1
|
Marunouchi T, Matsumura K, Sato K, Takeuchi S, Murakami T, Tanonaka K. Simvastatin and rosuvastatin attenuate necroptosis in rat failing hearts following myocardial infarction; the contribution of Hsp90 inhibition in cardiomyocytes to prevent necroptosis. Biochem Pharmacol 2025; 233:116792. [PMID: 39894308 DOI: 10.1016/j.bcp.2025.116792] [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: 07/23/2024] [Revised: 12/23/2024] [Accepted: 01/30/2025] [Indexed: 02/04/2025]
Abstract
Statins, a class of dyslipidemic drugs known as HMG-CoA reductase inhibitors, have emerged as promising compounds for the treatment of chronic heart failure. Nevertheless, the precise mechanism remains to be fully elucidated. Necroptosis is a programmed necrosis-like cell death and has been involved in the development of chronic heart failure. However, it is unclear whether statins exert beneficial effects on therapy for heart failure by inhibiting cardiomyocyte necroptosis. In this study, we administered statins to rats after myocardial infarction and evaluated their effects on the necroptosis pathway in the failing heart following myocardial infarction. Administration of simvastatin or rosuvastatin reduced cardiac dysfunction after myocardial infarction. Concomitantly, the statins prevented the activation of the necroptotic intracellular signaling pathway in myocardial tissue. Simvastatin, but not rosuvastatin treatment attenuated the interaction between Hsp90 and necroptosis-related proteins in myocardial tissue, suggesting that simvastatin prevents necroptosis via Hsp90 inhibition. Necroptosis induced in primary cultured cardiomyocytes isolated from neonatal rats was inhibited by pretreatment with simvastatin. Administration of simvastatin and rosuvastatin inhibited the infiltration of M1 macrophages into myocardial tissue and reduced the myocardial tissue content of tumor necrosis factor-α, an inflammatory cytokine that induces necroptosis, respectively. The findings in the study indicate that statins may prevent necroptosis of cardiomyocytes, attenuating the development of heart failure. However, the present study suggests that the cardioprotective mechanism of simvastatin is not same as that of rosuvastatin.
Collapse
Affiliation(s)
- Tetsuro Marunouchi
- Department of Molecular and Cellular Pharmacology, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392 Japan
| | - Kasumi Matsumura
- Department of Molecular and Cellular Pharmacology, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392 Japan
| | - Kaho Sato
- Department of Molecular and Cellular Pharmacology, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392 Japan
| | - Shiori Takeuchi
- Department of Molecular and Cellular Pharmacology, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392 Japan
| | - Takuma Murakami
- Department of Molecular and Cellular Pharmacology, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392 Japan
| | - Kouichi Tanonaka
- Department of Molecular and Cellular Pharmacology, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392 Japan.
| |
Collapse
|
2
|
Parlar MA, Mutlu H, Doğantekin B, Musaoğlu İS, Albayrakoğlu ND, Yavuz ML, Özbolat ZB, Kaplan M. The Association of Statin Therapy with Liver and Pancreatic Fat Fraction in Type 2 Diabetes Mellitus. Diagnostics (Basel) 2025; 15:426. [PMID: 40002577 PMCID: PMC11854770 DOI: 10.3390/diagnostics15040426] [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: 12/26/2024] [Revised: 02/02/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: It has been shown that the use of statins in patients with type 2 diabetes mellitus (T2DM) worsens hyperglycemia and hemoglobin A1c levels but may help in the preservation of pancreatic β-cell function. The potential role of a high pancreatic fat fraction (PFF) in this process has not yet been clarified. This study aimed to investigate whether the liver fat fraction (LFF) and PFF in T2DM patients is affected by statin therapy. Methods: This cross-sectional study involved a total of 140 T2DM patients, including both those who were receiving (n = 70) and those who were not receiving (n = 70) statin therapy. The mapping of the LFF and PFF utilizing the IDEAL-IQ sequence was conducted in magnetic resonance imaging. Results: In T2DM patients who used statins, the median PFF was higher compared to those who did not use statins (8.4 vs. 6.2%, p = 0.021), while the median LFF was found to be similar (8.4 vs. 8.9, p = 0.572). Variations in PFF were associated with the use of various statins (non-statin group: 6.2 vs. atovastatin: 8.7 vs. rosuvastatin: 3.2 vs. pitavastatin: 9.2, p = 0.004). The multivariable regression analysis indicated that insulin usage decreased log(LFF) by a factor of 0.16-fold (ꞵ ± SE = -0.16 ± 0.05, p = 0.010), and rosuvastatin usage reduced log(PFF) by 0.16-fold (ꞵ ± SE = -0.16 ± 0.07, p = 0.025), irrespective of other risk factors. Furthermore, the use of atorvastatin (ꞵ ± SE = 0.17 ± 0.06, p = 0.011) and pitavastatin (ꞵ ± SE = 0.19 ± 0.07, p = 0.008) were independently associated with an increase in log(PFF). Conclusions: In patients with T2DM, statin use did not show a significant effect on the liver fat fraction, but it caused differences in the pancreatic fat fraction. The observation of a lower pancreatic fat fraction in patients taking a rosuvastatin and atorvastatin dose of 40 mg/day suggests that different types and doses of statins may have varying effects on pancreatic fat accumulation.
Collapse
Affiliation(s)
- Mehmet Akif Parlar
- Department of Internal Medicine, Sultan 2. Abdülhamid Han Training and Research Hospital, University of Health Sciences, Selimiye Neighborhood, Tıbbiye Street, 34668 Istanbul, Turkey; (H.M.); (B.D.); (İ.S.M.); (N.D.A.); (M.K.)
| | - Hakan Mutlu
- Department of Internal Medicine, Sultan 2. Abdülhamid Han Training and Research Hospital, University of Health Sciences, Selimiye Neighborhood, Tıbbiye Street, 34668 Istanbul, Turkey; (H.M.); (B.D.); (İ.S.M.); (N.D.A.); (M.K.)
| | - Betül Doğantekin
- Department of Internal Medicine, Sultan 2. Abdülhamid Han Training and Research Hospital, University of Health Sciences, Selimiye Neighborhood, Tıbbiye Street, 34668 Istanbul, Turkey; (H.M.); (B.D.); (İ.S.M.); (N.D.A.); (M.K.)
| | - İsmail Serhat Musaoğlu
- Department of Internal Medicine, Sultan 2. Abdülhamid Han Training and Research Hospital, University of Health Sciences, Selimiye Neighborhood, Tıbbiye Street, 34668 Istanbul, Turkey; (H.M.); (B.D.); (İ.S.M.); (N.D.A.); (M.K.)
| | - Nisa Demirboşnak Albayrakoğlu
- Department of Internal Medicine, Sultan 2. Abdülhamid Han Training and Research Hospital, University of Health Sciences, Selimiye Neighborhood, Tıbbiye Street, 34668 Istanbul, Turkey; (H.M.); (B.D.); (İ.S.M.); (N.D.A.); (M.K.)
| | - Mustafa Lütfi Yavuz
- Department of Cardiology, Istanbul University Faculty of Medicine, 34093 Istanbul, Turkey;
| | - Zehra Buşra Özbolat
- Deparment of Chest Diseases, Çerkezköy State Hospital, Tekirdağ Provincial Health Directorate, 59100 Tekirdağ, Turkey;
| | - Mustafa Kaplan
- Department of Internal Medicine, Sultan 2. Abdülhamid Han Training and Research Hospital, University of Health Sciences, Selimiye Neighborhood, Tıbbiye Street, 34668 Istanbul, Turkey; (H.M.); (B.D.); (İ.S.M.); (N.D.A.); (M.K.)
| |
Collapse
|
3
|
Wang Q, Hartig SM, Ballantyne CM, Wu H. The multifaceted life of macrophages in white adipose tissue: Immune shift couples with metabolic switch. Immunol Rev 2024; 324:11-24. [PMID: 38683173 PMCID: PMC11262992 DOI: 10.1111/imr.13338] [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] [Indexed: 05/01/2024]
Abstract
White adipose tissue (WAT) is a vital endocrine organ that regulates energy balance and metabolic homeostasis. In addition to fat cells, WAT harbors macrophages with distinct phenotypes that play crucial roles in immunity and metabolism. Nutrient demands cause macrophages to accumulate in WAT niches, where they remodel the microenvironment and produce beneficial or detrimental effects on systemic metabolism. Given the abundance of macrophages in WAT, this review summarizes the heterogeneity of WAT macrophages in physiological and pathological conditions, including their alterations in quantity, phenotypes, characteristics, and functions during WAT growth and development, as well as healthy or unhealthy expansion. We will discuss the interactions of macrophages with other cell partners in WAT including adipose stem cells, adipocytes, and T cells in the context of various microenvironment niches in lean or obese condition. Finally, we highlight how adipose tissue macrophages merge immunity and metabolic changes to govern energy balance for the organism.
Collapse
Affiliation(s)
- Qun Wang
- Key Laboratory of Infection and Immunity of Shandong Province, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Sean M. Hartig
- Department of Medicine, Baylor College of Medicine, Houston, Texas, USA 77030
| | | | - Huaizhu Wu
- Department of Medicine, Baylor College of Medicine, Houston, Texas, USA 77030
| |
Collapse
|
4
|
Liu W, Yang L, Di C, Sun J, Liu P, Liu H. Nonstructural Protein A238L of the African Swine Fever Virus (ASFV) Enhances Antiviral Immune Responses by Activating the TBK1-IRF3 Pathway. Vet Sci 2024; 11:252. [PMID: 38921999 PMCID: PMC11209439 DOI: 10.3390/vetsci11060252] [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: 05/15/2024] [Revised: 05/26/2024] [Accepted: 05/29/2024] [Indexed: 06/27/2024] Open
Abstract
African swine fever virus (ASFV) is a double-stranded DNA virus with an envelope. ASFV has almost the largest genome among all DNA viruses, and its mechanisms of immune evasion are complex. Better understanding of the molecular mechanisms of ASFV genes will improve vaccine design. A238L, a nonstructural protein of ASFV, inhibits NF-κB activation by suppressing the HAT activity of p300. Whether A238L also affects the transcriptional activity of IRF3 remains unexplored. Here we first confirmed the ability of A238L to suppress NF-κB-activity in L929 cells. A238L inhibits the expression of proinflammatory cytokine genes. In contrast, A238L increased the phosphorylation levels of TBK1 and IRF3 in three different cell lines. A238L increases the IRF3-driven promoter activity and induces IRF3 nuclear translocation. Furthermore, A238L enhanced innate antiviral immunity in the absence or presence of poly d (A:T) or poly (I:C) stimulation, or herpes simplex virus type 1 (HSV-1) or Sendai virus (SeV) infection. This study reveals a previously unrecognized role of A238L in promoting antiviral immune responses by TBK1-IRF3 pathway activation.
Collapse
Affiliation(s)
- Wei Liu
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (W.L.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou 225009, China
| | - Lanlan Yang
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (W.L.)
| | - Chuanyuan Di
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (W.L.)
| | - Jing Sun
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (W.L.)
| | - Penggang Liu
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (W.L.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou 225009, China
| | - Huisheng Liu
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (W.L.)
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, College of Veterinary Medicine, Chinese Academy of Agricultural Sciences, Lanzhou University, Lanzhou 730000, China
| |
Collapse
|
5
|
Priscilla L, Yoo C, Jang S, Park S, Lim G, Kim T, Lee DY. Immunotherapy targeting the obese white adipose tissue microenvironment: Focus on non-communicable diseases. Bioact Mater 2024; 35:461-476. [PMID: 38404641 PMCID: PMC10884763 DOI: 10.1016/j.bioactmat.2024.01.027] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 01/14/2024] [Accepted: 01/30/2024] [Indexed: 02/27/2024] Open
Abstract
Obesity triggers inflammatory responses in the microenvironment of white adipose tissue, resulting in chronic systemic inflammation and the subsequent development of non-communicable diseases, including type 2 diabetes, coronary heart disease, and breast cancer. Current therapy approaches for obesity-induced non-communicable diseases persist in prioritizing symptom remission while frequently overlooking the criticality of targeting and alleviating inflammation at its source. Accordingly, this review highlights the importance of the microenvironment of obese white adipose tissue and the promising potential of employing immunotherapy to target it as an effective therapeutic approach for non-communicable diseases induced by obesity. Additionally, this review discusses the challenges and offers perspective about the immunotherapy targeting the microenvironment of obese white adipose tissue.
Collapse
Affiliation(s)
- Lia Priscilla
- Department of Bioengineering, College of Engineering, and BK FOUR Biopharmaceutical Innovation Leader for Education and Research Group, Hanyang University, Seoul, 04763, Republic of Korea
| | - Chaerim Yoo
- Department of Bioengineering, College of Engineering, and BK FOUR Biopharmaceutical Innovation Leader for Education and Research Group, Hanyang University, Seoul, 04763, Republic of Korea
| | - Seonmi Jang
- Department of Bioengineering, College of Engineering, and BK FOUR Biopharmaceutical Innovation Leader for Education and Research Group, Hanyang University, Seoul, 04763, Republic of Korea
| | - Sewon Park
- Department of Bioengineering, College of Engineering, and BK FOUR Biopharmaceutical Innovation Leader for Education and Research Group, Hanyang University, Seoul, 04763, Republic of Korea
| | - Gayoung Lim
- Department of Bioengineering, College of Engineering, and BK FOUR Biopharmaceutical Innovation Leader for Education and Research Group, Hanyang University, Seoul, 04763, Republic of Korea
| | - Taekyun Kim
- Department of Bioengineering, College of Engineering, and BK FOUR Biopharmaceutical Innovation Leader for Education and Research Group, Hanyang University, Seoul, 04763, Republic of Korea
| | - Dong Yun Lee
- Department of Bioengineering, College of Engineering, and BK FOUR Biopharmaceutical Innovation Leader for Education and Research Group, Hanyang University, Seoul, 04763, Republic of Korea
- Institute of Nano Science and Technology (INST) & Institute for Bioengineering and Biopharmaceutical Research (IBBR), Hanyang University, Seoul, 04763, Republic of Korea
- Elixir Pharmatech Inc., Seoul, 07463, Republic of Korea
| |
Collapse
|
6
|
Xu F, Wang C, Tao Q, Zhang J, Zhao M, Shi S, Zhu M, Tang C, Zhang L, Zhou C, Hu C. Stent-specific fat attenuation index is associated with target vessel revascularization after PCI. Eur Radiol 2024; 34:823-832. [PMID: 37624413 DOI: 10.1007/s00330-023-10111-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 02/26/2023] [Accepted: 04/14/2023] [Indexed: 08/26/2023]
Abstract
OBJECTIVES To explore the clinical relevance of stent-specific perivascular fat attenuation index (FAI) in patients with stent implantation. METHODS A total of 162 consecutive patients who underwent coronary computed tomography angiography (CCTA) following stent implantation were retrospectively included. The stent-specific FAI at 2 cm adjacent to the stent edge was calculated. The endpoints were defined as target vessel revascularization (TVR) on the stented vessel after CCTA and readmission times due to chest pain after stent implantation. Binary logistic regression analysis for TVR and ordinal regression models were conducted to identify readmission times (0, 1, and ≥ 2) with generalized estimating equations on a per-stent basis. RESULTS On a per-stent basis, 9 stents (4.5%) experienced TVR after PCI at a median 30 months' follow-up duration. Stent-specific FAI differed significantly among subgroups of patients with stent implantation and different readmission times (p = 0.002); patients with at least one readmission had higher stent-specific FAI than those without readmission (p < 0.001). Bifurcated stents (odds ratio [OR]: 11.192, p = 0.001) and stent-specific FAI (OR: 1.189, p = 0.04) were independently associated with TVR. With no readmission as a reference, stent-specific FAI (OR: 0.984, p = 0.007) was an independent predictor for hospital readmission times ≥ 2 (p = 0.003). CONCLUSION Non-invasive stent-specific FAI derived from CCTA was found to be associated with TVR, which was a promising imaging marker for functional assessment in patients who underwent stent implantation. CLINICAL RELEVANCE STATEMENT Noninvasive fat attenuation index adjacent to the stents edge derived from CCTA, an imaging marker reflecting the presence of inflammation acting on the neointimal tissue at the sites of coronary stenting, might be relevant clinically with target vessel revascularization. KEY POINTS • Non-invasive stent-specific FAI derived from CCTA was associated with TVR (OR: 1.189 [95% CI: 1.007-1.043], p = 0.04) in patients who underwent stent implantation. • Stent-specific FAI significantly differed among a subgroup of patients with chest pain after stent implantation and with different readmission times (p = 0.002); the patients with at least one readmission had higher stent-specific FAI than those without readmission (p < 0.001). • Non-invasive stent-specific FAI derived from CCTA could be used as an imaging maker for the functional assessment of patients following stent implantation.
