1
|
Koutaki D, Paltoglou G, Manou M, Vourdoumpa A, Ramouzi E, Tzounakou AM, Michos A, Bacopoulou F, Mantzou E, Zoumakis E, Papadopoulou M, Kassari P, Charmandari E. The Role of Secreted Frizzled-Related Protein 5 (Sfrp5) in Overweight and Obesity in Childhood and Adolescence. Nutrients 2024; 16:3133. [PMID: 39339733 PMCID: PMC11434931 DOI: 10.3390/nu16183133] [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: 07/14/2024] [Revised: 09/05/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
Background/Objective: Secreted frizzled-related protein 5 (Sfrp5) is an anti-inflammatory adipokine that has been implicated in the pathophysiology of obesity and its metabolic complications. Despite the fact that numerous studies have been carried out in adults, limited data on Sfrp5 exist for youth, especially in relation to overweight and obesity. Methods: In our study, we assessed the concentrations of Sfrp5, total oxidative (TOS) and antioxidative (TAS) status, high-sensitivity C-reactive protein (hs-CRP), and several cytokines (IL-1α, IL-1β, IL-2, IL-6, IL-8, IL-12, TNF-α) in 120 children and adolescents (mean age ± SE: 11.48 ± 0.25 years; 48 prepubertal, 72 pubertal; 74 males and 46 females) before and 1 year after the implementation of a personalized, structured, lifestyle intervention program of healthy diet, sleep, and physical exercise. Results: Based on the body mass index (BMI), participants were categorized as having morbid obesity (n = 63, 52.5%), obesity (n = 21, 17.5%), overweight (n = 22, 18.33%), or normal BMIs (n = 14, 11.67%), based on the International Obesity Task Force (IOTF) cut-off points. Following the 1-year lifestyle intervention program, a significant improvement in anthropometric measurements (BMI, BMI-z score, diastolic blood pressure, WHR, and WHtR), body-composition parameters, hepatic enzymes, lipid profile, inflammation markers, and the insulin-sensitivity profile (HbA1C, HOMA index) was observed in all subjects. Sfrp5 decreased in subjects with obesity (p < 0.01); however, it increased significantly (p < 0.05) in patients with morbid obesity. Linear regression analysis indicates that TNF-α and systolic blood pressure were the best positive predictors and hs-CRP was the best negative predictor for Sfpr5 concentration at initial assessment and glucose concentration for ΔSfrp5, while TNF-α and TAS were the best positive predictors for Sfpr5 concentration at annual assessment. Conclusions: These results indicate that Sfrp5 is associated with severe obesity and is increased following weight loss in children and adolescents with morbid obesity. It is also related to metabolic homeostasis, as well as inflammation and oxidative status.
Collapse
Affiliation(s)
- Diamanto Koutaki
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, “Aghia Sophia” Children’s Hospital, 11527 Athens, Greece; (D.K.); (G.P.); (M.M.); (A.V.); (E.R.); (A.-M.T.); (E.M.); (E.Z.); (M.P.); (P.K.)
| | - George Paltoglou
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, “Aghia Sophia” Children’s Hospital, 11527 Athens, Greece; (D.K.); (G.P.); (M.M.); (A.V.); (E.R.); (A.-M.T.); (E.M.); (E.Z.); (M.P.); (P.K.)
| | - Maria Manou
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, “Aghia Sophia” Children’s Hospital, 11527 Athens, Greece; (D.K.); (G.P.); (M.M.); (A.V.); (E.R.); (A.-M.T.); (E.M.); (E.Z.); (M.P.); (P.K.)
- Department of Hygiene and Epidemiology, School of Medicine, University of Ioannina, 45110 Ioannina, Greece
| | - Aikaterini Vourdoumpa
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, “Aghia Sophia” Children’s Hospital, 11527 Athens, Greece; (D.K.); (G.P.); (M.M.); (A.V.); (E.R.); (A.-M.T.); (E.M.); (E.Z.); (M.P.); (P.K.)
| | - Eleni Ramouzi
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, “Aghia Sophia” Children’s Hospital, 11527 Athens, Greece; (D.K.); (G.P.); (M.M.); (A.V.); (E.R.); (A.-M.T.); (E.M.); (E.Z.); (M.P.); (P.K.)
| | - Anastasia-Maria Tzounakou
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, “Aghia Sophia” Children’s Hospital, 11527 Athens, Greece; (D.K.); (G.P.); (M.M.); (A.V.); (E.R.); (A.-M.T.); (E.M.); (E.Z.); (M.P.); (P.K.)
| | - Athanasios Michos
- Division of Infectious Diseases, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, “Aghia Sophia” Children’s Hospital, 11527 Athens, Greece;
| | - Flora Bacopoulou
- University Research Institute of Maternal and Child Health and Precision Medicine, and UNESCO Chair on Adolescent Health Care, National and Kapodistrian University of Athens, “Aghia Sophia” Children’s Hospital, 11527 Athens, Greece;
| | - Emilia Mantzou
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, “Aghia Sophia” Children’s Hospital, 11527 Athens, Greece; (D.K.); (G.P.); (M.M.); (A.V.); (E.R.); (A.-M.T.); (E.M.); (E.Z.); (M.P.); (P.K.)
| | - Emmanouil Zoumakis
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, “Aghia Sophia” Children’s Hospital, 11527 Athens, Greece; (D.K.); (G.P.); (M.M.); (A.V.); (E.R.); (A.-M.T.); (E.M.); (E.Z.); (M.P.); (P.K.)
| | - Marina Papadopoulou
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, “Aghia Sophia” Children’s Hospital, 11527 Athens, Greece; (D.K.); (G.P.); (M.M.); (A.V.); (E.R.); (A.-M.T.); (E.M.); (E.Z.); (M.P.); (P.K.)
| | - Penio Kassari
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, “Aghia Sophia” Children’s Hospital, 11527 Athens, Greece; (D.K.); (G.P.); (M.M.); (A.V.); (E.R.); (A.-M.T.); (E.M.); (E.Z.); (M.P.); (P.K.)
- Division of Endocrinology and Metabolism, Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| | - Evangelia Charmandari
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, “Aghia Sophia” Children’s Hospital, 11527 Athens, Greece; (D.K.); (G.P.); (M.M.); (A.V.); (E.R.); (A.-M.T.); (E.M.); (E.Z.); (M.P.); (P.K.)
- Division of Endocrinology and Metabolism, Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| |
Collapse
|
2
|
Zhang M, Li G, Li K, Gao Z, Yin C, Zeng F, Yang H, Dong W, Zhou G, Pan W, Wang Y, Jin J. Prognostic significance of serum secreted frizzled-related protein 5 in patients with acute aortic dissection. Heliyon 2024; 10:e35905. [PMID: 39253195 PMCID: PMC11382199 DOI: 10.1016/j.heliyon.2024.e35905] [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: 02/15/2024] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 09/11/2024] Open
Abstract
Background Secreted frizzled-related protein 5 (SFRP5) is a novel adipokine that has been found to be closely associated with metabolic and cardiovascular diseases. We investigated serum SFRP5 levels during the acute phase and their predictive value for the prognosis of acute aortic dissection (AAD). Methods In total, 152 AAD patients and 164 controls were enrolled in this study. Serum SFRP5 levels were measured using an enzyme-linked immunosorbent assay (ELISA). AAD patients were divided into high-SFRP5 and low-SFRP5 groups based on the optimal cutoff value and followed up for prognosis. The primary endpoint was all-cause mortality, and the secondary endpoint focused on AAD-related events (including AAD-related mortality and unplanned reoperations). Results Serum SFRP5 levels were significantly higher in AAD patients than in non-AAD controls, regardless of whether they had Stanford type A or B AD. Multivariate logistic regression analysis revealed an independent association between SFRP5 and the presence of AAD (adjusted OR 1.267, 95 % CI 1.152-1.394; p < 0.001). The receiver operating characteristic curve demonstrated that the optimal cutoff value for SFRP5 to predict the presence of AAD was 10.26 ng/mL (AUC 0.7241, sensitivity 49.34 %, specificity 87.20 %). Notably, serum SFRP5 levels of patients in the death group were significantly higher than those in the survival group. Compared with patients in the low-SFRP5 group, those in the high-SFRP5 group exhibited a significantly increased risk of all-cause mortality (HR 9.540, 95 % CI 2.803-32.473; p < 0.001) and AAD-related events (HR 6.915, 95 % CI 2.361-20.254; p < 0.001) during the follow-up period. Conclusion Serum SFRP5 levels were significantly elevated in the acute phase of AAD, and high serum SFRP5 levels were independently associated with poor AAD prognosis. These results suggest that serum SFRP5 level during the acute phase may be an effective biomarker and therapeutic target for the prognosis of AAD.
