1
|
Valencia O, López C, Vanegas-Duarte E, Fillizola C, Bejarano Ramírez DF, Cortés Mejía NA, Vera Torres A. Risk Factors Related to the Development of Nonalcoholic Fatty Liver: A Systematic Review. Can J Gastroenterol Hepatol 2025; 2025:9964486. [PMID: 40264655 PMCID: PMC12014263 DOI: 10.1155/cjgh/9964486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 12/05/2024] [Indexed: 04/24/2025] Open
Abstract
Background: Nonalcoholic fatty liver disease (NAFLD) has a major impact on public health owing to its high morbidity and mortality due to its close relationship with several conditions, including metabolic syndrome, cirrhosis, and cancer. Therefore, this review aimed to systematically compile and summarize the scientific literature on early risk factors for NAFLD development. Methods: A systematic review of population-based cohort studies was conducted. Studies reporting the risk factors associated with nonalcoholic steatohepatitis (NASH) and NAFLD were screened. Results: The search yielded 987 unique records, of which 196 articles were selected after title and abstract screening. A total of 39 articles were read in full text after quality analysis using Downs and Black criteria; 10 of the studies were excluded due to heterogeneity or inconclusive results. Finally, 30 publications were included in this systematic review. The review revealed that clinical conditions such as obesity, weight change, psoriasis, polycystic ovary syndrome, diabetes, thyroid disorders, and elevated serum uric acid levels increase the risk of developing nonalcoholic fatty liver. In addition, lifestyle factors such as sedentary behavior, active or passive smoking, poor sleep quality, and consumption of carbonated beverages are associated with this condition. Conclusions: Evidence was found on the association between different clinical and lifestyle risk factors and NAFLD. This supports the need for preventive, diagnostic, and therapeutic strategies to improve the metabolic, hepatic, and oncological outcomes related to this condition.
Collapse
Affiliation(s)
- Omaira Valencia
- Population Health, Fundación Santa Fe de Bogota, Bogota, Colombia
| | - Carolina López
- Population Health, Fundación Santa Fe de Bogota, Bogota, Colombia
| | | | | | - Diana Fernanda Bejarano Ramírez
- Transplant and Hepatobiliary Surgery Department, Fundación Santa Fe de Bogotá, Bogotá, Colombia
- Epidemiology and Biostatistics Group, Graduate School of Epidemiology and Biostatistics, CES University, Medellín, Colombia
| | - Nicolás Andrés Cortés Mejía
- Division of Anesthesiology, Critical Care Medicine and Pain Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Alonso Vera Torres
- Transplant and Hepatobiliary Surgery Department, Fundación Santa Fe de Bogotá, Bogotá, Colombia
| |
Collapse
|
2
|
Mei Y, Li W, Wang B, Chen Z, Wu X, Lin Y, Wang M. Gut microbiota: an emerging target connecting polycystic ovarian syndrome and insulin resistance. Front Cell Infect Microbiol 2025; 15:1508893. [PMID: 40134784 PMCID: PMC11933006 DOI: 10.3389/fcimb.2025.1508893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 02/18/2025] [Indexed: 03/27/2025] Open
Abstract
Polycystic ovary syndrome (PCOS) is a highly heterogeneous metabolic disorder, with oligomenorrhea and hirsutism as patients' primary complaints. Hyperinsulinemia is a crucial pathophysiological mechanism in the development of PCOS, with 50-70% of patients exhibiting insulin resistance (IR). This condition not only exacerbates ovulatory dysfunction but also leads to various adverse metabolic outcomes, such as dyslipidemia and diabetes, and increases the risk of cardiovascular events both before and after menopause. Gut microbiota is a microbial community within the host that possesses significant metabolic potential and is shaped by external environmental factors, the neuro-immune network, and metabolism. Recent studies have shown that gut microbiota dysbiosis is closely related to the development and progression of PCOS. Despite the growing recognition of the potential role of gut microbiota in the pathogenesis and treatment of PCOS, its clinical application remains in its infancy. Currently, most clinical guidelines and expert consensus still emphasize traditional therapeutic approaches, such as hormonal treatments, lifestyle modifications, and insulin sensitizers. However, accumulating evidence suggests that gut microbiota may influence the metabolic and reproductive health of PCOS patients through various mechanisms. Therefore, understanding the role of gut microbiota between PCOS and IR is essential. This review describes the changes in the gut microbiota of IR-PCOS patients, examines the potential mechanisms by which the gut microbiota contributes to IR in PCOS patients, and updates the evidence supporting the gut microbiota as a potential metabolic regulatory target in IR-PCOS. In summary, gut microbiota dysbiosis may be involved in the development and progression of IR in PCOS patients, and improving gut microbiota may offer metabolic stability benefits.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Min Wang
- Department of Laboratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
3
|
Nakahara H, Ono A, Hayes CN, Shirane Y, Miura R, Fujii Y, Tamura Y, Uchikawa S, Fujino H, Nakahara T, Murakami E, Yamauchi M, Kawaoka T, Miki D, Tsuge M, Kobayashi T, Ohdan H, Arihiro K, Oka S. Multiomics Analysis of Liver Molecular Dysregulation Leading to Nonviral-Related Hepatocellular Carcinoma Development. J Proteome Res 2025; 24:1102-1117. [PMID: 39982271 PMCID: PMC11894656 DOI: 10.1021/acs.jproteome.4c00729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 12/26/2024] [Accepted: 02/10/2025] [Indexed: 02/22/2025]
Abstract
Chronic liver diseases exhibit diverse backgrounds, and it is believed that numerous factors contribute to progression to cancer. To achieve effective prevention of nonviral hepatocellular carcinoma, it is imperative to identify fundamental molecular abnormalities at the patient level. Utilizing cancer-adjacent liver tissues obtained from hepatocellular carcinoma patients (chronic liver disease), we conducted RNA-Seq and metabolome analyses. In the chronic liver disease cohort, upregulation of inflammation-associated signals was observed, concomitant with accumulation of acylcarnitine and fatty acid and depletion of NADP+, gamma-tocopherol, and dehydroisoandrosterone-3-sulfate-1 (DHEAS). To minimize heterogeneity, we performed multiomics clustering, successfully categorizing the chronic liver disease cases into two distinct subtypes. Subtype 1 demonstrated elevated inflammatory levels, whereas Subtype 2 included a disproportionately high proportion of elderly cases. Furthermore, RNA-Seq analysis revealed upregulation of inflammatory signals in Subtype 1, while both subtypes exhibited downregulation of fatty acid metabolism. Metabolome analysis indicated a tendency of increased acylcarnitine levels in Subtype 1 and augmented fatty acid accumulation in Subtype 2. Validation of differentially expressed genes using the Gene Expression Omnibus (GEO) data set revealed the potential for amelioration through supplementation with antioxidants such as epigallocatechin gallate (EGCG).
