1
|
Maher S, Atta S, Kamel M, A. Hammam O, Okasha H. Therapeutic Potential and Mechanistic Insights of a Novel Synthetic α-Lactalbumin-Derived Peptide for the Treatment of Liver Fibrosis. J Clin Exp Hepatol 2025; 15:102488. [PMID: 39868009 PMCID: PMC11755051 DOI: 10.1016/j.jceh.2024.102488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 12/09/2024] [Indexed: 01/28/2025] Open
Abstract
Background Liver fibrosis is a serious global health issue, but current treatment options are limited due to a lack of approved therapies capable of preventing or reversing established fibrosis. Aim This study investigated the antifibrotic effects of a synthetic peptide derived from α-lactalbumin in a mouse model of thioacetamide (TAA)-induced liver fibrosis. Methods In silico analyses were conducted to assess the physicochemical properties, pharmacophore features, and docking interactions of the peptide. Mice with induced fibrosis were treated with three different doses of the synthetic peptide (2.5, 5, or 10 μg/kg, twice weekly for 8 weeks). Immunohistochemistry, antioxidant enzyme levels, IGF-1 levels, and expression of fibrosis-related genes were assessed. Results Peptide interacted with human prothrombin's many sites with varying binding affinities. Besides, ligand similarity analysis identified 26 thrombin inhibitors with high Tanimoto scores. The peptide exhibited antifibrotic effects with dose-dependent improvements. The upregulated expression of IGF-1 in all treated groups compared with the pathological untreated group. In contrast, fibrotic markers such as TIMP, PDGF-α, and TGF-β were upregulated in the untreated pathological group but downregulated in the peptide-treated groups. The assessment of IGF-1 concentration in sera demonstrated that the peptide-treated groups exhibited an increase in IGF-1 levels. Histopathological examination of peptide-treated groups showed normal hepatic architecture with hepatocytes arranged in thin plates. Immunohistochemical results of high dose peptide-treated group showed a few numbers of positive αSMA with mild proliferating cell nuclear antigen expression. Conclusion The synthetic α-lactalbumin peptide shows promise as an antifibrotic therapy. Its safety and effectiveness are supported by in silico and in vivo analyses. The peptide's pharmacophore characteristics and potential as a thrombin inhibitor combine with its ability to downregulate fibrotic markers and maintain liver tissue integrity. These findings concluded the potential of this peptide as a promising therapeutic candidate for liver fibrosis, warranting further investigation.
Collapse
Affiliation(s)
- Sara Maher
- Immunology Department, Theodor Bilharz Research Institute, Giza, Egypt
| | - Shimaa Atta
- Immunology Department, Theodor Bilharz Research Institute, Giza, Egypt
| | - Manal Kamel
- Immunology Department, Theodor Bilharz Research Institute, Giza, Egypt
| | - Olfat A. Hammam
- Pathology Department, Theodor Bilharz Research Institute, Giza, Egypt
| | - Hend Okasha
- Biochemistry and Molecular Biology Department, Theodor Bilharz Research Institute, Giza, Egypt
| |
Collapse
|
2
|
Wu F, Zheng F, Li X, Wu D, Li H, Zeng Y, Tang Y, Liu S, Li A. Association between non-skimmed milk consumption and metabolic dysfunction-associated fatty liver disease in US adults: insights from NHANES data. BMC Gastroenterol 2025; 25:270. [PMID: 40251472 PMCID: PMC12007357 DOI: 10.1186/s12876-025-03834-x] [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: 01/30/2025] [Accepted: 04/01/2025] [Indexed: 04/20/2025] Open
Abstract
PURPOSE Previous studies on the association between non-skimmed milk consumption and non-alcoholic fatty liver disease (NAFLD) have reported inconsistent findings, with some suggesting an increased risk and others indicating a protective effect. Moreover, as the research focus has shifted globally from NAFLD to metabolic dysfunction-associated fatty liver disease (MAFLD), there remains limited evidence on the relationship between non-skimmed milk consumption and MAFLD. The objective of this cross-sectional study was to investigate this association using data from the National Health and Nutrition Examination Survey (NHANES). METHODS In this U.S. population-based study, adults with complete information on non-skimmed milk consumption and MAFLD diagnosis from the 2017-March 2020 Pre-Pandemic NHANES were included. MAFLD was defined using the controlled attenuation parameter (CAP). The association between non-skimmed milk consumption and MAFLD was assessed using weighted multivariable logistic regression, adjusting for potential confounders. Subgroup and sensitivity analyses were conducted to evaluate effect modifications and robustness. RESULTS The study involved 3,758 participants in total, 1,423 (37.87%) of whom had MAFLD according to the diagnosis. Frequent non-skimmed milk consumption was independently associated with higher MAFLD risk. Compared to the "Rarely" group (< 1 time/week), the "Sometimes" group (≥ 1 time/week but < 1 time/day) had an odds ratio (OR) of 1.67 (95% CI: 1.32-2.12, P = 0.004), and the "Often" group (≥ 1 time/day) had an OR of 1.36 (95% CI: 1.06-1.75, P = 0.046). Stratified analysis revealed that the association was significantly modified by education level (P for interaction = 0.010), with a stronger association observed among participants with higher education levels. Sensitivity analysis yielded consistent results, further supporting the robustness of the association. CONCLUSION Our findings suggest a significant association between frequent non-skimmed milk consumption and risk of MAFLD, particularly in highly educated individuals. These results highlight the importance of dietary modifications, specifically reducing non-skimmed milk intake, as a potential preventive strategy for MAFLD, especially in high-risk populations.
Collapse
Affiliation(s)
- Futao Wu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fuying Zheng
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xue Li
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Danzhu Wu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Honghao Li
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Gastroenterology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, China
| | - Yingyi Zeng
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yan Tang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Side Liu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
- Department of Gastroenterology, Zhuhai Peoples Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, China.
| | - Aimin Li
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
3
|
Fernández-Garza LE, Guillen-Silva F, Sotelo-Ibarra MA, Domínguez-Mendoza AE, Barrera-Barrera SA, Barrera-Saldaña HA. Growth hormone and aging: a clinical review. FRONTIERS IN AGING 2025; 6:1549453. [PMID: 40260058 PMCID: PMC12009952 DOI: 10.3389/fragi.2025.1549453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 03/18/2025] [Indexed: 04/23/2025]
Abstract
Aging is a complex biological process characterized by functional decline, reduced quality of life, and increased vulnerability to diseases such as type 2 diabetes, cardiovascular conditions, neurodegeneration, and cancer. Advances in medical technology have introduced the concept of aging therapies, with growth hormone (GH) and its primary mediator, insulin-like growth factor 1 (IGF-1), receiving considerable attention for their potential to counteract age-related physiological and metabolic changes. GH plays a multifaceted role in the human body, primarily influencing body composition by increasing muscle mass, reducing fat tissue, promoting bone formation, and regulating the metabolism of proteins, lipids, and glucose. Additional effects have been noted on endothelial function, cognitive performance, and circadian rhythms. This review examines the molecular mechanisms of GH in aging, its potential as an anti-aging therapy, and findings from clinical trials involving these hormones for this purpose. It also addresses the associated adverse effects, limitations, and controversies. While some studies report significant benefits, these therapies' long-term safety and efficacy in promoting healthy aging remain uncertain, highlighting the need for further research.
Collapse
Affiliation(s)
- Luis E. Fernández-Garza
- Innbiogem SC, Monterrey, Mexico
- Servicio de Medicina Interna, Hospital General de Zona No. 2, Instituto Mexicano del Seguro Social, Monterrey, Mexico
- LANSEIDI-CONAHCyT, Monterrey, Mexico
| | | | - Marcos A. Sotelo-Ibarra
- Innbiogem SC, Monterrey, Mexico
- Facultad de Ciencias Biológicas de la Universidad Autónoma de Nuevo León, Ciudad Universitaria, San Nicolás de Los Garza, Mexico
| | | | | | - Hugo A. Barrera-Saldaña
- Innbiogem SC, Monterrey, Mexico
- LANSEIDI-CONAHCyT, Monterrey, Mexico
- Facultad de Ciencias Biológicas de la Universidad Autónoma de Nuevo León, Ciudad Universitaria, San Nicolás de Los Garza, Mexico
- Facultad de Medicina de la Universidad Autónoma de Nuevo León, Monterrey, Mexico
- Dirección de Investigación Científica de Laboratorios Columbia, Coyoacán, Mexico
| |
Collapse
|
4
|
Liu Y, Xue H, Liu Y, Li H, Liang Q, Ma L, Zhao M, Liu J. Serum Insulin-Like Growth Factor 1 and the Prognosis of Patients With Advanced Liver Diseases: A Meta-Analysis. Clin Transl Gastroenterol 2025:01720094-990000000-00374. [PMID: 39981963 DOI: 10.14309/ctg.0000000000000829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 02/12/2025] [Indexed: 02/22/2025] Open
Abstract
INTRODUCTION Serum insulin-like growth factor 1 (IGF-1), a hepatocyte-derived cytokine, has been suggested to reflect hepatic function reserve. The aim of this systematic review and meta-analysis was to investigate the association between serum IGF-1 levels on the admission and prognosis of patients with advanced liver diseases. METHODS A thorough examination of the literature was conducted across various databases, namely PubMed, Embase, Web of Science, Wanfang, and CNKI, with the aim of identifying relevant cohort studies. The data were synthesized using the random-effects model, taking into account the potential impact of heterogeneity. RESULTS A total of 9 cohorts were included. Patients with a low serum level of IGF-1, as compared with those with a high IGF-1 at baseline, exhibited a significantly poorer transplant-free survival (risk ratio: 3.03, 95% confidence interval: 2.17 to 4.22, P < 0.001), with no significant heterogeneity observed ( P for Cochrane Q test = 0.92, I2 = 0%). A sensitivity analysis, which was conducted by excluding 1 study at a time, yielded consistent results (risk ratio: 2.94-3.24, P all < 0.05). In addition, consistent results were observed in further subgroup analyses based on various factors, including cutoffs of IGF-1, country of the study, patient diagnosis, methods for measuring serum IGF-1, follow-up duration, analytic model, and quality scores ( P for subgroup difference all > 0.05). DISCUSSION A diminished serum IGF-1 level on admission could potentially serve as an indicator for an unfavorable prognosis among patients afflicted with advanced liver disease, such as severe hepatitis and cirrhosis.
Collapse
Affiliation(s)
- Yihan Liu
- Zhoukou Central Hospital Affiliated to Xinxiang Medical University, Zhoukou, Henan, China
| | - Haojie Xue
- Zhoukou Central Hospital Affiliated to Xinxiang Medical University, Zhoukou, Henan, China
| | - Yang Liu
- Ward 1, Department of Gastroenterology, Zhoukou Central Hospital, Zhoukou, Henan, China
| | - Han Li
- Ward 1, Department of Gastroenterology, Zhoukou Central Hospital, Zhoukou, Henan, China
| | - Qian Liang
- Zhoukou Central Hospital, Zhoukou, Henan, China
| | - Longhui Ma
- Zhoukou Central Hospital, Zhoukou, Henan, China
| | - Ming Zhao
- Ward 1, Department of Gastroenterology, Zhoukou Central Hospital, Zhoukou, Henan, China
| | - Junying Liu
- Zhoukou Central Hospital, Zhoukou, Henan, China
| |
Collapse
|
5
|
Kineman RD, Del Rio-Moreno M, Waxman DJ. Liver-specific actions of GH and IGF1 that protect against MASLD. Nat Rev Endocrinol 2025; 21:105-117. [PMID: 39322791 DOI: 10.1038/s41574-024-01037-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/29/2024] [Indexed: 09/27/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD; also known as nonalcoholic fatty liver disease) is a chronic condition associated with metabolic syndrome, a group of conditions that includes obesity, insulin resistance, hyperlipidaemia and cardiovascular disease. Primary growth hormone (GH) deficiency is associated with MASLD, and the decline in circulating levels of GH with weight gain might contribute to the development of MASLD. Raising endogenous GH secretion or administering GH replacement therapy in the context of MASLD enhances insulin-like growth factor 1 (IGF1) production and reduces steatosis and the severity of liver injury. GH and IGF1 indirectly control MASLD progression by regulating systemic metabolic function. Evidence supports the proposal that GH and IGF1 also have a direct role in regulating liver metabolism and health. This Review focuses on how GH acts on the hepatocyte in a sex-dependent manner to limit lipid accumulation, reduce stress, and promote survival and regeneration. In addition, we discuss how GH and IGF1 might regulate non-parenchymal cells of the liver to control inflammation and fibrosis, which have a major effect on hepatocyte survival and regeneration. Development of a better understanding of how GH and IGF1 coordinate the functions of specific, individual liver cell types might provide insight into the aetiology of MASLD initiation and progression and suggest novel approaches for the treatment of MASLD.
Collapse
Affiliation(s)
- Rhonda D Kineman
- Department of Medicine, Section of Endocrinology, Diabetes, and Metabolism, University of Illinois at Chicago, Chicago, IL, USA.
- Jesse Brown VA Medical Center, Research and Development Division, Chicago, IL, USA.
| | - Mercedes Del Rio-Moreno
- Department of Medicine, Section of Endocrinology, Diabetes, and Metabolism, University of Illinois at Chicago, Chicago, IL, USA
- Jesse Brown VA Medical Center, Research and Development Division, Chicago, IL, USA
| | - David J Waxman
- Department of Biology and Bioinformatics Program, Boston University, Boston, MA, USA
| |
Collapse
|
6
|
Montero-Hidalgo AJ, Del Rio-Moreno M, Pérez-Gómez JM, Luque RM, Kineman RD. Update on regulation of GHRH and its actions on GH secretion in health and disease. Rev Endocr Metab Disord 2025:10.1007/s11154-025-09943-y. [PMID: 39838154 DOI: 10.1007/s11154-025-09943-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/06/2025] [Indexed: 01/23/2025]
Abstract
This review focuses on our current understanding of how growth hormone releasing hormone (GHRH): 1) stimulates GH release and synthesis from pituitary growth hormone (GH)-producing cells (somatotropes), 2) drives somatotrope proliferation, 3) is negatively regulated by somatostatin (SST), GH and IGF1, 4) is altered throughout lifespan and in response to metabolic challenges, and 5) analogues can be used clinically to treat conditions of GH excess or deficiency. Although a large body of early work provides an underpinning for our current understanding of GHRH, this review specifically highlights more recent work that was made possible by state-of-the-art analytical tools, receptor-specific agonists and antagonists, high-resolution in vivo and ex vivo imaging and the development of tissue (cell) -specific ablation mouse models, to paint a more detailed picture of the regulation and actions of GHRH.