Collapse
Affiliation(s)
- Mingle Zhang
- Department of Cardiology, The Second Affiliated Hospital (Xinqiao Hospital) of Army Medical University, Chongqing, 400037, China
- School of Medicine, Chongqing University, Chongqing, 400044, China
| | - Gaoshan Li
- Department of Cardiology, The Second Affiliated Hospital (Xinqiao Hospital) of Army Medical University, Chongqing, 400037, China
| | - Kunyan Li
- Department of Cardiology, The Second Affiliated Hospital (Xinqiao Hospital) of Army Medical University, Chongqing, 400037, China
| | - Zhichun Gao
- Department of Cardiology, The Second Affiliated Hospital (Xinqiao Hospital) of Army Medical University, Chongqing, 400037, China
| | - Chun Yin
- Department of Cardiology, The Second Affiliated Hospital (Xinqiao Hospital) of Army Medical University, Chongqing, 400037, China
| | - Fangzheng Zeng
- Department of Emergency Medicine, The Second Affiliated Hospital (Xinqiao Hospital) of Army Medical University, Chongqing, 400037, China
| | - Hao Yang
- Department of Cardiology, The Second Affiliated Hospital (Xinqiao Hospital) of Army Medical University, Chongqing, 400037, China
| | - Wang Dong
- Department of Cardiology, The Second Affiliated Hospital (Xinqiao Hospital) of Army Medical University, Chongqing, 400037, China
| | - Guiquan Zhou
- Department of Cardiology, The Second Affiliated Hospital (Xinqiao Hospital) of Army Medical University, Chongqing, 400037, China
| | - Wenxu Pan
- Department of Cardiology, The Second Affiliated Hospital (Xinqiao Hospital) of Army Medical University, Chongqing, 400037, China
| | - Ying Wang
- Department of Cardiology, The Second Affiliated Hospital (Xinqiao Hospital) of Army Medical University, Chongqing, 400037, China
| | - Jun Jin
- Department of Cardiology, The Second Affiliated Hospital (Xinqiao Hospital) of Army Medical University, Chongqing, 400037, China
| |
Collapse
|
3
|
Li R, Liu S, Yeo K, Edwards S, Li MY, Santos R, Rad SK, Wu F, Maddern G, Young J, Tomita Y, Townsend A, Fenix K, Hauben E, Price T, Smith E. Diagnostic and prognostic significance of circulating secreted frizzled-related protein 5 in colorectal cancer. Cancer Med 2024; 13:e7352. [PMID: 38872420 PMCID: PMC11176579 DOI: 10.1002/cam4.7352] [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: 03/27/2024] [Revised: 05/15/2024] [Accepted: 05/26/2024] [Indexed: 06/15/2024] Open
Abstract
BACKGROUND Secreted Frizzled-Related Protein 5 (SFRP5) modulates Wnt signalling pathways, affecting diverse biological processes. We assessed the diagnostic and prognostic value of circulating SFRP5 (cSFRP5) in colorectal cancer (CRC) METHODS: Plasma cSFRP5 concentrations were measured using enzyme-linked immunosorbent assay (ELISA) in healthy donors (n = 133), individuals diagnosed with CRC (n = 449), colorectal polyps (n = 85), and medical conditions in other organs including cancer, inflammation, and benign states (n = 64). RESULTS Patients with CRC, polyps, and other conditions showed higher cSFRP5 levels than healthy individuals (p < 0.0001). Receiver operating characteristic curves comparing healthy donors with medical conditions, polyps and CRC were 0.814 (p < 0.0001), 0.763 (p < 0.0001) and 0.762 (p < 0.0001), respectively. In CRC, cSFRP5 correlated with patient age (p < 0.0001), tumour stage (p < 0.0001), and histological differentiation (p = 0.0273). Levels, adjusted for patient age, sex, plasma age and collection institution, peaked in stage II versus I (p < 0.0001), III (p = 0.0002) and IV (p < 0.0001), were lowest in stage I versus III (p = 0.0002) and IV (p = 0.0413), with no difference between stage III and IV. Elevated cSFRP5 levels predicted longer overall survival in stages II-III CRC (univariate: HR 1.82, 95% CI: 1.02-3.26, p = 0.024; multivariable: HR 2.34, 95% CI: 1.12-4.88, p = 0.015). CONCLUSION This study confirms cSFRP5 levels are elevated in CRC compared to healthy control and reveals a correlation between elevated cSFRP5 and overall survival in stages II-III disease.
Collapse
Affiliation(s)
- Runhao Li
- Solid Tumour Group, Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville South, South Australia, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| | - Saifei Liu
- Solid Tumour Group, Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville South, South Australia, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| | - Kenny Yeo
- Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
- Discipline of Surgery, The University of Adelaide, Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville South, South Australia, Australia
| | - Suzanne Edwards
- School of Public Health, The University of Adelaide, Adelaide, South Australia, Australia
| | - Man Ying Li
- Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
- Discipline of Surgery, The University of Adelaide, Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville South, South Australia, Australia
| | - Ryan Santos
- Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
- Viral Immunology Group, The University of Adelaide and Basil Hetzel Institute for Translational Health Research, Woodville, South Australia, Australia
| | - Sima Kianpour Rad
- Solid Tumour Group, Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville South, South Australia, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| | - Fangmeinuo Wu
- Solid Tumour Group, Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville South, South Australia, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| | - Guy Maddern
- Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
- Discipline of Surgery, The University of Adelaide, Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville South, South Australia, Australia
| | - Joanne Young
- Solid Tumour Group, Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville South, South Australia, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
- Medical Oncology, The Queen Elizabeth Hospital, Woodville, South Australia, Australia
| | - Yoko Tomita
- Solid Tumour Group, Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville South, South Australia, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
- Medical Oncology, The Queen Elizabeth Hospital, Woodville, South Australia, Australia
| | - Amanda Townsend
- Solid Tumour Group, Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville South, South Australia, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
- Medical Oncology, The Queen Elizabeth Hospital, Woodville, South Australia, Australia
| | - Kevin Fenix
- Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
- Discipline of Surgery, The University of Adelaide, Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville South, South Australia, Australia
| | - Ehud Hauben
- Discipline of Surgery, The University of Adelaide, Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville South, South Australia, Australia
| | - Timothy Price
- Solid Tumour Group, Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville South, South Australia, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
- Medical Oncology, The Queen Elizabeth Hospital, Woodville, South Australia, Australia
| | - Eric Smith
- Solid Tumour Group, Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville South, South Australia, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
- Discipline of Surgery, The University of Adelaide, Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville South, South Australia, Australia
- Medical Oncology, The Queen Elizabeth Hospital, Woodville, South Australia, Australia
| |
Collapse
|
4
|
An F, Song J, Chang W, Zhang J, Gao P, Wang Y, Xiao Z, Yan C. Research Progress on the Mechanism of the SFRP-Mediated Wnt Signalling Pathway Involved in Bone Metabolism in Osteoporosis. Mol Biotechnol 2024; 66:975-990. [PMID: 38194214 DOI: 10.1007/s12033-023-01018-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 12/01/2023] [Indexed: 01/10/2024]
Abstract
Osteoporosis (OP) is a metabolic bone disease linked to an elevated fracture risk, primarily stemming from disruptions in bone metabolism. Present clinical treatments for OP merely alleviate symptoms. Hence, there exists a pressing need to identify novel targets for the clinical treatment of OP. Research indicates that the Wnt signalling pathway is modulated by serum-secreted frizzled-related protein 5 (SFRP5), potentially serving as a pivotal regulator in bone metabolism disorders. Moreover, studies confirm elevated SFRP5 expression in OP, with SFRP5 overexpression leading to the downregulation of Wnt and β-catenin proteins in the Wnt signalling pathway, as well as the expression of osteogenesis-related marker molecules such as RUNX2, ALP, and OPN. Conversely, the opposite has been reported when SFRP5 is knocked out, suggesting that SFRP5 may be a key factor involved in the regulation of bone metabolism via the Wnt signalling axis. However, the molecular mechanisms underlying the action of SFRP5-induced OP have yet to be comprehensively elucidated. This review focusses on the molecular structure and function of SFRP5 and the potential molecular mechanisms of the SFRP5-mediated Wnt signalling pathway involved in bone metabolism in OP, providing reasonable evidence for the targeted therapy of SFRP5 for the prevention and treatment of OP.
Collapse
Affiliation(s)
- Fangyu An
- Teaching Experiment Training Center, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Jiayi Song
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Weirong Chang
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Jie Zhang
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Peng Gao
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Yujie Wang
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Zhipan Xiao
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Chunlu Yan
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China.
| |
Collapse
|
5
|
Song Y, Ma Y, Zhang K, Zhang W, Xiong G, Qi T, Shi J, Qiu H, Zhang J, Han F, Kan C, Sun X. Secreted frizzled-related protein 5: A promising therapeutic target for metabolic diseases via regulation of Wnt signaling. Biochem Biophys Res Commun 2023; 677:70-76. [PMID: 37549604 DOI: 10.1016/j.bbrc.2023.08.008] [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: 07/03/2023] [Revised: 08/01/2023] [Accepted: 08/02/2023] [Indexed: 08/09/2023]
Abstract
Metabolic diseases pose a significant global health challenge, characterized by an imbalance in metabolism and resulting in various complications. Secreted frizzled-related protein 5 (SFRP5), an adipokine known for its anti-inflammatory properties, has gained attention as a promising therapeutic target for metabolic diseases. SFRP5 acts as a key regulator in the Wnt signaling pathway, exerting its influence on critical cellular functions including proliferation, differentiation, and migration. Its significance extends to the realm of adipose tissue biology, where it plays a central role in regulating inflammation, insulin resistance, adipogenesis, lipid metabolism, glucose homeostasis, and energy balance. By inhibiting Wnt signaling, SFRP5 facilitates adipocyte growth, promotes lipid accumulation, and contributes to a decrease in oxidative metabolism. Lifestyle interventions and pharmacological treatments have shown promise in increasing SFRP5 levels and protecting against metabolic abnormalities. SFRP5 is a pivotal player in metabolic diseases and presents itself as a promising therapeutic target. An overview of SFRP5 and its involvement in metabolic disorders and metabolism is provided in this comprehensive review. By elucidating these aspects, valuable insights can be gained to foster the development of effective strategies in combating metabolic diseases.