Collapse
Affiliation(s)
- Hikaru Nakahara
- Department
of Gastroenterology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
- Department
of Clinical and Molecular Genetics, Hiroshima
University, Hiroshima 734-8551, Japan
| | - Atsushi Ono
- Department
of Gastroenterology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | - C. Nelson Hayes
- Department
of Gastroenterology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | - Yuki Shirane
- Department
of Gastroenterology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | - Ryoichi Miura
- Department
of Gastroenterology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | - Yasutoshi Fujii
- Department
of Gastroenterology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
- Department
of Clinical Oncology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | - Yosuke Tamura
- Department
of Gastroenterology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | - Shinsuke Uchikawa
- Department
of Gastroenterology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | - Hatsue Fujino
- Department
of Gastroenterology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | - Takashi Nakahara
- Hiroshima
Prefectural Hospital Gastroenterology & Hepatology, Hiroshima 734-8530, Japan
| | - Eisuke Murakami
- Department
of Gastroenterology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | - Masami Yamauchi
- Department
of Clinical Oncology, Hiroshima Prefectural
Hospital, Hiroshima 734-8530, Japan
| | - Tomokazu Kawaoka
- Department
of Gastroenterology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | - Daiki Miki
- Department
of Gastroenterology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | - Masataka Tsuge
- Department
of Gastroenterology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
- Liver center, Hiroshima University Hospital, Hiroshima 734-8551, Japan
| | - Tsuyoshi Kobayashi
- Department
of Gastroenterological and Transplant Surgery, Graduate School of
Biomedical and Health Sciences, Hiroshima
University, Hiroshima 734-8551, Japan
| | - Hideki Ohdan
- Department
of Gastroenterological and Transplant Surgery, Graduate School of
Biomedical and Health Sciences, Hiroshima
University, Hiroshima 734-8551, Japan
| | - Koji Arihiro
- Department
of Anatomical Pathology, Hiroshima University
Hospital, Hiroshima 734-8551, Japan
| | - Shiro Oka
- Department
of Gastroenterology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| |
Collapse
|
4
|
Wang Y, Yang S, Zhang S, Yang Y, Li S, Zhang M, Li X, Bai H, Luo P, Yuan Y. The value of sex hormones and sex hormone-binding globulin in metabolic dysfunction-associated fatty liver disease among boys with obesity. Front Endocrinol (Lausanne) 2025; 16:1446049. [PMID: 39980852 PMCID: PMC11839430 DOI: 10.3389/fendo.2025.1446049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Accepted: 01/15/2025] [Indexed: 02/22/2025] Open
Abstract
Objective This study aims to investigate the relationship between metabolic dysfunction-associated fatty liver disease (MAFLD) and sex hormones and sex hormone-binding globulin (SHBG) in boys with obesity. Methods Retrospective analysis of metabolic indicators and sex hormone levels in boys with obesity who sought medical attention at the First People's Hospital of Lianyungang City from January 2020 to December 2023. Based on abdominal ultrasound results, they were categorized into a simple obesity group and MAFLD group, and differences between the two groups were compared. Utilizing logistic regression analysis to explore the risk factors for developing MAFLD, and through the construction of Receiver Operating Characteristic (ROC) curves, conducting a preliminary assessment of the diagnostic value for MAFLD. Results A total of 155 male children with obesity were included in the study, mean age of 11.07 ± 1.53 years. Children in the MAFLD group had higher levels of height[(159.49 ± 12.73)cm vs.(155.55 ± 10.50)cm], weight[(82.32 ± 18.75)kg vs.(68.28 ± 15.00)kg], BMI[(32.08 ± 4.49)kg/m2 vs.(27.85 ± 4.21)kg/m2],fasting insulin[33.42(24.07,43.93)uIU/ml vs.23.91(15.72,31.52)uIU/ml],HOMA-IR[7.27(5.26,10.71) vs.4.87(3.27,6.86)],fastingC-peptide[1409.00(1175.00,1668.00)pmol/L vs.1020.00(849.05,1303.00)pmol/L], WBC[(7.85 ± 1.80)×109/L vs.(7.15 ± 1.42)×109/L], HbA1c[5.40(5.30,5.70)% vs.(5.30(5.20,5.60)%],ALT[48.00(27.00,80.00)U/L vs.19.00(15.00,26.50)U/L], and AST[31.00(24.00,60.00)U/L vs.21.00(18.50, 26.00)U/L] compared to the simple obesity group (P<0.05). Children in the MAFLD group had lower levels of HDL[(1.05 ± 0.21)mmol/L vs.(1.16 ± 0.26)mmol/L], testosterone [42.41(30.33,143.28)ng/dl vs.125.41(23.41,221.57)ng/dl], and SHBG[13.20(9.10,17.30)nmol/l vs.19.60(13.50,29.85)nmol/l] compared to the simple obesity group (P<0.05). Logistic regression showed that BMI, testosterone, and SHBG were independent risk factors for MAFLD in boys, and ROC curve analysis indicated their potential value in the early diagnosis of MAFLD. Conclusion BMI, testosterone, and SHBG are independent risk factors for the occurrence of MAFLD in boys with obesity. To control the occurrence of MAFLD, it is essential to address the root cause of the high growth rate of obesity. The roles of testosterone and SHBG in MAFLD merit further research.
Collapse
Affiliation(s)
- Ying Wang
- Pediatric Endocrinology Department, The First People’s Hospital of Lianyungang, The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, China
| | - Shuyi Yang
- Pediatric Endocrinology Department, The First People’s Hospital of Lianyungang, The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, China
| | - Suming Zhang
- Pediatric Endocrinology Department, The First People’s Hospital of Lianyungang, The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, China
- Department of Pediatrics, Lianyungang Municipal Oriental Hospital, Lianyungang, China
| | - Ye Yang
- Pediatric Endocrinology Department, The First People’s Hospital of Lianyungang, The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, China
| | - Siqing Li
- Pediatric Endocrinology Department, The First People’s Hospital of Lianyungang, The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, China
- Pediatric Endocrinology Department, The First People’s Hospital of Lianyungang, Lianyungang Clinical College of Nanjing Medical University, Lianyungang, China
| | - Meiyu Zhang
- Pediatric Endocrinology Department, The First People’s Hospital of Lianyungang, The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, China
- Pediatric Endocrinology Department, The First People’s Hospital of Lianyungang, Lianyungang Clinical College of Nanjing Medical University, Lianyungang, China
| | - Xiaona Li
- Pediatric Endocrinology Department, The First People’s Hospital of Lianyungang, The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, China
| | - Hua Bai
- Pediatric Endocrinology Department, The First People’s Hospital of Lianyungang, The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, China
| | - Peiliang Luo
- Pediatric Endocrinology Department, The First People’s Hospital of Lianyungang, The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, China
| | - Yingdi Yuan
- Pediatric Endocrinology Department, The First People’s Hospital of Lianyungang, The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, China
- Pediatric Endocrinology Department, The First People’s Hospital of Lianyungang, Lianyungang Clinical College of Nanjing Medical University, Lianyungang, China
| |
Collapse
|
5
|
Lan Y, Jin B, Fan Y, Huang Y, Zhou J. The Circadian Rhythm Regulates the Hepato-ovarian Axis Linking Polycystic Ovary Syndrome and Non-alcoholic Fatty Liver Disease. Biochem Genet 2025:10.1007/s10528-024-11010-1. [PMID: 39826031 DOI: 10.1007/s10528-024-11010-1] [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: 09/26/2024] [Accepted: 12/17/2024] [Indexed: 01/20/2025]
Abstract
This study aimed to identify shared gene expression related to circadian rhythm disruption in polycystic ovary syndrome (PCOS) and non-alcoholic fatty liver disease (NAFLD) to discover common diagnostic biomarkers. Visceral fat RNA samples were collected from 12 PCOS and 14 non-PCOS patients, a sample size representing the clinical situation and sufficient to capture PCOS gene expression profiles. Along with liver transcriptome profiles from NAFLD patients, these data were analyzed to identify crosstalk circadian rhythm-related genes (CRRGs) between the diseases. Single-sample and single-gene gene set enrichment analyses explored immune infiltration and pathways associated with CRRGs. Diagnostic biomarkers were identified using a random forest algorithm and validated through nomograms and a mouse model. Seven crosstalk CRRGs (FOS, ACHE, FOSB, EGR1, NR4A1, DUSP1, and EGR3) were associated with clinical features, immunoinflammatory microenvironment, and metabolic pathways in both diseases. EGR1, DUSP1, and NR4A1 were identified as diagnostic biomarkers, exhibiting robust diagnostic capacity (AUC = 0.7679 for PCOS, AUG = 0.9981 for NAFLD). Nomogram validation showed excellent calibration, and independent datasets confirmed their discriminatory ability (AUC = 0.6528 for PCOS, AUC = 0.8275 for NAFLD). Additionally, ceRNA networks and androgen receptor binding sites were identified, suggesting their regulatory roles. Mouse model validation confirmed significant downregulation of EGR1, DUSP1, and NR4A1 in liver tissues, consistent with sequencing data. This study identifies crosstalk CRRGs and diagnostic biomarkers shared between PCOS and NAFLD, highlighting their roles in immune and metabolic dysregulation. These biomarkers offer the potential for improving diagnosis and guiding targeted treatments for both diseases.
Collapse
Affiliation(s)
- Yibing Lan
- Department of Gynecology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, China
| | - Bihui Jin
- Department of Reproductive Endocrinology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, China
| | - Yuhang Fan
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Yizhou Huang
- Department of Gynecology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, China
| | - Jianhong Zhou
- Department of Gynecology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, China.
| |
Collapse
|
6
|
Alhermi A, Perks H, Nigi V, Altahoo N, Atkin SL, Butler AE. The Role of the Liver in the Pathophysiology of PCOS: A Literature Review. Biomolecules 2025; 15:51. [PMID: 39858445 PMCID: PMC11764088 DOI: 10.3390/biom15010051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 12/29/2024] [Accepted: 12/30/2024] [Indexed: 01/27/2025] Open
Abstract
Polycystic ovary syndrome (PCOS) is the most common endocrine metabolic disorder found in women of reproductive age and is characterized by both metabolic and reproductive dysfunction. Women with PCOS commonly have insulin resistance, increased susceptibility to type 2 diabetes mellitus, dyslipidemia, hyperinsulinemia, increased cardiovascular risk, hepatic steatosis, infertility, and an overall reduction in physical and psychological well-being. Several previous studies have shown a causal association between PCOS and hepatic disorders, such as chronic liver disease (CLD) and nonalcoholic fatty liver disease (NAFLD), where PCOS was identified as contributing to the hepatic features. Whilst it is recognized that PCOS may contribute to hepatic dysfunction, there is also evidence that the liver may contribute to the features of PCOS. The purpose of this review is to discuss the current understanding regarding hepatic involvement in PCOS pathophysiology, the inflammatory markers and hepatokines involved in the development of PCOS, and the role of genetics in the occurrence of PCOS. This review illustrates that PCOS and NAFLD are both common disorders and that there is both genetic and metabolic linkage between the disorders. As such, whilst PCOS may contribute to NAFLD development, the converse may also be the case, with a potential bidirectional relationship between PCOS and liver disease.