Collapse
Affiliation(s)
- Antonio J Montero-Hidalgo
- Maimonides Institute for Biomedical Research of Córdoba (IMIBIC), Cordoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Cordoba, Spain
- Hospital Universitario Reina Sofía (HURS), Cordoba, Spain
| | - Mercedes Del Rio-Moreno
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Illinois at Chicago, Chicago, IL, USA
- Jesse Brown Veterans Affairs Medical Center, Research and Development Division Chicago, 820 S. Damen Ave., MP151, Rm 6215, Chicago, IL, USA
| | - Jesús M Pérez-Gómez
- Maimonides Institute for Biomedical Research of Córdoba (IMIBIC), Cordoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Cordoba, Spain
- Hospital Universitario Reina Sofía (HURS), Cordoba, Spain
| | - Raúl M Luque
- Maimonides Institute for Biomedical Research of Córdoba (IMIBIC), Cordoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Cordoba, Spain
- Hospital Universitario Reina Sofía (HURS), Cordoba, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de La Obesidad y Nutrición, Cordoba, CIBERobn, Spain
| | - Rhonda D Kineman
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Illinois at Chicago, Chicago, IL, USA.
- Jesse Brown Veterans Affairs Medical Center, Research and Development Division Chicago, 820 S. Damen Ave., MP151, Rm 6215, Chicago, IL, USA.
| |
Collapse
|
7
|
Mazaheri F, Hoseini R, Gharzi A. Vitamin D and exercise improve VEGF-B production and IGF-1 levels in diabetic rats: insights the role of miR-1 suppression. Sci Rep 2025; 15:1328. [PMID: 39779732 PMCID: PMC11711202 DOI: 10.1038/s41598-024-81230-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 11/25/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Type 2 Diabetes Mellitus (T2DM) is closely associated with the development of vascular damage in the heart. In this study, the researchers aimed to determine whether Aerobic Training (AT) and Vitamin D supplementation (Vit D) could alleviate heart complications and vascular damage caused by diabetes. The effects of an eight-week AT program and Vit D on the expression of miR-1, IGF-1 genes, and VEGF-B in the cardiomyocytes of rats with T2DM. METHODS This study was an experimental investigation. Fifty male Wistar rats were divided into 2 groups Non-Diabetic Obese Control (NC; n = 10), and diabetic (n = 40). The rats were then randomly divided into four groups: AT plus Vit D (AT + Vit D; n-=10), AT (n = 10), Vit D (Vit D; n = 10), and Control Diabetic (C; n = 10). The exercise groups underwent treadmill training for 8 weeks at an aerobic intensity equal to 50-60% of their maximal oxygen uptake (VO2max), which corresponded to a speed of 15-25 m/min at a 0% incline, for 30-60 min per day, 5 days per week. The Vit D and AT + Vit D groups received 5,000 international units (IU) of Vitamin D (combined with sesame oil) per week via a single-dose injection. Data were analyzed using one-way analysis of variance (ANOVA) followed by Tukey's post-hoc test for multiple comparisons among the groups. Paired data were analyzed using paired t-tests. RESULTS The results showed that BW, BMI, and FI significantly decreased in the AT + Vit D (p = 0.001 for all variables), AT (p = 0.001 for all variables), and Vit D (p = 0.001 for all variables) groups compared to baseline. In contrast, BW, BMI, and FI increased in the C (p = 0.001, p = 0.006, p = 0.020, respectively) and NC (p = 0.001 for all variables) groups. Significant differences were observed between the groups in terms of visceral fat, insulin, glucose, and HOMA-IR (p = 0.001 for all variables). Serum 25-hydroxyvitamin D levels varied significantly among the groups (p = 0.002). The AT + Vit D group showed significantly increased VEGF-B (p = 0.001 for both comparisons), upregulated IGF-1 (p = 0.001 for both comparisons), and downregulated miR-1 (p = 0.001 for both comparisons) compared to the AT and Vit D groups, respectively. CONCLUSIONS AT and Vit D increased the expression of IGF-1 and VEGF-B in the heart of T2DM rats while decreasing the expression of miR-1. These effects were more pronounced when AT and Vit D were combined. The study concludes that the combination of AT and Vit D has cardio-protective effects in T2DM rats, counteracting abnormal angiogenesis induced by diabetes. These effects are mediated, at least in part, by the upregulation of IGF-1 and VEGF-B, and the downregulation of miR-1.
Collapse
Affiliation(s)
- Fatemeh Mazaheri
- Department of Exercise Physiology, Faculty of Sport Sciences, Razi University, Kermanshah, Iran
| | - Rastegar Hoseini
- Department of Exercise Physiology, Faculty of Sport Sciences, Razi University, P.O.Box. 6714414971, Kermanshah, Iran.
| | - Ahmad Gharzi
- Department of Biology, Faculty of Science, Razi University, Kermanshah, Iran
| |
Collapse
|
8
|
Saaybi SR, Shiau H, Lee G, Orandi BJ, Gutierrez Sanchez LH. Treatment of rapid recurrence of severe steatosis with combined glucagon-like peptide-1 agonist and growth hormone therapy in a pediatric patient transplanted for metabolic dysfunction-associated steatohepatitis cirrhosis in the setting of hypopituitarism. Am J Transplant 2025:S1600-6135(25)00002-4. [PMID: 39793899 DOI: 10.1016/j.ajt.2025.01.002] [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: 07/22/2024] [Revised: 12/30/2024] [Accepted: 01/04/2025] [Indexed: 01/13/2025]
Abstract
The association between hypopituitarism and metabolic dysfunction-associated steatotic liver disease is increasingly recognized, although data about therapies targeting recurrence posttransplant is limited. An 8-year-old with hypopituitarism-associated metabolic dysfunction-associated steatotic liver disease underwent a liver transplant due to rapid progression of metabolic dysfunction-associated steatohepatitis. Hepatosteatosis recurred within weeks. Her therapeutic plan included a glucagon-like peptide-1 agonist and growth hormone replacement. Her transaminases normalized in 2.5 months, and her macrosteatosis significantly improved on the 1-year surveillance biopsy. This case highlights one of the youngest reported children with hypopituitarism to have undergone transplantation for rapidly progressing metabolic dysfunction-associated steatohepatitis and its recurrence post-operatively. We observed that steatosis improved with growth hormone replacement and glucagon-like peptide-1 agonist therapy. If started early, this combination could help delay recurrence of steatosis post-transplantation. Further research is needed to determine long-term effects and establish protocols.
Collapse
Affiliation(s)
- Stephanie R Saaybi
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
| | - Henry Shiau
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
| | - Goo Lee
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Babak John Orandi
- Department of Surgery, New York University, New York, New York, USA; Department of Medicine, New York University, New York, New York, USA
| | - Luz Helena Gutierrez Sanchez
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
9
|
Hartl L, Schwarz M, Simbrunner B, Jachs M, Wolf P, Bauer DJM, Scheiner B, Balcar L, Semmler G, Hofer BS, Dominik N, Marculescu R, Trauner M, Mandorfer M, Reiberger T. Insulin-like growth factor-1 in cirrhosis is linked to hepatic dysfunction and fibrogenesis and predicts liver-related mortality. Aliment Pharmacol Ther 2025; 61:88-98. [PMID: 39305115 PMCID: PMC11636078 DOI: 10.1111/apt.18289] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 04/23/2024] [Accepted: 09/09/2024] [Indexed: 12/13/2024]
Abstract
BACKGROUND AND AIMS We aimed to characterise insulin-like growth factor-1 (IGF-1) signalling in patients with advanced chronic liver disease (ACLD). METHODS Consecutive patients undergoing hepatic venous pressure gradient [HVPG] measurement were prospectively included. Clinical stages were defined as follows: probable ACLD (pACLD): liver stiffness ≥10 kPa and HVPG ≤5 mmHg, S0: mild PH (HVPG 6-9 mmHg), S1: clinically significant PH (CSPH), S2: CSPH with varices, S3: past variceal bleeding, S4: past/current non-bleeding hepatic decompensation and S5: further decompensation. RESULTS In total, 269 patients were included; 105 were compensated (pACLD: n = 18; S0: n = 30; S1: n = 20; S2: n = 37), and 164 were decompensated (S3: n = 11; S4: n = 89; S5: n = 64). Median levels of IGF-1 decreased with progressive cirrhosis (from pACLD: 88.5 ng/mL to S5: 51.0 ng/mL; p < 0.001). Patients with CSPH had significantly lower IGF-1 levels (63.5 ng/mL vs. 81.0 ng/mL; p = 0.001). IGF-1 showed an independent negative association with body mass index (BMI; aB: -1.56; p < 0.001), enhanced liver fibrosis (ELF) test (aB: -8.43; p < 0.001), MELD (aB: -1.13; p = 0.042) and age (per 10 years; aB: -6.87; p < 0.001). IGF-1 exhibited an excellent AUROC (0.856) for the prediction of liver-related death at 6 months of follow-up. Lower IGF-1 (per 10 ng/mL) was linked to higher risk of (further) decompensation (0.90; 95% CI: 0.83-0.98; p = 0.016), acute-on-chronic liver failure (ACLF; asHR: 0.80; 95% CI: 0.68-0.93; p = 0.004) and liver-related death (asHR: 0.76; 95% CI: 0.63-0.91; p = 0.004). CONCLUSION Decreased levels of IGF-1 reflect impaired hepatic function and fibrogenesis in patients with cirrhosis, which seems particularly relevant in obesity since low IGF-1 was independently linked to high BMI. Lower IGF-1 in cirrhosis predicts decompensation, ACLF and liver-related death.
Collapse
Affiliation(s)
- Lukas Hartl
- Division of Gastroenterology and Hepatology, Department of Medicine IIIMedical University of ViennaViennaAustria
- Vienna Hepatic Hemodynamic Lab, Division of Gastroenterology and Hepatology, Department of Medicine IIIMedical University of ViennaViennaAustria
- Clinical Research Group MOTION, Division of Gastroenterology and Hepatology, Department of Medicine IIIMedical University of ViennaViennaAustria
| | - Michael Schwarz
- Division of Gastroenterology and Hepatology, Department of Medicine IIIMedical University of ViennaViennaAustria
- Vienna Hepatic Hemodynamic Lab, Division of Gastroenterology and Hepatology, Department of Medicine IIIMedical University of ViennaViennaAustria
| | - Benedikt Simbrunner
- Division of Gastroenterology and Hepatology, Department of Medicine IIIMedical University of ViennaViennaAustria
- Vienna Hepatic Hemodynamic Lab, Division of Gastroenterology and Hepatology, Department of Medicine IIIMedical University of ViennaViennaAustria
- Clinical Research Group MOTION, Division of Gastroenterology and Hepatology, Department of Medicine IIIMedical University of ViennaViennaAustria
- Christian Doppler Lab for Portal Hypertension and Liver FibrosisMedical University of ViennaViennaAustria
| | - Mathias Jachs
- Division of Gastroenterology and Hepatology, Department of Medicine IIIMedical University of ViennaViennaAustria
- Vienna Hepatic Hemodynamic Lab, Division of Gastroenterology and Hepatology, Department of Medicine IIIMedical University of ViennaViennaAustria
- Clinical Research Group MOTION, Division of Gastroenterology and Hepatology, Department of Medicine IIIMedical University of ViennaViennaAustria
| | - Peter Wolf
- Division of Endocrinology, Department of Medicine IIIMedical University of ViennaViennaAustria
| | - David Josef Maria Bauer
- Division of Gastroenterology and Hepatology, Department of Medicine IIIMedical University of ViennaViennaAustria
- Vienna Hepatic Hemodynamic Lab, Division of Gastroenterology and Hepatology, Department of Medicine IIIMedical University of ViennaViennaAustria
| | - Bernhard Scheiner
- Division of Gastroenterology and Hepatology, Department of Medicine IIIMedical University of ViennaViennaAustria
- Vienna Hepatic Hemodynamic Lab, Division of Gastroenterology and Hepatology, Department of Medicine IIIMedical University of ViennaViennaAustria
- Clinical Research Group MOTION, Division of Gastroenterology and Hepatology, Department of Medicine IIIMedical University of ViennaViennaAustria
| | - Lorenz Balcar
- Division of Gastroenterology and Hepatology, Department of Medicine IIIMedical University of ViennaViennaAustria
- Vienna Hepatic Hemodynamic Lab, Division of Gastroenterology and Hepatology, Department of Medicine IIIMedical University of ViennaViennaAustria
- Clinical Research Group MOTION, Division of Gastroenterology and Hepatology, Department of Medicine IIIMedical University of ViennaViennaAustria
| | - Georg Semmler
- Division of Gastroenterology and Hepatology, Department of Medicine IIIMedical University of ViennaViennaAustria
- Vienna Hepatic Hemodynamic Lab, Division of Gastroenterology and Hepatology, Department of Medicine IIIMedical University of ViennaViennaAustria
- Clinical Research Group MOTION, Division of Gastroenterology and Hepatology, Department of Medicine IIIMedical University of ViennaViennaAustria
| | - Benedikt Silvester Hofer
- Division of Gastroenterology and Hepatology, Department of Medicine IIIMedical University of ViennaViennaAustria
- Vienna Hepatic Hemodynamic Lab, Division of Gastroenterology and Hepatology, Department of Medicine IIIMedical University of ViennaViennaAustria
- Clinical Research Group MOTION, Division of Gastroenterology and Hepatology, Department of Medicine IIIMedical University of ViennaViennaAustria
- Christian Doppler Lab for Portal Hypertension and Liver FibrosisMedical University of ViennaViennaAustria
| | - Nina Dominik
- Division of Gastroenterology and Hepatology, Department of Medicine IIIMedical University of ViennaViennaAustria
- Vienna Hepatic Hemodynamic Lab, Division of Gastroenterology and Hepatology, Department of Medicine IIIMedical University of ViennaViennaAustria
- Clinical Research Group MOTION, Division of Gastroenterology and Hepatology, Department of Medicine IIIMedical University of ViennaViennaAustria
| | - Rodrig Marculescu
- Department of Laboratory MedicineMedical University of ViennaViennaAustria
| | - Michael Trauner
- Division of Gastroenterology and Hepatology, Department of Medicine IIIMedical University of ViennaViennaAustria
- Clinical Research Group MOTION, Division of Gastroenterology and Hepatology, Department of Medicine IIIMedical University of ViennaViennaAustria
| | - Mattias Mandorfer
- Division of Gastroenterology and Hepatology, Department of Medicine IIIMedical University of ViennaViennaAustria
- Vienna Hepatic Hemodynamic Lab, Division of Gastroenterology and Hepatology, Department of Medicine IIIMedical University of ViennaViennaAustria
- Clinical Research Group MOTION, Division of Gastroenterology and Hepatology, Department of Medicine IIIMedical University of ViennaViennaAustria
| | - Thomas Reiberger
- Division of Gastroenterology and Hepatology, Department of Medicine IIIMedical University of ViennaViennaAustria
- Vienna Hepatic Hemodynamic Lab, Division of Gastroenterology and Hepatology, Department of Medicine IIIMedical University of ViennaViennaAustria
- Clinical Research Group MOTION, Division of Gastroenterology and Hepatology, Department of Medicine IIIMedical University of ViennaViennaAustria
- Christian Doppler Lab for Portal Hypertension and Liver FibrosisMedical University of ViennaViennaAustria
| |
Collapse
|
10
|
Choe Y, Lee YJ, Lee YA, Ko JS, Shin CH. Hepatopulmonary syndrome secondary to metabolic associated fatty liver disease in childhood - novel treatment with growth hormone replacement therapy: a case report and systematic review of literature. Front Endocrinol (Lausanne) 2024; 15:1407686. [PMID: 39502571 PMCID: PMC11534608 DOI: 10.3389/fendo.2024.1407686] [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: 03/27/2024] [Accepted: 07/17/2024] [Indexed: 11/08/2024] Open
Abstract
Objective Hepatopulmonary syndrome (HPS) is a rare complication of metabolic associated fatty liver disease (MAFLD) occurring subsequent to hypopituitarism, often developing after resection of hypothalamic or pituitary tumors. The aim of this study is to report an illustrative case of an HPS patient who was successfully treated with growth hormone replacement therapy, without liver transplantation which is conventionally regarded as the only treatment option. Additionally, we conducted a comprehensive review of published case reports of HPS in the pediatric population. Methods We systematically searched literature databases to identify case reports and case series of HPS associated with hypopituitarism diagnosed in childhood. The search included MEDLINE/PubMed, Scopus, Embase, and Google Scholar from 1990 to 2023. The review process adhered to the PRISMA checklist for comprehensive reporting and methodological transparency. Results An 18-year-old female, who had been followed up for MAFLD after craniopharyngioma resection, presented with cyanosis and progressive dyspnea. She was diagnosed with severe degree of HPS. The patient began treatment with recombinant human growth hormone, leading to a significant improvement in respiratory symptoms within 3 months, and normalization of lung shunt ratio after 6 months of therapy. In our systematic review, nine patients from nine studies across six countries were identified. The median age at diagnosis of hypopituitarism was 10.5 years (range 1-16 years), and HPS was diagnosed at a median interval of 7 years later (range 0-26 years). Half of the patients had not received growth hormone therapy after being diagnosed with hypopituitarism, which subsequently led to the diagnosis of HPS. Three patients underwent liver transplantation, but non-alcoholic steatohepatitis recurred in all cases. Six patients were successfully treated with growth hormone replacement therapy without undergoing liver transplantation. Conclusions HPS can occur in pediatric patients with MAFLD who have undergone resection of the tumor in the hypothalamus or pituitary gland. Our findings suggest that growth hormone replacement therapy can be a possible alternative to liver transplantation for HPS patients. However, further investigations need to be performed to validate the efficacy of growth hormone treatment in different causes of HPS cases.