Collapse
Affiliation(s)
- Yixin Song
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China
| | - Yujie Ma
- Department of Pathophysiology, School of Basic Medical Sciences, Weifang Medical University, Weifang, 261053, China
| | - Kexin Zhang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China
| | - Wenqiang Zhang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Department of Pathology, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China
| | - Guoji Xiong
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China
| | - Tongbing Qi
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China
| | - Junfeng Shi
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China
| | - Hongyan Qiu
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China
| | - Jingwen Zhang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China
| | - Fang Han
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Department of Pathology, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China
| | - Chengxia Kan
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China.
| | - Xiaodong Sun
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China.
| |
Collapse
|
6
|
Carena MC, Badi I, Polkinghorne M, Akoumianakis I, Psarros C, Wahome E, Kotanidis CP, Akawi N, Antonopoulos AS, Chauhan J, Sayeed R, Krasopoulos G, Srivastava V, Farid S, Walcot N, Douglas G, Channon KM, Casadei B, Antoniades C. Role of Human Epicardial Adipose Tissue-Derived miR-92a-3p in Myocardial Redox State. J Am Coll Cardiol 2023; 82:317-332. [PMID: 37468187 PMCID: PMC10368522 DOI: 10.1016/j.jacc.2023.05.031] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 05/08/2023] [Accepted: 05/09/2023] [Indexed: 07/21/2023]
Abstract
BACKGROUND Visceral obesity is directly linked to increased cardiovascular risk, including heart failure. OBJECTIVES This study explored the ability of human epicardial adipose tissue (EAT)-derived microRNAs (miRNAs) to regulate the myocardial redox state and clinical outcomes. METHODS This study screened for miRNAs expressed and released from human EAT and tested for correlations with the redox state in the adjacent myocardium in paired EAT/atrial biopsy specimens from patients undergoing cardiac surgery. Three miRNAs were then tested for causality in an in vitro model of cardiomyocytes. At a clinical level, causality/directionality were tested using genome-wide association screening, and the underlying mechanisms were explored using human biopsy specimens, as well as overexpression of the candidate miRNAs and their targets in vitro and in vivo using a transgenic mouse model. The final prognostic value of the discovered targets was tested in patients undergoing cardiac surgery, followed up for a median of 8 years. RESULTS EAT miR-92a-3p was related to lower oxidative stress in human myocardium, a finding confirmed by using genetic regulators of miR-92a-3p in the human heart and EAT. miR-92a-3p reduced nicotinamide adenine dinucleotide phosphate (NADPH)-oxidase-derived superoxide (O2.-) by targeting myocardial expression of WNT5A, which regulated Rac1-dependent activation of NADPH oxidases. Finally, high miR-92a-3p levels in EAT were independently related with lower risk of adverse cardiovascular events. CONCLUSIONS EAT-derived miRNAs exert paracrine effects on the human heart. Indeed miR-92a-3p suppresses the wingless-type MMTV integration site family, member 5a/Rac1/NADPH oxidase axis and improves the myocardial redox state. EAT-derived miR-92a-3p is related to improved clinical outcomes and is a rational therapeutic target for the prevention and treatment of obesity-related heart disease.
Collapse
Affiliation(s)
- Maria Cristina Carena
- Cardiovascular Medicine Division, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Ileana Badi
- Cardiovascular Medicine Division, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Murray Polkinghorne
- Cardiovascular Medicine Division, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Ioannis Akoumianakis
- Cardiovascular Medicine Division, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Costas Psarros
- Cardiovascular Medicine Division, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Elizabeth Wahome
- Cardiovascular Medicine Division, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Christos P Kotanidis
- Cardiovascular Medicine Division, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Nadia Akawi
- Cardiovascular Medicine Division, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom; Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Alexios S Antonopoulos
- Cardiovascular Medicine Division, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Jagat Chauhan
- Cardiovascular Medicine Division, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Rana Sayeed
- Cardiothoracic Surgery Department, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - George Krasopoulos
- Cardiothoracic Surgery Department, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Vivek Srivastava
- Cardiothoracic Surgery Department, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Shakil Farid
- Cardiothoracic Surgery Department, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Nicholas Walcot
- Cardiothoracic Surgery Department, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Gillian Douglas
- Cardiovascular Medicine Division, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Keith M Channon
- Cardiovascular Medicine Division, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom; Acute Multidisciplinary Imaging and Interventional Centre, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Barbara Casadei
- Cardiovascular Medicine Division, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Charalambos Antoniades
- Cardiovascular Medicine Division, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom; Acute Multidisciplinary Imaging and Interventional Centre, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
7
|
Kelly CJ, Chu M, Untaru R, Assadi-Khansari B, Chen D, Croft AJ, Horowitz JD, Boyle AJ, Sverdlov AL, Ngo DTM. Association of Circulating Plasma Secreted Frizzled-Related Protein 5 (Sfrp5) Levels with Cardiac Function. J Cardiovasc Dev Dis 2023; 10:274. [PMID: 37504530 PMCID: PMC10380407 DOI: 10.3390/jcdd10070274] [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: 05/02/2023] [Revised: 06/23/2023] [Accepted: 06/23/2023] [Indexed: 07/29/2023] Open
Abstract
Secreted frizzled-related protein 5 (SFRP5) is a novel anti-inflammatory adipokine that may play a role in cardiovascular development and disease. However, there is yet to be a comprehensive investigation into whether circulating SFRP5 can be a biomarker for cardiac function. Plasma SFRP5 levels were measured via ELISA in 262 patients admitted to a cardiology unit. Plasma SFRP5 levels were significantly lower in patients with a history of heart failure (HF), coronary artery disease (CAD), and atrial fibrillation (AF; p = 0.001). In univariate analyses, SFRP5 levels were also significantly positively correlated with left ventricular ejection fraction (LVEF) (r = 0.52, p < 0.001) and negatively correlated with E/E' (r = -0.30, p < 0.001). Patients with HF, CAD, low LVEF, low triglycerides, high CRP, and high eGFR were associated with lower SFRP5 levels independent of age, BMI, or diabetes after multivariate analysis (overall model r = 0.729, SE = 0.638). Our results show that low plasma SFRP5 levels are independently associated with the presence of HF, CAD, and, importantly, impaired LV function. These results suggest a potential role of SFRP5 as a biomarker, as well as a mediator of cardiac dysfunction independent of obesity and metabolic regulation.
Collapse
Affiliation(s)
- Conagh J Kelly
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan 2308, Australia
- Hunter Medical Research Institute, New Lambton Heights 2305, Australia
| | - Matthew Chu
- School of Medicine, University of Adelaide, Adelaide 5000, Australia
| | - Rossana Untaru
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan 2308, Australia
| | - Bahador Assadi-Khansari
- Hunter Medical Research Institute, New Lambton Heights 2305, Australia
- Hunter New England Local Health District, Newcastle 2305, Australia
| | - Dongqing Chen
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan 2308, Australia
- Hunter Medical Research Institute, New Lambton Heights 2305, Australia
| | - Amanda J Croft
- Hunter Medical Research Institute, New Lambton Heights 2305, Australia
- School of Medicine and Public Health, University of Newcastle, Callaghan 2308, Australia
| | - John D Horowitz
- School of Medicine, University of Adelaide, Adelaide 5000, Australia
| | - Andrew J Boyle
- Hunter Medical Research Institute, New Lambton Heights 2305, Australia
- Hunter New England Local Health District, Newcastle 2305, Australia
- School of Medicine and Public Health, University of Newcastle, Callaghan 2308, Australia
| | - Aaron L Sverdlov
- Hunter Medical Research Institute, New Lambton Heights 2305, Australia
- School of Medicine, University of Adelaide, Adelaide 5000, Australia
- Hunter New England Local Health District, Newcastle 2305, Australia
- School of Medicine and Public Health, University of Newcastle, Callaghan 2308, Australia
| | - Doan T M Ngo
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan 2308, Australia
- Hunter Medical Research Institute, New Lambton Heights 2305, Australia
- Hunter New England Local Health District, Newcastle 2305, Australia
| |
Collapse
|
8
|
Roy PK, Islam J, Lalhlenmawia H. Prospects of potential adipokines as therapeutic agents in obesity-linked atherogenic dyslipidemia and insulin resistance. Egypt Heart J 2023; 75:24. [PMID: 37014444 PMCID: PMC10073393 DOI: 10.1186/s43044-023-00352-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 03/28/2023] [Indexed: 04/05/2023] Open
Abstract
BACKGROUND In normal circumstances, AT secretes anti-inflammatory adipokines (AAKs) which regulates lipid metabolism, insulin sensitivity, vascular hemostasis, and angiogenesis. However, during obesity AT dysfunction occurs and leads to microvascular imbalance and secretes several pro-inflammatory adipokines (PAKs), thereby favoring atherogenic dyslipidemia and insulin resistance. Literature suggests decreased levels of circulating AAKs and increased levels of PAKs in obesity-linked disorders. Importantly, AAKs have been reported to play a vital role in obesity-linked metabolic disorders mainly insulin resistance, type-2 diabetes mellitus and coronary heart diseases. Interestingly, AAKs counteract the microvascular imbalance in AT and exert cardioprotection via several signaling pathways such as PI3-AKT/PKB pathway. Although literature reviews have presented a number of investigations detailing specific pathways involved in obesity-linked disorders, literature concerning AT dysfunction and AAKs remains sketchy. In view of the above, in the present contribution an effort has been made to provide an insight on the AT dysfunction and role of AAKs in modulating the obesity and obesity-linked atherogenesis and insulin resistance. MAIN BODY "Obesity-linked insulin resistance", "obesity-linked cardiometabolic disease", "anti-inflammatory adipokines", "pro-inflammatory adipokines", "adipose tissue dysfunction" and "obesity-linked microvascular dysfunction" are the keywords used for searching article. Google scholar, Google, Pubmed and Scopus were used as search engines for the articles. CONCLUSIONS This review offers an overview on the pathophysiology of obesity, management of obesity-linked disorders, and areas in need of attention such as novel therapeutic adipokines and their possible future perspectives as therapeutic agents.
Collapse
Affiliation(s)
- Probin Kr Roy
- Department of Pharmacy, Regional Institute of Paramedical and Nursing Sciences (RIPANS), Aizawl, Mizoram, 796017, India.
| | - Johirul Islam
- Coromandel International Limited, Hyderabad, Telangana, 500101, India
| | - Hauzel Lalhlenmawia
- Department of Pharmacy, Regional Institute of Paramedical and Nursing Sciences (RIPANS), Aizawl, Mizoram, 796017, India
| |
Collapse
|
9
|
Nappi F, Avtaar Singh SS. Distinctive Signs of Disease as Deterrents for the Endothelial Function: A Systematic Review. Metabolites 2023; 13:430. [PMID: 36984870 PMCID: PMC10057506 DOI: 10.3390/metabo13030430] [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: 01/31/2023] [Revised: 03/02/2023] [Accepted: 03/13/2023] [Indexed: 03/18/2023] Open
Abstract
Endothelial integrity plays a major role in homeostasis and is responsive to the numerous endogenous factors released. While its functional role in vascular tone is well described, its role in the pathophysiology of cardiovascular disease is of interest as a potential therapeutic target. We performed a systematic review to provide an overview of new therapeutic and diagnostic targets for the treatment of coronary artery disease related to endothelial dysfunction. Databases of PubMed, Ovid's version of MEDLINE, and EMBASE were interrogated with appropriate search terms. Inclusion criteria have been met by 28 studies that were included in the final systematic review. We identified inflammation, pulmonary hypertension, diabetes mellitus and Fabry disease as pathophysiological mechanisms and explored the therapeutic options related to these conditions including medications such as Canakinumab. Endothelial dysfunction has a key role in several different pathophysiological processes which can be targeted for therapeutic options. Ongoing research should be targeted at making the transition to clinical practice. Further research is also needed on understanding the amelioration of endothelial dysfunction with the use of cardiovascular medications.