Collapse
Affiliation(s)
- Abrar Alhermi
- School of Medicine, Royal College of Surgeons of Ireland, Busaiteen, Adliya P.O. Box 15503, Bahrain; (A.A.); (H.P.); (V.N.); (N.A.)
| | - Heather Perks
- School of Medicine, Royal College of Surgeons of Ireland, Busaiteen, Adliya P.O. Box 15503, Bahrain; (A.A.); (H.P.); (V.N.); (N.A.)
| | - Varsha Nigi
- School of Medicine, Royal College of Surgeons of Ireland, Busaiteen, Adliya P.O. Box 15503, Bahrain; (A.A.); (H.P.); (V.N.); (N.A.)
| | - Noor Altahoo
- School of Medicine, Royal College of Surgeons of Ireland, Busaiteen, Adliya P.O. Box 15503, Bahrain; (A.A.); (H.P.); (V.N.); (N.A.)
| | - Stephen L. Atkin
- Research Department, Royal College of Surgeons of Ireland, Busaiteen, Adliya P.O. Box 15503, Bahrain;
| | - Alexandra E. Butler
- Research Department, Royal College of Surgeons of Ireland, Busaiteen, Adliya P.O. Box 15503, Bahrain;
| |
Collapse
|
7
|
Fu Q, Fu L. Engineering nanosystems for regulating reproductive health in women. Theranostics 2025; 15:439-459. [PMID: 39744682 PMCID: PMC11671389 DOI: 10.7150/thno.102626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 11/05/2024] [Indexed: 01/11/2025] Open
Abstract
Reproductive health-related diseases have a significant impact on the well-being of millions of women worldwide, severely compromising their quality of life. Women encounter unique challenges in terms of reproductive health, including gynecological diseases and malignant neoplasms prior to pregnancy, as well as complications during pregnancy that greatly undermine their physical and mental health. Despite recent advancements in the field of female reproduction, substantial challenges still persist. To address these challenges, nanotechnology-based diagnostic and therapeutic strategies have emerged to provide intelligent detection and treatment for pathologies related to women's reproductive health. Although some progress has been made with nanotherapeutics in this domain, its application is still nascent due to the delicate and intricate nature of the female reproductive system. This review comprehensively presents the latest advancements in nanomedicine for regulating woman's reproductive health. Firstly, based on the time period of onset, nanomedicine applications are categorized into four subcategories: 1) preconception diseases such as polycystic ovary syndrome, endometriosis, and gynecologic malignancy treatment; 2) pregastrulation period diseases including placenta accreta spectrum disorders and ectopic pregnancy; 3) mid-term pregnancy diseases like preeclampsia; and 4) late pregnancy diseases such as deep vein thrombosis during pregnancy. The systematic introduction covers the progress made by nanomedicine in various disease areas. Finally, this article discusses the challenges faced by these nanomedicines from research to clinical translation while also highlighting future directions.
Collapse
Affiliation(s)
- Qinrui Fu
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Maternal and Child Health Care Hospital of Shandong Province Affiliated to Qingdao University, Qingdao University, Jinan 250014, China
- Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Lejun Fu
- School of Chemistry and Materials Science, Anhui Normal University, Wuhu 230022, China
| |
Collapse
|
8
|
Fodor Duric L, Belčić V, Oberiter Korbar A, Ćurković S, Vujicic B, Gulin T, Muslim J, Gulin M, Grgurević M, Catic Cuti E. The Role of SHBG as a Marker in Male Patients with Metabolic-Associated Fatty Liver Disease: Insights into Metabolic and Hormonal Status. J Clin Med 2024; 13:7717. [PMID: 39768643 PMCID: PMC11677371 DOI: 10.3390/jcm13247717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/11/2024] [Accepted: 12/12/2024] [Indexed: 01/03/2025] Open
Abstract
Background: Metabolic-associated fatty liver disease (MAFLD) is a spectrum of liver diseases linked to insulin resistance (IR), type 2 diabetes, and metabolic disorders. IR accelerates fat accumulation in the liver, worsening MAFLD. Regular physical activity and weight loss can improve liver function, reduce fat, and lower cardiovascular risk. This study examines the role of sex hormone-binding globulin (SHBG) in MAFLD, focusing on its potential as a biomarker and its relationship with insulin resistance. Methods: The study included 98 male patients (ages 30-55) with MAFLD, identified through systematic examinations, and 74 healthy male controls. All participants underwent abdominal ultrasound and blood tests after fasting, assessing markers such as glucose, liver enzymes (AST, ALT, γGT), lipids (cholesterol, triglycerides), insulin, SHBG, estradiol, and testosterone. SHBG levels were analyzed in relation to body mass index (BMI) and age. Results: A significant association was found between low SHBG levels and the presence of fatty liver. Individuals with MAFLD had lower SHBG levels compared to controls. BMI and age were key factors influencing SHBG, with higher BMI linked to lower SHBG in younger men, while SHBG remained stable in older individuals regardless of BMI. Conclusion: SHBG may serve as a valuable biomarker for early detection and risk assessment of MAFLD. The complex relationship between SHBG, BMI, and age highlights the importance of considering both hormonal and metabolic factors when assessing fatty liver risk. Our findings support the need for comprehensive metabolic evaluations in clinical practice.
Collapse
Affiliation(s)
- Ljiljana Fodor Duric
- School of Medicine, University of Catholica Croatica, 10000 Zagreb, Croatia
- Department of Nephrology and Arterial Hypertension, Medikol Polyclinic, 10000 Zagreb, Croatia
| | - Velimir Belčić
- Medikol Polyclinic, 10000 Zagreb, Croatia; (V.B.); (J.M.)
| | | | - Sanja Ćurković
- Faculty of Kinesiology, University of Zagreb, 10000 Zagreb, Croatia;
- Faculty of Agriculture, University of Zagreb, 10000 Zagreb, Croatia
| | - Bozidar Vujicic
- School of Medicine, University of Rijeka, 10000 Rijeka, Croatia;
- Department of Nephrology, Dialysis and Transplantation, University Hospital Center Rijeka, 10000 Rijeka, Croatia
| | - Tonko Gulin
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia;
- Department of Nephrology and Arterial Hypertension, University Hospital Center “Sestre Milosrdnice”, 10000 Zagreb, Croatia
| | - Jelena Muslim
- Medikol Polyclinic, 10000 Zagreb, Croatia; (V.B.); (J.M.)
| | - Matko Gulin
- Department of Radiology, University Hospital Center “Sestre Milosrdnice”, 10000 Zagreb, Croatia;
| | - Mladen Grgurević
- Department of Diabetes, Endocrinology and Metabolic Diseases Vuk Vrhovac, Merkur University Hospital, 10000 Zagreb, Croatia;
| | | |
Collapse
|
9
|
Cherubini A, Della Torre S, Pelusi S, Valenti L. Sexual dimorphism of metabolic dysfunction-associated steatotic liver disease. Trends Mol Med 2024; 30:1126-1136. [PMID: 38890029 DOI: 10.1016/j.molmed.2024.05.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 05/22/2024] [Accepted: 05/23/2024] [Indexed: 06/20/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most common chronic liver condition. MASLD is a sexually dimorphic condition, with its development and progression influenced by sex chromosomes and hormones. Estrogens typically protect against, whereas androgens promote, MASLD. Therapeutic approaches for a sex-specific personalized medicine include estrogen replacement, androgen blockers, and novel drugs targeting hormonal pathways. However, the interactions between hormonal factors and inherited genetic variation impacts MASLD risk, necessitating more tailored therapies. Understanding sex disparities and the role of estrogens could improve MASLD interventions and management, whereas clinical trials addressing sex differences are crucial for advancing personalized treatment. This review explores the underappreciated impact of sexual dimorphism in MASLD and discusses the potential therapeutic application of sex-related hormones.
Collapse
Affiliation(s)
- Alessandro Cherubini
- Department of Transfusion Medicine, Precision Medicine Lab, Biological Resource Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Sara Della Torre
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, 20133 Milan, Italy
| | - Serena Pelusi
- Department of Transfusion Medicine, Precision Medicine Lab, Biological Resource Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Luca Valenti
- Department of Transfusion Medicine, Precision Medicine Lab, Biological Resource Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
10
|
Shan GY, Wan H, Zhang YX, Cheng JY, Qiao DR, Liu YY, Shi WN, Li HJ. Pathogenesis and research progress of nonalcoholic fatty liver disease/nonalcoholic steatohepatitis. World J Hepatol 2024; 16:1142-1150. [PMID: 39474575 PMCID: PMC11514618 DOI: 10.4254/wjh.v16.i10.1142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 09/05/2024] [Accepted: 09/23/2024] [Indexed: 10/21/2024] Open
Abstract
In this editorial, we comment on the article by Mei et al. Nonalcoholic steatohepatitis (NASH) is a severe inflammatory subtype of nonalcoholic fatty liver disease (NAFLD) with pathological features including steatosis, hepatocellular damage, and varying degrees of fibrosis. With the epidemic of metabolic diseases and obesity, the prevalence of NAFLD in China has increased, and it is now similar to that in developed countries; thus, NAFLD has become a major chronic liver disease in China. Human epidemiological data suggest that estrogen has a protective effect on NASH in premenopausal women and that sex hormones influence the development of liver disease. This review focuses on the pathogenesis, treatment, and relationship between NASH and other diseases as well as on the relationship between NASH and sex hormone metabolism, with the aim of providing new strategies for the treatment of NASH.