Collapse
Affiliation(s)
- Feng Xu
- Department of Radiology, the First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
- Department of Medical Imaging, the Affiliated Suqian First People's Hospital of Nanjing Medical University, Suqian, 223800, Jiangsu, China
| | - Chengcheng Wang
- Department of Medical Imaging, the Affiliated Suqian First People's Hospital of Nanjing Medical University, Suqian, 223800, Jiangsu, China
| | - Qing Tao
- Department of Radiology, the First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Jian Zhang
- Department of Medical Imaging, the Affiliated Suqian First People's Hospital of Nanjing Medical University, Suqian, 223800, Jiangsu, China
| | - Mingming Zhao
- Department of Medical Imaging, the Affiliated Suqian First People's Hospital of Nanjing Medical University, Suqian, 223800, Jiangsu, China
| | - Shiwei Shi
- Department of Medical Imaging, the Affiliated Suqian First People's Hospital of Nanjing Medical University, Suqian, 223800, Jiangsu, China
| | - Mengmeng Zhu
- Department of Medical Imaging, the Affiliated Suqian First People's Hospital of Nanjing Medical University, Suqian, 223800, Jiangsu, China
| | - Chunxiang Tang
- Department of Radiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, China
| | - Longjiang Zhang
- Department of Radiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, China
| | - Changsheng Zhou
- Department of Radiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, China.
| | - Chunhong Hu
- Department of Radiology, the First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China.
- Institute of Medical Imaging, Soochow University, Jiangsu Province, Suzhou, 215006, China.
| |
Collapse
|
7
|
Song Y, Tan Y, Deng M, Shan W, Zheng W, Zhang B, Cui J, Feng L, Shi L, Zhang M, Liu Y, Sun Y, Yi W. Epicardial adipose tissue, metabolic disorders, and cardiovascular diseases: recent advances classified by research methodologies. MedComm (Beijing) 2023; 4:e413. [PMID: 37881786 PMCID: PMC10594046 DOI: 10.1002/mco2.413] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 09/12/2023] [Accepted: 09/25/2023] [Indexed: 10/27/2023] Open
Abstract
Epicardial adipose tissue (EAT) is located between the myocardium and visceral pericardium. The unique anatomy and physiology of the EAT determines its great potential in locally influencing adjacent tissues such as the myocardium and coronary arteries. Classified by research methodologies, this study reviews the latest research progress on the role of EAT in cardiovascular diseases (CVDs), particularly in patients with metabolic disorders. Studies based on imaging techniques demonstrated that increased EAT amount in patients with metabolic disorders is associated with higher risk of CVDs and increased mortality. Then, in-depth profiling studies indicate that remodeled EAT may serve as a local mediator of the deleterious effects of cardiometabolic conditions and plays a crucial role in CVDs. Further, in vitro coculture studies provided preliminary evidence that the paracrine effect of remodeled EAT on adjacent cardiomyocytes can promote the occurrence and progression of CVDs. Considering the important role of EAT in CVDs, targeting EAT might be a potential strategy to reduce cardiovascular risks. Several interventions have been proved effective in reducing EAT amount. Our review provides valuable insights of the relationship between EAT, metabolic disorders, and CVDs, as well as an overview of the methodological constructs of EAT-related studies.
Collapse
Affiliation(s)
- Yujie Song
- Department of Cardiovascular SurgeryXijing HospitalThe Fourth Military Medical UniversityXi'anChina
| | - Yanzhen Tan
- Department of Cardiovascular SurgeryXijing HospitalThe Fourth Military Medical UniversityXi'anChina
| | - Meng Deng
- Department of General MedicineXijing HospitalThe Fourth Military Medical UniversityXi'anChina
| | - Wenju Shan
- Department of General MedicineXijing HospitalThe Fourth Military Medical UniversityXi'anChina
| | - Wenying Zheng
- Department of Cardiovascular SurgeryXijing HospitalThe Fourth Military Medical UniversityXi'anChina
| | - Bing Zhang
- Department of Cardiovascular SurgeryXijing HospitalThe Fourth Military Medical UniversityXi'anChina
| | - Jun Cui
- Department of Cardiovascular SurgeryXijing HospitalThe Fourth Military Medical UniversityXi'anChina
| | - Lele Feng
- Department of Cardiovascular SurgeryXijing HospitalThe Fourth Military Medical UniversityXi'anChina
| | - Lei Shi
- Department of Cardiovascular SurgeryXijing HospitalThe Fourth Military Medical UniversityXi'anChina
| | - Miao Zhang
- Department of Cardiovascular SurgeryXijing HospitalThe Fourth Military Medical UniversityXi'anChina
| | - Yingying Liu
- Department of Cardiovascular SurgeryXijing HospitalThe Fourth Military Medical UniversityXi'anChina
| | - Yang Sun
- Department of General MedicineXijing HospitalThe Fourth Military Medical UniversityXi'anChina
| | - Wei Yi
- Department of Cardiovascular SurgeryXijing HospitalThe Fourth Military Medical UniversityXi'anChina
| |
Collapse
|
8
|
Christensen IB, Blom I, Dohlmann TL, Finger F, Helge JW, Gerhart-Hines Z, Dela F, Larsen S. Effect of Simvastatin Treatment on Mitochondrial Function and Inflammatory Status of Human White Adipose Tissue. J Clin Endocrinol Metab 2023; 108:e916-e922. [PMID: 37161534 DOI: 10.1210/clinem/dgad259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 02/20/2023] [Accepted: 05/08/2023] [Indexed: 05/11/2023]
Abstract
BACKGROUND Statin therapy has shown pleiotropic effects affecting both mitochondrial function and inflammatory status. However, few studies have investigated the concurrent effects of statin exposure on mitochondrial function and inflammatory status in human subcutaneous white adipose tissue. OBJECTIVES In a cross-sectional study, we investigated the effects of simvastatin on mitochondrial function and inflammatory status in subcutaneous white adipose tissue of 55 human participants: 38 patients (19 females/19 males) in primary prevention with simvastatin (> 40 mg/d, > 3 mo) and 17 controls (9 females/8 males) with elevated plasma cholesterol. The 2 groups were matched on age, body mass index, and maximal oxygen consumption. METHODS Anthropometrics and fasting biochemical characteristics were measured. Mitochondrial respiratory capacity was assessed in white adipose tissue by high-resolution respirometry. Subcutaneous white adipose tissue expression of the inflammatory markers IL-6, chemokine (C-C motif) ligand 2 (CCL2), CCL-5, tumor necrosis factor-α, IL-10, and IL-4 was analyzed by quantitative PCR. RESULTS Simvastatin-treated patients showed lower plasma cholesterol (P < .0001), low-density lipoprotein (P < .0001), and triglyceride levels (P = .0116) than controls. Simvastatin-treated patients had a lower oxidative phosphorylation capacity of mitochondrial complex II (P = .0001 when normalized to wet weight, P < .0001 when normalized to citrate synthase activity [intrinsic]), and a lower intrinsic mitochondrial electron transport system capacity (P = .0004). Simvastatin-treated patients showed higher IL-6 expression than controls (P = .0202). CONCLUSION Simvastatin treatment was linked to mitochondrial respiratory capacity in human subcutaneous white adipose tissue, but no clear link was found between statin exposure, respiratory changes, and inflammatory status of adipose tissue.
Collapse
Affiliation(s)
- Ida Bager Christensen
- Xlab, Center for Healthy Aging, Department of Biomedical Sciences, Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Ida Blom
- Xlab, Center for Healthy Aging, Department of Biomedical Sciences, Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Tine Lovsø Dohlmann
- Xlab, Center for Healthy Aging, Department of Biomedical Sciences, Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
- Department of Epidemiology Research, Statens Serum Institut, 2300 Copenhagen S, Denmark
| | - Fabian Finger
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Jørn W Helge
- Xlab, Center for Healthy Aging, Department of Biomedical Sciences, Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Zachary Gerhart-Hines
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Flemming Dela
- Xlab, Center for Healthy Aging, Department of Biomedical Sciences, Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
- Department of Geriatrics, Bispebjerg-Frederiksberg University Hospital, 2400 Copenhagen NV, Denmark
| | - Steen Larsen
- Xlab, Center for Healthy Aging, Department of Biomedical Sciences, Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
- Clinical Research Centre, Medical University of Bialystok, 15-089 Białystok, Poland
| |
Collapse
|
9
|
Qi M, Janssen I, Barinas-Mitchell E, Budoff M, Brooks MM, Karlamangla AS, Derby CA, Chang CCH, Shields KJ, El Khoudary SR. The quantity and quality of cardiovascular fat at mid-life and future cognitive performance among women: The SWAN cardiovascular fat ancillary study. Alzheimers Dement 2023; 19:4073-4083. [PMID: 37212597 PMCID: PMC11221406 DOI: 10.1002/alz.13133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 04/10/2023] [Accepted: 04/12/2023] [Indexed: 05/23/2023]
Abstract
INTRODUCTION Cardiovascular fat is a novel risk factor that may link to dementia. Fat volume and radiodensity are measurements of fat quantity and quality, respectively. Importantly, high fat radiodensity could indicate healthy or adverse metabolic processes. METHODS The associations of cardiovascular fat (including epicardial, paracardial, and thoracic perivascular adipose tissue [PVAT]) quantity and quality assessed at mean age of 51 with subsequent cognitive performance measured repeatedly over 16 years of follow-up were examined using mixed models among 531 women. RESULTS Higher thoracic PVAT volume was associated with a higher future episodic memory (β[standard error (SE)] = 0.08 [0.04], P = 0.033), while higher thoracic PVAT radiodensity with lower future episodic (β[SE] = -0.06 [0.03], P = 0.045) and working (β[SE] = -0.24 [0.08], P = 0.003) memories. The latter association is prominent at higher volume of thoracic PVAT. DISCUSSION Mid-life thoracic PVAT may have a distinct contribution to future cognition possibly due to its distinct adipose tissue type (brown fat) and anatomical proximity to the brain circulation. HIGHLIGHTS Higher mid-life thoracic perivascular adipose tissue (thoracic PVAT) volume is related to a better future episodic memory in women. Higher mid-life thoracic PVAT radiodensity is related to worse future working and episodic memories. Negative association of high thoracic PVAT radiodensity with working memory is prominent at higher thoracic PVAT volume. Mid-life thoracic PVAT is linked to future memory loss, an early sign of Alzheimer's disease. Mid-life women's epicardial and paracardial fat are not related to future cognition.
Collapse
Affiliation(s)
- Meiyuzhen Qi
- University of Pittsburgh School of Public Health, Department of Epidemiology, Pittsburgh, PA, 15261, USA
| | - Imke Janssen
- Rush University Rush Medical Center, Department of Preventive Medicine, Chicago, IL, 60612, USA
| | - Emma Barinas-Mitchell
- University of Pittsburgh School of Public Health, Department of Epidemiology, Pittsburgh, PA, 15261, USA
| | - Matthew Budoff
- Harbor-UCLA Medical Center, Lundquist Institute, Torrance, CA, 90509, USA
| | - Maria M. Brooks
- University of Pittsburgh School of Public Health, Department of Epidemiology, Pittsburgh, PA, 15261, USA
| | - Arun S. Karlamangla
- University of California Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Carol A. Derby
- Albert Einstein College of Medicine Department of Neurology and Department of Epidemiology & Public Health, Bronx, NY, 10461, USA
| | - Chung-Chou H. Chang
- University of Pittsburgh School of Public Health, Department of Epidemiology, Pittsburgh, PA, 15261, USA
| | | | - Samar R. El Khoudary
- University of Pittsburgh School of Public Health, Department of Epidemiology, Pittsburgh, PA, 15261, USA
| |
Collapse
|
10
|
González-Ibáñez L, Meneses ME, Sánchez-Tapia M, Pérez-Luna D, Torres N, Torre-Villalvazo I, Bonilla M, Petlacalco B, Castillo I, López-Barradas A, Macías A, Tovar AR, Martínez-Carrera D. Edible and medicinal mushrooms ( Pleurotus ostreatus, Ustilago maydis, Ganoderma lucidum) reduce endoplasmic reticulum stress and inflammation in adipose tissue of obese Wistar rats fed with a high fat plus saccharose diet. Food Funct 2023. [PMID: 37161495 DOI: 10.1039/d3fo00089c] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Obesity is an increasing global public health problem. A strategy to treat obesity is the use of functional foods. Edible and medicinal mushrooms contain diverse bioactive compounds showing important antihyperlipidemic, antioxidant, and prebiotic properties. We analysed the effects of adding (10%) of Pleurotus ostreatus (Po, basidiomata), Ganoderma lucidum (Gl, basidiomata), or Ustilago maydis (Um, galls), milled, to a high fat plus saccharose diet (HFD + S) for 6 months in a model of obesity with Wistar rats. We assessed weight gain, body composition, lipid parameters, endoplasmic reticulum stress (proteins and inflammatory markers: BiP, XBP-1, JNK, p-JNK, TNF-α), and adiponectin in subcutaneous adipose tissue (SAT). The consumption of edible and medicinal mushrooms decreased weight gain (-17.2-30.1%) and fat mass (-23.7-43.1%), maintained fat-free mass, reduced levels of serum biochemical parameters (TC: -40.1-44.1%, TG: -37.7-51.6%, LDL-C: -64.5-71.1%), and prevented adipocyte hypertrophy (-30.9-36.9%) and collagen deposition (-70.9-73.7%) in SAT. Compared with the HFD + S group, mushroom consumption by Wistar rats significantly reduced the expression of proteins associated with endoplasmic reticulum stress and inflammation (BiP: -72.2-88.2%; XBP-1: -71.5-81.8%; JNK: -71.2-90.0%; p-JNK: -37.3-81.0%; TNF-α: -80.7-91.5%), whereas significantly increased adiponectin protein expression (246.4-654.2%) in SAT. These effects outperformed those obtained through the commercial lipid-lowering drug atorvastatin, contributing synergistically to prevent further obesity-related dysfunctions, such as insulin resistance derived from inflammation and ER stress in adipose tissue. Bioactive compounds from edible, functional and medicinal mushrooms represent new emerging therapies for obesity treatments using natural products.