Collapse
Affiliation(s)
- Francesco Nappi
- Department of Cardiac Surgery, Centre Cardiologique du Nord, 93200 Saint-Denis, France
| | | |
Collapse
|
10
|
Regulation of pleiotropic physiological roles of nitric oxide signaling. Cell Signal 2023; 101:110496. [PMID: 36252791 DOI: 10.1016/j.cellsig.2022.110496] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 10/05/2022] [Accepted: 10/11/2022] [Indexed: 11/06/2022]
Abstract
Nitric Oxide (NO) is a highly diffusible, ubiquitous signaling molecule and a free radical that is naturally synthesized by our body. The pleiotropic effects of NO in biological systems are due to its reactivity with different molecules, such as molecular oxygen (O2), superoxide anion, DNA, lipids, and proteins. There are several contradictory findings in the literature pertaining to its role in oncology. NO is a Janus-faced molecule shown to have both tumor promoting and tumoricidal effects, which depend on its concentration, duration of exposure, and location. A high concentration is shown to have cytotoxic effects by triggering apoptosis, and at a low concentration, NO promotes angiogenesis, metastasis, and tumor progression. Upregulated NO synthesis has been implicated as a causal factor in several pathophysiological conditions including cancer. This dichotomous effect makes it highly challenging to discover its true potential in cancer biology. Understanding the mechanisms by which NO acts in different cancers helps to develop NO based therapeutic strategies for cancer treatment. This review addresses the physiological role of this molecule, with a focus on its bimodal action in various types of cancers.
Collapse
|
11
|
Sahu B, Bal NC. Adipokines from white adipose tissue in regulation of whole body energy homeostasis. Biochimie 2023; 204:92-107. [PMID: 36084909 DOI: 10.1016/j.biochi.2022.09.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 08/08/2022] [Accepted: 09/01/2022] [Indexed: 02/06/2023]
Abstract
Diseases originating from altered energy homeostasis including obesity, and type 2 diabetes are rapidly increasing worldwide. Research in the last few decades on animal models and humans demonstrates that the white adipose tissue (WAT) is critical for energy balance and more than just an energy storage site. WAT orchestrates the whole-body metabolism through inter-organ crosstalk primarily mediated by cytokines named "Adipokines". The adipokines influence metabolism and fuel selection of the skeletal muscle and liver thereby fine-tuning the load on WAT itself in physiological conditions like starvation, exercise and cold. In addition, adipokine secretion is influenced by various pathological conditions like obesity, inflammation and diabetes. In this review, we have surveyed the current state of knowledge on important adipokines and their significance in regulating energy balance and metabolic diseases. Furthermore, we have summarized the interplay of pro-inflammatory and anti-inflammatory adipokines in the modulation of pathological conditions.
Collapse
Affiliation(s)
- Bijayashree Sahu
- School of Biotechnology, KIIT University, Bhubaneswar, Odisha, 751024, India.
| | - Naresh C Bal
- School of Biotechnology, KIIT University, Bhubaneswar, Odisha, 751024, India.
| |
Collapse
|
12
|
Ding N, Zheng C. Secreted frizzled-related protein 5 promotes angiogenesis of human umbilical vein endothelial cells and alleviates myocardial injury in diabetic mice with myocardial infarction by inhibiting Wnt5a/JNK signaling. Bioengineered 2022; 13:11656-11667. [PMID: 35506262 PMCID: PMC9275896 DOI: 10.1080/21655979.2022.2070964] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
The purpose of this study is to investigate whether secreted frizzled-related protein 5 (SFRP5) affects the proliferation, migration and angiogenesis of human umbilical vein endothelial cells (HUVECs) induced by high glucose (HG). HUVECs were treated with different concentrations of glucose. MTT, wound healing, angiogenesis, and ELISA assays were used to detect cell cytotoxicity, migration, tube formation, and VEGF165 and VEGF165b levels, respectively. The mice model of type 2 diabetes mellitus (T2DM) complicated with myocardial infarction (MI) was established. SFRP5 was injected intrabitoneally for 2 weeks. cardiac output (CO), left ventricular ejection fraction (LVEF) and left ventricular shortening fraction (LVSF) were detected by echocardiography. Western blot was used to detect the protein levels of SFRP5, Wnt5a, JNK1/2/3, p-JNK1/2/3, TGF-β1, Caspase3, Bax, and Bcl-2. The expression of SFRP5 was declined in HG-induced HUVECs and T2DM-MI. Intervention of SFRP5 promoted the migration of HUVECs and angiogenesis, as evidenced by a lower expression of Bax and caspase3, but a higher expression of Bcl-2. Meanwhile, SFRP5 inhibition repress Wnt5a and p-JNK expression. Howerver, The JNK inhibitor (SP600125) enhanced the down-regulation of Wnt5a/JNK pathway proteins by SFRP5. SFRP5 intervention increased the levels of CO, LVSF, and LVEF in T2DM-MI mice. SFRP5 inhibited myocardial pathological injury and fibrosis in T2DM-MI mice and SFRP5 could down-regulate Wnt5a and p-JNK1/2/3 activation. SFRP5 promotes the proliferation, migration and angiogenesis of HUVECs induced by HG, and inhibits cardiac dysfunction, pathological damage, fibrosis, and myocardial angiogenesis in diabetic myocardial ischemia mice, which is achieved by inhibiting Wnt5a/JNK signaling.
Collapse
Affiliation(s)
- Nian Ding
- Clinical College of Traditional Chinese medicine, Hubei University of Chinese Medicine, Wuhan, China.,Medical Ward, Wuhan Hospital of Traditional Chinese Medicine, Wuhan, China
| | - Chenghong Zheng
- Clinical College of Traditional Chinese medicine, Hubei University of Chinese Medicine, Wuhan, China.,Medical Ward, Wuhan Hospital of Traditional Chinese Medicine, Wuhan, China
| |
Collapse
|
13
|
Mei R, Qin W, Zheng Y, Wan Z, Liu L. Role of Adipose Tissue Derived Exosomes in Metabolic Disease. Front Endocrinol (Lausanne) 2022; 13:873865. [PMID: 35600580 PMCID: PMC9114355 DOI: 10.3389/fendo.2022.873865] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 03/30/2022] [Indexed: 12/12/2022] Open
Abstract
Adipose tissues perform physiological functions such as energy storage and endocrine, whose dysfunction will lead to severe metabolic disorders. Accumulating evidences show that exosomes can meditate communications between different tissues by transporting nucleic acids, proteins and other biological factors. More importantly, exosomes secreted by adipose tissue function as critical contributing factors that elucidate specific mechanisms in metabolic disturbance such as obesity, adipose inflammation and diabetes etc. Adipose tissue is the major source of circulating exosomal miRNAs. miRNA secreted from adipose tissues not only altered in patients with metabolic disease, but also result in an increase in metabolic organ talk. Here we have reviewed the latest progress on the role of adipose tissue derived exosomes roles in metabolic disorders. Moreover, the current obstacles hindering exosome-based therapeutic strategies have also been discussed.
Collapse
Affiliation(s)
| | | | | | - Zhuo Wan
- *Correspondence: Zhuo Wan, ; Li Liu,
| | - Li Liu
- *Correspondence: Zhuo Wan, ; Li Liu,
| |
Collapse
|
14
|
Jung HN, Jung CH. The Role of Anti-Inflammatory Adipokines in Cardiometabolic Disorders: Moving beyond Adiponectin. Int J Mol Sci 2021; 22:ijms222413529. [PMID: 34948320 PMCID: PMC8707770 DOI: 10.3390/ijms222413529] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/09/2021] [Accepted: 12/13/2021] [Indexed: 02/07/2023] Open
Abstract
The global burden of obesity has multiplied owing to its rapidly growing prevalence and obesity-related morbidity and mortality. In addition to the classic role of depositing extra energy, adipose tissue actively interferes with the metabolic balance by means of secreting bioactive compounds called adipokines. While most adipokines give rise to inflammatory conditions, the others with anti-inflammatory properties have been the novel focus of attention for the amelioration of cardiometabolic complications. This review compiles the current evidence on the roles of anti-inflammatory adipokines, namely, adiponectin, vaspin, the C1q/TNF-related protein (CTRP) family, secreted frizzled-related protein 5 (SFRP5), and omentin-1 on cardiometabolic health. Further investigations on the mechanism of action and prospective human trials may pave the way to their clinical application as innovative biomarkers and therapeutic targets for cardiovascular and metabolic disorders.
Collapse
Affiliation(s)
- Han Na Jung
- Asan Medical Center, Department of Internal Medicine, University of Ulsan College of Medicine, Seoul 05505, Korea;
- Asan Diabetes Center, Asan Medical Center, Seoul 05505, Korea
| | - Chang Hee Jung
- Asan Medical Center, Department of Internal Medicine, University of Ulsan College of Medicine, Seoul 05505, Korea;
- Asan Diabetes Center, Asan Medical Center, Seoul 05505, Korea
- Correspondence:
| |
Collapse
|
15
|
Deng D, Han X, Diao Z, Liu W. Secreted Frizzled-Related Protein 5 Ameliorates Vascular Calcification in a Rat Model of Chronic Kidney Disease through the Wnt/β-Catenin Pathway. Kidney Blood Press Res 2021; 46:758-767. [PMID: 34469882 DOI: 10.1159/000517095] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 05/07/2021] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Vascular calcification (VC) is highly prevalent and a major cardiovascular risk factor in chronic kidney disease (CKD) patients. Secreted frizzled-related protein 5 (SFRP5), an inhibitor of the Wnt pathway, is an adipokine with a positive effect on metabolic and cardiovascular diseases. Our previous in vitro study showed that SFRP5 attenuates high phosphate-induced calcification in vascular smooth muscle cells by inhibiting the Wnt/β-catenin pathway. Therefore, we hypothesized that SFRP5 may protect against CKD-associated VC (CKD-VC) through the same signalling. METHODS The rat model of CKD with VC was induced by 0.75% adenine combined with 1.8% high phosphate diet, which were administered with adenovirus vectors of SFRP5. We evaluated the SFRP5 effect on VC by von Kossa staining and calcium content analysis and osteogenic markers by immunohistochemistry and Western blot. The components of Wnt/ß-catenin signalling were also evaluated. RESULTS SFRP5 local and serum levels were significantly decreased in the CKD-VC rat model compared with the control group. Adenovirus-mediated overexpression of SFRP5 significantly inhibited VC, which was due to suppression of CKD-induced expression of calcification and osteoblastic markers. Additionally, SFRP5 abrogated activation of the Wnt/β-catenin pathway that plays a major role in the pathogenesis of VC. The specificity of SFRP5 for inhibition of VC was confirmed using an empty adenovirus as a control. CONCLUSION Our results suggest that SFRP5 ameliorates VC of CKD rats by inhibiting the expression of calcification and osteoblastic markers as well as the Wnt/β-catenin pathway. Collectively, this study suggests that SFRP5 is a potential therapeutic target in CKD-VC.