Collapse
Affiliation(s)
- Guan-Yue Shan
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun 130061, Jilin Province, China
| | - Hui Wan
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun 130061, Jilin Province, China
| | - Yu-Xin Zhang
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun 130061, Jilin Province, China
| | - Jun-Ya Cheng
- Department of Bioengineering, Pharmacy School of Jilin University, Changchun 130061, Jilin Province, China
| | - Duan-Rui Qiao
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun 130061, Jilin Province, China
| | - Yi-Ying Liu
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun 130061, Jilin Province, China
| | - Wen-Na Shi
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun 130061, Jilin Province, China
| | - Hai-Jun Li
- Institute of Liver Diseases, Institute of Translational Medicine, The First Hospital of Jilin University, Changchun 130061, Jilin Province, China
| |
Collapse
|
11
|
Eid N, Bhatnagar P, Chan LL, Garcia-Macia M. Suppression of hepatic steatosis in non-alcoholic steatohepatitis model by modified Xiaoyao San formula: Evidence, mechanisms and perspective. World J Hepatol 2024; 16:1388-1392. [DOI: 10.4254/wjh.v16.i10.1388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 09/24/2024] [Accepted: 10/11/2024] [Indexed: 11/22/2024] Open
Abstract
In this letter, we comment on a recent publication by Mei et al, in the World Journal of Hepatology, investigating the hepatoprotective effects of the modified Xiaoyao San (MXS) formula in a male rat model of non-alcoholic steatohepatitis (NASH). The authors found that MXS treatment mitigated hepatic steatosis and inflammation in the NASH model, as evidenced by the reduction in lipid droplets (LDs), fibrosis markers and lipogenic factors. Interestingly, these hepatoprotective effects were associated with androgen upregulation (based on metabolomics analysis of male steroid hormone metabolites), adenosine 5’-monophosphate-activated protein kinase (AMPK) activation, and restoration of phosphatase and tensin homolog (PTEN) expression. However, the authors did not clearly discuss the relationships between MXS-induced hepatic steatosis reduction in the NASH model, and androgen upregulation, AMPK activation, and restoration of PTEN expression. This editorial emphasizes the reported mechanisms and explains how they act or interact with each other to reduce hepatic steatosis and inflammation in the NASH model. As a perspective, we propose additional mechanisms (such as autophagy/lipophagy activation in hepatocytes) for the clearance of LDs and suppression of hepatic steatosis by MXS in the NASH model. A proper understanding of the mechanisms of MXS-induced reduction of hepatic steatosis might help in the treatment of NASH and related diseases.
Collapse
Affiliation(s)
- Nabil Eid
- Department of Anatomy, Division of Human Biology, School of Medicine, IMU University, Kuala Lumpur 57000, Malaysia
| | - Payal Bhatnagar
- Department of Pharmaceutical Technology, School of pharmacy, IMU University, Kuala Lumpur 57000, Malaysia
| | - Li-Li Chan
- Department of Pathology and Pharmacology, School of Medicine, IMU University, Kuala Lumpur 57000, Malaysia
| | - Marina Garcia-Macia
- Institute of Functional Biology and Genomics, Department of Biochemistry and Molecular Biology, University of Salamanca, Salamanca 37007, Spain
| |
Collapse
|
12
|
Shan GY, Wan H, Zhang YX, Cheng JY, Qiao DR, Liu YY, Shi WN, Li HJ. Pathogenesis and research progress of nonalcoholic fatty liver disease/nonalcoholic steatohepatitis. World J Hepatol 2024; 16:1322-1330. [DOI: 10.4254/wjh.v16.i10.1322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 09/05/2024] [Accepted: 09/23/2024] [Indexed: 11/22/2024] Open
Abstract
In this editorial, we comment on the article by Mei et al. Nonalcoholic steatohepatitis (NASH) is a severe inflammatory subtype of nonalcoholic fatty liver disease (NAFLD) with pathological features including steatosis, hepatocellular damage, and varying degrees of fibrosis. With the epidemic of metabolic diseases and obesity, the prevalence of NAFLD in China has increased, and it is now similar to that in developed countries; thus, NAFLD has become a major chronic liver disease in China. Human epidemiological data suggest that estrogen has a protective effect on NASH in premenopausal women and that sex hormones influence the development of liver disease. This review focuses on the pathogenesis, treatment, and relationship between NASH and other diseases as well as on the relationship between NASH and sex hormone metabolism, with the aim of providing new strategies for the treatment of NASH.
Collapse
Affiliation(s)
- Guan-Yue Shan
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun 130061, Jilin Province, China
| | - Hui Wan
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun 130061, Jilin Province, China
| | - Yu-Xin Zhang
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun 130061, Jilin Province, China
| | - Jun-Ya Cheng
- Department of Bioengineering, Pharmacy School of Jilin University, Changchun 130061, Jilin Province, China
| | - Duan-Rui Qiao
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun 130061, Jilin Province, China
| | - Yi-Ying Liu
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun 130061, Jilin Province, China
| | - Wen-Na Shi
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun 130061, Jilin Province, China
| | - Hai-Jun Li
- Institute of Liver Diseases, Institute of Translational Medicine, The First Hospital of Jilin University, Changchun 130061, Jilin Province, China
| |
Collapse
|
13
|
Eid N, Bhatnagar P, Chan LL, Garcia-Macia M. Suppression of hepatic steatosis in non-alcoholic steatohepatitis model by modified Xiaoyao San formula: Evidence, mechanisms and perspective. World J Hepatol 2024; 16:1208-1212. [PMID: 39474573 PMCID: PMC11514612 DOI: 10.4254/wjh.v16.i10.1208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 09/24/2024] [Accepted: 10/11/2024] [Indexed: 10/21/2024] Open
Abstract
In this letter, we comment on a recent publication by Mei et al, in the World Journal of Hepatology, investigating the hepatoprotective effects of the modified Xiaoyao San (MXS) formula in a male rat model of non-alcoholic steatohepatitis (NASH). The authors found that MXS treatment mitigated hepatic steatosis and inflammation in the NASH model, as evidenced by the reduction in lipid droplets (LDs), fibrosis markers and lipogenic factors. Interestingly, these hepatoprotective effects were associated with androgen upregulation (based on metabolomics analysis of male steroid hormone metabolites), adenosine 5'-monophosphate-activated protein kinase (AMPK) activation, and restoration of phosphatase and tensin homolog (PTEN) expression. However, the authors did not clearly discuss the relationships between MXS-induced hepatic steatosis reduction in the NASH model, and androgen upregulation, AMPK activation, and restoration of PTEN expression. This editorial emphasizes the reported mechanisms and explains how they act or interact with each other to reduce hepatic steatosis and inflammation in the NASH model. As a perspective, we propose additional mechanisms (such as autophagy/lipophagy activation in hepatocytes) for the clearance of LDs and suppression of hepatic steatosis by MXS in the NASH model. A proper understanding of the mechanisms of MXS-induced reduction of hepatic steatosis might help in the treatment of NASH and related diseases.
Collapse
Affiliation(s)
- Nabil Eid
- Department of Anatomy, Division of Human Biology, School of Medicine, IMU University, Kuala Lumpur 57000, Malaysia.
| | - Payal Bhatnagar
- Department of Pharmaceutical Technology, School of pharmacy, IMU University, Kuala Lumpur 57000, Malaysia
| | - Li-Li Chan
- Department of Pathology and Pharmacology, School of Medicine, IMU University, Kuala Lumpur 57000, Malaysia
| | - Marina Garcia-Macia
- Institute of Functional Biology and Genomics, Department of Biochemistry and Molecular Biology, University of Salamanca, Salamanca 37007, Spain
| |
Collapse
|
14
|
Papadimitriou K, Mousiolis AC, Mintziori G, Tarenidou C, Polyzos SA, Goulis DG. Hypogonadism and nonalcoholic fatty liver disease. Endocrine 2024; 86:28-47. [PMID: 38771482 DOI: 10.1007/s12020-024-03878-1] [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: 01/15/2024] [Accepted: 05/12/2024] [Indexed: 05/22/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD), recently proposed to be renamed to metabolic dysfunction-associated steatotic liver disease (MASLD), is a major global public health concern, affecting approximately 25-30% of the adult population and possibly leading to cirrhosis, hepatocellular carcinoma, and liver transplantation. The liver is involved in the actions of sex steroids via their hepatic metabolism and production of the sex hormone-binding globulin (SHBG). Liver disease, including NAFLD, is associated with reproductive dysfunction in men and women, and the prevalence of NAFLD in patients with hypogonadism is considerable. A wide spectrum of possible pathophysiological mechanisms linking NAFLD and male/female hypogonadism has been investigated. As therapies targeting NAFLD may impact hypogonadism in men and women, and vice versa, treatments of the latter may affect NAFLD, and an insight into their pathophysiological pathways is imperative. This paper aims to elucidate the complex association between NAFLD and hypogonadism in men and women and discuss the therapeutic options and their impact on both conditions.