Collapse
Affiliation(s)
- Laura González-Ibáñez
- Centro de Biotecnología de Hongos Comestibles, Funcionales y Medicinales (CB-HCFM), Campus Puebla, Colegio de Postgraduados (CP), Boulevard Forjadores de Puebla no. 205, Puebla 72760, Mexico.
| | - María E Meneses
- Centro de Biotecnología de Hongos Comestibles, Funcionales y Medicinales (CB-HCFM), Campus Puebla, Colegio de Postgraduados (CP), Boulevard Forjadores de Puebla no. 205, Puebla 72760, Mexico.
- CONACYT-Colegio de Postgraduados (CP), Campus Puebla, Boulevard Forjadores de Puebla 205, Puebla 72760, Mexico
| | - Mónica Sánchez-Tapia
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Mexico City 14080, Mexico
| | - Daniel Pérez-Luna
- Centro de Biotecnología de Hongos Comestibles, Funcionales y Medicinales (CB-HCFM), Campus Puebla, Colegio de Postgraduados (CP), Boulevard Forjadores de Puebla no. 205, Puebla 72760, Mexico.
| | - Nimbe Torres
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Mexico City 14080, Mexico
| | - Iván Torre-Villalvazo
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Mexico City 14080, Mexico
| | - Myrna Bonilla
- Centro de Biotecnología de Hongos Comestibles, Funcionales y Medicinales (CB-HCFM), Campus Puebla, Colegio de Postgraduados (CP), Boulevard Forjadores de Puebla no. 205, Puebla 72760, Mexico.
| | - Beatriz Petlacalco
- Centro de Biotecnología de Hongos Comestibles, Funcionales y Medicinales (CB-HCFM), Campus Puebla, Colegio de Postgraduados (CP), Boulevard Forjadores de Puebla no. 205, Puebla 72760, Mexico.
| | - Ivan Castillo
- Centro de Biotecnología de Hongos Comestibles, Funcionales y Medicinales (CB-HCFM), Campus Puebla, Colegio de Postgraduados (CP), Boulevard Forjadores de Puebla no. 205, Puebla 72760, Mexico.
| | - Adriana López-Barradas
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Mexico City 14080, Mexico
| | - Antonio Macías
- Centro de Biotecnología de Hongos Comestibles, Funcionales y Medicinales (CB-HCFM), Campus Puebla, Colegio de Postgraduados (CP), Boulevard Forjadores de Puebla no. 205, Puebla 72760, Mexico.
| | - Armando R Tovar
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Mexico City 14080, Mexico
| | - Daniel Martínez-Carrera
- Centro de Biotecnología de Hongos Comestibles, Funcionales y Medicinales (CB-HCFM), Campus Puebla, Colegio de Postgraduados (CP), Boulevard Forjadores de Puebla no. 205, Puebla 72760, Mexico.
| |
Collapse
|
11
|
Furuta K, Tang X, Islam S, Tapia A, Chen ZB, Ibrahim SH. Endotheliopathy in the metabolic syndrome: Mechanisms and clinical implications. Pharmacol Ther 2023; 244:108372. [PMID: 36894027 PMCID: PMC10084912 DOI: 10.1016/j.pharmthera.2023.108372] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/25/2023] [Accepted: 02/28/2023] [Indexed: 03/09/2023]
Abstract
The increasing prevalence of the metabolic syndrome (MetS) is a threat to global public health due to its lethal complications. Nonalcoholic fatty liver disease (NAFLD) is the hepatic manifestation of the MetS characterized by hepatic steatosis, which is potentially progressive to the inflammatory and fibrotic nonalcoholic steatohepatitis (NASH). The adipose tissue (AT) is also a major metabolic organ responsible for the regulation of whole-body energy homeostasis, and thereby highly involved in the pathogenesis of the MetS. Recent studies suggest that endothelial cells (ECs) in the liver and AT are not just inert conduits but also crucial mediators in various biological processes via the interaction with other cell types in the microenvironment both under physiological and pathological conditions. Herein, we highlight the current knowledge of the role of the specialized liver sinusoidal endothelial cells (LSECs) in NAFLD pathophysiology. Next, we discuss the processes through which AT EC dysfunction leads to MetS progression, with a focus on inflammation and angiogenesis in the AT as well as on endothelial-to-mesenchymal transition of AT-ECs. In addition, we touch upon the function of ECs residing in other metabolic organs including the pancreatic islet and the gut, the dysregulation of which may also contribute to the MetS. Finally, we highlight potential EC-based therapeutic targets for human MetS, and NASH based on recent achievements in basic and clinical research and discuss how to approach unsolved problems in the field.
Collapse
Affiliation(s)
- Kunimaro Furuta
- Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, MN, USA; Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Xiaofang Tang
- Department of Diabetes Complications & Metabolism, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Shahidul Islam
- Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Alonso Tapia
- Department of Diabetes Complications & Metabolism, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Zhen Bouman Chen
- Department of Diabetes Complications & Metabolism, City of Hope Comprehensive Cancer Center, Duarte, CA, USA.
| | - Samar H Ibrahim
- Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, MN, USA; Division of Pediatric Gastroenterology & Hepatology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
12
|
Zivkovic S, Maric G, Cvetinovic N, Lepojevic-Stefanovic D, Bozic Cvijan B. Anti-Inflammatory Effects of Lipid-Lowering Drugs and Supplements-A Narrative Review. Nutrients 2023; 15:nu15061517. [PMID: 36986246 PMCID: PMC10053759 DOI: 10.3390/nu15061517] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/15/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023] Open
Abstract
Cardiovascular diseases (CVD) are the leading cause of death worldwide. Since the establishment of the "lipid hypothesis", according to which, cholesterol level is directly correlated to the risk of CVD, many different lipid-lowering agents have been introduced in clinical practice. A majority of these drugs, in addition to their lipid-lowering properties, may also exhibit some anti-inflammatory and immunomodulatory activities. This hypothesis was based on the observation that a decrease in lipid levels occurs along with a decrease in inflammation. Insufficient reduction in the inflammation during treatment with lipid-lowering drugs could be one of the explanations for treatment failure and recurrent CVD events. Thus, the aim of this narrative review was to evaluate the anti-inflammatory properties of currently available lipid-lowering medications including statins, ezetimibe, bile acid sequestrants (BAS), proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors, fibrates, omega-3 fatty acids, and niacin, as well as dietary supplements and novel drugs used in modern times.
Collapse
Affiliation(s)
- Stefan Zivkovic
- Department of Cardiovascular Disease, Zvezdara University Medical Center, 11000 Belgrade, Serbia
| | - Gorica Maric
- Faculty of Medicine, Institute of Epidemiology, University of Belgrade, Dr. Subotica 8, 11000 Belgrade, Serbia
| | - Natasa Cvetinovic
- Department of Cardiovascular Disease, University Medical Center "Dr Dragisa Misovic-Dedinje", 11000 Belgrade, Serbia
| | | | - Bojana Bozic Cvijan
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| |
Collapse
|
13
|
Kamal AHM, Chakrabarty JK, Chowdhury SM. Lipopolysaccharide and statin-mediated immune-responsive protein networks revealed in macrophages through affinity purification spacer-arm controlled cross-linking (AP-SPACC) proteomics. Mol Omics 2023; 19:48-59. [PMID: 36377691 DOI: 10.1039/d2mo00224h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Toll-like receptor 4 (TLR4), a pattern recognition receptor, is activated by lipopolysaccharides (LPS) and induces the MyD88 pathway, which subsequently produces pro-inflammatory cytokines through activation of transcriptional nuclear factor (NF)-κB. Statins have been widely prescribed to reduce cholesterol synthesis for patients with cardiovascular disease. Statins may have pleiotropic effects, which include anti- and pro-inflammatory effects on cells. The molecular mechanism of the sequential influence of LPS and statin on the innate immune system remains unknown. We employed affinity purification-spacer-arm controlled cross-linking (AP-SPACC) MS-based proteomics analysis to identify the LPS- and statin-LPS-responsive proteins and their networks. LPS-stimulated RAW 264.7 macrophage cells singly and combined with the drug statin used in this study. Two chemical cross-linkers with different spacer chain lengths were utilized to stabilize the weak and transient interactors. Proteomic analysis identified 1631 differentially expressed proteins. We identified 151 immune-response proteins through functional enrichment analysis and visualized their interaction networks. Selected candidate protein-coding genes were validated, specifically squamous cell carcinoma antigens recognized by T cells 3, sphingosine-1-phosphate lyase 1, Ras-related protein Rab-35, and tumor protein D52 protein-coding genes through transcript-level expression analysis. The expressions of those genes were significantly increased upon statin treatment and decreased in LPS-stimulated macrophage cells. Therefore, we presumed that the expression changes of genes occurred due to immune response during activation of inflammation. These results highlight the immune-responsive proteins network, providing a new platform for novel investigations and discovering future therapeutic targets for inflammatory diseases.
Collapse
Affiliation(s)
- Abu Hena Mostafa Kamal
- Department of Chemistry and Biochemistry, University of Texas at Arlington, TX, 76019, USA. .,Advanced Technology Cores, Dan L Duncan Comprehensive Cancer Center, Metabolomics Core, Baylor College of Medicine, Houston, TX, 77030, USA.,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jayanta K Chakrabarty
- Department of Chemistry and Biochemistry, University of Texas at Arlington, TX, 76019, USA. .,Quantitative Proteomics and Metabolomics Center, Columbia University, New York, NY, 10027, USA
| | - Saiful M Chowdhury
- Department of Chemistry and Biochemistry, University of Texas at Arlington, TX, 76019, USA.
| |
Collapse
|
14
|
Afrin S, El Sabah M, Manzoor A, Miyashita-Ishiwata M, Reschke L, Borahay MA. Adipocyte coculture induces a pro-inflammatory, fibrotic, angiogenic, and proliferative microenvironment in uterine leiomyoma cells. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166564. [PMID: 36181981 PMCID: PMC9719372 DOI: 10.1016/j.bbadis.2022.166564] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 09/13/2022] [Accepted: 09/23/2022] [Indexed: 10/14/2022]
Abstract
OBJECTIVE Obesity and its consequences are among the biggest challenges facing the healthcare system. Uterine leiomyomas are the most common gynecologic tumors. The risk of leiomyoma increases with obesity, but the underlying mechanisms of this association remain unclear. The aim of the present study to determine the cellular and molecular mechanisms by which adipocyte contributes to both leiomyoma tumor initiation and promotion. METHODS Primary myometrium and leiomyoma cells were isolated from patients who underwent a hysterectomy or myomectomy. Pro-inflammatory, fibrotic, and angiogenic factors were measured using a multiplex cytokine array in human primary and immortalized myometrial and leiomyoma cells cocultured with human adipocyte (SW872) cells, or in animal ELT3 leiomyoma cells cocultured with 3 T3-L1 adipocytes. The free fatty acids (FFAs) and fatty acid-binding protein 4 (FABP4) levels were measured using immunofluorescence assays. Other protein abundances were determined using western blots. The expression levels of TNF-α, MCP-1, phospho-NF-κB, TGFβ3 and VEGF-A in lean and obese in different leiomyoma patients were determined by immunofluorescence staining. RESULTS Adipocytes promote inflammation, fibrosis, and angiogenesis in uterine leiomyoma cells by upregulating associated factors, such as IL-1β, TNF-α, MCP-1, GM-CSF, TGF-βs, PLGF, VEGF, HB-EGF, G-CSF and FGF2. Coculture led to the transfer of FFAs and FABP4 from adipocytes to leiomyoma cells, suggesting that adipocytes may modulate metabolic activity in these tumor cells. Increased levels of FFA and FABP4 expressions were detected in obese leiomyoma tissue compared to lean. The adipocyte-leiomyoma cell interaction increased the phospho-NF-κB level, which plays a key role in inflammation, restructuring metabolic pathways, and angiogenesis. Obese leiomyoma patients expressed a higher amount of TNF-α, MCP-1, phospho-NF-κB, TGFβ3 and VEGF-A than lean leiomyoma patients, consistent with in vitro findings. Furthermore, we found that adipocyte secretory factors enhance leiomyoma cell proliferation by increasing PCNA abundance. Finally, the inhibition of the inflammatory factors TNF-α, MCP-1, and NF-κB abrogated the adipocyte coculture-induced proliferation of leiomyoma cells. CONCLUSIONS Adipocytes release inflammatory, fibrotic, and angiogenic factors, along with FFAs, which contribute to a tumor-friendly microenvironment that may promote leiomyoma growth and can represent a new target for leiomyoma prevention and treatment.