Collapse
Affiliation(s)
- Dai Deng
- Department of Nephrology, Beijing Friendship Hospital, Faculty of Kidney Diseases, Capital Medical University, Beijing, China
| | - Xue Han
- Department of Nephrology, Beijing Friendship Hospital, Faculty of Kidney Diseases, Capital Medical University, Beijing, China
| | - Zongli Diao
- Department of Nephrology, Beijing Friendship Hospital, Faculty of Kidney Diseases, Capital Medical University, Beijing, China
| | - Wenhu Liu
- Department of Nephrology, Beijing Friendship Hospital, Faculty of Kidney Diseases, Capital Medical University, Beijing, China
| |
Collapse
|
16
|
Koutaki D, Michos A, Bacopoulou F, Charmandari E. The Emerging Role of Sfrp5 and Wnt5a in the Pathogenesis of Obesity: Implications for a Healthy Diet and Lifestyle. Nutrients 2021; 13:nu13072459. [PMID: 34371968 PMCID: PMC8308727 DOI: 10.3390/nu13072459] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 07/05/2021] [Accepted: 07/09/2021] [Indexed: 01/03/2023] Open
Abstract
In recent decades, the prevalence of obesity has risen dramatically worldwide among all age groups. Obesity is characterized by excess fat accumulation and chronic low-grade inflammation. The adipose tissue functions as a metabolically active endocrine organ secreting adipokines. A novel duo of adipokines, the anti-inflammatory secreted frizzled-related protein 5 (Sfrp5) and the proinflammatory wingless type mouse mammary tumor virus (MMTV) integration site family member 5A (Wnt5a), signal via the non-canonical Wnt pathway. Recent evidence suggests that Sfpr5 and Wnt5a play a key role in the pathogenesis of obesity and its metabolic complications. This review summarizes the current knowledge on the novel regulatory system of anti-inflammatory Sfrp5 and pro-inflammatory Wnt5a, and their relation to obesity and obesity-related complications. Future studies are required to investigate the potential role of Sfrp5 and Wnt5a as biomarkers for monitoring the response to lifestyle interventions and for predicting the development of cardiometabolic risk factors. These adipokines may also serve as novel therapeutic targets for obesity-related disorders.
Collapse
Affiliation(s)
- Diamanto Koutaki
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, “Aghia Sophia” Children’s Hospital, 11527 Athens, Greece;
| | - Athanasios Michos
- Division of Infectious Diseases, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, “Aghia Sophia” Children’s Hospital, 11527 Athens, Greece;
| | - Flora Bacopoulou
- University Research Institute of Maternal and Child Health & Precision Medicine, and UNESCO Chair on Adolescent Health Care, National and Kapodistrian University of Athens, “Aghia Sophia” Children’s Hospital, 11527 Athens, Greece;
| | - Evangelia Charmandari
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, “Aghia Sophia” Children’s Hospital, 11527 Athens, Greece;
- Division of Endocrinology and Metabolism, Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
- Correspondence: ; Tel./Fax: +30-213-2013-384
| |
Collapse
|
17
|
Wnt5a promotes renal tubular inflammation in diabetic nephropathy by binding to CD146 through noncanonical Wnt signaling. Cell Death Dis 2021; 12:92. [PMID: 33462195 PMCID: PMC7814016 DOI: 10.1038/s41419-020-03377-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 12/20/2020] [Accepted: 12/21/2020] [Indexed: 12/15/2022]
Abstract
Immune and inflammatory factors have emerged as key pathophysiological mechanisms in the progression of diabetic renal injury. Noncanonical Wnt5a signaling plays an essential role in obesity- or diabetes-induced metabolic dysfunction and inflammation, but its explicit molecular mechanisms and biological function in diabetic nephropathy (DN) remain unknown. In this study, we found that the expression of Wnt5a and CD146 in the kidney and the level of soluble form of CD146 (sCD146) in serum and urine samples were upregulated in DN patients compared to controls, and this alteration was correlated with the inflammatory process and progression of renal impairment. Blocking the activation of Wnt5a signaling with the Wnt5a antagonist Box5 prevented JNK phosphorylation and high glucose-induced inflammatory responses in db/db mice and high glucose-treated HK-2 cells. Similar effects were observed by silencing Wnt5a with small-interfering RNA (siRNA) in cultured HK-2 cells. Knockdown of CD146 blocked Wnt5a-induced expression of proinflammatory cytokines and activation of JNK, which suggests that CD146 is essential for the activation of the Wnt5a pathway. Finally, we confirmed that Wnt5a directly interacted with CD146 to activate noncanonical Wnt signaling in HK-2 cells. Taken together, our findings suggest that by directly binding to CD146, Wnt5a-induced noncanonical signaling is a contributing mechanism for renal tubular inflammation in diabetic nephropathy. The concentration of sCD146 in serum and urine could be a potential biomarker to predict renal outcomes in DN patients.
Collapse
|
18
|
Wei X, Liu Q, Guo S, Wu Y. Role of Wnt5a in periodontal tissue development, maintenance, and periodontitis: Implications for periodontal regeneration (Review). Mol Med Rep 2021; 23:167. [PMID: 33398377 PMCID: PMC7821221 DOI: 10.3892/mmr.2020.11806] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 11/25/2020] [Indexed: 02/05/2023] Open
Abstract
The periodontium is a highly dynamic microenvironment constantly adapting to changing external conditions. In the processes of periodontal tissue formation and remodeling, certain molecules may serve an essential role in maintaining periodontal homeostasis. Wnt family member 5a (Wnt5a), as a member of the Wnt family, has been identified to have extensive biological roles in development and disease, predominantly through the non‑canonical Wnt signaling pathway or through interplay with the canonical Wnt signaling pathway. An increasing number of studies has also demonstrated that it serves crucial roles in periodontal tissues. Wnt5a participates in the development of periodontal tissues, maintains a non‑mineralized state of periodontal ligament, and regulates bone homeostasis. In addition, Wnt5a is involved in the pathogenesis of periodontitis. Recently, it has been shown to serve a positive role in the regeneration of integrated periodontal complex. The present review article focuses on recent research studies of Wnt5a and its functions in development, maintenance, and pathological disorders of periodontal tissues, as well as its potential effect on periodontal regeneration.
Collapse
Affiliation(s)
- Xiuqun Wei
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Qian Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Shujuan Guo
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yafei Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
19
|
Wang B, Pan Y, Yang G, Cui Z, Yu W, Liu H, Bai B. Sfrp5/Wnt5a and leptin/adiponectin levels in the serum and the periarterial adipose tissue of patients with peripheral arterial occlusive disease. Clin Biochem 2020; 87:46-51. [PMID: 33188773 DOI: 10.1016/j.clinbiochem.2020.11.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 10/15/2020] [Accepted: 11/03/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND Leptin, adiponectin, secreted frizzled-related protein 5 (Sfrp5) and wingless-type family member 5a (Wnt5a) are novel adipokines that are involved in insulin sensitivity and atherosclerosis. The aim of the present study was to investigate the serum and periarterial adipose tissue leptin/adiponectin and Sfrp5/Wnt5a levels in patients with peripheral arterial occlusive disease (PAOD). METHODS A total of 75 patients with PAOD and 39 control subjects were recruited. The serum concentrations of leptin, adiponectin, Sfrp5 and Wnt5a were measured by ELISAs, and the leptin, adiponectin, Sfrp5 and Wnt5a levels in the periarterial adipose tissue were observed by western blotting. RESULTS The serum Sfrp5 levels were significantly lower in the patients with PAOD than in the control subjects (p < 0.001) and Wnt5a levels were higher in the patients with PAOD (p < 0.001). The serum leptin levels were significantly higher in the patients with PAOD than in the control subjects (p < 0.001), and adiponectin levels were significantly lower in the patients with PAOD (p < 0.001). The serum Sfrp5 levels were associated with ABI (rs = 0.274; p = 0.018), Wnt5a (rs = -0.409; p < 0.001), adiponectin (rs = 0.244; p = 0.035) and Leptin/Adiponetin ratio (rs = -0.244; p = 0.037). The adiponectin and Sfrp5 protein levels were decreased in the periarterial adipose tissue of patients with PAOD compared with control subjects. The leptin and Wnt5a protein levels were increased in the periarterial adipose tissue of patients with PAOD compared with control subjects. CONCLUSION We demonstrated that the adiponectin and Sfrp5 levels in the serum and periarterial adipose tissue were significantly lower in the patients with PAOD than in the control subjects. The leptin and Wnt5a levels in the serum and periarterial adipose tissue were significantly higher in the patients with PAOD than in the control subjects.
