1
|
Chen F, Wang Z, Yao H, Liu Q, Gan Y, Xu S, Bao H, Jin Y, Hu YP, Gao J, Song S, Wang MJ. Large-scale manufacturing of human gallbladder epithelial cell products and derived hepatocytes via a chemically defined approach. Trends Biotechnol 2025:S0167-7799(25)00136-2. [PMID: 40399214 DOI: 10.1016/j.tibtech.2025.04.009] [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: 09/17/2024] [Revised: 03/26/2025] [Accepted: 04/08/2025] [Indexed: 05/23/2025]
Abstract
Manufacturing sufficient quantities of high-quality hepatocytes holds significant promise for the treatment of liver diseases and drug screening. Here, we developed a chemically defined, animal-free method for the large-scale production of human gallbladder epithelial cells (hGBECs) under good manufacturing practice conditions, enabling their clinical application. The cell products were characterized for growth ability, phenotype, freeze-thaw viability, genetic stability, biological contamination, tumorigenicity, and acute toxicity to ensure quality control and biological safety. We also provide a protocol for generating functional hepatocytes from hGBECs. The derived hepatocytes demonstrated typical liver functions, including albumin secretion, urea production, and drug metabolism. In addition, these cells were used in drug toxicity testing. We conducted further functional experiments on Cu2+ transport and alcohol metabolism. Transplantation of these cells in vivo was able to rescue mice from liver failure. This large-scale, convenient strategy for manufacturing hGBECs serves as a biobank for clinical applications and provides a valuable model for studying liver diseases.
Collapse
Affiliation(s)
- Fei Chen
- Department of Cell Biology, Second Military Medical University (Naval Medical University), Shanghai, China.
| | - Zijun Wang
- Department of Cell Biology, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Hao Yao
- Department of Hematology, People's Liberation Army The General Hospital of Western Theater Command, Chengdu, China
| | - Qinggui Liu
- Department of Cell Biology, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Yuwen Gan
- Department of Cell Biology, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Shoujia Xu
- Department of Cell Biology, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Haili Bao
- Department of General Surgery, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yiqiang Jin
- Shanghai Baixian Biotechnology Co., Ltd., Shanghai, China; Shanghai Angecon Biotechnology Co., Ltd., Shanghai, China
| | - Yi-Ping Hu
- Department of Cell Biology, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Junling Gao
- Department of Cell Biology, Second Military Medical University (Naval Medical University), Shanghai, China.
| | - Shaohua Song
- Department of General Surgery, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China; Transplantation Center, Changzheng Hospital, Naval Medical University, Shanghai, China.
| | - Min-Jun Wang
- Department of Cell Biology, Second Military Medical University (Naval Medical University), Shanghai, China.
| |
Collapse
|
2
|
Saiprayong K, Chupradit K, Sasithong P, Suwanpitak S, Muneekaew S, Thongsin N, Srisantitham J, Wattanapanitch M. Development of 2LTRZFP-expressing induced pluripotent stem cells as a potential anti-HIV-1 gene therapy against viral integration. J Leukoc Biol 2025; 117:qiaf018. [PMID: 39946247 DOI: 10.1093/jleuko/qiaf018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 11/29/2024] [Accepted: 02/12/2025] [Indexed: 04/26/2025] Open
Abstract
Highly active antiretroviral drug is the standard treatment for HIV-1 infection to suppress the viral load. However, this treatment does not completely eradicate the virus; it simply decreases the viral load to undetectable levels. The development of a novel therapy to cure the disease is essential. Previously, we developed an engineered zinc finger protein (ZFP) that specifically binds to the 2-LTR-circle junction (2LTRZFP), the target site for viral integrase, preventing HIV-1 integration in human CD34+ hematopoietic stem/progenitor cells (HSPCs) and macrophages. Although the transduction efficiency of 2LTRZFP was ∼50%, purifying and expanding the 2LTRZFP-expressing HSPCs proved difficult. In addition, the batch-to-batch variability in transduction efficiency could have a major impact on the therapeutic efficacy. In this study, we introduced the 2LTRZFP into human induced pluripotent stem cells (iPSCs) followed by clonal isolation and functional validation of the 2LTRZFP. Upon the HIV-1 challenge, the 2LTRZFP protein was found to inhibit the viral integration in iPSCs, iPSC-derived HSPCs, and macrophages. The engineered iPSC clone could be differentiated into functional macrophages, as evidenced by M1 and M2 polarization, and phagocytosis. Our finding revealed that the 2LTRZFP did not perturb the macrophage differentiation process. Therefore, the 2LTRZFP-expressing iPSCs could provide an unlimited supply of HIV-1-resistant HSPCs for transplantation, potentially leading to HIV-1-resistant blood cells. The knowledge obtained from this study will provide a cornerstone for HIV-1 gene therapy using HSPC transplantation as a sustainable HIV-1 treatment in the future.