Collapse
Affiliation(s)
- Sadia Afrin
- Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Malak El Sabah
- Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ahmed Manzoor
- School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Mariko Miyashita-Ishiwata
- Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Lauren Reschke
- Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Mostafa A Borahay
- Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
15
|
Ako S, Teper Y, Ye L, Sinnett-Smith J, Hines OJ, Rozengurt E, Eibl G. Statins Inhibit Inflammatory Cytokine Production by Macrophages and Acinar-to-Ductal Metaplasia of Pancreatic Cells. GASTRO HEP ADVANCES 2022; 1:640-651. [PMID: 36313271 PMCID: PMC9615480 DOI: 10.1016/j.gastha.2022.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 04/15/2022] [Indexed: 11/21/2022]
Abstract
BACKGROUND AND AIMS Animal data show that the presence of an oncogenic Kras mutation in pancreatic acinar cells leads to acinar-to-ductal metaplasia (ADM), pancreatic intraepithelial neoplasia (PanIN), and pancreatic ductal adenocarcinoma (PDAC). Inflammatory macrophages play an important role in the formation of ADMs and transition to PanINs. Epidemiologically, statins are associated with a reduced risk of PDAC. We investigated whether statins inhibit inflammatory cytokine production in macrophages and whether this leads to reduced ADM formation. METHODS The efficacy of statins on inflammatory cytokine production in 2 macrophage cell lines was measured by real-time polymerase chain reaction and enzyme-linked immunosorbent assay. The effect of macrophage-conditioned medium on ADM in primary pancreatic acinar cells was investigated. Mouse pancreatic tissue samples were analyzed for macrophage numbers, cytokine levels, and neoplastic/dysplastic area. RESULTS Lipophilic statins prevented inflammatory cytokine production in Raw264.7 and J774A.1 cells stimulated by lipopolysaccharide. The inhibitory effect of statins was mediated by inhibition of mevalonate and geranylgeranyl pyrophosphate synthesis and disruption of the actin cytoskeleton but not by a reduction in intracellular cholesterol. Treatment of macrophages with lipophilic statins also blocked ADM formation of primary pancreatic acinar cells. Furthermore, oral administration of simvastatin was associated with a reduction in the number of intrapancreatic macrophages, decreased inflammatory cytokine levels in the pancreas, and attenuated ADM/PanIN formation in mice. CONCLUSION Our data support the hypothesis that statins oppose early PDAC development by their effects on macrophages and ADM formation. The inhibitory actions of statins on macrophages may collaborate with direct inhibitory effects on transformed pancreatic epithelial cells, which cumulatively may reduce early PDAC development and progression.
Collapse
Affiliation(s)
- Soichiro Ako
- Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Yaroslav Teper
- Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Linda Ye
- Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, California
| | - James Sinnett-Smith
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Oscar J. Hines
- Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Enrique Rozengurt
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Guido Eibl
- Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, California
| |
Collapse
|
16
|
Abstract
Statins are 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors used worldwide to manage dyslipidaemia and thus limit the development of atherosclerotic disease and its complications. These atheroprotective drugs are now known to exert pleiotropic actions outside of their cholesterol-lowering activity, including altering immune cell function. Macrophages are phagocytic leukocytes that play critical functional roles in the pathogenesis of atherosclerosis and are directly targeted by statins. Early studies documented the anti-inflammatory effects of statins on macrophages, but emerging evidence suggests that these drugs can also enhance pro-inflammatory macrophage responses, creating an unresolved paradox. This review comprehensively examines the in vitro, in vivo, and clinical literature to document the statin-induced changes in macrophage polarization and immunomodulatory functions, explore the underlying mechanisms involved, and offer potential explanations for this paradox. A better understanding of the immunomodulatory actions of statins on macrophages should pave the way for the development of novel therapeutic approaches to manage atherosclerosis and other chronic diseases and conditions characterised by unresolved inflammation.
Collapse
|
17
|
Park CS, Hwang IC, Park JJ, Park JH, Park JB, Cho GY. Determinants of the survival benefit associated with statins in patients with acute heart failure. ESC Heart Fail 2021; 8:5424-5435. [PMID: 34612019 PMCID: PMC8712823 DOI: 10.1002/ehf2.13637] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 07/12/2021] [Accepted: 09/19/2021] [Indexed: 12/29/2022] Open
Abstract
Aims The benefit of statins in patients with heart failure (HF) remains controversial and the mechanism of action is largely speculative. We investigated the determinants of the survival benefit associated with statins in HF patients. Methods and results We enrolled 1680 acute HF patients receiving statins and 2157 patients not receiving statins admitted between 2009 and 2016. The left ventricular (LV) global longitudinal strain (GLS) was assessed as a measure of myocardial contractility. The primary outcome was 5 year all‐cause mortality. Statin therapy was independently associated with improved survival in patients with HF with preserved ejection fraction (HFpEF) [adjusted hazard ratio (HR) 0.781, 95% confidence interval (CI) 0.621–0.981, P = 0.034], but not in those with HF with reduced EF (HFrEF) (adjusted HR 0.881, 95% CI 0.712–1.090, P = 0.244). Mortality reduction associated with statin therapy was significant in patients with ischaemic HF (adjusted HR 0.775, 95% CI 0.607–0.989, P = 0.040), but not in those with non‐ischaemic HF (adjusted HR 0.895, 95% CI 0.734–1.092, P = 0.275). The relative magnitude of survival benefit with statin therapy increased as LV‐EF and LV‐GLS increased, with a steeper dose–response relationship in patients with ischaemic HF. In the subgroup of patients with ischaemic HF, survival benefit with statin therapy was confined to those ≤75 years of age. Conclusions Our study suggests that the survival benefit of statins is confined to patients with HFpEF and those with ischaemic HF. Myocardial contractility may modulate the prognostic effects of statins in HF patients, particularly when the aetiology is ischaemic rather than non‐ischaemic.
Collapse
Affiliation(s)
- Chan Soon Park
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea.,Cardiovascular Center, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, Republic of Korea
| | - In-Chang Hwang
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea.,Cardiovascular Center, Seoul National University Bundang Hospital, 173 Gumi-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Jin Joo Park
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea.,Cardiovascular Center, Seoul National University Bundang Hospital, 173 Gumi-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Jae-Hyeong Park
- Department of Cardiology in Internal Medicine, Chungnam National University Hospital, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Jun-Bean Park
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea.,Cardiovascular Center, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, Republic of Korea
| | - Goo-Yeong Cho
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea.,Cardiovascular Center, Seoul National University Bundang Hospital, 173 Gumi-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, Republic of Korea
| |
Collapse
|
18
|
Li H, Chen C, Wang DW. Inflammatory Cytokines, Immune Cells, and Organ Interactions in Heart Failure. Front Physiol 2021; 12:695047. [PMID: 34276413 PMCID: PMC8281681 DOI: 10.3389/fphys.2021.695047] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 05/25/2021] [Indexed: 12/20/2022] Open
Abstract
Despite mounting evidence demonstrating the significance of inflammation in the pathophysiological mechanisms of heart failure (HF), most large clinical trials that target the inflammatory responses in HF yielded neutral or even worsening outcomes. Further in-depth understanding about the roles of inflammation in the pathogenesis of HF is eagerly needed. This review summarizes cytokines, cardiac infiltrating immune cells, and extracardiac organs that orchestrate the complex inflammatory responses in HF and highlights emerging therapeutic targets.
Collapse
Affiliation(s)
- Huihui Li
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chen Chen
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
19
|
Goupille C, Ouldamer L, Pinault M, Guimares C, Arbion F, Jourdan ML, Frank PG. Identification of a Positive Association between Mammary Adipose Cholesterol Content and Indicators of Breast Cancer Aggressiveness in a French Population. J Nutr 2021; 151:1119-1127. [PMID: 33831951 DOI: 10.1093/jn/nxaa432] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 08/26/2020] [Accepted: 12/10/2020] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Several studies have recently highlighted important roles for adipose tissue in cancer. However, few have examined adipose tissue cholesterol, and no study has been performed in breast adipose tissue associated with breast tumors. OBJECTIVES The present work was designed to determine if breast adipose tissue cholesterol from the tumor-surrounding area is associated with breast cancer aggressiveness. METHODS Between 2009 and 2011, 215 breast adipose tissue samples were collected at the Tours University Hospital (France) during surgery of women (aged 28-89 y) with invasive breast cancer. Associations of free cholesterol (FC), esterified cholesterol (EC), and total cholesterol (TC) amounts with clinical variables (age, BMI, and treated or untreated hypercholesterolemia) and tumor aggressiveness parameters [phenotype, grade, presence of inflammatory breast cancer (IBC), and multifocality] were tested using Student's t test and after ANOVA. RESULTS The predominant form of cholesterol in adipose tissue was FC, and 50% of patients had no detectable EC. The adipose tissue FC content (μg/mg total lipid) was 18% greater in patients >70 y old than in those 40-49 y old (P < 0.05) and the TC content tended to be 12% greater in untreated hypercholesterolemic patients than in normocholesterolemic patients (P = 0.06). Breast adipose cholesterol concentrations were increased in tissues obtained from patients with human-epidermal-growth-factor-receptor-2 (HER2) phenotype (+13% FC; P < 0.05 compared with luminal A), IBC (+15% FC; P = 0.06 compared with noninflammatory tumors), as well as with multifocal triple-negative tumors (+34% FC, P < 0.05; +30% TC, P < 0.05, compared with unifocal triple-negative tumors). Among patients with triple-negative tumors, hypercholesterolemia was significantly more common (P < 0.05) in patients with multifocal tumors (64%) than in patients with unifocal tumors (25%). CONCLUSIONS This study is the first of this magnitude that analyzes cholesterol concentrations in adipose tissue from female breast cancer patients. An increase in breast adipose tissue cholesterol content may contribute to breast cancer aggressiveness (HER2 phenotype, multifocality of triple-negative tumors, and IBC).
Collapse
Affiliation(s)
- Caroline Goupille
- CHRU de Tours, Hôpital Bretonneau, Service de Gynécologie, Tours, France.,Laboratoire "Nutrition, Growth and Cancer", Université de Tours, INSERM UMR1069, Tours, France
| | - Lobna Ouldamer
- CHRU de Tours, Hôpital Bretonneau, Service de Gynécologie, Tours, France.,Laboratoire "Nutrition, Growth and Cancer", Université de Tours, INSERM UMR1069, Tours, France
| | - Michelle Pinault
- Laboratoire "Nutrition, Growth and Cancer", Université de Tours, INSERM UMR1069, Tours, France
| | - Cyrille Guimares
- Laboratoire "Nutrition, Growth and Cancer", Université de Tours, INSERM UMR1069, Tours, France
| | - Flavie Arbion
- CHRU de Tours, Hôpital Bretonneau, Service de Pathologie, Tours, France
| | - Marie L Jourdan
- CHRU de Tours, Hôpital Bretonneau, Service de Gynécologie, Tours, France.,Laboratoire "Nutrition, Growth and Cancer", Université de Tours, INSERM UMR1069, Tours, France
| | - Philippe G Frank
- Laboratoire "Nutrition, Growth and Cancer", Université de Tours, INSERM UMR1069, Tours, France.,French Network for Nutrition and Cancer Research (NACRe network), France.,SGS France Life Services, Saint Benoît, France
| |
Collapse
|
20
|
Jichitu A, Bungau S, Stanescu AMA, Vesa CM, Toma MM, Bustea C, Iurciuc S, Rus M, Bacalbasa N, Diaconu CC. Non-Alcoholic Fatty Liver Disease and Cardiovascular Comorbidities: Pathophysiological Links, Diagnosis, and Therapeutic Management. Diagnostics (Basel) 2021; 11:689. [PMID: 33921359 PMCID: PMC8069361 DOI: 10.3390/diagnostics11040689] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/09/2021] [Accepted: 04/11/2021] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) has a growing prevalence in recent years. Its association with cardiovascular disease has been intensively studied, and certain correlations have been identified. The connection between these two entities has lately aroused interest regarding therapeutic management. In order to find the best therapeutic options, a detailed understanding of the pathophysiology that links (NAFLD) to cardiovascular comorbidities is needed. This review focuses on the pathogenic mechanisms that are behind these two diseases and on the therapeutic management available at this time.
Collapse
Affiliation(s)
- Alexandra Jichitu
- Clinical Emergency Hospital of Bucharest, 105402 Bucharest, Romania; (A.J.); (C.C.D.)
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania;
| | - Ana Maria Alexandra Stanescu
- Department 5, Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Cosmin Mihai Vesa
- Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania; (C.M.V.); (C.B.)
| | - Mirela Marioara Toma
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania;
| | - Cristiana Bustea
- Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania; (C.M.V.); (C.B.)
| | - Stela Iurciuc
- Department of Cardiology, Faculty of Medicine, “Victor Babeş” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Marius Rus
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania;
| | - Nicolae Bacalbasa
- Department 13, Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
- Department of Surgery, “Ion Cantacuzino” Clinical Hospital, 030167 Bucharest, Romania
| | - Camelia Cristina Diaconu
- Clinical Emergency Hospital of Bucharest, 105402 Bucharest, Romania; (A.J.); (C.C.D.)
- Department 5, Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| |
Collapse
|
21
|
Kauerova S, Bartuskova H, Muffova B, Janousek L, Fronek J, Petras M, Poledne R, Kralova Lesna I. Statins Directly Influence the Polarization of Adipose Tissue Macrophages: A Role in Chronic Inflammation. Biomedicines 2021; 9:biomedicines9020211. [PMID: 33669779 PMCID: PMC7923086 DOI: 10.3390/biomedicines9020211] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 02/07/2021] [Accepted: 02/17/2021] [Indexed: 02/06/2023] Open
Abstract
Statins represent one of the most widely used classes of drugs in current medicine. In addition to a substantial decrease in atherogenic low density lipoprotein (LDL) particle concentrations, several large trials have documented their potent anti-inflammatory activity. Based on our preliminary data, we showed that statins are able to decrease the proportion of pro-inflammatory macrophages (CD14+16+CD36high) in visceral adipose tissue in humans. In the present study including 118 healthy individuals (living kidney donors), a very close relationship between the pro-inflammatory macrophage proportion and LDL cholesterol levels was found. This was confirmed after adjustment for the most important risk factors. The effect of statins on the proportion of pro-inflammatory macrophages was also confirmed in an experimental model of the Prague hereditary hypercholesterolemia rat. A direct anti-inflammatory effect of fluvastatin on human macrophage polarization in vitro was documented. Based on modifying the LDL cholesterol concentrations, statins are suggested to decrease the cholesterol inflow through the lipid raft of macrophages in adipose tissue and hypercholesterolemia to enhance the pro-inflammatory macrophage phenotype polarization. On the contrary, due to their opposite effect, statins respond with anti-inflammatory activity, affecting the whole organism.
Collapse
Affiliation(s)
- Sona Kauerova
- Laboratory for Atherosclerosis Research, Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic; (H.B.); (R.P.); (I.K.L.)
- Correspondence: ; Tel.: +420-236-05-5446
| | - Hana Bartuskova
- Laboratory for Atherosclerosis Research, Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic; (H.B.); (R.P.); (I.K.L.)