Collapse
Affiliation(s)
- Kasiani Papadimitriou
- Unit of Reproductive Endocrinology, First Department of Obstetrics and Gynecology, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece.
| | - Athanasios C Mousiolis
- Unit of Reproductive Endocrinology, First Department of Obstetrics and Gynecology, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Gesthimani Mintziori
- Unit of Reproductive Endocrinology, First Department of Obstetrics and Gynecology, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | - Stergios A Polyzos
- First Laboratory of Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Dimitrios G Goulis
- Unit of Reproductive Endocrinology, First Department of Obstetrics and Gynecology, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
15
|
Marycz K, Wiatrak B, Irwin-Houston JM, Marcinkowska K, Mularczyk M, Bourebaba L. Sex hormone binding globulin (SHBG) modulates mitochondrial dynamics in PPARγ-depleted equine adipose derived stromal cells. J Mol Med (Berl) 2024; 102:1015-1036. [PMID: 38874666 PMCID: PMC11269461 DOI: 10.1007/s00109-024-02459-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 03/28/2024] [Accepted: 06/05/2024] [Indexed: 06/15/2024]
Abstract
Peroxisome proliferator-activated receptor gamma (PPARγ) is a transcription factor that promotes adipogenesis, lipid uptake and storage, insulin sensitivity, and glucose metabolism. Hence, defects in PPARγ have been associated to the development of metabolic disorders. Sex hormone-binding globulin (SHBG) is a glycoprotein primarily produced in the liver that regulates the bioavailability of sex hormones. Alike PPARγ, low SHBG levels have been correlated with insulin resistance and associated endocrine abnormalities. Therefore, this study aimed to verify whether SHBG may restore depleted PPARγ functions and thus serve as a new candidate for the management of metabolic conditions. A model of equine adipose-derived stromal cells (EqASCs) has been used, in which a PPARγ silencing and SHBG treatment have been achieved to determine the changes in cell viability, premature senescence, oxidative stress, and mitochondrial functions. Obtained data demonstrated that loss in PPARγ triggers cell apoptosis which is not reversed by SHBG application. Moreover, PPARγ knockdown cells exhibited premature senescence, which has been substantially alleviated by SHBG concomitantly to increased BAX/BCL2 ratio, suggesting a possible effect on senescence-induced apoptosis resistance. Interestingly, PPARγ silencing induced a significant alteration in mitochondrial membrane potential as well as the expression of dynamics and metabolism-related markers. SHBG treatment enabled to ameliorate the transmembrane potential, to normalize the expression levels of key dynamics and metabolism mediators, and to restore the protein levels of PINK, which is critically involved in mitochondria recycling machinery. Presented data suggest that SHBG may provide new mechanistic insights into the regulation of PPARγ functions, and thus offers a preliminary picture on a possible SHBG-PPARγ metabolic crosstalk. KEY MESSAGES : PPARγ is a transcription factor that tightly regulates cell metabolism. Low SHBG levels correlate with insulin resistance and associated endocrine abnormalities. PPARγ silencing reduces cell viability, triggers premature senescence and profound mitochondrial failure in equine ASCs. SHBG protein reverses senescent phenotype and apoptosis resistance of PPARγ- ASCs. SHBG improves mitochondrial dynamics and metabolism following PPARγ knockdown. SHBG might serve as a PPARγ potential mimicking agent for the modulation of ASCs metabolic processes.
Collapse
Affiliation(s)
- Krzysztof Marycz
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Norwida 27B, 50-375, Wrocław, Poland
- International Institute of Translational Medicine, Jesionowa 11, Malin, 55-114, Wisznia Mała, Poland
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California, Davis, Davis, CA, 95516, USA
| | - Benita Wiatrak
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Norwida 27B, 50-375, Wrocław, Poland
| | - Jennifer M Irwin-Houston
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Norwida 27B, 50-375, Wrocław, Poland
| | - Klaudia Marcinkowska
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Norwida 27B, 50-375, Wrocław, Poland
| | - Malwina Mularczyk
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Norwida 27B, 50-375, Wrocław, Poland
- International Institute of Translational Medicine, Jesionowa 11, Malin, 55-114, Wisznia Mała, Poland
| | - Lynda Bourebaba
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Norwida 27B, 50-375, Wrocław, Poland.
| |
Collapse
|
16
|
Bin DH, Liu F, Peng KP, Zhan M, Tan Y, Liu Q, Tang W, Mo ZN, Peng XJ, Tian GX. The relationship between follicle-stimulating hormone and metabolic dysfunction-associated fatty liver disease in men. Nutr Diabetes 2024; 14:52. [PMID: 38991999 PMCID: PMC11239811 DOI: 10.1038/s41387-024-00314-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 06/19/2024] [Accepted: 07/04/2024] [Indexed: 07/13/2024] Open
Abstract
OBJECTIVES The present study aimed to investigate the relationship between male hormones and metabolic dysfunction-associated fatty liver disease (MAFLD) in males. METHODS Data from the Fangchenggang Area Male Health and Examination Survey (FAMHES) were used to analyze the male hormone levels between MAFLD patients and controls. Univariate and multivariate logistic regression analyses were performed to identify risk factors for MAFLD. Receiver operating characteristic curve analysis was used to assess the diagnostic performance of male hormones for MAFLD. RESULT A total of 1578 individuals were included, with 482 individuals (30.54%) of MAFLD, including 293 (18.57%) with mild disease and 189 (11.98%) with moderate-to-severe disease. The MAFLD patients were significantly older than those without MAFLD. The LH, FSH, and SHBG levels in the MAFLD patients were significantly greater than those in the control group. Age, FSH, LH, SHBG, and estradiol were all risk factors for MAFLD. Age, FSH, and LH were risk factors for moderate-to-severe MAFLD. FSH was an independent risk factor for MAFLD and moderate-to-severe MAFLD. FSH showed an excellent diagnostic value, with an AUC of 0.992 alone and 0.996 after adjusting age. CONCLUSIONS Our findings indicate that FSH may be a potential diagnostic and predictive biomarker for MAFLD.
Collapse
Affiliation(s)
- Dong-Hua Bin
- Department of Anus and Intesine, The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Fang Liu
- Department of Ultrasoud, The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Ke-Ping Peng
- Department of Otorhinolaryngology-Head and Neck surgery, The first Hospital, Hunan University of Chinese Medicine, Changsha, China
| | - Min Zhan
- Department of Anus and Intesine, The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Yan Tan
- Department of Ultrasoud, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Qiao Liu
- Department of Ultrasoud, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Wang Tang
- Department of Ultrasoud, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Zeng-Nan Mo
- Centre for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
| | - Xiong-Jun Peng
- Department of Medical Equipment, The Second Xiangya Hospital, Central South University, Changsha, China.
| | - Gui-Xiang Tian
- Department of Ultrasoud, The Second Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
17
|
Hu R, Long S, Luo M, Tang B, Tan T, Dong W, Wang Q, Zhang J. Hyperglycemia Inhibits Hepatic SHBG Synthesis Through the NGBR-AMPK-HNF4 Pathway in Rats with Polycystic Ovary Syndrome Induced by Letrozole in Combination with a High-Fat Diet. Mol Nutr Food Res 2024; 68:e2300915. [PMID: 38862276 DOI: 10.1002/mnfr.202300915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 02/25/2024] [Indexed: 06/13/2024]
Abstract
SCOPE Polycystic ovary syndrome (PCOS) is closely related to non-alcoholic fatty liver disease (NAFLD), and sex hormone-binding globulin (SHBG) is a glycoprotein produced by the liver. Hepatic lipogenesis inhibits hepatic SHBG synthesis, which leads to hyperandrogenemia and ovarian dysfunction in PCOS. Therefore, this study aims to characterize the mechanism whereby liver lipogenesis inhibits SHBG synthesis. METHODS AND RESULTS This study establishes a rat model of PCOS complicated by NAFLD using a high-fat diet in combination with letrozole and performs transcriptomic analysis of the liver. Transcriptomic analysis of the liver shows that the expression of neurite growth inhibitor-B receptor (NgBR), hepatocyte nuclear factor 4α (HNF4α), and SHBG is low. Meantime, HepG2 cells are treated with palmitic acid (PA) to model NAFLD in vitro, which causes decreases in the expression of NgBR, HNF4α, and SHBG. However, the expression of HNF4α and SHBG is restored by treatment with the AMP-activated protein kinase (AMPK) agonist AICAR. CONCLUSIONS NgBR regulates the expression of HNF4α by activating the AMPK signaling pathway, thereby affecting the synthesis of SHBG in the liver. Further mechanistic studies regarding the effect of liver fat on NGBR expression are warranted.