Collapse
Affiliation(s)
- Biyu Wang
- The Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yu Pan
- The Department of Medical Administration, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Guang Yang
- The Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zong'ao Cui
- The Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wenping Yu
- The Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hui Liu
- The Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bing Bai
- The Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
20
|
Recinella L, Orlando G, Ferrante C, Chiavaroli A, Brunetti L, Leone S. Adipokines: New Potential Therapeutic Target for Obesity and Metabolic, Rheumatic, and Cardiovascular Diseases. Front Physiol 2020; 11:578966. [PMID: 33192583 PMCID: PMC7662468 DOI: 10.3389/fphys.2020.578966] [Citation(s) in RCA: 147] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 10/14/2020] [Indexed: 12/11/2022] Open
Abstract
Besides its role as an energy storage organ, adipose tissue can be viewed as a dynamic and complex endocrine organ, which produces and secretes several adipokines, including hormones, cytokines, extracellular matrix (ECM) proteins, and growth and vasoactive factors. A wide body of evidence showed that adipokines play a critical role in various biological and physiological functions, among which feeding modulation, inflammatory and immune function, glucose and lipid metabolism, and blood pressure control. The aim of this review is to summarize the effects of several adipokines, including leptin, diponectin, resistin, chemerin, lipocalin-2 (LCN2), vaspin, omentin, follistatin-like 1 (FSTL1), secreted protein acidic and rich in cysteine (SPARC), secreted frizzled-related protein 5 (SFRP5), C1q/TNF-related proteins (CTRPs), family with sequence similarity to 19 member A5 (FAM19A5), wingless-type inducible signaling pathway protein-1 (WISP1), progranulin (PGRN), nesfatin-1 (nesfatin), visfatin/PBEF/NAMPT, apelin, retinol binding protein 4 (RPB4), and plasminogen activator inhibitor-1 (PAI-1) in the regulation of insulin resistance and vascular function, as well as many aspects of inflammation and immunity and their potential role in managing obesity-associated diseases, including metabolic, osteoarticular, and cardiovascular diseases.
Collapse
Affiliation(s)
| | | | | | | | - Luigi Brunetti
- Department of Pharmacy, Gabriele d’Annunzio University, Chieti, Italy
| | | |
Collapse
|
21
|
Kim JA, Choi KM. Newly Discovered Adipokines: Pathophysiological Link Between Obesity and Cardiometabolic Disorders. Front Physiol 2020; 11:568800. [PMID: 32982804 PMCID: PMC7492654 DOI: 10.3389/fphys.2020.568800] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 08/14/2020] [Indexed: 12/21/2022] Open
Abstract
With the increasing prevalence of obesity, obesity-related problems such as cardiometabolic disorders (CMD), are also rapidly increasing. To prevent and alleviate the progressive course of CMD, it is important to discover the pathophysiological mechanisms between obesity and CMD. Adipose tissue is now recognized as an active endocrine organ that releases adipokines. Adipokines play a pivotal role in chronic low-grade inflammation, oxidative stress, and impaired insulin signaling, contributing to metabolic derangement and leading to CMD. Recent studies have provided substantial evidence supporting the association between adipokines and CMD. In this review, we highlight the pathophysiological action of adipokines in CMD that includes metabolic syndrome, type 2 diabetes, non-alcoholic fatty liver disease, and cardiovascular diseases. We focused on translational and clinical research of novel adipokines associated with metabolic and cardiovascular regulation. Exploration of the role of these adipokines connecting obesity and CMD may provide a perspective on adipokine-based therapeutic implications for CMD.
Collapse
Affiliation(s)
- Jung A Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Korea University, Seoul, South Korea
| | - Kyung Mook Choi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Korea University, Seoul, South Korea
| |
Collapse
|
22
|
Wu J, Zheng H, Liu X, Chen P, Zhang Y, Luo J, Kuang J, Li J, Yang Y, Ma T, Yang Y, Huang X, Liang G, Liang D, Hu Y, Wu JHY, Arnott C, Mai W, Huang Y. Prognostic Value of Secreted Frizzled-Related Protein 5 in Heart Failure Patients With and Without Type 2 Diabetes Mellitus. Circ Heart Fail 2020; 13:e007054. [PMID: 32842761 DOI: 10.1161/circheartfailure.120.007054] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 06/12/2020] [Indexed: 12/22/2022]
Abstract
BACKGROUND Patients with heart failure (HF) with diabetes mellitus have distinct biomarker profiles compared with those without diabetes mellitus. SFRP5 (secreted frizzled-related protein 5) is an anti-inflammatory adipokine with an important suppressing role on the development of type 2 diabetes mellitus (T2DM). This study aimed to evaluate the prognostic value of SFRP5 in patients with HF with and without T2DM. METHODS The study included 833 consecutive patients with HF, 312 (37.5%) of whom had T2DM. Blood samples were collected at presentation, and SFRP5 levels were measured. The primary outcome was the composite end points of first occurrence of HF rehospitalization or all-cause mortality during follow-up. RESULTS During median follow-up of 2.1 years, 335 (40.2%) patients in the cohort experienced the composite primary outcome. After adjustment for multiple risk factors, each doubling of SFRP5 level was associated with a 21% decreased risk of primary outcomes in the overall study population (P<0.001). Subgroup analyses showed that the association between level of SFPR5 and primary outcomes may be stronger in patients with T2DM (hazard ratio, 0.69 [95% CI, 0.61-0.79]) than in patients without T2DM (hazard ratio, 0.89 [95% CI, 0.79-1.01]; interaction P=0.006). Similar associations were observed when taking SFRP5 as a categorical variable. Addition of SFRP5 significantly improved discrimination and reclassification of the incident primary outcomes beyond clinical risk factors and N-terminal pro-B-type natriuretic peptide in all patients with HF and those with T2DM (all P<0.01). CONCLUSIONS SFRP5 is an independent novel biomarker for risk stratification in HF, especially in HF with T2DM.
Collapse
Affiliation(s)
- Jiandi Wu
- Department of Cardiology, Affiliated Foshan Hospital (J.W., D.L.), Southern Medical University, Foshan, China
| | - Haoxiao Zheng
- Department of Cardiology, Shunde Hospital (H.Z., X.L., J. Luo, X.H., Y. Hu, Y. Huang), Southern Medical University, Foshan, China
| | - Xinyue Liu
- Department of Cardiology, Shunde Hospital (H.Z., X.L., J. Luo, X.H., Y. Hu, Y. Huang), Southern Medical University, Foshan, China
| | - Peisong Chen
- Department of Laboratory medicine (P.C.), the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yunlong Zhang
- Key Laboratory of Neuroscience, School of Basic Medical Sciences, Guangzhou Medical University, China (Y.Z.)
| | - Jianjin Luo
- Department of Cardiology, Shunde Hospital (H.Z., X.L., J. Luo, X.H., Y. Hu, Y. Huang), Southern Medical University, Foshan, China
- Department of Cardiology, the Second Hospital of Zhaoqing, Guangdong, China (J. Luo, G.L.)
| | - Jian Kuang
- Department of Cardiology (J.K., W.M.), the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), China (J.K., W.M.)
| | - Jingwei Li
- Department of Cardiology, People's Liberation Army General Hospital, Beijing, China (J. Li)
| | - Yu Yang
- Department of Geriatrics, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China (Yu Yang)
| | - Tianyi Ma
- Department of Cardiology, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Changsha, China (T.M.)
| | - Yanhua Yang
- Department of Cardiology, Dongguan people's Hospital (Yanhua Yang), Southern Medical University, Foshan, China
| | - Xiaohui Huang
- Department of Cardiology, Shunde Hospital (H.Z., X.L., J. Luo, X.H., Y. Hu, Y. Huang), Southern Medical University, Foshan, China
| | - Guoquan Liang
- Department of Cardiology, the Second Hospital of Zhaoqing, Guangdong, China (J. Luo, G.L.)
| | - Donglian Liang
- Department of Cardiology, Affiliated Foshan Hospital (J.W., D.L.), Southern Medical University, Foshan, China
| | - Yunzhao Hu
- Department of Cardiology, Shunde Hospital (H.Z., X.L., J. Luo, X.H., Y. Hu, Y. Huang), Southern Medical University, Foshan, China
| | - Jason H Y Wu
- The George Institute for Global Health, Faculty of Medicine, University of New South Wales, Sydney, Australia (J.H.Y.W., C.A., Y. Huang)
| | - Clare Arnott
- The George Institute for Global Health, Faculty of Medicine, University of New South Wales, Sydney, Australia (J.H.Y.W., C.A., Y. Huang)
- Department of Cardiology, Royal Prince Alfred Hospital, Sydney, NSW, Australia (C.A.)
- Charles Perkins Centre, University of Sydney, NSW, Australia (C.A.)
| | - Weiyi Mai
- Department of Cardiology (J.K., W.M.), the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), China (J.K., W.M.)
| | - Yuli Huang
- Department of Cardiology, Shunde Hospital (H.Z., X.L., J. Luo, X.H., Y. Hu, Y. Huang), Southern Medical University, Foshan, China
- The George Institute for Global Health, Faculty of Medicine, University of New South Wales, Sydney, Australia (J.H.Y.W., C.A., Y. Huang)
| |
Collapse
|
23
|
Shao Y, Saredy J, Yang WY, Sun Y, Lu Y, Saaoud F, Drummer C, Johnson C, Xu K, Jiang X, Wang H, Yang X. Vascular Endothelial Cells and Innate Immunity. Arterioscler Thromb Vasc Biol 2020; 40:e138-e152. [PMID: 32459541 PMCID: PMC7263359 DOI: 10.1161/atvbaha.120.314330] [Citation(s) in RCA: 188] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In addition to the roles of endothelial cells (ECs) in physiological processes, ECs actively participate in both innate and adaptive immune responses. We previously reported that, in comparison to macrophages, a prototypic innate immune cell type, ECs have many innate immune functions that macrophages carry out, including cytokine secretion, phagocytic function, antigen presentation, pathogen-associated molecular patterns-, and danger-associated molecular patterns-sensing, proinflammatory, immune-enhancing, anti-inflammatory, immunosuppression, migration, heterogeneity, and plasticity. In this highlight, we introduce recent advances published in both ATVB and many other journals: (1) several significant characters classify ECs as novel immune cells not only in infections and allograft transplantation but also in metabolic diseases; (2) several new receptor systems including conditional danger-associated molecular pattern receptors, nonpattern receptors, and homeostasis associated molecular patterns receptors contribute to innate immune functions of ECs; (3) immunometabolism and innate immune memory determine the innate immune functions of ECs; (4) a great induction of the immune checkpoint receptors in ECs during inflammations suggests the immune tolerogenic functions of ECs; and (5) association of immune checkpoint inhibitors with cardiovascular adverse events and cardio-oncology indicates the potential contributions of ECs as innate immune cells.