Collapse
Affiliation(s)
- Yunsoo Choe
- Department of Pediatrics, Hanyang University Guri Hospital, Guri, Republic of Korea
- Department of Pediatrics, Hanyang University College of Medicine, Seoul, Republic of Korea
| | - Yun Jeong Lee
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Young Ah Lee
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jae Sung Ko
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Choong Ho Shin
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
11
|
Ji Y, Duan Y, Li Y, Lu Q, Liu D, Yang Y, Chang R, Tian J, Yao W, Yin J, Gao X. A long-acting FGF21 attenuates metabolic dysfunction-associated steatohepatitis-related fibrosis by modulating NR4A1-mediated Ly6C phenotypic switch in macrophages. Br J Pharmacol 2024; 181:2923-2946. [PMID: 38679486 DOI: 10.1111/bph.16378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 02/17/2024] [Accepted: 03/04/2024] [Indexed: 05/01/2024] Open
Abstract
BACKGROUND AND PURPOSE Because of the absence of effective therapies for metabolic dysfunction-associated steatohepatitis (MASH), there is a rising interest in fibroblast growth factor 21 (FGF21) analogues due to their potential anti-fibrotic activities in MASH treatment. PsTag-FGF21, a long-acting FGF21 analogue, has demonstrated promising therapeutic effects in several MASH mouse models. However, its efficacy and mechanism against MASH-related fibrosis remain less well defined, compared with the specific mechanisms through which FGF21 improves glucose and lipid metabolism. EXPERIMENTAL APPROACH The effectiveness of PsTag-FGF21 was evaluated in two MASH-fibrosis models. Co-culture systems involving macrophages and hepatic stellate cells (HSCs) were employed for further assessment. Hepatic macrophages were selectively depleted by administering liposome-encapsulated clodronate via tail vein injections. RNA sequencing and cytokine profiling were conducted to identify key factors involved in macrophage-HSC crosstalk. KEY RESULTS We first demonstrated the significant attenuation of hepatic fibrosis by PsTag-FGF21 in two MASH-fibrosis models. Furthermore, we highlighted the crucial role of macrophage phenotypic switch in PsTag-FGF21-induced HSC deactivation. FGF21 was demonstrated to regulate macrophages in a PsTag-FGF21-like manner. NR4A1, a nuclear factor which is notably down-regulated in human livers with MASH, was identified as a mediator responsible for PsTag-FGF21-induced phenotypic switch. Transcriptional control over insulin-like growth factor 1, a crucial factor in macrophage-HSC crosstalk, was exerted by the intrinsically disordered region domain of NR4A1. CONCLUSION AND IMPLICATIONS Our results have elucidated the previously unclear mechanisms through which PsTag-FGF21 treats MASH-related fibrosis and identified NR4A1 as a potential therapeutic target for fibrosis.
Collapse
Affiliation(s)
- Yue Ji
- Jiangsu Key Laboratory of Draggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yiliang Duan
- Jiangsu Key Laboratory of Draggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yuanyuan Li
- Jiangsu Key Laboratory of Draggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Qingzhou Lu
- Jiangsu Key Laboratory of Draggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Dingkang Liu
- Jiangsu Key Laboratory of Draggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yifan Yang
- Jiangsu Key Laboratory of Draggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Ruilong Chang
- Jiangsu Key Laboratory of Draggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Jing Tian
- Jiangsu Key Laboratory of Draggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Wenbing Yao
- Jiangsu Key Laboratory of Draggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Jun Yin
- Jiangsu Key Laboratory of Draggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Xiangdong Gao
- Jiangsu Key Laboratory of Draggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
12
|
Benfica LF, Brito LF, do Bem RD, de Oliveira LF, Mulim HA, Braga LG, Cyrillo JNSG, Bonilha SFM, Mercadante MEZ. Detection and characterization of copy number variation in three differentially-selected Nellore cattle populations. Front Genet 2024; 15:1377130. [PMID: 38694873 PMCID: PMC11061390 DOI: 10.3389/fgene.2024.1377130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 04/05/2024] [Indexed: 05/04/2024] Open
Abstract
Introduction: Nellore cattle (Bos taurus indicus) is the main beef cattle breed raised in Brazil. This breed is well adapted to tropical conditions and, more recently, has experienced intensive genetic selection for multiple performance traits. Over the past 43 years, an experimental breeding program has been developed in the Institute of Animal Science (IZ, Sertaozinho, SP, Brazil), which resulted in three differentially-selected lines known as Nellore Control (NeC), Nellore Selection (NeS), and Nellore Traditional (NeT). The primary goal of this selection experiment was to determine the response to selection for yearling weight (YW) and residual feed intake (RFI) on Nellore cattle. The main objectives of this study were to: 1) identify copy number variation (CNVs) in Nellore cattle from three selection lines; 2) identify and characterize CNV regions (CNVR) on these three lines; and 3) perform functional enrichment analyses of the CNVR identified. Results: A total of 14,914 unique CNVs and 1,884 CNVRs were identified when considering all lines as a single population. The CNVRs were non-uniformly distributed across the chromosomes of the three selection lines included in the study. The NeT line had the highest number of CNVRs (n = 1,493), followed by the NeS (n = 823) and NeC (n = 482) lines. The CNVRs covered 23,449,890 bp (0.94%), 40,175,556 bp (1.61%), and 63,212,273 bp (2.54%) of the genome of the NeC, NeS, and NeT lines, respectively. Two CNVRs were commonly identified between the three lines, and six, two, and four exclusive regions were identified for NeC, NeS, and NeT, respectively. All the exclusive regions overlap with important genes, such as SMARCD3, SLC15A1, and MAPK1. Key biological processes associated with the candidate genes were identified, including pathways related to growth and metabolism. Conclusion: This study revealed large variability in CNVs and CNVRs across three Nellore lines differentially selected for YW and RFI. Gene annotation and gene ontology analyses of the exclusive CNVRs to each line revealed specific genes and biological processes involved in the expression of growth and feed efficiency traits. These findings contribute to the understanding of the genetic mechanisms underlying the phenotypic differences among the three Nellore selection lines.
Collapse
Affiliation(s)
- Lorena F. Benfica
- Department of Animal Sciences, Purdue University, West Lafayette, IN, United States
- Department of Animal Science, Faculty of Agricultural and Veterinary Sciences, Sao Paulo State University, Jaboticabal, São Paulo, Brazil
| | - Luiz F. Brito
- Department of Animal Sciences, Purdue University, West Lafayette, IN, United States
| | - Ricardo D. do Bem
- Department of Animal Science, Faculty of Agricultural and Veterinary Sciences, Sao Paulo State University, Jaboticabal, São Paulo, Brazil
| | | | - Henrique A. Mulim
- Department of Animal Sciences, Purdue University, West Lafayette, IN, United States
| | - Larissa G. Braga
- Department of Animal Science, Faculty of Agricultural and Veterinary Sciences, Sao Paulo State University, Jaboticabal, São Paulo, Brazil
- Department of Animal Biosciences, University of Guelph, Guelph, ON, Canada
| | | | - Sarah F. M. Bonilha
- Beef Cattle Research Center, Institute of Animal Science, Sertaozinho, São Paulo, Brazil
| | - Maria Eugenia Z. Mercadante
- Department of Animal Science, Faculty of Agricultural and Veterinary Sciences, Sao Paulo State University, Jaboticabal, São Paulo, Brazil
- Beef Cattle Research Center, Institute of Animal Science, Sertaozinho, São Paulo, Brazil
| |
Collapse
|
13
|
Dutta K, Bhatt SP, Madan S, Ansari IA, Tyagi K, Pandey S, Misra A. Acanthosis nigricans independently predicts hepatic fibrosis in people with type 2 diabetes in North India. Prim Care Diabetes 2024; 18:224-229. [PMID: 38245384 DOI: 10.1016/j.pcd.2024.01.003] [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: 09/25/2023] [Revised: 01/02/2024] [Accepted: 01/04/2024] [Indexed: 01/22/2024]
Abstract
BACKGROUND Acanthosis nigricans (AN) is a skin condition characterized by hyperpigmentation and thickening, often found in individuals with insulin resistance. Despite this well-established association, the potential link between AN and hepatic fibrosis in people with type 2 diabetes (T2D) has yet to be thoroughly explored. METHODOLOGY We recruited a total of 300 people with T2D, half of whom had AN (n, 150), and the other half without AN (n, 150). We evaluated body composition, biochemistry, and hepatic fat analysis (using the controlled attenuation parameter, CAP), as well as assessments of hepatic stiffness (using the kilopascal, kPa) using Fibroscan. We used multivariable regression analysis to find independent predictors of AN and their relationship to hepatic fibrosis. Furthermore, we developed a prediction equation and AUC for hepatic fibrosis. RESULTS Upon comparison between AN vs. NAN group, following were significatly higher; weight, BMI, hepatic transaminases, liver span, CAP, and kPa. After adjusting for age, weight, body mass index, diabetes duration, and specific anti-hyperglycaemic drugs (gliclazide, DPP-4 inhibitors, pioglitazone, and Glucagon-like peptide-1 receptor agonists), adjusted OR for AN were, liver span, 1.78 (95% CI: 0.91-3.49, p = 0.09), CAP, 7.55 (95% CI: 0.93-61.1, p = 0.05), and kPa, 2.47 (95% CI: 1.50-4.06, p = 0.001). A ROC analysis of predictive score for hepatic fibrosis showed optimal sensitivity and specificity at a score cut-off of 25.2 (sensitivity 62%, specificity 63%), with an AUC of 0.6452 (95% CI: 0.61235-0.76420). CONCLUSION Acanthosis nigricans has the potential to be used as an easy-to-identify clinical marker for risk of hepatic fat and fibrosis in Asian Indians with T2D, allowing for early detection and management strategies.