- Department of Physiology, Faculty of Science, Charles University, 128 00 Prague, Czech Republic
| | - Barbora Muffova
- Department of Immunology, Faculty of Science, Charles University, 128 00 Prague, Czech Republic;
| | - Libor Janousek
- Department of Transplantation Surgery, Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic; (L.J.); (J.F.)
| | - Jiri Fronek
- Department of Transplantation Surgery, Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic; (L.J.); (J.F.)
| | - Marek Petras
- Department of Epidemiology and Biostatistics, Third Faculty of Medicine, Charles University, 100 00 Prague, Czech Republic;
| | - Rudolf Poledne
- Laboratory for Atherosclerosis Research, Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic; (H.B.); (R.P.); (I.K.L.)
| | - Ivana Kralova Lesna
- Laboratory for Atherosclerosis Research, Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic; (H.B.); (R.P.); (I.K.L.)
- Department of Anesthesia and Intensive Medicine, First Faculty of Medicine, Charles University and University Military Hospital, 169 02 Prague, Czech Republic
| |
Collapse
|
22
|
Targeting perivascular and epicardial adipose tissue inflammation: therapeutic opportunities for cardiovascular disease. Clin Sci (Lond) 2020; 134:827-851. [PMID: 32271386 DOI: 10.1042/cs20190227] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 03/20/2020] [Accepted: 03/30/2020] [Indexed: 02/07/2023]
Abstract
Major shifts in human lifestyle and dietary habits toward sedentary behavior and refined food intake triggered steep increase in the incidence of metabolic disorders including obesity and Type 2 diabetes. Patients with metabolic disease are at a high risk of cardiovascular complications ranging from microvascular dysfunction to cardiometabolic syndromes including heart failure. Despite significant advances in the standards of care for obese and diabetic patients, current therapeutic approaches are not always successful in averting the accompanying cardiovascular deterioration. There is a strong relationship between adipose inflammation seen in metabolic disorders and detrimental changes in cardiovascular structure and function. The particular importance of epicardial and perivascular adipose pools emerged as main modulators of the physiology or pathology of heart and blood vessels. Here, we review the peculiarities of these two fat depots in terms of their origin, function, and pathological changes during metabolic deterioration. We highlight the rationale for pharmacological targeting of the perivascular and epicardial adipose tissue or associated signaling pathways as potential disease modifying approaches in cardiometabolic syndromes.
Collapse
|
23
|
Characterization of glucose uptake metabolism in visceral fat by 18 F-FDG PET/CT reflects inflammatory status in metabolic syndrome. PLoS One 2020; 15:e0228602. [PMID: 32027706 PMCID: PMC7004347 DOI: 10.1371/journal.pone.0228602] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 01/18/2020] [Indexed: 02/06/2023] Open
Abstract
Objective The inflammatory activity of visceral adipose tissue (VAT) is elevated in metabolic syndrome (MS), and associated with vulnerability to atherosclerosis. Inflammation can be assessed by glucose uptake in atherosclerotic plaques. We investigated whether the glucose uptake of VAT, assessed by 18F-fluorodeoxyglucose positron emission tomography/computed tomography (18F-FDG PET/CT), is associated with systemic inflammatory status, and related to the number of MS components. Methods 18F-FDG PET/CT was performed in a total of 203 participants: 59 without MS component; M(0), 92 with one or two MS components; M(1–2), and 52 with MS. Glucose uptake in VAT was evaluated using the mean standardized uptake value (SUVmean) and the maximum SUV (SUVmax). Glucose uptakes of immune-related organs such as the spleen and bone marrow (BM) were evaluated using the SUVmax. Results VAT SUVmax correlated with high-sensitivity C-reactive protein (hsCRP) and the SUVmax of spleen and BM, which reflect the status of systemic inflammation. Both hsCRP and the SUVmax of the spleen and BM were higher in the MS group than in the M(1–2) or M(0) groups. In VAT, SUVmax increased with increasing number of MS components, while SUVmean decreased. Conclusions The SUVmax and SUVmean of VAT assessed by 18F-FDG PET/CT reflected inflammation-driven unique glucose metabolism in the VAT of MS patients, distinct from that of atherosclerotic plaques.
Collapse
|
24
|
Packer M. Atrial Fibrillation and Heart Failure With Preserved Ejection Fraction in Patients With Nonalcoholic Fatty Liver Disease. Am J Med 2020; 133:170-177. [PMID: 31622581 DOI: 10.1016/j.amjmed.2019.09.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 09/24/2019] [Accepted: 09/24/2019] [Indexed: 02/07/2023]
Abstract
The most common causes of chronic liver disease in the developed world-nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH)-are the hepatic manifestations of an insulin-resistant state that is linked to visceral adiposity and systemic inflammation. NAFLD and NASH lead to an expansion of epicardial adipose tissue and the release of proinflammatory adipocytokines that cause microcirculatory dysfunction and fibrosis of the adjoining myocardium, resulting in atrial fibrillation as well as heart failure with a preserved ejection fraction (HFpEF). Inflammatory changes in the left atrium lead to electroanatomical remodeling; thus, NAFLD and NASH markedly increase the risk of atrial fibrillation. Simultaneously, patients with NAFLD or NASH commonly show diastolic dysfunction or latent HFpEF. Interventions include 1) weight loss by caloric restriction, bariatric surgery, or intensive exercise, and 2) drugs that ameliorate fat-mediated inflammation in both the liver and heart (eg, statins, metformin, sodium-glucose cotransporter 2 inhibitors, glucagon-like peptide-1 receptor agonists, and pioglitazone). Patients with NAFLD or NASH commonly have an inflammation-related atrial and ventricular myopathy, which may contribute to symptoms and long-term outcomes.
Collapse
Affiliation(s)
- Milton Packer
- Baylor Heart and Vascular Institute, Baylor University Medical Center, Dallas, Tex; Imperial College, London, UK.
| |
Collapse
|
25
|
Yang J, Fukuchi KI. TIR-Domain-Containing Adaptor-Inducing Interferon- β (TRIF) Is Involved in Glucose Metabolism in Adipose Tissue through the Insulin/AKT Signaling Pathway. Int J Endocrinol 2020; 2020:6942307. [PMID: 33376487 PMCID: PMC7744180 DOI: 10.1155/2020/6942307] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 11/09/2020] [Accepted: 11/25/2020] [Indexed: 01/06/2023] Open
Abstract
Obesity significantly increases the risk of developing type 2 diabetes mellitus and other metabolic diseases. Obesity is associated with chronic low-grade inflammation in white adipose tissues, which is thought to play an essential role in developing insulin resistance. Many lines of evidence indicate that toll-like receptors (TLRs) and their downstream signaling pathways are involved in development of chronic low-grade inflammation and insulin resistance, which are associated with obesity. Mice lacking molecules positively involved in the TLR signaling pathways are generally protected from high-fat diet-induced inflammation and insulin resistance. In this study, we have determined the effects of genetic deficiency of toll/interleukin-1 receptor-domain-containing adaptor-inducing interferon-β (TRIF) on food intake, bodyweight, glucose metabolism, adipose tissue macrophage polarization, and insulin signaling in normal chow diet-fed mice to investigate the role of the TRIF-dependent TLR signaling in adipose tissue metabolism and inflammation. TRIF deficiency (TRIF-/-) increased food intake and bodyweight. The significant increase in bodyweight in TRIF-/- mice was discernible as early as 24 weeks of age and sustained thereafter. TRIF-/- mice showed impaired glucose tolerance in glucose tolerance tests, but their insulin tolerance tests were similar to those in TRIF+/+ mice. Although no difference was found in the epididymal adipose mass between the two groups, the percentage of CD206+ M2 macrophages in epididymal adipose tissue decreased in TRIF-/- mice compared with those in TRIF+/+ mice. Furthermore, activation of epididymal adipose AKT in response to insulin stimulation was remarkably diminished in TRIF-/- mice compared with TRIF+/+ mice. Our results indicate that the TRIF-dependent TLR signaling contributes to maintaining insulin/AKT signaling and M2 macrophages in epididymal adipose tissue under a normal chow diet and provide new evidence that TLR4-targeted therapies for type 2 diabetes require caution.
Collapse
Affiliation(s)
- Junling Yang
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, 1 Illini Drive, Peoria, IL 61605, USA
| | - Ken-Ichiro Fukuchi
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, 1 Illini Drive, Peoria, IL 61605, USA
| |
Collapse
|
26
|
Gong M, Wen S, Nguyen T, Wang C, Jin J, Zhou L. Converging Relationships of Obesity and Hyperuricemia with Special Reference to Metabolic Disorders and Plausible Therapeutic Implications. Diabetes Metab Syndr Obes 2020; 13:943-962. [PMID: 32280253 PMCID: PMC7125338 DOI: 10.2147/dmso.s232377] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 03/09/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Obesity and hyperuricemia mutually influence metabolic syndrome. This study discusses the metabolic relationships between obesity and hyperuricemia in terms of pathophysiology, complications, and treatments. METHODS We searched for preclinical or clinical studies on the pathophysiology, complications, and therapy of obesity and hyperuricemia on the PubMed database. RESULTS In this systemic review, we summarized our searching results on topics of pathophysiology, complications and therapeutic strategy. In pathophysiology, we firstly introduce genetic variations for obesity, hyperuricemia and their relationships by genetic studies. Secondly, we talk about the epigenetic influences on obesity and hyperuricemia. Thirdly, we describe the central metabolic regulation and the role of hyperuricemia. Then, we refer to the character of adipose tissue inflammation and oxidative stress in the obesity and hyperuricemia. In the last part of this topic, we reviewed the critical links of gut microbiota in the obesity and hyperuricemia. In the following part, we review the pathophysiology of major complications in obesity and hyperuricemia including insulin resistance and type 2 diabetes mellitus, chronic kidney disease, cardiovascular diseases, and cancers. Finally, we recapitulate the therapeutic strategies especially the novel pharmaceutic interventions for obesity and hyperuricemia, which concurrently show the mutual metabolic influences between two diseases. CONCLUSION The data reviewed here delineate the metabolic relationships between obesity and hyperuricemia, and provide a comprehensive overview of the therapeutic targets for the management of metabolic syndromes.
Collapse
Affiliation(s)
- Min Gong
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai201399, People’s Republic of China
| | - Song Wen
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai201399, People’s Republic of China
| | - Thiquynhnga Nguyen
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai201399, People’s Republic of China
| | - Chaoxun Wang
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai201399, People’s Republic of China
| | - Jianlan Jin
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai201399, People’s Republic of China
| | - Ligang Zhou
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai201399, People’s Republic of China
- Correspondence: Ligang Zhou Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai201399, ChinaTel +8613611927616 Email
| |
Collapse
|
27
|
Bi X, Jiang B, Zhou J, Luo L, Yin Z. Phosphorylated Hsp27 prevents LPS-induced excessive inflammation in THP-1 cells via suppressing ROS-mediated upregulation of CBP. Cell Biol Int 2020; 44:253-267. [PMID: 31483551 DOI: 10.1002/cbin.11228] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 08/31/2019] [Indexed: 01/24/2023]
Abstract
Heat shock protein 27 (Hsp27) is a member of the small heat shock protein family expressed at high levels to protect cells against heat shock and other conditions of stress. Hsp27 has been indicated in the regulation of inflammation signaling pathway, and Hsp27 phosphorylation is vital for efficient control of host-defense response in early stages of lipopolysaccharide (LPS)-stimulated inflammation. The notion that CREB-binding protein (CBP) is involved in the regulation of two major transcription factors, nuclear factor-κB (NF-κB) and AP-1, suggests that CBP, as a coactivator protein, may also play an important role in the cellular response to inflammation. Here, we explored the mechanism underlying the regulatory relationships between Hsp27 and CBP in THP-1 cells, and found that phosphorylated Hsp27 was critical to the protein level of CBP. Furthermore, in exploring the signaling mechanisms underlying its action, we found that p38MAPK-MK2-Hsp27 regulated NF-κB via CBP, which acted as a multi-protein complex assembly scaffold. Finally, we demonstrated that phosphorylated Hsp27 reduced reactive oxygen species accumulation thereby significantly repressed LPS-induced excessive increase of CBP. Taken together, our data demonstrated that Hsp27, in its phosphorylation state, plays a critical role in controlling LPS-induced inflammatory response by modulating CBP.
Collapse
Affiliation(s)
- Xiaowen Bi
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, Jiangsu, P.R. China
| | - Baolin Jiang
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, Jiangsu, P.R. China
| | - Jinyi Zhou
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, Jiangsu, P.R. China
| | - Lan Luo
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, P.R. China
| | - Zhimin Yin
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, Jiangsu, P.R. China
| |
Collapse
|
28
|
Obesity-Induced Adipose Tissue Inflammation as a Strong Promotional Factor for Pancreatic Ductal Adenocarcinoma. Cells 2019; 8:cells8070673. [PMID: 31277269 PMCID: PMC6678863 DOI: 10.3390/cells8070673] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 06/28/2019] [Accepted: 07/02/2019] [Indexed: 02/07/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is expected to soon become the second leading cause of cancer related deaths in the United States. This may be due to the rising obesity prevalence, which is a recognized risk factor for PDAC. There is great interest in deciphering the underlying driving mechanisms of the obesity–PDAC link. Visceral adiposity has a strong correlation to certain metabolic diseases and gastrointestinal cancers, including PDAC. In fact, our own data strongly suggest that visceral adipose tissue inflammation is a strong promoter for PDAC growth and progression in a genetically engineered mouse model of PDAC and diet-induced obesity. In this review, we will discuss the relationship between obesity-associated adipose tissue inflammation and PDAC development, with a focus on the key molecular and cellular components in the dysfunctional visceral adipose tissue, which provides a tumor permissive environment.
Collapse
|
29
|
Raggi P, Gadiyaram V, Zhang C, Chen Z, Lopaschuk G, Stillman AE. Statins Reduce Epicardial Adipose Tissue Attenuation Independent of Lipid Lowering: A Potential Pleiotropic Effect. J Am Heart Assoc 2019; 8:e013104. [PMID: 31190609 PMCID: PMC6645620 DOI: 10.1161/jaha.119.013104] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Background High epicardial adipose tissue (EAT) attenuation (Hounsfield units [HUs]) on computed tomography is considered a marker of inflammation and is associated with an increased risk of cardiovascular events. Statins reduce the volume of EAT, but it is unknown whether they affect EAT HUs. Methods and Results We reviewed the chest computed tomographic scans of 420 postmenopausal women randomized to either 80 mg of atorvastatin or 40 mg of pravastatin daily and rescanned after 1 year to measure change in coronary artery calcium score. EAT HUs were measured near the proximal right coronary artery and remote from any area of coronary artery calcium. Computed tomographic images were also queried for subcutaneous adipose tissue (SubQ) attenuation (HUs) change over time. The mean patients’ age was 65±6 years. The baseline EAT HU value was higher than the SubQ HU value (−89.4±24.0 HU versus −123.3±30.4 HU; P<0.001). The EAT HU value decreased significantly in the entire cohort (−5.4±29.7 HU [−6% change]; P<0.001), but equally in the patients given atorvastatin and pravastatin (−6.35+31 HU and −4.55+28 HU; P=0.55). EAT HU change was not associated with change in total cholesterol, low‐density lipoprotein cholesterol, coronary artery calcium, and EAT volume (all P=not significant). Change in high‐density lipoprotein cholesterol was marginally associated with EAT HU change (P=0.07). Statin treatment did not induce a change in SubQ HUs. Conclusions Statins induced a decrease in EAT HUs over time, independent of intensity of low‐density lipoprotein cholesterol lowering. The positive effect on EAT and the neutral effect on SubQ suggest that statins induced a decrease in metabolic activity in EAT by reduction in cellularity, vascularity, or inflammation. The clinical significance of the observed change in EAT HUs remains to be demonstrated.