Collapse
Affiliation(s)
- Ying Shao
- Centers of Inflammation, Translational & Clinical Lung Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140
| | - Jason Saredy
- Metabolic Disease Research, Cardiovascular Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140
| | - William Y. Yang
- Metabolic Disease Research, Cardiovascular Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140
| | - Yu Sun
- Centers of Inflammation, Translational & Clinical Lung Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140
| | - Yifan Lu
- Centers of Inflammation, Translational & Clinical Lung Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140
| | - Fatma Saaoud
- Centers of Inflammation, Translational & Clinical Lung Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140
| | - Charles Drummer
- Centers of Inflammation, Translational & Clinical Lung Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140
| | - Candice Johnson
- Centers of Inflammation, Translational & Clinical Lung Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140
| | - Keman Xu
- Centers of Inflammation, Translational & Clinical Lung Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140
| | - Xiaohua Jiang
- Centers of Inflammation, Translational & Clinical Lung Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140
- Metabolic Disease Research, Cardiovascular Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140
| | - Hong Wang
- Metabolic Disease Research, Cardiovascular Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140
| | - Xiaofeng Yang
- Centers of Inflammation, Translational & Clinical Lung Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140
- Metabolic Disease Research, Cardiovascular Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140
| |
Collapse
|
24
|
Oh YJ, Kim H, Kim AJ, Ro H, Chang JH, Lee HH, Chung W, Jun HS, Jung JY. Reduction of Secreted Frizzled-Related Protein 5 Drives Vascular Calcification through Wnt3a-Mediated Rho/ROCK/JNK Signaling in Chronic Kidney Disease. Int J Mol Sci 2020; 21:ijms21103539. [PMID: 32429518 PMCID: PMC7278993 DOI: 10.3390/ijms21103539] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 05/14/2020] [Accepted: 05/16/2020] [Indexed: 12/20/2022] Open
Abstract
Vascular calcification (VC) is commonly associated with bone loss in patients with chronic kidney disease (CKD). The Wingless-related integration site (Wnt) regulates osteoblast activation through canonical signaling pathways, but the common pathophysiology of these pathways during VC and bone loss has not been identified. A rat model of adenine-induced CKD with VC was used in this study. The rats were fed 0.75% adenine (2.5% protein, 0.92% phosphate) with or without intraperitoneal injection of calcitriol (0.08 µg/kg/day) for 4 weeks. Angiotensin II (3 µM)-induced VC was achieved in high phosphate medium (3 mM) through its effect on vascular smooth muscle cells (VSMCs). In an mRNA profiler polymerase chain reaction assay of the Wnt signaling pathway, secreted frizzled-related protein 5 (sFRP5) levels were significantly decreased in the CKD rat model compared with the control group. The repression of sFRP5 on VSMC trans-differentiation was mediated through Rho/Rho-associated coiled coil containing protein kinase (ROCK) and c-Jun N-terminal kinase (JNK) pathways activated by Wnt3a. In a proof of concept study conducted with patients with CKD, serum sFRP5 concentrations were significantly lower in subjects with VC than in those without VC. Our findings suggest that repression of sFRP5 is associated with VC in the CKD environment via activation of the noncanonical Wnt pathway, and thus that sFRP5 might be a novel therapeutic target for VC in CKD.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/blood
- Adaptor Proteins, Signal Transducing/genetics
- Adenine/toxicity
- Adipokines/genetics
- Adipokines/metabolism
- Animals
- Cells, Cultured
- Core Binding Factor Alpha 1 Subunit/genetics
- Core Binding Factor Alpha 1 Subunit/metabolism
- Disease Models, Animal
- Gene Expression Profiling
- Humans
- JNK Mitogen-Activated Protein Kinases/genetics
- JNK Mitogen-Activated Protein Kinases/metabolism
- Male
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Osteoblasts/drug effects
- Osteoblasts/metabolism
- Osteogenesis/drug effects
- Osteogenesis/genetics
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins/metabolism
- Rats
- Rats, Sprague-Dawley
- Renal Insufficiency, Chronic/chemically induced
- Renal Insufficiency, Chronic/genetics
- Renal Insufficiency, Chronic/metabolism
- Vascular Calcification/chemically induced
- Vascular Calcification/genetics
- Vascular Calcification/metabolism
- Wnt Signaling Pathway/drug effects
- Wnt Signaling Pathway/genetics
- rho-Associated Kinases/genetics
- rho-Associated Kinases/metabolism
Collapse
Affiliation(s)
- Yun Jung Oh
- Department of Internal Medicine, Graduate School of Medicine, Gachon University, Incheon 21936, Korea;
- Division of Nephrology, Department of Internal Medicine, Cheju Halla General Hospital, Jeju 63127, Korea
| | - Hyunsook Kim
- Division of Nephrology, Gachon Advanced Institute for Health Sciences and Technology, Incheon 21999, Korea;
| | - Ae Jin Kim
- Division of Nephrology, Department of Internal Medicine, Gil Medical Center, Incheon 21565, Korea; (A.J.K.); (H.R.); (J.H.C.); (H.H.L.); (W.C.)
- Division of Nephrology, Department of Internal Medicine, College of Medicine, Gachon University, Incheon 21565, Korea
| | - Han Ro
- Division of Nephrology, Department of Internal Medicine, Gil Medical Center, Incheon 21565, Korea; (A.J.K.); (H.R.); (J.H.C.); (H.H.L.); (W.C.)
- Division of Nephrology, Department of Internal Medicine, College of Medicine, Gachon University, Incheon 21565, Korea
| | - Jae Hyun Chang
- Division of Nephrology, Department of Internal Medicine, Gil Medical Center, Incheon 21565, Korea; (A.J.K.); (H.R.); (J.H.C.); (H.H.L.); (W.C.)
- Division of Nephrology, Department of Internal Medicine, College of Medicine, Gachon University, Incheon 21565, Korea
| | - Hyun Hee Lee
- Division of Nephrology, Department of Internal Medicine, Gil Medical Center, Incheon 21565, Korea; (A.J.K.); (H.R.); (J.H.C.); (H.H.L.); (W.C.)
- Division of Nephrology, Department of Internal Medicine, College of Medicine, Gachon University, Incheon 21565, Korea
| | - Wookyung Chung
- Division of Nephrology, Department of Internal Medicine, Gil Medical Center, Incheon 21565, Korea; (A.J.K.); (H.R.); (J.H.C.); (H.H.L.); (W.C.)
- Division of Nephrology, Department of Internal Medicine, College of Medicine, Gachon University, Incheon 21565, Korea
| | - Hee-Sook Jun
- College of Pharmacy, Gachon University, Incheon 21936, Korea;
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Korea
| | - Ji Yong Jung
- Division of Nephrology, Gachon Advanced Institute for Health Sciences and Technology, Incheon 21999, Korea;
- Division of Nephrology, Department of Internal Medicine, Gil Medical Center, Incheon 21565, Korea; (A.J.K.); (H.R.); (J.H.C.); (H.H.L.); (W.C.)
- Division of Nephrology, Department of Internal Medicine, College of Medicine, Gachon University, Incheon 21565, Korea
- Correspondence: ; Tel.: +82-32-458-2621; Fax: +82-32-460-3431
| |
Collapse
|
25
|
Tong S, Du Y, Ji Q, Dong R, Cao J, Wang Z, Li W, Zeng M, Chen H, Zhu X, Zhou Y. Expression of Sfrp5/Wnt5a in human epicardial adipose tissue and their relationship with coronary artery disease. Life Sci 2020; 245:117338. [PMID: 31981630 DOI: 10.1016/j.lfs.2020.117338] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 01/03/2020] [Accepted: 01/18/2020] [Indexed: 12/11/2022]
Abstract
Secreted frizzled-related protein 5 (Sfrp5) primarily acts in combination with wingless-type family member 5a (Wnt5a), to inhibits chronic inflammation and repress atherosclerosis and other metabolic disorders. Epicardial adipose tissue (EAT), surrounding the heart and coronary arteries, has been found to be highly related to the progression of coronary artery disease through adipokines production. However, little is known about EAT-derived Sfrp5 and Wnt5a in humans. We aimed to investigate whether the EAT-derived Sfrp5/Wnt5a levels are altered in patients with CAD. Fifty-eight patients with CAD and 29 patients without CAD who underwent cardiac surgery were enrolled. Serum samples and paired adipose biopsies from EAT and subcutaneous adipose tissue (SAT) were collected, and Sfrp5 and Wnt5a levels were detected. Correlation and multivariate regression analyses were performed to determine the relationship between Sfrp5/Wnt5a expression and CAD and other clinical risk factors. According to the results, the CAD group had lower Sfrp5 and higher Wnt5a levels in EAT and serum (all p < 0.05). Serum Sfrp5 levels were significantly lower in CAD patients with impaired myocardial function. EAT Sfrp5 mRNA levels and serum Sfrp5 levels were both negatively associated with the presence of CAD, after adjustment for known biomarkers, EAT mRNA and serum Wnt5a levels correlated positively with the presence of CAD. Thus, we concluded that low Sfrp5 and high Wnt5a levels are associated with the presence of CAD, independent of other conventional risk factors.
