret Street, Pecs, 7624, Hungary
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Akos Koller
- Department of Neurosurgery, Medical School, University of Pecs, 2 Ret Street, Pecs, 7624, Hungary
| | - Gergely Orsi
- ELKH-PTE Clinical Neuroscience MR Research Group, Eötvös Lóránd Research Network (ELKH), Pecs, Hungary
- Department of Neurology, Medical School, University of Pecs, Pecs, Hungary
| | - Gabor Perlaki
- ELKH-PTE Clinical Neuroscience MR Research Group, Eötvös Lóránd Research Network (ELKH), Pecs, Hungary
- Department of Neurology, Medical School, University of Pecs, Pecs, Hungary
| | - Attila Schwarcz
- Department of Neurosurgery, Medical School, University of Pecs, 2 Ret Street, Pecs, 7624, Hungary
| | - Andras Buki
- Department of Neurosurgery, Medical School, University of Pecs, 2 Ret Street, Pecs, 7624, Hungary
| | - Zoltan Ungvari
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Peter J Toth
- Department of Neurosurgery, Medical School, University of Pecs, 2 Ret Street, Pecs, 7624, Hungary.
- Institute for Translational Medicine, Medical School, University of Pecs, Pecs, Hungary.
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
50
|
Elbaset MA, Nasr M, Ibrahim BMM, Ahmed-Farid OAH, Bakeer RM, Hassan NS, Ahmed RF. Curcumin nanoemulsion counteracts hepatic and cardiac complications associated with high-fat/high-fructose diet in rats. J Food Biochem 2022; 46:e14442. [PMID: 36165438 DOI: 10.1111/jfbc.14442] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 09/05/2022] [Accepted: 09/15/2022] [Indexed: 01/14/2023]
Abstract
The relationship between the incidence of cardiovascular abnormalities and non-alcoholic fatty liver disease (NAFLD) has long been postulated. Curcumin (CUR) is a potential anti-atherosclerotic agent but its poor water solubility hinders its pharmacological use. Therefore, the present study aimed to investigate the effect of formulation of CUR nanoemulsion prepared using the spontaneous emulsification technique on high fat high fructose (HFHF)-induced hepatic and cardiac complications. Fifty Wistar rats were divided into five groups. CUR nanoemulsion at doses of 5 and 10 mg/kg and conventional powdered CUR at a dose of 50 mg/kg were orally administered daily to rats for two weeks, and compared with normal control and HFHF control. Results revealed that the high dose level of CUR nanoemulsion was superior to conventional CUR in ameliorating the HFHF-induced insulin resistance status and hyperlipidemia, with beneficial impact on rats' recorded electrocardiogram (ECG), serum aspartate aminotransferase (ALT) and alanine aminotransferase (AST) levels, leptin, adiponectin, creatine phosphokinase, lactate dehydrogenase and cardiac troponin-I. In addition, hepatic and cardiac oxidative and nitrosative stresses, oxidative DNA damage and disrupted cellular energy statuses were counteracted. Results were also confirmed by histopathological examination. PRACTICAL APPLICATIONS: The use of curcumin nanoemulsion could be beneficial in combating hepatic and cardiac complications resulting from HFHF diets.
Collapse
Affiliation(s)
- Marawan Abd Elbaset
- Department of Pharmacology, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt
| | - Maha Nasr
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Bassant M M Ibrahim
- Department of Pharmacology, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt
| | - Omar A H Ahmed-Farid
- Department of Physiology, National Organization for Drug Control and Research, Giza, Egypt
| | - Rofanda M Bakeer
- Department of Pathology, Faculty of Medicine, Helwan University, Helwan, Egypt
| | - Nabila S Hassan
- Department of Pathology, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt
| | - Rania F Ahmed
- Department of Pharmacology, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt
| |
Collapse
|