Collapse
Affiliation(s)
- Paolo Raggi
- 1 Mazankowski Alberta Heart Institute and Department of Medicine University of Alberta Edmonton Alberta Canada
| | | | - Chao Zhang
- 3 Department of Biostatistics and Bioinformatics Emory University Atlanta GA
| | - Zhengjia Chen
- 3 Department of Biostatistics and Bioinformatics Emory University Atlanta GA
| | - Gary Lopaschuk
- 4 Department of Pediatrics University of Alberta Edmonton Alberta Canada
| | - Arthur E Stillman
- 1 Mazankowski Alberta Heart Institute and Department of Medicine University of Alberta Edmonton Alberta Canada.,5 Division of Cardiothoracic Imaging Department of Radiology and Imaging Sciences Emory University Atlanta GA
| |
Collapse
|
30
|
Anti-inflammatory activity of elicited soybean ( Glycine max) extract on Balb/C mice ( Mus musculus) with high-fat and -fructose diet. Cent Eur J Immunol 2019; 44:7-14. [PMID: 31114431 PMCID: PMC6526585 DOI: 10.5114/ceji.2019.84010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 06/08/2017] [Indexed: 12/13/2022] Open
Abstract
Obesity causes adipocyte hypertrophy, which leads to cell death. Consequently, macrophages and lymphocytes infiltrate into the adipose tissue and elevate pro-inflammatory cytokine production through TLR activation. This study aimed to determine the efficacy of soybean extract, which was elicited by Saccharomyces cerevisiae and light, as an anti-inflammatory agent in mice with a high-fat and -fructose diet (HFFD). The elicited soybean extract (ESE) was administered orally to mice for four weeks after being given an HFFD for 20 weeks. Three different doses were used: (1) low-dose (78 mg/kg BW); (2) normal dose (104 mg/kg BW); and (3) high dose (130 mg/kg BW). HFFD mice model treated with simvastatin 2.8 mg/kg BW considered as drug control. After 24 weeks, the lymphocytes were isolated and the relative number of CD4+TLR3+ T, CD4+TLR4+ T, CD4+TNF-α+ T, and CD4+IFN-γ+ T cells were analysed using flow cytometry. The results showed that the HFFD mouse model had an increased number of CD4+TLR3+ T, CD4+TLR4+ T, CD4+TNF-α+ T, and CD4+IFN-γ+ T cells. ESE administration decreased the relative number of CD4+TLR3+ T, CD4+TLR4+ T, CD4+TNF-α+ T, and CD4+IFN-γ+ T cells. The normal dose of ESE is the most effective dose in suppressing inflammation compared to positive controls. ESE 104 mg/kg BW can be considered as an alternative herbal medicine that may suppress inflammation in HFFD mice.
Collapse
|
31
|
Fan L, Zhou P, Hong Q, Chen AX, Liu GY, Yu KD, Shao ZM. Toll-like receptor 3 acts as a suppressor gene in breast cancer initiation and progression: a two-stage association study and functional investigation. Oncoimmunology 2019; 8:e1593801. [PMID: 31069157 PMCID: PMC6492959 DOI: 10.1080/2162402x.2019.1593801] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 02/28/2019] [Accepted: 03/05/2019] [Indexed: 12/29/2022] Open
Abstract
Toll-like receptor 3 (TLR3) is a receptor recognizing double-stranded RNA (dsRNA) from viruses as well as from lytic mammalian cells. In the present study, we performed a two-stage association study (n = 3,551) and found that the minor alleles of two SNPs (the T-allele of rs5743312 and the T-allele of rs3775296) conferred increased risks of breast cancer incidence. The adjusted odds ratios (ORs) were 2.281 (P = 7.01 × 10-5) and 2.086 (P = 8.69 × 10-5), respectively. Specifically, the susceptibility variants within TLR3 were significantly associated with larger tumor size (adjusted P-values: 0.004 for rs5743312 and 0.004 for rs3775296). Furthermore, we investigated the biological function of the TLR3 protein in breast cancer cell lines. Notably, the stable expression of TLR3 directly inhibited cell proliferation both in vitro and in vivo. We also verified that TLR3 conferred less invasive phenotypes on breast cancer cells by regulating the mRNA expression of a panel of genes. TLR3-mediated inhibition of proliferation was caused by downregulation of the EGFR/PI3K/AKT pathway. In summary, our findings strongly suggest that common genetic changes in the TLR3 gene may influence breast cancer susceptibility and development, and TLR3 plays a negative regulatory role in the initiation and progression of human breast cancer cells, at least in part by downregulating the EGFR/PI3K/AKT pathway.
Collapse
Affiliation(s)
- Lei Fan
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Peng Zhou
- Department of Ophthalmology, Parkway Health, Shanghai, P.R.China
| | - Qi Hong
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Ao-Xiang Chen
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Guang-Yu Liu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Ke-Da Yu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Zhi-Ming Shao
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| |
Collapse
|
32
|
Singh P, Zhang Y, Sharma P, Covassin N, Soucek F, Friedman PA, Somers VK. Statins decrease leptin expression in human white adipocytes. Physiol Rep 2019; 6. [PMID: 29372612 PMCID: PMC5789723 DOI: 10.14814/phy2.13566] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 12/09/2017] [Indexed: 12/26/2022] Open
Abstract
Statin use is associated with increased calorie intake and consequent weight gain. It is speculated that statin‐dependent improvements in lipid profile may undermine the perceived need to follow lipid‐lowering and other dietary recommendations leading consequently to increased calorie intake. However, increases in calorie intake in statin users may also be related to statin‐dependent decreases in satiety factors such as leptin, an adipocyte‐derived adipokine. The objective of our study was to examine the direct effects of statins on leptin expression. Adipocytes are the main source of circulating leptin. Therefore, we examined the effects of atorvastatin and simvastatin on leptin expression in cultured human white adipocytes. We show that treatment of white adipocytes with simvastatin and atorvastatin decreases leptin mRNA expression (simvastatin: P = 0.008, atorvastatin: P = 0.03) and leptin secretion (simvastatin: P = 0.0001, atorvastatin: P = 0.0001). Both simvastatin and atorvastatin mediate decreases in leptin expression via extracellular‐signal‐regulated kinases 1/2 and peroxisome proliferator‐activated receptor gamma pathways (simvastatin: P = 0.01, atorvastatin: P = 0.026). Additionally, statin treatment also induced expected increases in adiponectin, while decreasing monocyte chemoattractant protein 1 (MCP1) mRNA. Furthermore, statins increased secretion of both total as well as high molecular weight adiponectin while decreasing MCP1 secretion. To conclude, statins act directly on human white adipocytes to regulate adipokine secretion and decrease leptin expression. Leptin is an important satiety factor. Hence, statin‐dependent decreases in leptin may contribute, at least in part, to increases in food intake in statin users.
Collapse
Affiliation(s)
- Prachi Singh
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Yuebo Zhang
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Pragya Sharma
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Naima Covassin
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Filip Soucek
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota.,ICRC - Department of Cardiovascular Diseases, St. Anne's University Hospital, Brno, Czech Republic
| | - Paul A Friedman
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Virend K Somers
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
33
|
Qi XY, Qu SL, Xiong WH, Rom O, Chang L, Jiang ZS. Perivascular adipose tissue (PVAT) in atherosclerosis: a double-edged sword. Cardiovasc Diabetol 2018; 17:134. [PMID: 30305178 PMCID: PMC6180425 DOI: 10.1186/s12933-018-0777-x] [Citation(s) in RCA: 140] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 10/06/2018] [Indexed: 02/06/2023] Open
Abstract
Perivascular adipose tissue (PVAT), the adipose tissue that surrounds most of the vasculature, has emerged as an active component of the blood vessel wall regulating vascular homeostasis and affecting the pathogenesis of atherosclerosis. Although PVAT characteristics resemble both brown and white adipose tissues, recent evidence suggests that PVAT develops from its own distinct precursors implying a closer link between PVAT and vascular system. Under physiological conditions, PVAT has potent anti-atherogenic properties mediated by its ability to secrete various biologically active factors that induce non-shivering thermogenesis and metabolize fatty acids. In contrast, under pathological conditions (mainly obesity), PVAT becomes dysfunctional, loses its thermogenic capacity and secretes pro-inflammatory adipokines that induce endothelial dysfunction and infiltration of inflammatory cells, promoting atherosclerosis development. Since PVAT plays crucial roles in regulating key steps of atherosclerosis development, it may constitute a novel therapeutic target for the prevention and treatment of atherosclerosis. Here, we review the current literature regarding the roles of PVAT in the pathogenesis of atherosclerosis.
Collapse
Affiliation(s)
- Xiao-Yan Qi
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, 421001 China
| | - Shun-Lin Qu
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, 421001 China
| | - Wen-Hao Xiong
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, 421001 China
| | - Oren Rom
- Cardiovascular Research Center, University of Michigan, Ann Arbor, MI USA
| | - Lin Chang
- Cardiovascular Research Center, University of Michigan, Ann Arbor, MI USA
| | - Zhi-Sheng Jiang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, 421001 China
| |
Collapse
|
34
|
Sunjaya AP, Sunjaya AF, Halim S, Ferdinal F. Risk and Benefits of Statins in Glucose Control Management of Type II Diabetes. Int J Angiol 2018; 27:121-131. [PMID: 30154630 DOI: 10.1055/s-0036-1572523] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Worldwide statins are considered to be the first-line pharmacological treatment for dyslipidemia and reducing the risk of coronary heart disease. However, recently various studies have shown its adverse effect on glucose control among diabetic patients and the U.S. Food and Drug Administration have revised statin drug labels to include information that increases in fasting serum glucose and glycated hemoglobin levels have been reported. This systematic review objective is to evaluate the risks and benefits of statins in glucose control management of type 2 diabetes patients based on the 44 published journal articles included and obtained through MEDLINE full text, PubMed, Science Direct, Pro Quest, SAGE, Taylor and Francis Online, Google Scholar, High Wire, and Elsevier Clinical Key. Statins were found to affect glucose control through several ways, namely, by affecting insulin production and secretion by β-pancreatic cells, insulin resistance, insulin uptake by the muscles and adipocytes and production of adipokines. Current evidence available shows that most of the statins give unfavorable side effects with regards to glucose control among diabetic patients. A dose-dependent and time-dependent effect was also observed in some statins which may be present among other statins as well.
Collapse
Affiliation(s)
| | | | - Samuel Halim
- Department of Internal Medicine, Faculty of Medicine, Tarumanagara University, Jakarta, Indonesia
| | - Frans Ferdinal
- Department of Biochemistry, Faculty of Medicine, Tarumanagara University, Jakarta, Indonesia
| |
Collapse
|
35
|
Xu XQ, Guo L, Wang X, Liu Y, Liu H, Zhou RH, Gu J, Liu JB, Xu P, Zhou L, Ho WZ. Human Cervical Epithelial Cells Release Antiviral Factors and Inhibit HIV Replication in Macrophages. J Innate Immun 2018; 11:29-40. [PMID: 30032138 DOI: 10.1159/000490586] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 06/03/2018] [Indexed: 12/11/2022] Open
Abstract
The female reproductive tract is a major site of HIV sexual transmission. We here examined whether human cervical epithelial cells (HCEs) can be immunologically activated and produce antiviral factors against HIV. We demonstrated that HCEs (End1/E6E7 cells) possess the functional toll-like receptor (TLR)3 signaling system, which could be activated by Poly I:C and induce multiple cellular HIV restriction factors. The treatment of primary human macrophages with supernatant (SN) from TLR3-activated End1/E6E7 cell cultures resulted in HIV inhibition. This SN-mediated HIV inhibition was mainly through the induction of interferons (IFN)-β and IFN-λs, as the antibodies to IFN-β or IFN-λs receptor could effectively block the SN-mediated anti-HIV effect. Further studies showed that the incubation of macrophages with SN from the activated cervical epithelial cell cultures induced the expression of a number of IFN-stimulated genes (ISGs), including IFN-stimulated gene (ISG15), ISG56, 2', 5'-oligoadenylate synthetase 1 (OAS 1), OAS 2, Myxovirus Resistance A (MxA), MxB, and Guanylate-binding protein 5 (GBP5). In addition, TLR3-activated cells produced the CC chemokines [regulated on activation, normal T cell expressed and secreted (RANTES), Human macrophage inflammatory protein 1 alpha (MIP-1α), MIP-1β] the ligands of HIV entry co-receptor CCR5. These observations support further studies on HCEs as potentially crucial and alternative targets for immunological intervention to control and prevent HIV sexual transmission.