Collapse
Affiliation(s)
- Koel Dutta
- Fortis C-DOC Center of Excellence for Diabetes, Metabolic Diseases, and Endocrinology, B-16, Chirag Enclave, New Delhi, India
| | - Surya Prakash Bhatt
- Fortis C-DOC Center of Excellence for Diabetes, Metabolic Diseases, and Endocrinology, B-16, Chirag Enclave, New Delhi, India; National Diabetes, Obesity and Cholesterol Foundation (N-DOC), SDA, New Delhi, India; Department of Pulmonary, Critical Care and Sleep Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Swati Madan
- National Diabetes, Obesity and Cholesterol Foundation (N-DOC), SDA, New Delhi, India
| | - Irshad Ahmad Ansari
- Fortis C-DOC Center of Excellence for Diabetes, Metabolic Diseases, and Endocrinology, B-16, Chirag Enclave, New Delhi, India
| | - Kanika Tyagi
- Fortis C-DOC Center of Excellence for Diabetes, Metabolic Diseases, and Endocrinology, B-16, Chirag Enclave, New Delhi, India
| | - Shivam Pandey
- Department of Biostatistics, All India Institute of Medical Sciences, New Delhi, India
| | - Anoop Misra
- Fortis C-DOC Center of Excellence for Diabetes, Metabolic Diseases, and Endocrinology, B-16, Chirag Enclave, New Delhi, India; National Diabetes, Obesity and Cholesterol Foundation (N-DOC), SDA, New Delhi, India; Diabetes Foundation (India), SDA, New Delhi, India.
| |
Collapse
|
14
|
Ichikawa A, Miki D, Hayes CN, Teraoka Y, Nakahara H, Tateno C, Ishida Y, Chayama K, Oka S. Multi-omics analysis of a fatty liver model using human hepatocyte chimeric mice. Sci Rep 2024; 14:3362. [PMID: 38336825 PMCID: PMC10858249 DOI: 10.1038/s41598-024-53890-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 02/06/2024] [Indexed: 02/12/2024] Open
Abstract
We developed a fatty liver mouse model using human hepatocyte chimeric mice. As transplanted human hepatocytes do not respond to mouse growth hormone (GH) and tend to accumulate fat, we hypothesized that addition of human GH would alter lipid metabolism and reduce accumulation of fat in the liver even when fed a high-fat diet. Six uPA/SCID chimeric mice were fed a high-fat GAN diet to induce fatty liver while six were fed a normal CRF1 diet, and GH was administered to three mice in each group. The mice were euthanized at 8 weeks, and human hepatocytes were extracted for RNA-Seq, DIA proteomics, and metabolomics analysis. Abdominal echocardiography revealed that the degree of fatty liver increased significantly in mice fed GAN diet (p < 0.001) and decreased significantly in mice treated with GH (p = 0.026). Weighted gene correlation network analysis identified IGF1 and SEMA7A as eigengenes. Administration of GH significantly reduced triglyceride levels and was strongly associated with metabolism of amino acids. MiBiOmics analysis identified perilipin-2 as a co-inertia driver. Results from multi-omics analysis revealed distinct gene expression and protein/metabolite profiles in each treatment group when mice were fed a high-fat or normal diet with or without administration of GH.
Collapse
Affiliation(s)
- Akemi Ichikawa
- Department of Gastroenterology, Graduate School of Biomedical and Health Science, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
- Pfizer, Inc., Tokyo, Japan
| | - Daiki Miki
- Department of Gastroenterology, Graduate School of Biomedical and Health Science, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - C Nelson Hayes
- Department of Gastroenterology, Graduate School of Biomedical and Health Science, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan.
| | - Yuji Teraoka
- Department of Gastroenterology, Graduate School of Biomedical and Health Science, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Hikaru Nakahara
- Department of Gastroenterology, Graduate School of Biomedical and Health Science, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
- Department of Clinical and Molecular Genetics, Hiroshima University, Hiroshima, Japan
| | | | - Yuji Ishida
- PhoenixBio Co., Ltd., Higashihiroshima, Japan
| | - Kazuaki Chayama
- Collaborative Research Laboratory of Medical Innovation, Hiroshima University, Hiroshima, Japan
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Shiro Oka
- Department of Gastroenterology, Graduate School of Biomedical and Health Science, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| |
Collapse
|
15
|
Isaacs S, Isaacs A. Endocrinology for the Hepatologist. CURRENT HEPATOLOGY REPORTS 2024; 23:99-109. [DOI: 10.1007/s11901-024-00639-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 01/09/2024] [Indexed: 01/03/2025]
|
16
|
Brie B, Sarmento-Cabral A, Pascual F, Cordoba-Chacon J, Kineman RD, Becu-Villalobos D. Modifications of the GH Axis Reveal Unique Sexually Dimorphic Liver Signatures for Lcn13, Asns, Hamp2, Hao2, and Pgc1a. J Endocr Soc 2024; 8:bvae015. [PMID: 38370444 PMCID: PMC10872697 DOI: 10.1210/jendso/bvae015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Indexed: 02/20/2024] Open
Abstract
Growth hormone (GH) modifies liver gene transcription in a sexually dimorphic manner to meet liver metabolic demands related to sex; thus, GH dysregulation leads to sex-biased hepatic disease. We dissected the steps of the GH regulatory cascade modifying GH-dependent genes involved in metabolism, focusing on the male-predominant genes Lcn13, Asns, and Cyp7b1, and the female-predominant genes Hao2, Pgc1a, Hamp2, Cyp2a4, and Cyp2b9. We explored mRNA expression in 2 settings: (i) intact liver GH receptor (GHR) but altered GH and insulin-like growth factor 1 (IGF1) levels (NeuroDrd2KO, HiGH, aHepIGF1kd, and STAT5bCA mouse lines); and (ii) liver loss of GHR, with or without STAT5b reconstitution (aHepGHRkd, and aHepGHRkd + STAT5bCA). Lcn13 was downregulated in males in most models, while Asns and Cyp7b1 were decreased in males by low GH levels or action, or constant GH levels, but unexpectedly upregulated in both sexes by the loss of liver Igf1 or constitutive Stat5b expression. Hao, Cyp2a4, and Cyp2b9 were generally decreased in female mice with low GH levels or action (NeuroDrd2KO and/or aHepGHRkd mice) and increased in HiGH females, while in contrast, Pgc1a was increased in female NeuroDrd2KO but decreased in STAT5bCA and aHepIGF1kd females. Bioinformatic analysis of RNAseq from aHepGHRkd livers stressed the greater impact of GHR loss on wide gene expression in males and highlighted that GH modifies almost completely different gene signatures in each sex. Concordantly, we show that altering different steps of the GH cascade in the liver modified liver expression of Lcn13, Asns, Cyp7b1, Hao2, Hamp2, Pgc1a, Cyp2a4, and Cyp2b9 in a sex- and gene-specific manner.
Collapse
Affiliation(s)
- Belen Brie
- Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), 1428 Ciudad de Buenos Aires, Argentina
| | - Andre Sarmento-Cabral
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Florencia Pascual
- Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), 1428 Ciudad de Buenos Aires, Argentina
| | - Jose Cordoba-Chacon
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Rhonda Denise Kineman
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Illinois at Chicago, Chicago, IL 60612, USA
- Research and Development Division, Jesse Brown Veterans Affairs Medical Center, Chicago, IL 60612, USA
| | - Damasia Becu-Villalobos
- Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), 1428 Ciudad de Buenos Aires, Argentina
| |
Collapse
|
17
|
Cheng J, Wang W. Association of Dietary Acid Load with Nonalcoholic Fatty Liver Disease and Advanced Liver Fibrosis in US Adults: Evidence from NHANES 1999-2018. Risk Manag Healthc Policy 2023; 16:2819-2832. [PMID: 38145208 PMCID: PMC10749110 DOI: 10.2147/rmhp.s437425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 12/10/2023] [Indexed: 12/26/2023] Open
Abstract
Background Evidence for an association between dietary acid load (DAL) and non-alcoholic fatty liver disease (NAFLD) is scarce and controversial. We aimed to address whether an association exists between DAL and NAFLD/advanced liver fibrosis (AHF) among US adults in a nationally representative study. Methods This was a cross-sectional study. We included adult participants from the National Health and Nutrition Examination Survey 1999-2018. Potential renal acid load (PRAL) and estimated net endogenous acid production (NEAP) was calculated from the literature and NAFLD/AHF was diagnosed by noninvasive markers. We comprehensively explored these relationships using multivariate adjusted regression models, restricted cubic spline, stratification analysis, and sensitivity analysis. Results We enrolled a total of 18,855 participants. All DAL metrics were positively and nonlinearly associated with NAFLD (all p-values < 0.0001), whereas NEAPF and NEAPR may be associated with AHF. In the stratified analysis, we found that the correlation between DAL and NAFLD exists in all ages and genders, but the effect of DAL seems to be more obvious in middle-aged, elderly and women. Similarly, we found that the effect of DAL on AHF was more significant in 45-60-year-olds and women. Sensitivity analyses revealed stability of all results. Conclusion DAL including PRAL and NEAP were positively associated with NAFLD in a large nationally representative cross-sectional study. NEAPF and NEAPR may be associated with increased odds of AHF. Adjustment for diet-dependent DAL requires age- and sex-specific strategies. Future prospective studies are needed to validate our findings.
Collapse
Affiliation(s)
- Jin Cheng
- Department of Medicine, Wuhan University of Science and Technology, Wuhan, 430000, People’s Republic of China
- Department of Gastroenterology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, 441021, People’s Republic of China
| | - Wei Wang
- Department of Gastroenterology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, 441021, People’s Republic of China
| |
Collapse
|
18
|
Rampersaud A, Connerney J, Waxman DJ. Plasma growth hormone pulses induce male-biased pulsatile chromatin opening and epigenetic regulation in adult mouse liver. eLife 2023; 12:RP91367. [PMID: 38091606 PMCID: PMC10721219 DOI: 10.7554/elife.91367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2023] Open
Abstract
Sex differences in plasma growth hormone (GH) profiles, pulsatile in males and persistent in females, regulate sex differences in hepatic STAT5 activation linked to sex differences in gene expression and liver disease susceptibility, but little is understood about the fundamental underlying, GH pattern-dependent regulatory mechanisms. Here, DNase-I hypersensitivity site (DHS) analysis of liver chromatin accessibility in a cohort of 18 individual male mice established that the endogenous male rhythm of plasma GH pulse-stimulated liver STAT5 activation induces dynamic, repeated cycles of chromatin opening and closing at several thousand liver DHS and comprises a novel mechanism conferring male bias to liver chromatin accessibility. Strikingly, a single physiological replacement dose of GH given to hypophysectomized male mice restored, within 30 min, liver STAT5 activity and chromatin accessibility at 83% of the dynamic, pituitary hormone-dependent male-biased DHS. Sex-dependent transcription factor binding patterns and chromatin state analysis identified key genomic and epigenetic features distinguishing this dynamic, STAT5-driven mechanism of male-biased chromatin opening from a second GH-dependent mechanism operative at static male-biased DHS, which are constitutively open in male liver. Dynamic but not static male-biased DHS adopt a bivalent-like epigenetic state in female liver, as do static female-biased DHS in male liver, albeit using distinct repressive histone marks in each sex, namely, H3K9me3 at male-biased DHS in female liver and H3K27me3 at female-biased DHS in male liver. Moreover, sex-biased H3K36me3 marks are uniquely enriched at static sex-biased DHS, which may serve to keep these sex-dependent hepatocyte enhancers free of H3K27me3 repressive marks and thus constitutively open. Pulsatile chromatin opening stimulated by endogenous, physiological hormone pulses is thus one of two distinct GH-determined mechanisms for establishing widespread sex differences in hepatic chromatin accessibility and epigenetic regulation, both closely linked to sex-biased gene transcription and the sexual dimorphism of liver function.
Collapse
Affiliation(s)
- Andy Rampersaud
- Department of Biology and Bioinformatics Program, Boston UniversityBostonUnited States
| | - Jeannette Connerney
- Department of Biology and Bioinformatics Program, Boston UniversityBostonUnited States
| | - David J Waxman
- Department of Biology and Bioinformatics Program, Boston UniversityBostonUnited States
| |
Collapse
|
19
|
Liu Z, Zhao W, Cao C, Wang Y, Xiao L, Wang X, Jin C, Xiao J. Pituitary stalk interruption syndrome and liver cirrhosis associated with diabetes and an inactivating KCNJ11 gene mutation: a case report and literature review. Front Endocrinol (Lausanne) 2023; 14:1297146. [PMID: 38152125 PMCID: PMC10751576 DOI: 10.3389/fendo.2023.1297146] [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: 09/19/2023] [Accepted: 11/28/2023] [Indexed: 12/29/2023] Open
Abstract
Background Pituitary stalk interruption syndrome (PSIS) is a congenital disease commonly found in patients with combined pituitary hormone deficiency (CPHD). Most PSIS patients manifest growth retardation and delayed puberty. We report a rare case of PSIS with tall stature, liver cirrhosis and diabetes, possibly caused by an inactivating KCNJ11 gene mutation. Case presentation A 37-year-old female patient initially presented with liver cirrhosis and diabetes, without any secondary sexual characteristics. Endocrine investigation indicated CPHD. Small anterior pituitary, invisible pituitary stalk and no eutopic posterior lobe hypersignal in the sella turcica viewed in magnetic resonance imaging (MRI) confirmed the diagnosis of PSIS. Despite receiving no growth hormone or sex hormone therapy, she reached a final height of 186 cm. Liver histopathology revealed nonalcoholic fatty cirrhosis. Genetic testing identified a heterozygous p.Arg301Cys mutation in the KCNJ11 gene. Conclusion This is a rare case of PSIS with liver cirrhosis and diabetes associated with an inactivating KCNJ11 gene mutation. It's supposed that early hyperinsulinism caused by the KCNJ11 gene mutation, as well as delayed epiphyseal closure due to estrogen deficiency, contributed to the patient's exceptionally tall stature. Untreated growth hormone deficiency (GHD) resulted in increased visceral fat, leading to nonalcoholic fatty liver disease (NAFLD) and cirrhosis. The decline in β cell function with age, combined with NAFLD, may have played a role in the development of diabetes.