Collapse
Affiliation(s)
- Rao Hu
- The First Affiliated Hospital, Gynecology &Obstetrics and Reproductive Medical Center, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Institute of Applied Anatomy and Reproductive Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan, 421001, China
| | - Shuanglian Long
- The First Affiliated Hospital, Gynecology &Obstetrics and Reproductive Medical Center, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Institute of Applied Anatomy and Reproductive Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan, 421001, China
| | - Min Luo
- The First Affiliated Hospital, Gynecology &Obstetrics and Reproductive Medical Center, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Institute of Applied Anatomy and Reproductive Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan, 421001, China
| | - Bowen Tang
- Institute of Applied Anatomy and Reproductive Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan, 421001, China
| | - Tao Tan
- Institute of Applied Anatomy and Reproductive Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan, 421001, China
| | - Weilei Dong
- The First Affiliated Hospital, Gynecology &Obstetrics and Reproductive Medical Center, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Qian Wang
- The First Affiliated Hospital, Gynecology &Obstetrics and Reproductive Medical Center, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Jiaming Zhang
- The First Affiliated Hospital, Gynecology &Obstetrics and Reproductive Medical Center, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| |
Collapse
|
18
|
Mladenović D, Vesković M, Šutulović N, Hrnčić D, Stanojlović O, Radić L, Macut JB, Macut D. Adipose-derived extracellular vesicles - a novel cross-talk mechanism in insulin resistance, non-alcoholic fatty liver disease, and polycystic ovary syndrome. Endocrine 2024; 85:18-34. [PMID: 38285412 DOI: 10.1007/s12020-024-03702-w] [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: 11/17/2023] [Accepted: 01/14/2024] [Indexed: 01/30/2024]
Abstract
Obesity is the best described risk factor for the development of non-alcoholic fatty liver disease (NAFLD)/metabolic dysfunction associated steatotic liver disease (MASLD) and polycystic ovary syndrome (PCOS) while the major pathogenic mechanism linking these entities is insulin resistance (IR). IR is primarily caused by increased secretion of proinflammatory cytokines, adipokines, and lipids from visceral adipose tissue. Increased fatty acid mobilization results in ectopic fat deposition in the liver which causes endoplasmic reticulum stress, mitochondrial dysfunction, and oxidative stress resulting in increased cytokine production and subsequent inflammation. Similarly, IR with hyperinsulinemia cause hyperandrogenism, the hallmark of PCOS, and inflammation in the ovaries. Proinflammatory cytokines from both liver and ovaries aggravate IR thus providing a complex interaction between adipose tissue, liver, and ovaries in inducing metabolic abnormalities in obese subjects. Although many pathogenic mechanisms of IR, NAFLD/MASLD, and PCOS are known, there is still no effective therapy for these entities suggesting the need for further evaluation of their pathogenesis. Extracellular vesicles (EVs) represent a novel cross-talk mechanism between organs and include membrane-bound vesicles containing proteins, lipids, and nucleic acids that may change the phenotype and function of target cells. Adipose tissue releases EVs that promote IR, the development of all stages of NAFLD/MASLD and PCOS, while mesenchymal stem cell-derived AVs may alleviate metabolic abnormalities and may represent a novel therapeutic device in NAFLD/MASLD, and PCOS. The purpose of this review is to summarize the current knowledge on the role of adipose tissue-derived EVs in the pathogenesis of IR, NAFLD/MASLD, and PCOS.
Collapse
Affiliation(s)
- Dušan Mladenović
- Institute of Pathophysiology "Ljubodrag Buba Mihailovic", Faculty of Medicine, University of Belgrade, Belgrade, Serbia.
| | - Milena Vesković
- Institute of Pathophysiology "Ljubodrag Buba Mihailovic", Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Nikola Šutulović
- Laboratory for Neurophysiology, Institute of Medical Physiology "Richard Burian", Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Dragan Hrnčić
- Laboratory for Neurophysiology, Institute of Medical Physiology "Richard Burian", Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Olivera Stanojlović
- Laboratory for Neurophysiology, Institute of Medical Physiology "Richard Burian", Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Lena Radić
- Clinic for Endocrinology, Diabetes and Metabolic Diseases, University Clinical Centre of Serbia, Belgrade, Serbia
| | - Jelica Bjekić Macut
- University of Belgrade Faculty of Medicine, Department of Endocrinology, UMC Bežanijska kosa, Belgrade, Serbia
| | - Djuro Macut
- University of Belgrade Faculty of Medicine, Clinic for Endocrinology, Diabetes and Metabolic Diseases, University Clinical Centre of Serbia, Belgrade, Serbia
| |
Collapse
|
19
|
Yan Z, Zheng Z, Xia T, Ni Z, Dou Y, Liu X. Causal relationship between gut microbiome and sex hormone-binding globulin: A bidirectional two-sample Mendelian randomization study. Am J Reprod Immunol 2024; 91:e13824. [PMID: 38356386 DOI: 10.1111/aji.13824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 01/23/2024] [Accepted: 02/01/2024] [Indexed: 02/16/2024] Open
Abstract
PROBLEM Currently, there is a variety of evidence linking the gut microbiota to changes in sex hormones. In contrast, the causal relationship between SHBG, a carrier of sex hormones, and the gut microbiota is unclear. METHOD OF STUDY Bidirectional two-sample Mendelian randomization (MR) analysis was used to detect the causal effect between SHBG and the gut microbiome. Summary statistics of genome-wide association studies (GWASs) for the gut microbiome and SHBG were obtained from public datasets. Inverse-variance weighting (IVW), weighted median, weighted mode, MR-Egger and simple mode methods were used to operate the MR analysis. F-statistics and sensitivity analyses performed to evaluate bias and reliability. RESULTS When we set gut microbiome as exposure and SHBG as outcome, we identified nine causal relationships. In males, Coprobacter (PIVW = 2.01 × 10-6 ), Ruminococcus2 (PIVW = 3.40 × 10-5 ), Barnesiella (PIVW = 2.79 × 10-2 ), Actinobacteria (PIVW = 3.25 × 10-2 ) and Eubacterium fissicatena groups (PIVW = 3.64 × 10-2 ) were associated with lower SHBG levels; Alphaproteobacteria (PIVW = 1.61 × 10-2 ) is associated with higher SHBG levels. In females, Lachnoclostridium (PIVW = 9.75 × 10-3 ) and Defluviitaleaceae UCG011 (PIVW = 3.67 × 10-2 ) were associated with higher SHBG levels; Victivallaceae (PIVW = 2.23 × 10-2 ) was associated with lower SHBG levels. According to the results of reverse MR analysis, three significant causal effect of SHBG was found on gut microbiota. In males, Dorea (PIVW = 4.17 × 10-2 ) and Clostridiales (PIVW = 4.36 × 10-2 ) were associated with higher SHBG levels. In females, Lachnoclostridium (PIVW = 7.44 × 10-4 ) was associated with higherr SHBG levels. No signifcant heterogeneity of instrumental variables or horizontal pleiotropy was found in bidirectional two-sample MR analysis. CONCLUSIONS This study may provide new insights into the causal relationship between the gut microbiome and sex hormone-binding protein levels, as well as new treatment and prevention strategies for diseases such as abnormal changes in sex hormones.
Collapse
Affiliation(s)
- Ziqiao Yan
- Department of Traditional Chinese Medicine, The First Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Zheng Zheng
- Department of Gynecology, Guang'anmen Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, China
| | - Tiantian Xia
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Zhexin Ni
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Yongqi Dou
- Department of Traditional Chinese Medicine, The First Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Xinmin Liu
- Department of Gynecology, Guang'anmen Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, China
| |
Collapse
|
20
|
Manzhalii E, Tatarchuk T, Tutchenko T, Kosei N, Mnevets R. Relationships between nonalcoholic fatty liver disease and polycystic ovary syndrome. REPRODUCTIVE ENDOCRINOLOGY 2023:40-45. [DOI: 10.18370/2309-4117.2023.70.40-45] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Polycystic ovary syndrome (PCOS) and non-alcoholic fatty liver disease (NAFLD) are two common non-infectious pathologies. Their frequency and medico-social significance have increased significantly over the past decades and reflect the effects of radical changes in human lifestyle on human health in a transgenerational aspect. Due to absence a complete understanding of the etiology and pathogenesis of PCOS and NAFLD, modern medicine still does not have etiopathogenetic methods of treating these frequent diseases. Considering epidemiological and pathogenetic data PCOS and NAFLD can be regarded as related pathologies, which creates the prospect of improving the effectiveness of their management if the joint approach to scientific research and implementation of their results in medical practice is applied.The purpose of the review is to summarize the current scientific data on the common pathophysiological and clinical relationships between PCOS and NAFLD, which can contribute to improving the effectiveness management of both diseases.Pathogenetic relationships between PCOS and NAFLD are bidirectional. According to current data, it seems possible to consider PCOS as a significant risk factor for the development of NAFLD in women of reproductive age both in the presence of excess weight and with normal body weight.As is known, that liver is involved in the clearance of androgens by the formation of sex steroid-binding globulin, as well as in the inactivation of insulin. Disruption of these metabolic processes due to liver pathology can lead to the development or strengthening of biochemical and clinical hyperandrogenism and the development of hyperinsulinemia and insulin resistance (IR).Conclusions. The presence of fatty liver and especially steatohepatitis deepens the hormonal disorders inherent in PCOS (primarily, an increase of free androgens, IR and ovulatory dysfunction), as well as potentiates and accelerates the development of cardiometabolic complications, including atherogenic dyslipidemia, dysglycemia, metabolic syndrome and cardiovascular complications. Early detection of NAFLD in women with PCOS, regardless of the presence of obesity, may improve the effectiveness of PCOS management and prevention of its cardiometabolic risks.