Collapse
Affiliation(s)
- Xi-Qiu Xu
- Institute of Medical Virology, State Key Laboratory of Virology, Wuhan University School of Basic Medical Sciences, Wuhan, China
| | - Le Guo
- Institute of Medical Virology, State Key Laboratory of Virology, Wuhan University School of Basic Medical Sciences, Wuhan, China
| | - Xu Wang
- Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, USA
| | - Yu Liu
- Institute of Medical Virology, State Key Laboratory of Virology, Wuhan University School of Basic Medical Sciences, Wuhan, China
| | - Hang Liu
- Institute of Medical Virology, State Key Laboratory of Virology, Wuhan University School of Basic Medical Sciences, Wuhan, China
| | - Run-Hong Zhou
- Institute of Medical Virology, State Key Laboratory of Virology, Wuhan University School of Basic Medical Sciences, Wuhan, China
| | - Jun Gu
- Institute of Medical Virology, State Key Laboratory of Virology, Wuhan University School of Basic Medical Sciences, Wuhan, China
| | - Jin-Biao Liu
- Animal Biosafety Level III Laboratory at the Center for Animal Experiment, Wuhan University School of Medicine, Wuhan, China
| | - Pei Xu
- School of Medicine (Shenzhen), Sun Yat-sen University, Guangzhou, China
| | - Li Zhou
- Animal Biosafety Level III Laboratory at the Center for Animal Experiment, Wuhan University School of Medicine, Wuhan,
| | - Wen-Zhe Ho
- Institute of Medical Virology, State Key Laboratory of Virology, Wuhan University School of Basic Medical Sciences, Wuhan, China.,Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
36
|
Statin therapy modulates thickness and inflammatory profile of human epicardial adipose tissue. Int J Cardiol 2018; 274:326-330. [PMID: 30454723 DOI: 10.1016/j.ijcard.2018.06.106] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 06/12/2018] [Accepted: 06/27/2018] [Indexed: 01/29/2023]
Abstract
BACKGROUND Epicardial adipose tissue (EAT) thickness and pro-inflammatory status has been shown to be associated with several cardiac diseases, including aortic stenosis (AS). Thus, cardiac visceral fat could represent a potential new target for drugs. In the present study we evaluate the effect of statin therapy on EAT accumulation and inflammation. METHODS Echocardiographic EAT thickness was assessed in 193 AS patients taking (n.87) and not taking (n.106) statins, undergoing cardiac surgery. To explore the association between statin therapy and EAT inflammation, EAT biopsies were obtained for cytokines immunoassay determination in EAT secretomes. An in vitro study was also conducted and the modulation of EAT and subcutaneous adipose tissue (SCAT) secretomes by atorvastatin was assessed in paired biopsies. RESULTS Statin therapy was significantly associated with lower EAT thickness (p < 0.0001) and with lower levels of EAT-secreted inflammatory mediators (p < 0.0001). Of note, there was a significant correlation between EAT thickness and its pro-inflammatory status. In vitro, atorvastatin showed a direct anti-inflammatory effect on EAT which was significantly higher compared to the SCAT response to statin incubation (p < 0.0001). CONCLUSIONS The present study indicates a robust association between statin therapy and reduced EAT accumulation in patients with AS. The present data also suggest a direct relationship between EAT thickness and its inflammatory status, both modulated by statin therapy. The in vitro results support the hypothesis of a direct action of statins on EAT secretory profile. Overall our data suggest EAT as a potential new therapeutic target for statin therapy.
Collapse
|
37
|
Packer M. Epicardial Adipose Tissue May Mediate Deleterious Effects of Obesity and Inflammation on the Myocardium. J Am Coll Cardiol 2018; 71:2360-2372. [PMID: 29773163 DOI: 10.1016/j.jacc.2018.03.509] [Citation(s) in RCA: 406] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 02/20/2018] [Accepted: 03/18/2018] [Indexed: 02/07/2023]
Abstract
Epicardial adipose tissue has unique properties that distinguish it from other depots of visceral fat. Rather than having distinct boundaries, the epicardium shares an unobstructed microcirculation with the underlying myocardium, and in healthy conditions, produces cytokines that nourish the heart. However, in chronic inflammatory disorders (especially those leading to heart failure with preserved ejection fraction), the epicardium becomes a site of deranged adipogenesis, leading to the secretion of proinflammatory adipokines that can cause atrial and ventricular fibrosis. Accordingly, in patients at risk of heart failure with preserved ejection fraction, drugs that promote the accumulation or inflammation of epicardial adipocytes may lead to heart failure, whereas treatments that ameliorate the proinflammatory characteristics of epicardial fat may reduce the risk of heart failure. These observations suggest that epicardial adipose tissue is a transducer of the adverse effects of systemic inflammation and metabolic disorders on the heart, and thus, represents an important target for therapeutic interventions.
Collapse
Affiliation(s)
- Milton Packer
- Baylor Heart and Vascular Institute, Baylor University Medical Center, Dallas, Texas.
| |
Collapse
|
38
|
The Anti-Inflammatory Effects of Shinbaro3 Is Mediated by Downregulation of the TLR4 Signalling Pathway in LPS-Stimulated RAW 264.7 Macrophages. Mediators Inflamm 2018; 2018:4514329. [PMID: 29849490 PMCID: PMC5907526 DOI: 10.1155/2018/4514329] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 01/11/2018] [Accepted: 01/29/2018] [Indexed: 02/07/2023] Open
Abstract
Shinbaro3, a formulation derived from the hydrolysed roots of Harpagophytum procumbens var. sublobatum (Engl.) Stapf, has been clinically used in the pharamacopuncture treatment of arthritis in Korea. In the present study, Shinbaro3 inhibited NO generation in LPS-induced RAW 264.7 cells in a dose-dependent manner. Shinbaro3 also downregulated the mRNA and protein expression of inflammatory mediators in a dose-dependent manner. Three mechanisms explaining the effects of Shinbaro3 in RAW 264.7 cells were identified as follows: (1) inhibition of the extracellular signal-regulated kinase 1 and 2 (ERK1/2), stress-activated protein kinase (SAPK)/c-Jun N-terminal protein kinase (JNK), and p38 mitogen-activated protein kinase (MAPK) pathways; (2) suppression of IκB kinase-α/β (IKK-α/β) phosphorylation and nuclear factor-kappa B (NF-κB) subunits in the NF-κB pathway, which are involved in MyD88-dependent signalling; and (3) downregulation of IFN-β mRNA expression via inhibition of interferon regulatory factor 3 (IRF3) and Janus-activated kinase 1 (JAK1)/signal transducer and activator of transcription 1 (STAT1) phosphorylation, which is involved in TRIF-dependent signalling. Shinbaro3 exerted anti-inflammatory effects in LPS-stimulated RAW 264.7 macrophage cells through modulation of the TLR4/MyD88 pathways, suggesting that Shinbaro3 is a novel anti-inflammatory therapeutic candidate in the field of pharmacopuncture.
Collapse
|
39
|
Boutari C, Mantzoros CS. Inflammation: A key player linking obesity with malignancies. Metabolism 2018; 81:A3-A6. [PMID: 29309747 DOI: 10.1016/j.metabol.2017.12.015] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 12/28/2017] [Accepted: 12/28/2017] [Indexed: 12/16/2022]
Affiliation(s)
- Chrysoula Boutari
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| | - Christos S Mantzoros
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
40
|
Packer M. Are the effects of drugs to prevent and to treat heart failure always concordant? The statin paradox and its implications for understanding the actions of antidiabetic medications. Eur J Heart Fail 2018; 20:1100-1105. [PMID: 29566300 DOI: 10.1002/ejhf.1183] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 01/30/2018] [Accepted: 02/20/2018] [Indexed: 12/11/2022] Open
Abstract
Most treatments for chronic heart failure are effective both in preventing its onset and reducing its progression. However, statins prevent the development of heart failure, but they do not decrease morbidity and mortality in those with established heart failure. This apparent discordance cannot be explained by an effect to prevent interval myocardial infarctions. Instead, it seems that the disease that statins were preventing in trials of patients with a metabolic disorder was different from the disease that they were treating in trials of chronic heart failure. The most common phenotype of heart failure in patients with obesity and diabetes is heart failure with a preserved ejection fraction (HFpEF). In this disorder, the anti-inflammatory effects of statins might ameliorate myocardial fibrosis and cardiac filling abnormalities, but these actions may have little relevance to patients with heart failure and a reduced ejection fraction (HFrEF), whose primary derangement is cardiomyocyte loss and stretch. These distinctions may explain why statins were ineffective in trials that focused on HFrEF, but have been reported to produce favourable effects in observational studies of HFpEF. Similarly, selective cytokine antagonists were ineffective in HFrEF, but have been associated with benefits in HFpEF. These observations may have important implications for our understanding of the effects of antihyperglycaemic medications. Glucagon-like peptide-1 receptor agonists have had neutral effects on heart failure events in people at risk for HFpEF, but have exerted deleterious actions in HFrEF. Similarly, sodium-glucose co-transporter 2 inhibitors, which exert anti-inflammatory effects and reduce heart failure events in patients who are prone to HFpEF, may not be effective in HFrEF. The distinctions between HFrEF and HFpEF may explain why the effects of drugs on heart failure events in diabetes trials may not be relevant to their use in patients with systolic dysfunction.
Collapse
Affiliation(s)
- Milton Packer
- Baylor Heart and Vascular Institute, Baylor University Medical Center, Dallas, TX, USA
| |
Collapse
|
41
|
Giles JT, Ferrante AW, Broderick R, Zartoshti A, Rose J, Downer K, Zhang HZ, Winchester RJ. Adipose Tissue Macrophages in Rheumatoid Arthritis: Prevalence, Disease-Related Indicators, and Associations With Cardiometabolic Risk Factors. Arthritis Care Res (Hoboken) 2018; 70:175-184. [PMID: 28388816 DOI: 10.1002/acr.23253] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 04/04/2017] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Adipose tissue macrophages (ATMs) are a potent source of inflammatory cytokines, with profound effects on adipose tissue function, yet their potential role in rheumatoid arthritis (RA) pathobiology is largely unstudied. METHODS Periumbilical subcutaneous adipose tissue was obtained from 36 RA patients and 22 non-RA controls frequency matched on demographics and body mass index. Samples were stained for the macrophage marker CD68, and the average proportions of ATMs, crown-like structures (periadipocyte aggregates of 3 or more ATMs), and fibrosis were compared between groups. RESULTS The adjusted proportion of ATMs among all nucleated cells was 76% higher in RA than in non-RA samples (37.7 versus 21.3%, respectively; P < 0.001), and the adjusted average number of crown-like structures was more than 1.5-fold higher in the RA group than in controls (0.58 versus 0.23 crown-like structure/high-power field, respectively; P = 0.001). ATMs were significantly more abundant in early RA and in those with anti-cyclic citrullinated peptide seropositivity. Users of methotrexate, leflunomide, and tumor necrosis factor inhibitors had a significantly lower proportion of ATMs compared with nonusers. Crown-like structures were significantly higher in patients with rheumatoid factor seropositivity and in those with C-reactive protein levels ≥10 mg/liter, and significantly lower among those treated with statins. Linear ATMs were significantly associated with whole-body insulin resistance, but not with serum lipids. CONCLUSIONS ATMs and crown-like structures were more abundant in RA patients and were associated with systemic inflammation, autoimmunity, and whole-body insulin resistance, suggesting possible contributions to the RA disease process. Lower levels of ATMs and crown-like structures associated with specific RA treatments suggest that adipose tissue inflammation may be ameliorated by immunomodulation.
Collapse
Affiliation(s)
- Jon T Giles
- Columbia University College of Physicians and Surgeons, New York, New York
| | - Antony W Ferrante
- Columbia University College of Physicians and Surgeons, New York, New York
| | - Rachel Broderick
- Columbia University College of Physicians and Surgeons, New York, New York
| | - Afshin Zartoshti
- Columbia University College of Physicians and Surgeons, New York, New York
| | - Janine Rose
- Columbia University College of Physicians and Surgeons, New York, New York
| | - Kendall Downer
- Columbia University College of Physicians and Surgeons, New York, New York
| | - Hui-Zhu Zhang
- Columbia University College of Physicians and Surgeons, New York, New York
| | | |
Collapse
|
42
|
Dias S, Paredes S, Ribeiro L. Drugs Involved in Dyslipidemia and Obesity Treatment: Focus on Adipose Tissue. Int J Endocrinol 2018; 2018:2637418. [PMID: 29593789 PMCID: PMC5822899 DOI: 10.1155/2018/2637418] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 09/28/2017] [Accepted: 10/11/2017] [Indexed: 12/15/2022] Open
Abstract
Metabolic syndrome can be defined as a state of disturbed metabolic homeostasis characterized by visceral obesity, atherogenic dyslipidemia, arterial hypertension, and insulin resistance. The growing prevalence of metabolic syndrome will certainly contribute to the burden of cardiovascular disease. Obesity and dyslipidemia are main features of metabolic syndrome, and both can present with adipose tissue dysfunction, involved in the pathogenic mechanisms underlying this syndrome. We revised the effects, and underlying mechanisms, of the current approved drugs for dyslipidemia and obesity (fibrates, statins, niacin, resins, ezetimibe, and orlistat; sibutramine; and diethylpropion, phentermine/topiramate, bupropion and naltrexone, and liraglutide) on adipose tissue. Specifically, we explored how these drugs can modulate the complex pathways involved in metabolism, inflammation, atherogenesis, insulin sensitivity, and adipogenesis. The clinical outcomes of adipose tissue modulation by these drugs, as well as differences of major importance for clinical practice between drugs of the same class, were identified. Whether solutions to these issues will be found in further adjustments and combinations between drugs already in use or necessarily in new advances in pharmacology is not known. To better understand the effect of drugs used in dyslipidemia and obesity on adipose tissue not only is challenging for physicians but could also be the next step to tackle cardiovascular disease.
Collapse
Affiliation(s)
- Sofia Dias
- Department of Biomedicine, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Sílvia Paredes
- Department of Endocrinology, Hospital de Braga, 4710-243 Braga, Portugal
- Department of Public Health and Forensic Sciences, and Medical Education, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Laura Ribeiro
- Department of Biomedicine, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- Department of Public Health and Forensic Sciences, and Medical Education, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- I3S-Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal
| |
Collapse
|
43
|
Han KH. Functional Implications of HMG-CoA Reductase Inhibition on Glucose Metabolism. Korean Circ J 2018; 48:951-963. [PMID: 30334382 PMCID: PMC6196158 DOI: 10.4070/kcj.2018.0307] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Accepted: 09/27/2018] [Indexed: 02/06/2023] Open
Abstract
HMG-CoA reductase inhibitors, i.e. statins, are effective in reducing cardiovascular disease events but also in cardiac-related and overall mortality. Statins are in general well-tolerated, but currently the concerns are raised if statins may increase the risk of new-onset diabetes mellitus (NOD). In this review, the possible effects of statins on organs/tissues being involved in glucose metabolism, i.e. liver, pancreas, adipose tissue, and muscles, had been discussed. The net outcome seems to be inconsistent and often contradictory, which may be largely affected by in vitro experimental settings or/and in vivo animal conditions. The majority of studies point out statin-induced changes of regulations of isoprenoid metabolites and cell-associated cholesterol contents as predisposing factors related to the statin-induced NOD. On the other hand, it should be considered that dysfunctions of isoprenoid pathway and mitochondrial ATP production and the cholesterol homeostasis are already developed under (pre)diabetic and hypercholesterolemic conditions. In order to connect the basic findings with the clinical manifestation more clearly, further research efforts are needed.