Collapse
|
20
|
Takahashi Y. Nonalcoholic fatty liver disease and adult growth hormone deficiency: An under-recognized association? Best Pract Res Clin Endocrinol Metab 2023; 37:101816. [PMID: 37643935 DOI: 10.1016/j.beem.2023.101816] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Growth hormone (GH) plays an essential role not only in promoting growth in children, but also in many important metabolic processes in adults. One of the major metabolic functions of GH is its stimulatory effects on the liver in generating approximately 80% of circulating insulin-like growth factor 1 (IGF-1). Adult growth hormone deficiency (GHD) is an established clinical entity defined as a defect in endogenous GH secretion that is frequently associated with central obesity, loss of muscle mass, decreased bone mass, and impaired quality of life. Nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH) are conditions that are often under-recognized in adults with GHD, and accordingly some studies have shown that GH and IGF-1 levels are decreased in patients with NAFLD. Furthermore, it has been reported that it can progress to end-stage liver cirrhosis in some adults and children with GHD. Due to their underlying mechanisms of action, GH and IGF-1 can act on hepatocytes, macrophages, and hepatic stellate cells to mitigate progression to steatosis and fibrosis. It is, thus, important to recognize NAFLD/NASH as important complications in adult and childhood GHD. Therefore, careful and thorough evaluation of NAFLD/NASH in adults with GHD and the consideration for GH replacement therapy is crucial in these patients, together with management of other metabolic risk factors, such as obesity and dyslipidemia. This review will focus on recent reports on the role of GH and IGF-1 in the liver and its clinical significance in the regulation of hepatic function.
Collapse
Affiliation(s)
- Yutaka Takahashi
- Department of Diabetes and Endocrinology, Nara Medical University, Japan.
| |
Collapse
|
21
|
Ahmed A, Justo S, Yaghootkar H. Genetic scores associated with favourable and unfavourable adiposity have consistent effect on metabolic profile and disease risk across diverse ethnic groups. Diabet Med 2023; 40:e15213. [PMID: 37638553 DOI: 10.1111/dme.15213] [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: 07/18/2023] [Revised: 08/21/2023] [Accepted: 08/24/2023] [Indexed: 08/29/2023]
Abstract
AIM This study aims to investigate the associations between genetic risk scores (GRS) for favourable and unfavourable adiposity and a wide range of adiposity-related outcomes across diverse populations. METHODS We utilised previously identified variants associated with favourable (36 variants) and unfavourable (38 variants) adiposity to create GRS for each adiposity phenotype. We used summary statistics from 39 outcomes generated by the Pan-UKB genome-wide association studies Version 0.3, incorporating covariates such as age, sex and principal components in six populations: European (n = 420,531), African (6636), American (980), Central/South Asian (8876), East Asian (2709) and Middle Eastern (1599). RESULTS The favourable adiposity GRS was associated with a healthy metabolic profile, including lower risk of type 2 diabetes, lower liver enzyme levels, lower blood pressure, higher HDL-cholesterol, lower triglycerides, higher apolipoprotein A, lower apolipoprotein B, higher testosterone, lower calcium and lower insulin-like growth factor 1 generally consistently across all the populations. In contrast, the unfavourable adiposity GRS was associated with an adverse metabolic profile, including higher risk of type 2 diabetes, higher random glucose levels, higher HbA1c, lower HDL-cholesterol, higher triglycerides, higher liver enzyme levels, lower testosterone, and higher C-reactive protein generally consistently across all the populations. CONCLUSION The study provides evidence that the genetic scores associated with favourable and unfavourable adiposity have consistent effects on metabolic profiles and disease risk across diverse ethnic groups. These findings deepen our understanding of distinct adiposity subtypes and their impact on metabolic health.
Collapse
Affiliation(s)
- Altayeb Ahmed
- Department of Life Sciences, Centre for Inflammation Research and Translational Medicine, Brunel University London, London, UK
- Joseph Banks Laboratories, College of Health and Science, University of Lincoln, Lincoln, Lincolnshire, UK
| | - Stephen Justo
- Department of Life Sciences, Centre for Inflammation Research and Translational Medicine, Brunel University London, London, UK
| | - Hanieh Yaghootkar
- Joseph Banks Laboratories, College of Health and Science, University of Lincoln, Lincoln, Lincolnshire, UK
| |
Collapse
|
22
|
Dichtel LE, Corey KE, Haines MS, Chicote ML, Lee H, Kimball A, Colling C, Simon TG, Long MT, Husseini J, Bredella MA, Miller KK. Growth Hormone Administration Improves Nonalcoholic Fatty Liver Disease in Overweight/Obesity: A Randomized Trial. J Clin Endocrinol Metab 2023; 108:e1542-e1550. [PMID: 37379033 PMCID: PMC10655511 DOI: 10.1210/clinem/dgad375] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 06/05/2023] [Accepted: 06/20/2023] [Indexed: 06/29/2023]
Abstract
CONTEXT Overweight and obesity are associated with relative growth hormone (GH) deficiency, which has been implicated in the development of nonalcoholic fatty liver disease (NAFLD). NAFLD is a progressive disease without effective treatments. OBJECTIVE We hypothesized that GH administration would reduce hepatic steatosis in individuals with overweight/obesity and NAFLD. METHODS In this 6-month randomized, double-blind, placebo-controlled trial of low-dose GH administration, 53 adults aged 18 to 65 years with BMI ≥25 kg/m2 and NAFLD without diabetes were randomized to daily subcutaneous GH or placebo, targeting insulin-like growth factor 1 (IGF-1) to the upper normal quartile. The primary endpoint was intrahepatic lipid content (IHL) by proton magnetic resonance spectroscopy (1H-MRS) assessed before treatment and at 6 months. RESULTS Subjects were randomly assigned to a treatment group (27 GH; 26 placebo), with 41 completers (20 GH and 21 placebo) at 6 months. Reduction in absolute % IHL by 1H-MRS was significantly greater in the GH vs placebo group (mean ± SD: -5.2 ± 10.5% vs 3.8 ± 6.9%; P = .009), resulting in a net mean treatment effect of -8.9% (95% CI, -14.5 to -3.3%). All side effects were similar between groups, except for non-clinically significant lower extremity edema, which was more frequent in the GH vs placebo group (21% vs 0%, P = .02). There were no study discontinuations due to worsening of glycemic status, and there were no significant differences in change in glycemic measures or insulin resistance between the GH and placebo groups. CONCLUSION GH administration reduces hepatic steatosis in adults with overweight/obesity and NAFLD without worsening glycemic measures. The GH/IGF-1 axis may lead to future therapeutic targets for NAFLD.
Collapse
Affiliation(s)
- Laura E Dichtel
- Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02114, USA
| | - Kathleen E Corey
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02114, USA
| | - Melanie S Haines
- Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02114, USA
| | - Mark L Chicote
- Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Hang Lee
- Biostatistics Center, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02114, USA
| | - Allison Kimball
- Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02114, USA
| | - Caitlin Colling
- Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02114, USA
| | - Tracey G Simon
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02114, USA
| | - Michelle T Long
- Section of Gastroenterology, Boston Medical Center, Boston University School of Medicine, Boston, MA 02118, USA
| | - Jad Husseini
- Department of Radiology, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02114, USA
| | - Miriam A Bredella
- Department of Radiology, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02114, USA
| | - Karen K Miller
- Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
23
|
Rampersaud A, Connerney J, Waxman DJ. Plasma Growth Hormone Pulses Induce Male-biased Pulsatile Chromatin Opening and Epigenetic Regulation in Adult Mouse Liver. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.21.554153. [PMID: 37662275 PMCID: PMC10473588 DOI: 10.1101/2023.08.21.554153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Sex-differences in plasma growth hormone (GH) profiles, pulsatile in males and persistent in females, regulate sex differences in hepatic STAT5 activation linked to sex differences in gene expression and liver disease susceptibility, but little is understood about the fundamental underlying, GH pattern-dependent regulatory mechanisms. Here, DNase hypersensitivity site (DHS) analysis of liver chromatin accessibility in a cohort of 18 individual male mice established that the endogenous male rhythm of plasma GH pulse-stimulated liver STAT5 activation induces dynamic, repeated cycles of chromatin opening and closing at several thousand liver DHS and comprises a novel mechanism conferring male bias to liver chromatin accessibility. Strikingly, a single physiological replacement dose of GH given to hypophysectomized male mice restored, within 30 min, liver STAT5 activity and chromatin accessibility at 83% of the pituitary hormone-dependent dynamic male-biased DHS. Sex-dependent transcription factor binding patterns and chromatin state analysis identified key genomic and epigenetic features distinguishing this dynamic, STAT5-driven mechanism of male-biased chromatin opening from a second GH-dependent mechanism operative at static male-biased DHS, which are constitutively open in male liver. Dynamic but not static male-biased DHS adopt a bivalent-like epigenetic state in female liver, as do static female-biased DHS in male liver, albeit using distinct repressive histone marks in each sex, namely, H3K27me3 at female-biased DHS in male liver, and H3K9me3 at male-biased DHS in female liver. Moreover, sex-biased H3K36me3 marks are uniquely enriched at static sex-biased DHS, which may serve to keep these sex-dependent hepatocyte enhancers free of H3K27me3 repressive marks and thus constitutively open. Pulsatile chromatin opening stimulated by endogenous, physiological hormone pulses is thus one of two distinct GH-determined mechanisms for establishing widespread sex differences in hepatic chromatin accessibility and epigenetic regulation, both closely linked to sex-biased gene transcription and the sexual dimorphism of liver function.
Collapse
Affiliation(s)
- Andy Rampersaud
- Department of Biology and Bioinformatics Program, Boston University, Boston, MA 02215 USA
| | - Jeannette Connerney
- Department of Biology and Bioinformatics Program, Boston University, Boston, MA 02215 USA
| | - David J Waxman
- Department of Biology and Bioinformatics Program, Boston University, Boston, MA 02215 USA
| |
Collapse
|
24
|
Zaripova LN, Midgley A, Christmas SE, Beresford MW, Pain C, Baildam EM, Oldershaw RA. Mesenchymal Stem Cells in the Pathogenesis and Therapy of Autoimmune and Autoinflammatory Diseases. Int J Mol Sci 2023; 24:16040. [PMID: 38003230 PMCID: PMC10671211 DOI: 10.3390/ijms242216040] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/27/2023] [Accepted: 10/31/2023] [Indexed: 11/26/2023] Open
Abstract
Mesenchymal stem cells (MSCs) modulate immune responses and maintain self-tolerance. Their trophic activities and regenerative properties make them potential immunosuppressants for treating autoimmune and autoinflammatory diseases. MSCs are drawn to sites of injury and inflammation where they can both reduce inflammation and contribute to tissue regeneration. An increased understanding of the role of MSCs in the development and progression of autoimmune disorders has revealed that MSCs are passive targets in the inflammatory process, becoming impaired by it and exhibiting loss of immunomodulatory activity. MSCs have been considered as potential novel cell therapies for severe autoimmune and autoinflammatory diseases, which at present have only disease modifying rather than curative treatment options. MSCs are emerging as potential therapies for severe autoimmune and autoinflammatory diseases. Clinical application of MSCs in rare cases of severe disease in which other existing treatment modalities have failed, have demonstrated potential use in treating multiple diseases, including rheumatoid arthritis, systemic lupus erythematosus, myocardial infarction, liver cirrhosis, spinal cord injury, multiple sclerosis, and COVID-19 pneumonia. This review explores the biological mechanisms behind the role of MSCs in autoimmune and autoinflammatory diseases. It also covers their immunomodulatory capabilities, potential therapeutic applications, and the challenges and risks associated with MSC therapy.
Collapse
Affiliation(s)
- Lina N. Zaripova
- Institute of Fundamental and Applied Medicine, National Scientific Medical Center, 42 Abylai Khan Avenue, Astana 010000, Kazakhstan;
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, William Henry Duncan Building, 6 West Derby Street, Liverpool L7 8TX, UK
| | - Angela Midgley
- Department of Women and Children’s Health, Institute of Life Course and Medical Sciences, University of Liverpool, Institute in the Park, Alder Hey Children’s NHS Foundation Trust, Liverpool L14 5AB, UK; (A.M.); (M.W.B.); (C.P.)
| | - Stephen E. Christmas
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection and Global Health, Faculty of Health and Life Sciences, University of Liverpool, The Ronald Ross Building, 8 West Derby Street, Liverpool L69 7BE, UK;
| | - Michael W. Beresford
- Department of Women and Children’s Health, Institute of Life Course and Medical Sciences, University of Liverpool, Institute in the Park, Alder Hey Children’s NHS Foundation Trust, Liverpool L14 5AB, UK; (A.M.); (M.W.B.); (C.P.)
- Department of Paediatric Rheumatology, Alder Hey Children’s NHS Foundation Trust, East Prescott Road, Liverpool L14 5AB, UK
| | - Clare Pain
- Department of Women and Children’s Health, Institute of Life Course and Medical Sciences, University of Liverpool, Institute in the Park, Alder Hey Children’s NHS Foundation Trust, Liverpool L14 5AB, UK; (A.M.); (M.W.B.); (C.P.)
- Department of Paediatric Rheumatology, Alder Hey Children’s NHS Foundation Trust, East Prescott Road, Liverpool L14 5AB, UK
| | - Eileen M. Baildam
- Department of Paediatric Rheumatology, The Alexandra Hospital, Mill Lane, Cheadle SK8 2PX, UK;
| | - Rachel A. Oldershaw
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, William Henry Duncan Building, 6 West Derby Street, Liverpool L7 8TX, UK
| |
Collapse
|
25
|
Chang J, Chang Y, Cho Y, Jung HS, Park DI, Park SK, Ham SY, Wild SH, Byrne CD, Ryu S. Metabolic-associated fatty liver disease is associated with colorectal adenomas in young and older Korean adults. Liver Int 2023; 43:2548-2559. [PMID: 37735984 DOI: 10.1111/liv.15738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 09/03/2023] [Accepted: 09/05/2023] [Indexed: 09/23/2023]
Abstract
BACKGROUND AND AIMS Given that the majority of colorectal cancers (CRCs) develop from high-risk adenomas, identifying risk factors for high-risk adenomas is important. The relationship between metabolic dysfunction-associated fatty liver disease (MAFLD) and the risk of colorectal adenoma in young adults remains unclear. We aimed to evaluate this relationship in adults <50 (younger) and ≥50 (older) years of age. METHODS This cross-sectional study included 184 792 Korean adults (80% <50 years of age) who all underwent liver ultrasound and colonoscopy. Participants were grouped into those with and without MAFLD and classified by adenoma presence into no adenoma, low-risk adenoma, or high-risk adenoma (defined as ≥3 adenomas, any ≥10 mm, or adenoma with high-grade dysplasia/villous features). RESULTS The prevalence of low- and high-risk adenomas among young and older adults was 9.6% and 0.8% and 22.3% and 4.8%, respectively. MAFLD was associated with an increased prevalence of low- and high-risk adenomas in young and older adults. Young adults with MAFLD had a 1.30 (95% CIs 1.26-1.35) and 1.40 (1.23-1.59) times higher prevalence of low- and high-risk adenomas, respectively, compared to those without MAFLD. These associations were consistent even in lean adults (BMI < 23 kg/m2 ) and those without a family history of CRC. CONCLUSIONS MAFLD is associated with an increased prevalence of low- and high-risk adenomas in Korean adults, regardless of age or obesity status. Whether reducing metabolic risk factors, such as MAFLD, reduces the risk of precancerous lesions and ultimately reduces the risk of early-onset CRC requires further investigation.