Collapse
Affiliation(s)
- Shan Tong
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Capital Medical University, Beijing 100029, China; Center of Geriatrics, Hainan General Hospital, Hainan 580000, China
| | - Yu Du
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Capital Medical University, Beijing 100029, China
| | - Qingwei Ji
- Emergency & Critical Care Center, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Ran Dong
- Department of Cardiac Surgery Center, 11th ward, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Jian Cao
- Department of Cardiac Surgery Center, 11th ward, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Zhijian Wang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Capital Medical University, Beijing 100029, China
| | - Wei Li
- Center of Geriatrics, Hainan General Hospital, Hainan 580000, China
| | - Min Zeng
- Center of Geriatrics, Hainan General Hospital, Hainan 580000, China
| | - Hongying Chen
- The Jackson Clinics, Physical Therapy, Middleburg, VA 20117, USA
| | - Xiaogang Zhu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Capital Medical University, Beijing 100029, China
| | - Yujie Zhou
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Capital Medical University, Beijing 100029, China.
| |
Collapse
|
26
|
Wang D, Zhang Y, Shen C. Research update on the association between SFRP5, an anti-inflammatory adipokine, with obesity, type 2 diabetes mellitus and coronary heart disease. J Cell Mol Med 2020; 24:2730-2735. [PMID: 32004418 PMCID: PMC7077606 DOI: 10.1111/jcmm.15023] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 12/18/2019] [Accepted: 01/10/2020] [Indexed: 12/15/2022] Open
Abstract
Secreted frizzled-related protein 5 (SFRP5), an anti-inflammatory adipokine secreted by adipocytes, has been demonstrated to exert its anti-inflammatory effect via antagonizing the non-canonical wingless-type family member 5A (WNT5A) signalling pathways. The WNT5A protein, as a potent pro-inflammatory signalling molecule, is strongly involved in a variety of inflammatory disorders such as obesity, type 2 diabetes mellitus (T2DM) and atherosclerosis. In this review, we systematically outlined the current understanding on the roles of SFRP5 in the pathogenesis of three inflammatory diseases including obesity, T2DM and coronary heart disease (CHD). Our review might stimulate future research using SFRP5 as a promising novel therapeutic target for the treatment of obesity, T2DM and CHD.
Collapse
Affiliation(s)
- Di Wang
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yaping Zhang
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Chengxing Shen
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
27
|
He X, Ma H. Correlation Between Circulating Levels of Secreted Frizzled-Related Protein 5 and Type 2 Diabetic Patients and Subjects with Impaired-Glucose Regulation. Diabetes Metab Syndr Obes 2020; 13:1243-1250. [PMID: 32368117 PMCID: PMC7183773 DOI: 10.2147/dmso.s242657] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 03/09/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Secreted frizzled-related protein 5 (SFRP5) is a recently identified adipokine; however, its functions during pathogenesis of T2DM and obesity remain unclear. This research attempted to investigate associations between circulating SFRP5 and obesity/T2DM. MATERIALS AND METHODS According to diagnosis, 107 patients were assigned as impaired-glucose regulation (IGR) and 111 patients newly-diagnosed as T2DM were assigned as the T2DM group. Meanwhile, 132 subjects with normal-glucose tolerance (NGT) were assigned as the NGT group. Differences in plasma SFRP5 levels among three groups were compared. Correlation between SFRP5 levels and different metabolic markers was analyzed. Multiple-linear stepwise regression analyses were performed to determine independent factors for SFRP5. Patients in the T2DM group were administrated with metformin for 12 weeks. Meanwhile, changes in plasma SFRP5 levels were also analyzed. RESULTS Plasma SFRP5 level of the IGR group was significantly lower compared to the NGT group (219.1±39.7 pg/mL vs 236.7±72.6 pg/mL, P<0.05), however, that of the T2DM group was significantly lower compared to the IGR group (203.5±42.1 pg/mL vs 219.1±39.7 pg/mL, P<0.01). Level of plasma SFRP5 was negatively correlated with fasting plasma glucose, BMI, waist circumference (WC), normalized WC (waist-to-height ratio) (WHtR), 2h plasma glucose, fasting insulin, glycosylated hemoglobin (HbA1c), fasting C-peptide, HOMA-IR, and hs-CRP (P<0.01). Among the above factors, HbA1c and fasting insulin levels (FIns) were two independent factors. Plasma SFRP5 levels were increased after 12-week metformin treatment (201.0±34.8 pg/mL vs 213.1±34.4 pg/mL, P<0.05), while insulin resistance was alleviated (ln(HOMA-IR): 1.35±0.55 vs 1.07±0.49, P<0.01). CONCLUSION Metformin reduced circulating levels of secreted frizzled-related protein 5 and improved pathophysiological parameters of T2DM.
Collapse
Affiliation(s)
- Xiaoyan He
- Special Medical Department, Beijing Jiangong Hospital, Beijing100054, People’s Republic of China
- Correspondence: Xiaoyan He Special Medical Department, Beijing Jiangong Hospital, Beijing100054, People’s Republic of China Email
| | - Huijuan Ma
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang050051, People’s Republic of China
| |
Collapse
|
28
|
Lu HS, Schmidt AM, Hegele RA, Mackman N, Rader DJ, Weber C, Daugherty A. Annual Report on Sex in Preclinical Studies: Arteriosclerosis, Thrombosis, and Vascular Biology Publications in 2018. Arterioscler Thromb Vasc Biol 2019; 40:e1-e9. [PMID: 31869272 DOI: 10.1161/atvbaha.119.313556] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Hong S Lu
- From the Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky, Lexington (H.S.L., A.D.)
| | - Ann Marie Schmidt
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University Langone Medical Center, New York, NY (A.M.S.)
| | - Robert A Hegele
- Department of Medicine and Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada (R.A.H.)
| | - Nigel Mackman
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC (N.M.)
| | - Daniel J Rader
- Departments of Medicine and Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia (D.J.R.)
| | - Christian Weber
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität (LMU) and German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany (C.W.)
| | - Alan Daugherty
- From the Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky, Lexington (H.S.L., A.D.)
| |
Collapse
|
29
|
Funcke JB, Scherer PE. Beyond adiponectin and leptin: adipose tissue-derived mediators of inter-organ communication. J Lipid Res 2019; 60:1648-1684. [PMID: 31209153 PMCID: PMC6795086 DOI: 10.1194/jlr.r094060] [Citation(s) in RCA: 203] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 06/17/2019] [Indexed: 01/10/2023] Open
Abstract
The breakthrough discoveries of leptin and adiponectin more than two decades ago led to a widespread recognition of adipose tissue as an endocrine organ. Many more adipose tissue-secreted signaling mediators (adipokines) have been identified since then, and much has been learned about how adipose tissue communicates with other organs of the body to maintain systemic homeostasis. Beyond proteins, additional factors, such as lipids, metabolites, noncoding RNAs, and extracellular vesicles (EVs), released by adipose tissue participate in this process. Here, we review the diverse signaling mediators and mechanisms adipose tissue utilizes to relay information to other organs. We discuss recently identified adipokines (proteins, lipids, and metabolites) and briefly outline the contributions of noncoding RNAs and EVs to the ever-increasing complexities of adipose tissue inter-organ communication. We conclude by reflecting on central aspects of adipokine biology, namely, the contribution of distinct adipose tissue depots and cell types to adipokine secretion, the phenomenon of adipokine resistance, and the capacity of adipose tissue to act both as a source and sink of signaling mediators.
Collapse
Affiliation(s)
- Jan-Bernd Funcke
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX
| | - Philipp E Scherer
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX
| |
Collapse
|
30
|
Tong S, Ji Q, Du Y, Zhu X, Zhu C, Zhou Y. Sfrp5/Wnt Pathway: A Protective Regulatory System in Atherosclerotic Cardiovascular Disease. J Interferon Cytokine Res 2019; 39:472-482. [PMID: 31199714 PMCID: PMC6660834 DOI: 10.1089/jir.2018.0154] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Accepted: 03/21/2019] [Indexed: 12/19/2022] Open
Abstract
Adipose tissue stores energy and is the largest endocrine organ in the body, producing several adipokines. However, among these adipokines, few play a role in the positive metabolism that promotes good health. Secreted frizzled-related protein (Sfrp)-5, an antagonist that directly binds to Wnt, has attracted interest due to its favorable effects on atherosclerotic cardiovascular disease (ASCVD). This review focuses on Sfrp5 biology and the roles of the Sfrp5/Wnt system in ASCVD.
Collapse
Affiliation(s)
- Shan Tong
- Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Beijing Institute of Heart Lung and Blood Vessel Disease, Clinical Center for Coronary Heart Disease, Capital Medical University, Beijing, China
- Department of Geriatric Medicine and Gerontology, Hainan General Hospital, Hainan, China
| | - Qingwei Ji
- Emergency and Critical Care Center, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yu Du
- Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Beijing Institute of Heart Lung and Blood Vessel Disease, Clinical Center for Coronary Heart Disease, Capital Medical University, Beijing, China
| | - Xiaogang Zhu
- Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Beijing Institute of Heart Lung and Blood Vessel Disease, Clinical Center for Coronary Heart Disease, Capital Medical University, Beijing, China
| | - Caizhong Zhu
- Department of Geriatric Medicine and Gerontology, Hainan General Hospital, Hainan, China
| | - Yujie Zhou
- Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Beijing Institute of Heart Lung and Blood Vessel Disease, Clinical Center for Coronary Heart Disease, Capital Medical University, Beijing, China
| |
Collapse
|
31
|
Adipose Tissue-Derived Signatures for Obesity and Type 2 Diabetes: Adipokines, Batokines and MicroRNAs. J Clin Med 2019; 8:jcm8060854. [PMID: 31208019 PMCID: PMC6617388 DOI: 10.3390/jcm8060854] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 06/07/2019] [Accepted: 06/11/2019] [Indexed: 12/13/2022] Open
Abstract
: Obesity is one of the main risk factors for type 2 diabetes mellitus (T2DM). It is closely related to metabolic disturbances in the adipose tissue that primarily functions as a fat reservoir. For this reason, adipose tissue is considered as the primary site for initiation and aggravation of obesity and T2DM. As a key endocrine organ, the adipose tissue communicates with other organs, such as the brain, liver, muscle, and pancreas, for the maintenance of energy homeostasis. Two different types of adipose tissues-the white adipose tissue (WAT) and brown adipose tissue (BAT)-secrete bioactive peptides and proteins, known as "adipokines" and "batokines," respectively. Some of them have beneficial anti-inflammatory effects, while others have harmful inflammatory effects. Recently, "exosomal microRNAs (miRNAs)" were identified as novel adipokines, as adipose tissue-derived exosomal miRNAs can affect other organs. In the present review, we discuss the role of adipose-derived secretory factors-adipokines, batokines, and exosomal miRNA-in obesity and T2DM. It will provide new insights into the pathophysiological mechanisms involved in disturbances of adipose-derived factors and will support the development of adipose-derived factors as potential therapeutic targets for obesity and T2DM.
Collapse
|