Collapse
Affiliation(s)
- Ki Hoon Han
- Department of Internal Medicine, College of Medicine Ulsan University, Asan Medical Center, Seoul, Korea.
| |
Collapse
|
44
|
Hannibal TD, Schmidt-Christensen A, Nilsson J, Fransén-Pettersson N, Hansen L, Holmberg D. Deficiency in plasmacytoid dendritic cells and type I interferon signalling prevents diet-induced obesity and insulin resistance in mice. Diabetologia 2017; 60:2033-2041. [PMID: 28660492 PMCID: PMC6448810 DOI: 10.1007/s00125-017-4341-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 05/19/2017] [Indexed: 12/30/2022]
Abstract
AIMS/HYPOTHESIS Obesity is associated with glucose intolerance and insulin resistance and is closely linked to the increasing prevalence of type 2 diabetes. In mouse models of diet-induced obesity (DIO) and type 2 diabetes, an increased fat intake results in adipose tissue expansion and the secretion of proinflammatory cytokines. The innate immune system not only plays a crucial role in obesity-associated chronic low-grade inflammation but it is also proposed to play a role in modulating energy metabolism. However, little is known about how the modulation of metabolism by the immune system may promote increased adiposity in the early stages of increased dietary intake. Here we aimed to define the role of type I IFNs in DIO and insulin resistance. METHODS Mice lacking the receptor for IFN-α (IFNAR-/-) and deficient in plasmacytoid dendritic cells (pDCs) (B6.E2-2 fl/fl .Itgax-cre) were fed a diet with a high fat content or normal chow. The mice were analysed in vivo and in vitro using cellular, biochemical and molecular approaches. RESULTS We found that the development of obesity was inhibited by an inability to respond to type I IFNs. Furthermore, the development of obesity and insulin resistance in this model was associated with pDC recruitment to the fatty tissues and liver of obese mice (a 4.3-fold and 2.7-fold increase, respectively). Finally, we demonstrated that the depletion of pDCs protects mice from DIO and from developing obesity-associated metabolic complications. CONCLUSIONS/INTERPRETATION Our results provide genetic evidence that pDCs, via type I IFNs, regulate energy metabolism and promote the development of obesity.
Collapse
Affiliation(s)
- Tine D Hannibal
- Department of Experimental Medical Sciences, Lund University, Biomedical Center, CRC, 205 02, Malmö, Sweden
- Department of Immunology & Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anja Schmidt-Christensen
- Department of Experimental Medical Sciences, Lund University, Biomedical Center, CRC, 205 02, Malmö, Sweden
| | - Julia Nilsson
- Department of Experimental Medical Sciences, Lund University, Biomedical Center, CRC, 205 02, Malmö, Sweden
| | - Nina Fransén-Pettersson
- Department of Experimental Medical Sciences, Lund University, Biomedical Center, CRC, 205 02, Malmö, Sweden
- Department of Immunology & Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lisbeth Hansen
- Department of Experimental Medical Sciences, Lund University, Biomedical Center, CRC, 205 02, Malmö, Sweden
- Department of Immunology & Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Dan Holmberg
- Department of Experimental Medical Sciences, Lund University, Biomedical Center, CRC, 205 02, Malmö, Sweden.
- Department of Immunology & Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
45
|
Allott EH, Howard LE, Vidal AC, Moreira DM, Castro-Santamaria R, Andriole GL, Freedland SJ. Statin Use, Serum Lipids, and Prostate Inflammation in Men with a Negative Prostate Biopsy: Results from the REDUCE Trial. Cancer Prev Res (Phila) 2017; 10:319-326. [PMID: 28487295 DOI: 10.1158/1940-6207.capr-17-0019] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 02/21/2017] [Accepted: 03/20/2017] [Indexed: 01/02/2023]
Abstract
Statin use is associated with lower advanced prostate cancer risk. In addition to cholesterol lowering, statins have systemic anti-inflammatory properties. However, their effect on histologic prostate inflammation is not well understood, particularly among men at increased prostate cancer risk but with a negative prostate biopsy. We examined associations between serum lipid levels, statin use, and histologic prostate inflammation using data from 6,655 men with a negative baseline prostate biopsy in the REduction by DUtasteride of prostate Cancer Events (REDUCE) trial. Statin use and lipid levels [total cholesterol, low-density lipoprotein (LDL), high-density lipoprotein (HDL), triglycerides] were assessed at baseline. Inflammation was assessed by central review. Logistic regression was used to examine the effects of lipids and statin use on presence and extent of chronic and acute prostate inflammation [none, moderate (<20%), severe (≥20% biopsy cores)]. Chronic and acute inflammation affected 77% and 15% of men, respectively. Men with high HDL (≥60 vs. <40 mg/dL) had reduced presence of acute inflammation [OR, 0.79; 95% confidence interval (CI), 0.63-0.99] and were less likely to have severe acute inflammation (OR, 0.66; 95% CI, 0.45-0.97), but there were no other associations between lipids and inflammation. Statin users had reduced presence of chronic inflammation (OR, 0.81; 95% CI, 0.69-0.95) and were less likely to have severe chronic (OR, 0.80; 95% CI, 0.68-0.95) and severe acute inflammation (OR, 0.73; 95% CI, 0.53-1.00), relative to non-users. Given the possible role for inflammation in prostate cancer, the inverse association between statins and prostate inflammation suggests a mechanism linking statins with lower advanced prostate cancer risk. Cancer Prev Res; 10(6); 319-26. ©2017 AACR.
Collapse
Affiliation(s)
- Emma H Allott
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Lauren E Howard
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, North Carolina
| | - Adriana C Vidal
- Samuel Oschin Comprehensive Cancer Institute, Cedars Sinai Medical Center, Los Angeles, California
| | - Daniel M Moreira
- Department of Urology, University of Illinois at Chicago, Chicago, Illinois
| | | | - Gerald L Andriole
- Washington University School of Medicine in St. Louis, St. Louis, Missouri
| | - Stephen J Freedland
- Samuel Oschin Comprehensive Cancer Institute, Cedars Sinai Medical Center, Los Angeles, California. .,Division of Urology, Veterans Affairs Medical Center, Durham, North Carolina
| |
Collapse
|
46
|
Effects of pitavastatin and pravastatin on markers of immune activation and arterial inflammation in HIV. AIDS 2017; 31:797-806. [PMID: 28252528 DOI: 10.1097/qad.0000000000001427] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
OBJECTIVE Persistent immune activation is thought to contribute to increased cardiovascular disease risk in HIV and statins may help modulate systemic immune activation. We aimed to compare the effects of two key statins on markers of systemic immune activation and arterial inflammation in the HIV population. DESIGN Double-blind, active-controlled, parallel-group comparative trial performed in 45 sites. METHODS Two hundred and fifty-two antiretroviral therapy-treated HIV-infected participants with dyslipidemia were randomized (1 : 1) to pitavastatin 4 mg daily vs. pravastatin 40 mg daily in the HIV-infected patieNts and TREatment with PItavastatin vs. pravastatin for Dyslipidemia (INTREPID) trial. In this analysis of the INTREPID trial, we assessed markers of immune activation and arterial inflammation using a modified intent-to-treat population. This trial is registered with ClinicalTrials.gov (NCT01301066). RESULTS One hundred and twenty-six participants were randomized to receive pitavastatin and 126 to pravastatin. Ninety-nine participants in the pitavastatin group and 91 participants in the pravastatin group completed the study. Median age was 50 (45, 56) years [median (interquartile range)]. Baseline, low-density lipoprotein-cholestrol (LDL-C) was 153 (135, 171) mg/dl, log HIV-1 viral load was 1.1 ± 0.2 copies/ml, and CD4 cell count was 580 (439, 794) cells/μl. At week 52, the pitavastatin group had a significantly greater reduction (% change) compared with pravastatin in soluble CD14 (sCD14), (-10.0 vs. 0.6%, P = 0.02), oxidized LDL (oxLDL) (-26.9 vs. -17.5%, P = 0.02), and lipoprotein-associated phospholipase 2 (Lp-PLA2) (-26.6 vs. -15.5%, P = 0.005) (pitavastatin vs. pravastatin). CONCLUSION Fifty-two weeks of pitavastatin 4 mg daily (vs. pravastatin 40 mg daily) led to a greater reduction in select markers of immune activation and arterial inflammation (sCD14, oxLDL, and LpPLA2) among HIV-infected participants. Further work is needed to assess whether immune-modulatory effects of pitavastatin reduce cardiovascular disease risk in HIV.
Collapse
|
47
|
Akoumianakis I, Tarun A, Antoniades C. Perivascular adipose tissue as a regulator of vascular disease pathogenesis: identifying novel therapeutic targets. Br J Pharmacol 2016; 174:3411-3424. [PMID: 27976387 DOI: 10.1111/bph.13666] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Revised: 10/04/2016] [Accepted: 10/28/2016] [Indexed: 12/23/2022] Open
Abstract
Adipose tissue (AT) is an active endocrine organ with the ability to dynamically secrete a wide range of adipocytokines. Importantly, its secretory profile is altered in various cardiovascular disease states. AT surrounding vessels, or perivascular AT (PVAT), is recognized in particular as an important local regulator of vascular function and dysfunction. Specifically, PVAT has the ability to sense vascular paracrine signals and respond by secreting a variety of vasoactive adipocytokines. Due to the crucial role of PVAT in regulating many aspects of vascular biology, it may constitute a novel therapeutic target for the prevention and treatment of vascular disease pathogenesis. Signalling pathways in PVAT, such as those using adiponectin, H2 S, glucagon-like peptide 1 or pro-inflammatory cytokines, are among the potential novel pharmacological therapeutic targets of PVAT. LINKED ARTICLES This article is part of a themed section on Molecular Mechanisms Regulating Perivascular Adipose Tissue - Potential Pharmacological Targets? To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.20/issuetoc.
Collapse
Affiliation(s)
- Ioannis Akoumianakis
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford
| | - Akansha Tarun
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford
| | - Charalambos Antoniades
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford
| |
Collapse
|
48
|
Soybean-derived Bowman-Birk Inhibitor (BBI) Inhibits HIV Replication in Macrophages. Sci Rep 2016; 6:34752. [PMID: 27734899 PMCID: PMC5062087 DOI: 10.1038/srep34752] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 09/16/2016] [Indexed: 01/03/2023] Open
Abstract
The Bowman-Birk inhibitor (BBI), a soybean-derived protease inhibitor, is known to have anti-inflammatory effect in both in vitro and in vivo systems. Macrophages play a key role in inflammation and immune activation, which is implicated in HIV disease progression. Here, we investigated the effect of BBI on HIV infection of peripheral blood monocyte-derived macrophages. We demonstrated that BBI could potently inhibit HIV replication in macrophages without cytotoxicity. Investigation of the mechanism(s) of BBI action on HIV showed that BBI induced the expression of IFN-β and multiple IFN stimulated genes (ISGs), including Myxovirus resistance protein 2 (Mx2), 2',5'-oligoadenylate synthetase (OAS-1), Virus inhibitory protein (viperin), ISG15 and ISG56. BBI treatment of macrophages also increased the expression of several known HIV restriction factors, including APOBEC3F, APOBEC3G and tetherin. Furthermore, BBI enhanced the phosphorylation of IRF3, a key regulator of IFN-β. The inhibition of IFN-β pathway by the neutralization antibody to type I IFN receptor (Anti-IFNAR) abolished BBI-mediated induction of the anti-HIV factors and inhibition of HIV in macrophages. These findings that BBI could activate IFN-β-mediated signaling pathway, initialize the intracellular innate immunity in macrophages and potently inhibit HIV at multiple steps of viral replication cycle indicate the necessity to further investigate BBI as an alternative and cost-effective anti-HIV natural product.
Collapse
|
49
|
Choi Y, Abdelmegeed MA, Akbar M, Song BJ. Dietary walnut reduces hepatic triglyceride content in high-fat-fed mice via modulation of hepatic fatty acid metabolism and adipose tissue inflammation. J Nutr Biochem 2016; 30:116-25. [PMID: 27012628 DOI: 10.1016/j.jnutbio.2015.12.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 12/02/2015] [Accepted: 12/08/2015] [Indexed: 12/30/2022]
Abstract
In this study, we evaluated the protective effects of dietary walnuts on high-fat diet (HFD)-induced fatty liver and studied the underlying mechanisms. Male C57BL/6J mice were fed either a regular rodent chow or HFD (45% energy-derived) with or without walnuts (21.5% energy-derived) for 20weeks. Walnut supplementation did not change HFD-induced increase in body weight or visceral fat mass. However, dietary walnuts significantly decreased the amounts of hepatic triglyceride (TG) observed in HFD-fed mice. The addition of walnuts significantly altered the levels of proteins, involved in the hepatic lipid homeostasis, including AMP-activated protein kinase, fatty acid synthase and peroxisome proliferator-activated receptor-α. Since adipocyte inflammation and apoptosis are reportedly important in regulating hepatic fat accumulation, we also evaluated the protective effects of walnuts on adipose tissue injury. Real-time polymerase chain reaction results revealed that adipose tissues isolated from mice fed the HFD+walnut diets showed significantly decreased levels of macrophage infiltration with suppressed expression of proinflammatory genes compared to those significantly elevated in mice fed HFD alone. These improvements also coincided with reduction of HFD-induced apoptosis of adipocytes by dietary walnuts. However, the supplemented walnuts did not significantly alter HFD-induced peripheral glucose intolerance or insulin resistance despite a trend of improvement. Collectively, these results demonstrate that the protective effects of walnuts against HFD-induced hepatic TG accumulation in mice are mediated, at least partially, by modulating the key proteins in hepatic lipid homeostasis and suppression of the genes related to adipose tissue inflammation and macrophage infiltration as well as prevention of adipocyte apoptosis.
Collapse
Affiliation(s)
- Youngshim Choi
- Section of Molecular Pharmacology and Toxicology, Laboratory of Membrane Biochemistry and Biophysics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA
| | - Mohamed A Abdelmegeed
- Section of Molecular Pharmacology and Toxicology, Laboratory of Membrane Biochemistry and Biophysics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA
| | - Mohammed Akbar
- Section of Molecular Pharmacology and Toxicology, Laboratory of Membrane Biochemistry and Biophysics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA
| | - Byoung-Joon Song
- Section of Molecular Pharmacology and Toxicology, Laboratory of Membrane Biochemistry and Biophysics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA.
| |
Collapse
|
50
|
Ko W, Sohn JH, Jang JH, Ahn JS, Kang DG, Lee HS, Kim JS, Kim YC, Oh H. Inhibitory effects of alternaramide on inflammatory mediator expression through TLR4-MyD88-mediated inhibition of NF-кB and MAPK pathway signaling in lipopolysaccharide-stimulated RAW264.7 and BV2 cells. Chem Biol Interact 2016; 244:16-26. [DOI: 10.1016/j.cbi.2015.11.024] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2015] [Revised: 11/11/2015] [Accepted: 11/20/2015] [Indexed: 01/06/2023]
|