Collapse
Affiliation(s)
- Jiwon Chang
- Total Healthcare Center, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Yoosoo Chang
- Center for Cohort Studies, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Department of Occupational and Environmental Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Department of Clinical Research Design & Evaluation, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul, Republic of Korea
| | - Yoosun Cho
- Total Healthcare Center, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Center for Cohort Studies, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hyun-Suk Jung
- Total Healthcare Center, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Department of Clinical Research Design & Evaluation, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul, Republic of Korea
| | - Dong-Il Park
- Division of Gastroenterology, Department of Medicine, School of Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University, Seoul, Republic of Korea
| | - Soo-Kyung Park
- Division of Gastroenterology, Department of Medicine, School of Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University, Seoul, Republic of Korea
| | - Soo-Youn Ham
- Department of Radiology, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Sarah H Wild
- Usher Institute, University of Edinburgh, Edinburgh, UK
| | - Christopher D Byrne
- Nutrition and Metabolism, Faculty of Medicine, University of Southampton, Southampton, UK
- National Institute for Health and Care Research Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| | - Seungho Ryu
- Center for Cohort Studies, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Department of Occupational and Environmental Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Department of Clinical Research Design & Evaluation, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul, Republic of Korea
| |
Collapse
|
26
|
Elnagdy M, Wang Y, Rodriguez W, Zhang J, Bauer P, Wilkey DW, Merchant M, Pan J, Farooqui Z, Cannon R, Rai S, Maldonado C, Barve S, McClain CJ, Gobejishvili L. Increased expression of phosphodiesterase 4 in activated hepatic stellate cells promotes cytoskeleton remodeling and cell migration. J Pathol 2023; 261:361-371. [PMID: 37735782 PMCID: PMC10653049 DOI: 10.1002/path.6194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 06/24/2023] [Accepted: 07/30/2023] [Indexed: 09/23/2023]
Abstract
Activation and transdifferentiation of hepatic stellate cells (HSC) into migratory myofibroblasts is a key process in liver fibrogenesis. Cell migration requires an active remodeling of the cytoskeleton, which is a tightly regulated process coordinated by Rho-specific guanine nucleotide exchange factors (GEFs) and the Rho family of small GTPases. Rho-associated kinase (ROCK) promotes assembly of focal adhesions and actin stress fibers by regulating cytoskeleton organization. GEF exchange protein directly activated by cAMP 1 (EPAC1) has been implicated in modulating TGFβ1 and Rho signaling; however, its role in HSC migration has never been examined. The aim of this study was to evaluate the role of cAMP-degrading phosphodiesterase 4 (PDE4) enzymes in regulating EPAC1 signaling, HSC migration, and fibrogenesis. We show that PDE4 protein expression is increased in activated HSCs expressing alpha smooth muscle actin and active myosin light chain (MLC) in fibrotic tissues of human nonalcoholic steatohepatitis cirrhosis livers and mouse livers exposed to carbon tetrachloride. In human livers, TGFβ1 levels were highly correlated with PDE4 expression. TGFβ1 treatment of LX2 HSCs decreased levels of cAMP and EPAC1 and increased PDE4D expression. PDE4 specific inhibitor, rolipram, and an EPAC-specific agonist decreased TGFβ1-mediated cell migration in vitro. In vivo, targeted delivery of rolipram to the liver prevented fibrogenesis and collagen deposition and decreased the expression of several fibrosis-related genes, and HSC activation. Proteomic analysis of mouse liver tissues identified the regulation of actin cytoskeleton by the kinase effectors of Rho GTPases as a major pathway impacted by rolipram. Western blot analyses confirmed that PDE4 inhibition decreased active MLC and endothelin 1 levels, key proteins involved in cytoskeleton remodeling and contractility. The current study, for the first time, demonstrates that PDE4 enzymes are expressed in hepatic myofibroblasts and promote cytoskeleton remodeling and HSC migration. © 2023 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Mohamed Elnagdy
- University of Louisville Alcohol Research Center, University of Louisville, Kentucky, USA
- Hepatobiology and Toxicology Center, University of Louisville, Kentucky, USA
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, Kentucky, USA
| | - Yali Wang
- University of Louisville Alcohol Research Center, University of Louisville, Kentucky, USA
- Hepatobiology and Toxicology Center, University of Louisville, Kentucky, USA
- Department of Medicine, School of Medicine, University of Louisville, Kentucky, USA
| | - Walter Rodriguez
- University of Louisville Alcohol Research Center, University of Louisville, Kentucky, USA
- Hepatobiology and Toxicology Center, University of Louisville, Kentucky, USA
- Department of Medicine, School of Medicine, University of Louisville, Kentucky, USA
| | - JingWen Zhang
- University of Louisville Alcohol Research Center, University of Louisville, Kentucky, USA
- Hepatobiology and Toxicology Center, University of Louisville, Kentucky, USA
- Department of Medicine, School of Medicine, University of Louisville, Kentucky, USA
| | - Philip Bauer
- Department of Physiology, School of Medicine, University of Louisville, Kentucky, USA
- EndoProtech, Inc., Louisville, Kentucky, USA
| | - Daniel W. Wilkey
- Hepatobiology and Toxicology Center, University of Louisville, Kentucky, USA
- Department of Medicine, School of Medicine, University of Louisville, Kentucky, USA
| | - Michael Merchant
- University of Louisville Alcohol Research Center, University of Louisville, Kentucky, USA
- Hepatobiology and Toxicology Center, University of Louisville, Kentucky, USA
- Department of Medicine, School of Medicine, University of Louisville, Kentucky, USA
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, Kentucky, USA
| | - Jianmin Pan
- Department of Bioinformatics and Biostatistics, School of Public Health and Information Sciences, University of Louisville, Kentucky, USA
| | - Zainab Farooqui
- Department of Medicine, School of Medicine, University of Louisville, Kentucky, USA
| | - Robert Cannon
- Department of Surgery, School of Medicine, University of Louisville, Kentucky, USA
| | - Shesh Rai
- University of Louisville Alcohol Research Center, University of Louisville, Kentucky, USA
- Hepatobiology and Toxicology Center, University of Louisville, Kentucky, USA
- Department of Bioinformatics and Biostatistics, School of Public Health and Information Sciences, University of Louisville, Kentucky, USA
| | - Claudio Maldonado
- Department of Physiology, School of Medicine, University of Louisville, Kentucky, USA
- EndoProtech, Inc., Louisville, Kentucky, USA
| | - Shirish Barve
- University of Louisville Alcohol Research Center, University of Louisville, Kentucky, USA
- Hepatobiology and Toxicology Center, University of Louisville, Kentucky, USA
- Department of Medicine, School of Medicine, University of Louisville, Kentucky, USA
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, Kentucky, USA
| | - Craig J. McClain
- University of Louisville Alcohol Research Center, University of Louisville, Kentucky, USA
- Hepatobiology and Toxicology Center, University of Louisville, Kentucky, USA
- Department of Medicine, School of Medicine, University of Louisville, Kentucky, USA
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, Kentucky, USA
- Robley Rex VA Medical Center, Louisville, Kentucky, USA
| | - Leila Gobejishvili
- University of Louisville Alcohol Research Center, University of Louisville, Kentucky, USA
- Hepatobiology and Toxicology Center, University of Louisville, Kentucky, USA
- Department of Medicine, School of Medicine, University of Louisville, Kentucky, USA
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, Kentucky, USA
- Department of Physiology, School of Medicine, University of Louisville, Kentucky, USA
| |
Collapse
|
27
|
Cai CL, Marcelino M, Aranda JV, Beharry KD. Comparison of hyperoxia or normoxia resolution of intermittent hypoxia and intermittent hyperoxia episodes on liver histopathology, IGF-1, IGFBP-3, and GHBP in neonatal rats. Growth Horm IGF Res 2023; 72-73:101559. [PMID: 37708588 DOI: 10.1016/j.ghir.2023.101559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 08/23/2023] [Accepted: 09/06/2023] [Indexed: 09/16/2023]
Abstract
OBJECTIVE Extremely low gestational age neonates requiring oxygen therapy for chronic lung disease experience repeated fluctuations in arterial oxygen saturation, or intermittent hypoxia (IH), during the first few weeks of life. These events are associated with a high risk for reduced growth, hypertension, and insulin resistance in later life. This study tested the hypothesis that IH, or intermittent hyperoxia have similar negative effects on the liver; somatic growth; and liver insulin-like growth factor (IGF)-I, IGF binding protein (BP)-3, and growth hormone binding protein (GHBP), regardless of resolution in normoxia or hyperoxia between episodes. DESIGN Newborn rats on the first day of life (P0) were exposed to two IH paradigms: 1) hyperoxia (50% O2) with brief hypoxia (12% O2); or 2) normoxia (21% O2) with hypoxia (12% O2); intermittent hypoxia (50% O2/21% O2); hyperoxia only (50% O2); or room air (RA, 21% O2). Pups were euthanized on P14 or placed in RA until P21. Controls remained in RA from P0-P21. Growth, liver histopathology, apoptosis, IGFI, IGFBP-3, and GHBP were assessed. RESULTS Pathological findings of the liver hepatocytes, including cellular swelling, steatosis, apoptosis, necrosis and focal sinusoid congestion were seen in the IH and intermittent hyperoxia groups, and were particularly severe in the 21-12% O2 group during exposure (P14) with no significant improvements during recovery/reoxygenation (P21). These effects were associated with induction of HIF1α, and reductions in liver IGFI, IGFBP-3, and GHBP. CONCLUSIONS Exposure to IH or intermittent hyperoxia during the first few weeks of life regardless of resolution in RA or hyperoxia is detrimental to the immature liver. These findings may suggest that interventions to prevent frequent fluctuations in oxygen saturation during early neonatal life remain a high priority.
Collapse
Affiliation(s)
- Charles L Cai
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, State University of New York, Downstate Medical Center, Brooklyn, NY 11203, USA
| | - Matthew Marcelino
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, State University of New York, Downstate Medical Center, Brooklyn, NY 11203, USA
| | - Jacob V Aranda
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, State University of New York, Downstate Medical Center, Brooklyn, NY 11203, USA; Department of Ophthalmology; State University of New York, Downstate Medical Center, Brooklyn, NY 11203, USA
| | - Kay D Beharry
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, State University of New York, Downstate Medical Center, Brooklyn, NY 11203, USA; Department of Ophthalmology; State University of New York, Downstate Medical Center, Brooklyn, NY 11203, USA.
| |
Collapse
|
28
|
Vázquez-Borrego MC, Del Río-Moreno M, Pyatkov M, Sarmento-Cabral A, Mahmood M, Pelke N, Wnek M, Cordoba-Chacon J, Waxman DJ, Puchowicz MA, McGuinness OP, Kineman RD. Direct and systemic actions of growth hormone receptor (GHR)-signaling on hepatic glycolysis, de novo lipogenesis and insulin sensitivity, associated with steatosis. Metabolism 2023; 144:155589. [PMID: 37182789 PMCID: PMC10843389 DOI: 10.1016/j.metabol.2023.155589] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/28/2023] [Accepted: 05/08/2023] [Indexed: 05/16/2023]
Abstract
BACKGROUND Evidence is accumulating that growth hormone (GH) protects against the development of steatosis and progression of non-alcoholic fatty liver disease (NAFLD). GH may control steatosis indirectly by altering systemic insulin sensitivity and substrate delivery to the liver and/or by the direct actions of GH on hepatocyte function. APPROACH To better define the hepatocyte-specific role of GH receptor (GHR) signaling on regulating steatosis, we used a mouse model with adult-onset, hepatocyte-specific GHR knockdown (aHepGHRkd). To prevent the reduction in circulating insulin-like growth factor 1 (IGF1) and the subsequent increase in GH observed after aHepGHRkd, subsets of aHepGHRkd mice were treated with adeno-associated viral vectors (AAV) driving hepatocyte-specific expression of IGF1 or a constitutively active form of STAT5b (STAT5bCA). The impact of hepatocyte-specific modulation of GHR, IGF1 and STAT5b on carbohydrate and lipid metabolism was studied across multiple nutritional states and in the context of hyperinsulinemic:euglycemic clamps. RESULTS Chow-fed male aHepGHRkd mice developed steatosis associated with an increase in hepatic glucokinase (GCK) and ketohexokinase (KHK) expression and de novo lipogenesis (DNL) rate, in the post-absorptive state and in response to refeeding after an overnight fast. The aHepGHRkd-associated increase in hepatic KHK, but not GCK and steatosis, was dependent on hepatocyte expression of carbohydrate response element binding protein (ChREBP), in re-fed mice. Interestingly, under clamp conditions, aHepGHRkd also increased the rate of DNL and expression of GCK and KHK, but impaired insulin-mediated suppression of hepatic glucose production, without altering plasma NEFA levels. These effects were normalized with AAV-mediated hepatocyte expression of IGF1 or STAT5bCA. Comparison of the impact of AAV-mediated hepatocyte IGF1 versus STAT5bCA in aHepGHRkd mice across multiple nutritional states, indicated the restorative actions of IGF1 are indirect, by improving systemic insulin sensitivity, independent of changes in the liver transcriptome. In contrast, the actions of STAT5b are due to the combined effects of raising IGF1 and direct alterations in the hepatocyte gene program that may involve suppression of BCL6 and FOXO1 activity. However, the direct and IGF1-dependent actions of STAT5b cannot fully account for enhanced GCK activity and lipogenic gene expression observed after aHepGHRkd, suggesting other GHR-mediated signals are involved. CONCLUSION These studies demonstrate hepatocyte GHR-signaling controls hepatic glycolysis, DNL, steatosis and hepatic insulin sensitivity indirectly (via IGF1) and directly (via STAT5b). The relative contribution of these indirect and direct actions of GH on hepatocytes is modified by insulin and nutrient availability. These results improve our understanding of the physiologic actions of GH on regulating adult metabolism to protect against NAFLD progression.