Collapse
Affiliation(s)
- E.H. Manzhalii
- O.O. Bogomolets National Medical University; Ukrainian Liver Foundation; Global Longevity Institute; Clinic Verum Expert, Kyiv
| | - T.F. Tatarchuk
- SI “O.M. Lukyanova Institute of Pediatrics, Obstetrics and Gynecology of the NAMS of Ukraine”;State Scientific Institution “Center for Innovative Medical Technologies of the NAS of Ukraine”, Kyiv
| | - T.M. Tutchenko
- SI “O.M. Lukyanova Institute of Pediatrics, Obstetrics and Gynecology of the NAMS of Ukraine”; SSI “Center for Innovative Medical Technologies of the NAS of Ukraine”; “Dila” Medical Laboratory, Kyiv
| | - N.V. Kosei
- SI “O.M. Lukyanova Institute of Pediatrics, Obstetrics and Gynecology of the NAMS of Ukraine”; SSI “Center for Innovative Medical Technologies of the NAS of Ukraine”, Kyiv
| | - R.O. Mnevets
- ESC “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv; SI “O.M. Lukyanova Institute of Pediatrics, Obstetrics and Gynecology of the NAMS of Ukraine”, Kyiv, Ukraine
| |
Collapse
|
21
|
Ciarambino T, Crispino P, Guarisco G, Giordano M. Gender Differences in Insulin Resistance: New Knowledge and Perspectives. Curr Issues Mol Biol 2023; 45:7845-7861. [PMID: 37886939 PMCID: PMC10605445 DOI: 10.3390/cimb45100496] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/11/2023] [Accepted: 09/14/2023] [Indexed: 10/28/2023] Open
Abstract
Insulin resistance is the main mechanism in a whole series of pathological conditions, which are not only of metabolic interest but also of a systemic type. This phenomenon means that the body's cells become less sensitive to the hormone insulin, leading to higher levels of insulin in the blood. Insulin resistance is a phenomenon that can be found in both men and women and in particular, in the latter, it is found mainly after menopause. Premenopause, hormonal fluctuations during the menstrual cycle, and the presence of estrogen can affect insulin sensitivity. Androgens, such as testosterone, are typically higher in men and can contribute to insulin resistance. In both sexes, different human body types affect the distribution and location of body fat, also influencing the development of diabetes and cardiovascular disease. Insulin resistance is also associated with some neurological and neurogenerative disorders, polycystic ovary syndrome, atherosclerosis, and some of the main neoplastic pathologies. A healthy lifestyle, including regular physical activity, a balanced diet, and self-maintenance, can help to prevent the onset of insulin resistance, regardless of gender, although the different habits between men and women greatly affect the implementation of preventative guidelines that help in fighting the manifestations of this metabolic disorder. This review may help to shed light on gender differences in metabolic diseases by placing a necessary focus on personalized medical management and by inspiring differentiated therapeutic approaches.
Collapse
Affiliation(s)
- Tiziana Ciarambino
- Internal Medicine Department, Hospital of Marcianise, 81100 Caserta, Italy
| | - Pietro Crispino
- Internal Medicine Department, Hospital of Latina, 04100 Latina, Italy;
| | - Gloria Guarisco
- Diabetology, University Sapienza of Rome, Hospital of Latina, 04100 Latina, Italy;
| | - Mauro Giordano
- Internal Medicine Department, University of Campania, L. Vanvitelli, 81100 Naples, Italy;
| |
Collapse
|
22
|
Giannouli A, Stefanaki C, Kouskoutis C, Konidari M, Mani I, Konidari K, Markantonis SL, Mantzou A, Dourakis SP, Deligeoroglou E, Bacopoulou F. Hepatokine Profile in Adolescents with Polycystic Ovary Syndrome: A Case-Control Study. J Clin Med 2023; 12:5744. [PMID: 37685811 PMCID: PMC10489115 DOI: 10.3390/jcm12175744] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 08/29/2023] [Accepted: 08/30/2023] [Indexed: 09/10/2023] Open
Abstract
The current guidelines suggest routine screening for non-alcoholic fatty liver disease (NAFLD) in patients with polycystic ovary syndrome (PCOS). Hepatokines seem to be promising surrogate endpoints for the diagnosis and severity of NAFLD. PCOS has its onset in adolescence and its metabolic sequalae begin during the same period. There are scarce data on the hepatokine profile of adolescent PCOS patients. This case-control study examined the serum profile of the hepatokines sex hormone-binding globulin (SHBG), selenoprotein P, fibroblast growth factor 21 (FGF21), and fetuin A in a sample of adolescent PCOS patients, and their association to metabolic and hormonal parameters. The selenoprotein P and SHBG serum concentrations were significantly decreased in PCOS patients vs. the controls (median (IQR), 2.47 (0.40) vs. 2.66 (0.36) μg/mL, p = 0.025; mean ± SD, 41.71 ± 19.41 vs. 54.94 ± 22.12 nmol/L, p = 0.011, respectively), whereas selenoprotein P was significantly and positively associated with testosterone (r = 0.325, p = 0.007) and the free androgen index (r = 0.361, p = 0.002). The SHBG demonstrated multiple significant negative correlations with adverse metabolic parameters. Among the PCOS patients, the FGF21 concentrations were significantly higher in those with NAFLD, whereas a 1 pg/mL increase in the FGF21 concentration increased the odds of NAFLD diagnosis by liver ultrasound by 1%, suggesting FGF21 as a potential biomarker for hepatic disease in females with PCOS in adolescence. Fetuin A was the least differentiated hepatokine between the PCOS patients and controls with the least associations with metabolic and hormonal parameters.
Collapse
Affiliation(s)
- Aikaterini Giannouli
- Center for Adolescent Medicine and UNESCO Chair in Adolescent Health Care, First Department of Pediatrics, Medical School, National and Kapodistrian University of Athens, Aghia Sophia Children’s Hospital, 11527 Athens, Greece; (A.G.); (C.S.); (K.K.)
| | - Charikleia Stefanaki
- Center for Adolescent Medicine and UNESCO Chair in Adolescent Health Care, First Department of Pediatrics, Medical School, National and Kapodistrian University of Athens, Aghia Sophia Children’s Hospital, 11527 Athens, Greece; (A.G.); (C.S.); (K.K.)
| | - Christos Kouskoutis
- Laboratory of Biopharmaceutics and Pharmacokinetics, Department of Pharmaceutical Technology, Faculty of Pharmacy, National and Kapodistrian University of Athens, 15771 Athens, Greece; (C.K.); (S.L.M.)
| | - Marianna Konidari
- Department of Radiology, Medical School, National and Kapodistrian University of Athens, Aretaieio Hospital, 11528 Athens, Greece;
| | - Iliana Mani
- Second Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, Hippokratio Hospital, 11527 Athens, Greece; (I.M.); (S.P.D.)
| | - Konstantina Konidari
- Center for Adolescent Medicine and UNESCO Chair in Adolescent Health Care, First Department of Pediatrics, Medical School, National and Kapodistrian University of Athens, Aghia Sophia Children’s Hospital, 11527 Athens, Greece; (A.G.); (C.S.); (K.K.)
| | - Sophia L. Markantonis
- Laboratory of Biopharmaceutics and Pharmacokinetics, Department of Pharmaceutical Technology, Faculty of Pharmacy, National and Kapodistrian University of Athens, 15771 Athens, Greece; (C.K.); (S.L.M.)
| | - Aimilia Mantzou
- Division of Endocrinology, Metabolism, and Diabetes, First Department of Pediatrics, School of Medicine, National and Kapodistrian University of Athens, Aghia Sophia Children’s Hospital, 11527 Athens, Greece;
| | - Spyridon P. Dourakis
- Second Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, Hippokratio Hospital, 11527 Athens, Greece; (I.M.); (S.P.D.)
| | - Efthymios Deligeoroglou
- Department of Pediatric & Adolescent Gynecology, Mitera Children’s Hospital, 15123 Athens, Greece;
| | - Flora Bacopoulou
- Center for Adolescent Medicine and UNESCO Chair in Adolescent Health Care, First Department of Pediatrics, Medical School, National and Kapodistrian University of Athens, Aghia Sophia Children’s Hospital, 11527 Athens, Greece; (A.G.); (C.S.); (K.K.)