Collapse
Affiliation(s)
- Mari C Vázquez-Borrego
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Illinois at Chicago, Chicago, IL, United States of America; Research and Development Division, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, United States of America
| | - Mercedes Del Río-Moreno
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Illinois at Chicago, Chicago, IL, United States of America; Research and Development Division, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, United States of America
| | - Maxim Pyatkov
- Department of Biology & Bioinformatics Program, Boston University, Boston, MA, United States of America
| | - André Sarmento-Cabral
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Illinois at Chicago, Chicago, IL, United States of America; Research and Development Division, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, United States of America
| | - Mariyah Mahmood
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Illinois at Chicago, Chicago, IL, United States of America; Research and Development Division, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, United States of America
| | - Natalie Pelke
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Illinois at Chicago, Chicago, IL, United States of America; Research and Development Division, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, United States of America
| | - Magdalena Wnek
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Illinois at Chicago, Chicago, IL, United States of America; Research and Development Division, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, United States of America
| | - Jose Cordoba-Chacon
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Illinois at Chicago, Chicago, IL, United States of America; Research and Development Division, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, United States of America
| | - David J Waxman
- Department of Biology & Bioinformatics Program, Boston University, Boston, MA, United States of America
| | - Michelle A Puchowicz
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - Owen P McGuinness
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, United States of America
| | - Rhonda D Kineman
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Illinois at Chicago, Chicago, IL, United States of America; Research and Development Division, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, United States of America.
| |
Collapse
|
29
|
Shimonty A, Bonewald LF, Huot JR. Metabolic Health and Disease: A Role of Osteokines? Calcif Tissue Int 2023; 113:21-38. [PMID: 37193929 DOI: 10.1007/s00223-023-01093-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 05/01/2023] [Indexed: 05/18/2023]
Abstract
Maintenance of skeletal health is tightly regulated by osteocytes, osteoblasts, and osteoclasts via coordinated secretion of bone-derived factors, termed osteokines. Disruption of this coordinated process due to aging and metabolic disease promotes loss of bone mass and increased risk of fracture. Indeed, growing evidence demonstrates that metabolic diseases, including type 2 diabetes, liver disease and cancer are accompanied by bone loss and altered osteokine levels. With the persistent prevalence of cancer and the growing epidemic of metabolic disorders, investigations into the role of inter-tissue communication during disease progression are on the rise. While osteokines are imperative for bone homeostasis, work from us and others have identified that osteokines possess endocrine functions, exerting effects on distant tissues including skeletal muscle and liver. In this review we first discuss the prevalence of bone loss and osteokine alterations in patients with type 2 diabetes, non-alcoholic fatty liver disease, non-alcoholic steatohepatitis, cirrhosis, and cancer. We then discuss the effects of osteokines in mediating skeletal muscle and liver homeostasis, including RANKL, sclerostin, osteocalcin, FGF23, PGE2, TGF-β, BMPs, IGF-1 and PTHrP. To better understand how inter-tissue communication contributes to disease progression, it is essential that we include the bone secretome and the systemic roles of osteokines.
Collapse
Affiliation(s)
- Anika Shimonty
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Lynda F Bonewald
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA
- Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Joshua R Huot
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA.
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA.
- Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA.
- Department of Kinesiology, School of Health and Human Sciences, Indiana University Purdue University Indianapolis, Indianapolis, IN, USA.
| |
Collapse
|
30
|
Lu R, Liu Y, Hong T. Epidemiological characteristics and management of nonalcoholic fatty liver disease/nonalcoholic steatohepatitis in China: A narrative review. Diabetes Obes Metab 2023; 25 Suppl 1:13-26. [PMID: 36775938 DOI: 10.1111/dom.15014] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/27/2023] [Accepted: 02/08/2023] [Indexed: 02/14/2023]
Abstract
AIM With industrialization and spread of the westernized lifestyle, the number of people affected by non-alcoholic fatty liver disease (NAFLD)/non-alcoholic steatohepatitis (NASH) is growing rapidly in China; this has become a major public health concern. To better understand the burden and characteristics of NAFLD/NASH in China, we aim to perform a narrative review of the literature published in this field. MATERIALS AND METHODS We carried out a comprehensive electronic search of five English-language and three Chinese-language databases, to identify studies regarding NAFLD or NASH published from inception to November 30, 2022. Epidemiological studies of NAFLD/NASH in China were particularly noticed and summarized. We also searched the www. CLINICALTRIALS gov and www.chictr.org.cn websites for the registered trials on the treatment of the disease led by Chinese investigators or located in China. RESULTS The increasing rate of NAFLD prevalence in China is strikingly high, reaching more than twice that in western countries. The prevalence of NAFLD is nearly 30% of the general Chinese population, making it the leading cause of chronic liver diseases. The prevalence of NAFLD/NASH varies between provinces/regions, age groups, sexes, and individuals with different metabolic profiles. NAFLD co-exists in many Chinese patients with chronic hepatitis B. Since 2020, more Chinese studies have used the term metabolic-associated fatty liver disease (MAFLD), emphasizing the underlying metabolic disorders that occur concurrently with this disease. Several clinical trials involving lifestyle interventions, antidiabetic drugs, or traditional Chinese medicines, registered by Chinese investigators, have been completed or are ongoing. Moreover, several innovative targeted therapies developed in China are revolutionizing the treatment of NAFLD/NASH. CONCLUSIONS NAFLD has cast a heavy burden on the Chinese healthcare system. Chinese scholars are making efforts to achieve the optimal management of this disease.
Collapse
Affiliation(s)
- Ran Lu
- Department of Endocrinology and Metabolism, Peking University Third Hospital, Beijing, China
| | - Ye Liu
- Department of Endocrinology and Metabolism, Peking University Third Hospital, Beijing, China
| | - Tianpei Hong
- Department of Endocrinology and Metabolism, Peking University Third Hospital, Beijing, China
| |
Collapse
|
31
|
Pal Chaudhary S, Reyes S, Chase ML, Govindan A, Zhao L, Luther J, Bhan I, Bethea E, Franses JW, Paige Walsh E, Anne Dageford L, Kimura S, Elias N, Yeh H, Markman J, Bozorgzadeh A, Tanabe K, Ferrone C, Zhu AX, Andersson K, Thiim M, Antonio Catalano O, Kambadakone A, Vagefi PA, Qadan M, Pratt D, Hashemi N, Corey KE, Misdraji J, Goyal L, Clark JW. Resection of NAFLD/NASH-related Hepatocellular Carcinoma (HCC): Clinical Features and Outcomes Compared with HCC Due to Other Etiologies. Oncologist 2023; 28:341-350. [PMID: 36763374 PMCID: PMC10078904 DOI: 10.1093/oncolo/oyac251] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 10/19/2022] [Indexed: 02/11/2023] Open
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH) are the leading causes of hepatocellular carcinoma (HCC) worldwide. Limited data exist on surgical outcomes for NAFLD/NASH-related HCC compared with other HCC etiologies. We evaluated differences in clinicopathological characteristics and outcomes of patients undergoing surgical resection for NAFLD/NASH-associated HCC compared with other HCC etiologies. METHODS Demographic, clinicopathological features, and survival outcomes of patients with surgically resected HCC were collected. NAFLD activity score (NAS) and fibrosis score were assessed by focused pathologic review in a subset of patients. RESULTS Among 492 patients screened, 260 met eligibility (NAFLD/NASH [n = 110], and other etiologies [n = 150]). Median age at diagnosis was higher in the NAFLD/NASH HCC cohort compared with the other etiologies cohort (66.7 vs. 63.4 years, respectively, P = .005), with an increased percentage of female patients (36% vs. 18%, P = .001). NAFLD/NASH-related tumors were more commonly >5 cm (66.0% vs. 45%, P = .001). There were no significant differences in rates of lymphovascular or perineural invasion, histologic grade, or serum AFP levels. The NAFLD/NASH cohort had lower rates of background liver fibrosis, lower AST and ALT levels, and higher platelet counts (P < .01 for all). Median overall survival (OS) was numerically shorter in NAFLD/NASH vs other etiology groups, however, not statistically significant. CONCLUSIONS Patients with NAFLD/NASH-related HCC more commonly lacked liver fibrosis and presented with larger HCCs compared with patients with HCC from other etiologies. No differences were seen in rates of other high-risk features or survival. With the caveat of sample size and retrospective analysis, this supports a similar decision-making approach regarding surgical resection for NAFLD/NASH and other etiology-related HCCs.
Collapse
Affiliation(s)
- Surendra Pal Chaudhary
- Division of Oncology, Mass General Cancer Center and Harvard Medical School, Boston, MA, USA
| | | | | | | | - Lei Zhao
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jay Luther
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Irun Bhan
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Emily Bethea
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Joseph W Franses
- Division of Oncology, Mass General Cancer Center and Harvard Medical School, Boston, MA, USA
| | - Elizabeth Paige Walsh
- Division of Oncology, Mass General Cancer Center and Harvard Medical School, Boston, MA, USA
| | - Leigh Anne Dageford
- Transplantation Unit, Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Shoko Kimura
- Transplantation Unit, Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Nahel Elias
- Transplantation Unit, Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Heidi Yeh
- Transplantation Unit, Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - James Markman
- Transplantation Unit, Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Adel Bozorgzadeh
- Transplantation Unit, Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Kenneth Tanabe
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Cristina Ferrone
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Andrew X Zhu
- Jiahui Health, Jiahui International Cancer Center, Shanghai, People's Republic of China
| | - Karin Andersson
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Michael Thiim
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Onofrio Antonio Catalano
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Avinash Kambadakone
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Parsia A Vagefi
- Division of Surgical Transplantation, University of Texas Southwestern, Dallas, TX, USA
| | - Motaz Qadan
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Daniel Pratt
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Nikroo Hashemi
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Kathleen E Corey
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Joseph Misdraji
- Department of Pathology, Yale New Haven Hospital, Yale University, New Haven, CT, USA
| | - Lipika Goyal
- Division of Oncology, Mass General Cancer Center and Harvard Medical School, Boston, MA, USA
| | - Jeffrey W Clark
- Division of Oncology, Mass General Cancer Center and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
32
|
The Role of Growth Hormone and Insulin Growth Factor 1 in the Development of Non-Alcoholic Steato-Hepatitis: A Systematic Review. Cells 2023; 12:cells12040517. [PMID: 36831184 PMCID: PMC9954460 DOI: 10.3390/cells12040517] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 01/30/2023] [Accepted: 02/02/2023] [Indexed: 02/08/2023] Open
Abstract
Diabetic and obese patients have a high prevalence of non-alcoholic fatty liver disease (NAFLD). This condition groups a spectrum of conditions varying from simple steatosis to non-alcoholic steatohepatitis (NASH), with or without fibrosis. Multiple factors are involved in the development of NAFLD. However, details about its pathogenesis and factors that promote the progression to NASH are still missing. Growth hormone (GH) and insulin-like growth factor 1 (IGF-1) regulate metabolic, immune, and hepatic stellate cell functions. Increasing evidence suggests they may have roles in the progression from NAFLD to NASH. Following the PRISMA reporting guidelines, we conducted a systematic review to evaluate all clinical and experimental studies published in the literature correlating GH and IGF-1 to inflammation and fibrosis in NAFLD and NASH. Our results showed that GH and IGF-1 have a fundamental role in the pathogenesis of NASH, acting in slightly different ways to produce a synergic effect. Indeed, GH may mediate its protective effect in the pathogenesis of NASH by regulating lipogenesis pathways, while IGF-1 has the same effect by regulating cholesterol transport. Therefore, they could be used as therapeutic strategies in preventing NAFLD progression to NASH.
Collapse
|
33
|
Arlien-Søborg MC, Madsen MA, Dal J, Krusenstjerna-Hafstrøm T, Ringgaard S, Skou N, Høgild M, Jørgensen JOL. Ectopic lipid deposition and insulin resistance in patients with GH disorders before and after treatment. Eur J Endocrinol 2023; 188:6984866. [PMID: 36651164 DOI: 10.1093/ejendo/lvac014] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 12/02/2022] [Accepted: 12/08/2022] [Indexed: 01/13/2023]
Abstract
OBJECTIVES Insulin resistance is associated with ectopic lipid deposition. Growth hormone (GH) status also modulates ectopic lipid accumulation, but how this associates with insulin resistance in patients with GH disorders is not well established. DESIGN AND METHODS Twenty-one patients diagnosed with acromegaly and 12 patients with adult GH deficiency (GHD) were studied at diagnosis and after treatment. A reference group of 12 subjects was included. Each study day comprised assessment of body composition with dual-energy X-ray absorptiometry, ectopic lipid deposition in the liver by MR spectroscopy, and Homeostatic Model Assessment for Insulin Resistance (HOMA-IR). RESULTS Disease control of acromegaly decreased lean body mass (LBM) (P < .000) and increased the percentage of total body fat (TBF) (P < .000). GH replacement increased LBM in the GHD patients (P = .007) and decreased the percentage of TBF (P = .010). The intrahepatic lipid (IHL) content increased after disease control in acromegaly (P = .004), whereas IHL did not change significantly after GH replacement in GHD (P = .34). Insulin resistance (HOMA-IR) improved after disease control of acromegaly (P < .000) and remained unaltered after GH replacement in the GHD patients (P = .829). CONCLUSIONS GH status is a significant modulator of body composition and insulin sensitivity.GH excess reduces total fat mass and intrahepatic lipid content together with induction of insulin resistance.The data support the notion that GH-induced insulin resistance is unassociated with hepatic lipid accumulation.
Collapse
Affiliation(s)
- Mai C Arlien-Søborg
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Medical Research Laboratory, Aarhus University Hospital, Aarhus, Denmark
| | - Michael Alle Madsen
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Nuclear Medicine & PET Centre, Aarhus University Hospital, Aarhus, Denmark
| | - Jakob Dal
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Endocrinology, Aalborg University Hospital, Aalborg, Denmark
| | | | - Steffen Ringgaard
- Department of Clinical Medicine, The MR Research Centre, Aarhus University Hospital, Aarhus, Denmark
| | - Nickolaj Skou
- Department of Radiology, Aarhus University Hospital, Aarhus, Denmark
| | - Morten Høgild
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Medical Research Laboratory, Aarhus University Hospital, Aarhus, Denmark
| | - Jens Otto Lunde Jørgensen
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Medical Research Laboratory, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
34
|
Altalebi RR, Al-Hussaniy HA, Al-Tameemi ZS, Al-Zobaidy MAH, Albu-Rghaif AH, Alkuraishy HM, Hedeab GM, Azam F, Al-Samydai AM, Naji MA. Non-alcoholic fatty liver disease: relation to juvenile obesity, lipid profile, and hepatic enzymes. J Med Life 2023; 16:42-47. [PMID: 36873135 PMCID: PMC9979179 DOI: 10.25122/jml-2022-0091] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 06/07/2022] [Indexed: 03/07/2023] Open
Abstract
The prevalence of juvenile obesity is increasing, reaching epidemic proportions, presenting a link not only to NAFLD (non-alcoholic fatty liver disease) but to abnormal lipid profiles and liver enzyme abnormalities. Liver ultrasonography is a sensitive and specific tool for the recognition of NAFLD. This study aims to assess the association between NAFLD and juvenile obesity and to determine the other related changes in a set of indicators, including lipid profile abnormalities and serum transaminases. The sample included 470 obese and 210 non-obese individuals aged 6-16. Anthropometric measures were assessed, with the serum lipid profile and liver transaminases, and abdominal ultrasonography was used to detect NAFLD. Fatty liver was found in 38% of the obese subjects and none of the non-obese subjects. Within obese subjects, mean body mass index (BMI) and waist circumference increased significantly in patients with NAFLD compared to those without fatty liver. Moreover, LDL (low-density lipoprotein), CHOL (cholesterol), and serum liver enzymes were significantly higher in the presence of NAFLD. In conclusion, NAFLD commonly associates with juvenile obesity, relating to obesity and the abnormal lipid profile (including elevated CHOL and LDL) among obese people, reflecting elevated liver transaminases, which increase the risk of cirrhosis.
Collapse
Affiliation(s)
| | - Hany Akeel Al-Hussaniy
- Department of Pharmacy, Bilad Alrafidain University College, Diyala, Iraq.,Dr. Hany Akeel Institute, Iraqi Medical Research Center, Baghdad, Iraq
| | - Zahraa Salam Al-Tameemi
- Department of Pharmacy, Bilad Alrafidain University College, Diyala, Iraq.,Dr. Hany Akeel Institute, Iraqi Medical Research Center, Baghdad, Iraq
| | | | | | - Hayder Mutter Alkuraishy
- Department of Clinical Pharmacology, College of Medicine, Almustansria University, Baghdad, Iraq
| | - Gomaa Mostafa Hedeab
- Pharmacology Department & Health Research Unit, Medical College, Jouf University, Jouf, Saudi Arabia.,Pharmacology Department, Faculty of Medicine, Beni-Suef University, Beni-Suef, Egypt
| | - Faizul Azam
- Department of Pharmaceutical Chemistry and Pharmacognosy, Unaizah College of Pharmacy, Qassim University, Uniazah, Saudi Arabia
| | - Ali Mahmoud Al-Samydai
- Pharmacological and Diagnostic Research Centre, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Meena Akeel Naji
- Dr. Hany Akeel Institute, Iraqi Medical Research Center, Baghdad, Iraq
| |
Collapse
|
35
|
Mosca A, Della Volpe L, Alisi A, Panera N, Maggiore G, Vania A. The Role of the GH/IGF1 Axis on the Development of MAFLD in Pediatric Patients with Obesity. Metabolites 2022; 12:metabo12121221. [PMID: 36557260 PMCID: PMC9788441 DOI: 10.3390/metabo12121221] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 11/27/2022] [Accepted: 12/02/2022] [Indexed: 12/10/2022] Open
Abstract
The anomalies of the Growth Hormone (GH)/Insulin-like Growth Factor-1 (IGF1) axis are associated with a higher prevalence of Metabolic Associated Fatty Liver Disease (MAFLD) and with a more rapid progression towards fibrosis, cirrhosis, and end-stage liver disease. A total of 191 adolescents with obesity [12−18 years] were consecutively enrolled between January 2014 and December 2020 and underwent liver biopsy to diagnose MAFLD severity. In all patients GH, IGF1 and Insulin-like Growth Factor-Binding Protein 3 (IGFBP3) were measured. Patients with inflammation and ballooning have significantly lower values of GH and IGF1 than those without (GH: 5.4 vs. 7.5 ng/mL; IGF1 245 vs. 284 ng/mL, p < 0.05). GH and IGF1 were also negatively correlated with fibrosis’ degree (r = −0.51, p = 0.001, and r = −0.45, p = 0.001, respectively). Only GH correlated with TNF-a (r = −0.29, p = 0.04) and lobular inflammation (r = −0.36, p = 0.02). At multivariate regression, both GH and IGF1 values, after adjustment for age, sex and BMI, were negatively associated with HOMA-IR but above all with fibrosis (GH→β = −2.3, p = 0.001, IGF1→β = −2.8, p = 0.001). Even in the pediatric population, a reduction of GH input in the liver directly promotes development of de novo hepatic lipogenesis, steatosis, fibrosis and inflammation. The possible role of recombinant GH administration in adolescents with obesity and severe MAFLD deserves to be studied.
Collapse
Affiliation(s)
- Antonella Mosca
- Hepatogastroenterology, Nutrition, Digestive Endoscopy and Liver Transplant Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy
- Correspondence:
| | - Luca Della Volpe
- Hepatogastroenterology, Nutrition, Digestive Endoscopy and Liver Transplant Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy
| | - Anna Alisi
- Pathology Unit, Department of Diagnostic and Laboratory Medicine, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy
| | - Nadia Panera
- Pathology Unit, Department of Diagnostic and Laboratory Medicine, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy
| | - Giuseppe Maggiore
- Hepatogastroenterology, Nutrition, Digestive Endoscopy and Liver Transplant Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy
| | | |
Collapse
|
36
|
Das L, Sahoo J, Dahiya N, Taneja S, Bhadada SK, Bhat MH, Singh P, Suri V, Laway BA, Dutta P. Long-term hepatic and cardiac health in patients diagnosed with Sheehan's syndrome. Pituitary 2022; 25:971-981. [PMID: 36243797 DOI: 10.1007/s11102-022-01282-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/30/2022] [Indexed: 11/25/2022]
Abstract
PURPOSE Sheehan's syndrome (SS) is characterised by chronic pituitary insufficiency following a vascular insult to the pituitary in the peripartum period. There is a lack of substantial evidence on the long-term hepatic and cardiac consequences in these patients, following hormone replacement. METHODS Patients with a diagnosis of SS were recruited for the study. Detailed clinico-biochemical and radiological evaluation were performed in all patients (n = 60). Hepatic and cardiac complications were assessed using fibroscan and echocardiography (2D speckle-tracking) respectively, in a subset of patients (n = 29) as well as age-and BMI-matched controls (n = 26). Controlled attenuation parameter (for steatosis) and liver stiffness measurement (for fibrosis) were used to define non-alcoholic fatty liver disease (NAFLD). Diastolic cardiac function was evaluated using standard criteria and systolic function by ejection fraction and global longitudinal strain (GLS). RESULTS The mean age of the cohort was 42.7 ± 11.6 years. Multiple (≥ 2) hormone deficiencies were present in 68.8% of patients, with hypothyroidism (91.4%), hypocortisolism (88.3%), and growth hormone (GH) deficiency (85.7%) being the most common. At a mean follow-up of 9.8 ± 6.8 years, NAFLD was present in 63% of patients, with 51% having severe steatosis, which was predicted by the presence of GH deficiency and higher body mass index. Though the ejection fraction was similar, increased left ventricular GLS (18.8 vs. 7.7%) was present in a significantly higher number of patients versus controls. CONCLUSION NAFLD, especially severe hepatic steatosis, is highly prevalent in SS. Subclinical cardiac systolic dysfunction (impaired GLS) is also more common, but of mild intensity.
Collapse
Affiliation(s)
- Liza Das
- Department of Endocrinology, Post Graduate Institute of Medical Education and Research (PGIMER), Nehru Extension Block, Chandigarh, 160012, India
| | - Jayaprakash Sahoo
- Department of Endocrinology, Jawaharlal Institute of Post Graduate Medical Education and Research (JIPMER), Puducherry, India
| | | | - Sunil Taneja
- Department of Hepatology, PGIMER, Chandigarh, India
| | - Sanjay Kumar Bhadada
- Department of Endocrinology, Post Graduate Institute of Medical Education and Research (PGIMER), Nehru Extension Block, Chandigarh, 160012, India
| | - Mohammad Hayat Bhat
- Department of Internal Medicine and Endocrinology, Government Medical College, Srinagar, India
| | | | - Vanita Suri
- Department of Obstetrics and Gynaecology, PGIMER, Chandigarh, India
| | - Bashir Ahmad Laway
- Department of Endocrinology, Sher-I-Kashmir Institute of Medical Sciences (SKIMS), Soura, Srinagar, India.
| | - Pinaki Dutta
- Department of Endocrinology, Post Graduate Institute of Medical Education and Research (PGIMER), Nehru Extension Block, Chandigarh, 160012, India.
| |
Collapse
|
37
|
Abstract
Growth hormone orchestrates a complex, sex-dependent balancing act.
Collapse
Affiliation(s)
- David J Waxman
- Department of Biology and Bioinformatics Program, Boston University, Boston, MA, USA
| | - Rhonda D Kineman
- Department of Medicine, Section of Endocrinology, Diabetes, and Metabolism, University of Illinois at Chicago, Chicago, IL, USA
- Jesse Brown VA Medical Center, Chicago, IL, USA
| |
Collapse
|
38
|
Important Hormones Regulating Lipid Metabolism. Molecules 2022; 27:molecules27207052. [PMID: 36296646 PMCID: PMC9607181 DOI: 10.3390/molecules27207052] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/07/2022] [Accepted: 10/10/2022] [Indexed: 11/17/2022] Open
Abstract
There is a wide variety of kinds of lipids, and complex structures which determine the diversity and complexity of their functions. With the basic characteristic of water insolubility, lipid molecules are independent of the genetic information composed by genes to proteins, which determine the particularity of lipids in the human body, with water as the basic environment and genes to proteins as the genetic system. In this review, we have summarized the current landscape on hormone regulation of lipid metabolism. After the well-studied PI3K-AKT pathway, insulin affects fat synthesis by controlling the activity and production of various transcription factors. New mechanisms of thyroid hormone regulation are discussed, receptor α and β may mediate different procedures, the effect of thyroid hormone on mitochondria provides a new insight for hormones regulating lipid metabolism. Physiological concentration of adrenaline induces the expression of extrapituitary prolactin in adipose tissue macrophages, which promotes fat weight loss. Manipulation of hormonal action has the potential to offer a new therapeutic horizon for the global burden of obesity and its associated complications such as morbidity and mortality.
Collapse
|
39
|
Al-Massadi O, Parini P, Fernø J, Luquet S, Quiñones M. Metabolic actions of the growth hormone-insulin growth factor-1 axis and its interaction with the central nervous system. Rev Endocr Metab Disord 2022; 23:919-930. [PMID: 35687272 DOI: 10.1007/s11154-022-09732-x] [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] [Accepted: 05/02/2022] [Indexed: 10/18/2022]
Abstract
The growth hormone/insulin growth factor-1 axis is a key endocrine system that exerts profound effects on metabolism by its actions on different peripheral tissues but also in the brain. Growth hormone together with insulin growth factor-1 perform metabolic adjustments, including regulation of food intake, energy expenditure, and glycemia. The dysregulation of this hepatic axis leads to different metabolic disorders including obesity, type 2 diabetes or liver disease. In this review, we discuss how the growth hormone/insulin growth factor-1 axis regulates metabolism and its interactions with the central nervous system. Finally, we state our vision for possible therapeutic uses of compounds based in the components of this hepatic axis.
Collapse
Affiliation(s)
- Omar Al-Massadi
- Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Travesía da Choupana s/n, 15706, Santiago de Compostela, Spain.
- CIBER de Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, 15706, Santiago de Compostela, Spain.
| | - Paolo Parini
- Department of Laboratory Medicine, Division of Clinical Chemistry, Karolinska Institute, Stockholm, Sweden
- Department of Medicine, Metabolism Unit, Karolinska Institute at Karolinska University Hospital Huddinge, Stockholm, Sweden
- Patient Area Nephrology and Endocrinology, Inflammation and Infection Theme, Karolinska University Hospital, Stockholm, Sweden
| | - Johan Fernø
- Hormone Laboratory, Haukeland University Hospital, Bergen, Norway
| | - Serge Luquet
- Unité de Biologie Fonctionnelle et Adaptative, Univ Paris Diderot, Sorbonne Paris Cité, CNRS UMR 8251, F-75205, Paris, France
| | - Mar Quiñones
- Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Travesía da Choupana s/n, 15706, Santiago de Compostela, Spain.
- CIBER de Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, 15706, Santiago de Compostela, Spain.
- Unité de Biologie Fonctionnelle et Adaptative, Univ Paris Diderot, Sorbonne Paris Cité, CNRS UMR 8251, F-75205, Paris, France.
| |
Collapse
|
40
|
Affiliation(s)
- Damasia Becu-Villalobos
- Instituto de Biología y Medicina Experimental CONICET, Buenos Aires,Argentina
- Academia Nacional de Ciencias de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|