| |
Collapse
|
23
|
Sakr HF, Sirasanagandla SR, Das S, Bima AI, Elsamanoudy AZ. Insulin Resistance and Hypertension: Mechanisms Involved and Modifying Factors for Effective Glucose Control. Biomedicines 2023; 11:2271. [PMID: 37626767 PMCID: PMC10452601 DOI: 10.3390/biomedicines11082271] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 07/18/2023] [Accepted: 08/01/2023] [Indexed: 08/27/2023] Open
Abstract
Factors such as aging, an unhealthy lifestyle with decreased physical activity, snacking, a standard Western diet, and smoking contribute to raising blood pressure to a dangerous level, increasing the risk of coronary artery disease and heart failure. Atherosclerosis, or aging of the blood vessels, is a physiological process that has accelerated in the last decades by the overconsumption of carbohydrates as the primary sources of caloric intake, resulting in increased triglycerides and VLDL-cholesterol and insulin spikes. Classically, medications ranging from beta blockers to angiotensin II blockers and even calcium channel blockers were used alone or in combination with lifestyle modifications as management tools in modern medicine to control arterial blood pressure. However, it is not easy to control blood pressure or the associated complications. A low-carbohydrate, high-fat (LCHF) diet can reduce glucose and insulin spikes, improve insulin sensitivity, and lessen atherosclerosis risk factors. We reviewed articles describing the etiology of insulin resistance (IR) and its impact on arterial blood pressure from databases including PubMed, PubMed Central, and Google Scholar. We discuss how the LCHF diet is beneficial to maintaining arterial blood pressure at normal levels, slowing down the progression of atherosclerosis, and reducing the use of antihypertensive medications. The mechanisms involved in IR associated with hypertension are also highlighted.
Collapse
Affiliation(s)
- Hussein F. Sakr
- Department of Physiology, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat 123, Oman
| | - Srinivasa Rao Sirasanagandla
- Department of Human and Clinical Anatomy, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat 123, Oman; (S.R.S.); (S.D.)
| | - Srijit Das
- Department of Human and Clinical Anatomy, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat 123, Oman; (S.R.S.); (S.D.)
| | - Abdulhadi I. Bima
- Department of Clinical Biochemistry, Faculty of Medicine, King Abdulaziz University, Jeddah 21465, Saudi Arabia; (A.I.B.); (A.Z.E.)
| | - Ayman Z. Elsamanoudy
- Department of Clinical Biochemistry, Faculty of Medicine, King Abdulaziz University, Jeddah 21465, Saudi Arabia; (A.I.B.); (A.Z.E.)
| |
Collapse
|
24
|
Lonardo A. The heterogeneity of metabolic syndrome presentation and challenges this causes in its pharmacological management: a narrative review focusing on principal risk modifiers. Expert Rev Clin Pharmacol 2023; 16:891-911. [PMID: 37722710 DOI: 10.1080/17512433.2023.2259306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 09/12/2023] [Indexed: 09/20/2023]
Abstract
INTRODUCTION Metabolic syndrome (MetS), i.e. the cluster of cardiometabolic risk factors comprising visceral obesity, impaired glucose metabolism, arterial hypertension and atherogenic dyslipidemia, is prevalent globally and exacts a heavy toll on health care expenditures. AREAS COVERED The pathophenotypes of individual traits of the MetS in adults are discussed first, with strong emphasis on nonalcoholic fatty liver disease (NAFLD) and sex differences. Next, I discuss recent studies on phenotypic and outcome heterogeneity of the MetS, highlighting the role of NAFLD, sex, reproductive status, and depressive disorders. In the second half of the article, the therapeutic implications of the variable MetS types and features are analyzed, focusing on the most recent developments, and guidelines. EXPERT OPINION I have identified physiological, pathological, social and medical sources of phenotypical heterogeneity in the MetS and its constitutive traits. Improved understanding of these variables may be utilized in the setting of future precision medicine approaches in the field of metabolic disorders and target organ damage.
Collapse
Affiliation(s)
- Amedeo Lonardo
- Operating Unit of Metabolic Syndrome, Azienda Ospedaliero-Unversitaria di Modena, Ospedale Civile di Baggiovara, Baggiovara, Modena, Italy
| |
Collapse
|
25
|
Hua X, Zhang H, Yang W, Liu G, Zhang S, Wang Y. SFI, a sex hormone binding globulin based nomogram for predicting non-alcoholic fatty liver disease in the Chinese population. Front Endocrinol (Lausanne) 2023; 14:1176019. [PMID: 37334312 PMCID: PMC10276183 DOI: 10.3389/fendo.2023.1176019] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 05/10/2023] [Indexed: 06/20/2023] Open
Abstract
Background The purpose of this study is to establish a novel nomogram model for accurate detection of non-alcoholic fatty liver disease (NAFLD) in the Chinese population based on sex hormone binding globulin (SHBG) and other routine laboratory tests. Methods A total of 1417 participants (1003 testing and 414 validations) were enrolled into the study. Risk factors independently associated with NAFLD were identified and incorporated in the new nomogram, SFI. The performance of nomogram was assessed by analysis of receiver operating characteristic (ROC) curve, calibration curve, and decision curve. Results We formulated a new nomogram incorporating four independent factors: SHBG, body mass index (BMI), ALT/AST, and triglycerides (TG). The nomogram achieved good indexes of area under ROC 0.898 (95% confidence interval 0.865-0.926) in predicting NAFLD, which was significantly superior to previously reported models of FLI, HSI, LFS, and LAP. The calibration curve and decision curve demonstrated high performance and clinical utility of the nomogram in predicting NAFLD. Conclusion The nomogram SFI has high performance in predicting NAFLD in Chinese population and may be used as a cost-effective screening model to assess NAFLD in the general population.
Collapse
Affiliation(s)
- Xiaomin Hua
- Department of Health Care, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Shandong, China
| | - Heping Zhang
- Department of Cardiac Surgery, the Affiliated Hospital of Qingdao University, Shandong, China
| | - Wenru Yang
- Department of Health Care, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Shandong, China
| | - Guotao Liu
- Department of Health Care, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Shandong, China
| | - Suhua Zhang
- Department of Health Care, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Shandong, China
| | - Yingcui Wang
- Department of Health Care, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Shandong, China
- Department of Cardiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Shandong, China
| |
Collapse
|
26
|
Icariin Alleviates Nonalcoholic Fatty Liver Disease in Polycystic Ovary Syndrome by Improving Liver Fatty Acid Oxidation and Inhibiting Lipid Accumulation. MOLECULES (BASEL, SWITZERLAND) 2023; 28:molecules28020517. [PMID: 36677577 PMCID: PMC9861792 DOI: 10.3390/molecules28020517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/03/2023] [Accepted: 01/03/2023] [Indexed: 01/06/2023]
Abstract
(1) Background: Icariin is the main component of the Chinese herb Epimedium. A number of studies have shown that it alleviates abnormal lipid metabolism. However, it is not clear whether and how icariin can ameliorate hepatic steatosis with polycystic ovary syndrome (PCOS). This study was designed to explore the anti-hepatosteatosis effect of icariin in rats with polycystic ovary syndrome. (2) Methods: Female Sprague Dawley(SD)rats were treated with a high-fat diet and letrozole for 21 days to make nonalcoholic fatty liver disease (NAFLD) in the polycystic ovary syndrome model. Then model rats were treated with icariin (by gavage, once daily) for 28 days. Serum hormones and biochemical variables were determined by ELISA or enzyme. RNA-sequence analysis was used to enrich related target pathways. Then, quantitative Real-time PCR (qRT-PCR) and Western blot were performed to verify target genes and proteins. (3) Results: Icariin treatment reduced excess serum levels of Testosterone (T), Estradiol (E2), Luteinizing hormone (LH), Follicle-stimulating hormone (FSH), LH/FSH ratio, insulin, triglycerides (TG), and aspartate aminotransferase (AST) in high-fat diet (HFD) and letrozole fed rats. Meanwhile, icariin ameliorated HFD and letrozole-induced fatty liver, as evidenced by a reduction in excess triglyceride accumulation, vacuolization, and Oil Red O staining area in the liver of model rats. Results of RNA-sequencing, western blotting, and qRT-PCR analyses indicated that icariin up-regulated fatty acid translocase (CD36), in mitochondria, and peroxisome proliferator-activated receptor α (PPARα) expression, which led to the enhancement of fatty acid oxidation molecules, such as cytochrome P450, family 4, subfamily a, polypeptide 3 (CYP4A3), carnitine palmitoyltransferase 1 α (CPT1α), acyl-CoA oxidase 1 (ACOX1), medium-chain acyl-CoA dehydrogenase (MCAD), and long-chain acyl-CoA dehydrogenase (LCAD). Besides, icariin reduced lipid synthesis, which elicited stearoyl-Coenzyme A desaturase 1 (SCD1), fatty acid synthase (FASN), and acetyl-CoA (ACC). (4) Conclusion: Icariin showed an ameliorative effect on hepatic steatosis induced by HFD and letrozole, which was associated with improved fatty acid oxidation and reduced lipid accumulation in the liver.
Collapse
|
27
|
Singeap AM, Huiban L. Endocrinopathies in Nonalcoholic Fatty Liver Disease. ESSENTIALS OF NON-ALCOHOLIC FATTY LIVER DISEASE 2023:183-193. [DOI: 10.1007/978-3-031-33548-8_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|