Collapse
Affiliation(s)
- Kritayaporn Saiprayong
- Siriraj Center for Regenerative Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd, Bangkok 10700, Thailand
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd, Bangkok 10700, Thailand
| | - Koollawat Chupradit
- Siriraj Center for Regenerative Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd, Bangkok 10700, Thailand
| | - Pasut Sasithong
- Siriraj Center for Regenerative Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd, Bangkok 10700, Thailand
- Biomedical Sciences Graduate Program, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd, Bangkok 10700, Thailand
| | - Siriwal Suwanpitak
- Siriraj Center for Regenerative Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd, Bangkok 10700, Thailand
| | - Saitong Muneekaew
- Siriraj Center for Regenerative Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd, Bangkok 10700, Thailand
| | - Nontaphat Thongsin
- Siriraj Center for Regenerative Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd, Bangkok 10700, Thailand
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd, Bangkok 10700, Thailand
| | - Jakkrapatra Srisantitham
- Siriraj Center for Regenerative Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd, Bangkok 10700, Thailand
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd, Bangkok 10700, Thailand
| | - Methichit Wattanapanitch
- Siriraj Center for Regenerative Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd, Bangkok 10700, Thailand
| |
Collapse
|
3
|
Peng X, Lian Z, O'Brien V, Xiao J, Litchfield BA, Perrard XYD, Xu L, Ni J, Mukherjee A, Simmons T, Dong H, Mullick AE, Crooke R, Pownall HJ, Simon SI, Ballantyne CM, Wu H. Foamy monocytes and atherogenesis in mice with combined hyperlipidemia and effects of antisense knockdown of apoCIII. J Lipid Res 2025; 66:100763. [PMID: 39988193 PMCID: PMC11981816 DOI: 10.1016/j.jlr.2025.100763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 02/14/2025] [Accepted: 02/18/2025] [Indexed: 02/25/2025] Open
Abstract
Hypertriglyceridemia (HTG), particularly in combined hyperlipidemia, increases risk for atherosclerotic cardiovascular disease, but the underlying mechanisms remain incompletely understood. We sought to determine contributions of circulating monocytes to atherosclerosis associated with HTG in combined hyperlipidemia, created by transgenic expression of human apoCIII in Ldlr-/- mice (Ldlr-/-ApoCIIItg) fed Western high-fat diet (WD). Tissue culture with THP-1 and primary human monocytes was used to examine effects of triglyceride (TG)-rich lipoproteins on monocytes. Ldlr-/-ApoCIIItg mice were also treated with apoCIII antisense oligonucleotide (ASO) and examined for foamy monocytes and atherosclerosis. Compared to Ldlr-/- mice, Ldlr-/-ApoCIIItg mice fed WD had early and persistent increases in lipid accumulation within monocytes and enhanced atherosclerosis. Ldlr-/-ApoCIIItg mice versus Ldlr-/- mice had higher levels of CD11c, CD36, and cytokines in foamy monocytes, with increases in foamy monocyte adhesion to vascular cell adhesion molecule-1 and oxidized LDL uptake. Monocytes took up TG-rich lipoprotein in vivo and in vitro and changed phenotypes. Foamy monocytes infiltrated into atherosclerotic lesions, and specific and sustained depletion of CD11c+ (foamy) monocytes profoundly reduced atherosclerosis in Ldlr-/-ApoCIIItg mice on WD. Treatment with apoCIII ASO lowered plasma TG and cholesterol levels, improved foamy monocyte phenotypes, and reduced atherosclerosis in Ldlr-/-ApoCIIItg mice. In conclusion, HTG in combined hyperlipidemia accelerates atherosclerosis, in part, by increasing foamy monocyte formation and infiltration into atherosclerotic plaques. Treatment with apoCIII ASO is a potential new therapy for improving monocyte phenotypes and reducing atherosclerosis in combined hyperlipidemia.
Collapse
Affiliation(s)
- Xueying Peng
- Department of Clinical Pharmacy, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, Zhejiang, PR China; Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Zeqin Lian
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Veronica O'Brien
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Jing Xiao
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | | | | | - Lu Xu
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Jing Ni
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Aparna Mukherjee
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Timothy Simmons
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Henry Dong
- Department of Pediatrics, Children's Hospital of Pittsburgh UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | | | - Henry J Pownall
- Center for Bioenergetics, Houston Methodist Research Institute, Houston, TX, USA
| | - Scott I Simon
- Department of Biomedical Engineering, University of California, Davis, CA, USA
| | - Christie M Ballantyne
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA; Center for Cardiometabolic Disease Prevention, Baylor College of Medicine, Houston, TX, USA
| | - Huaizhu Wu
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
4
|
Barakat H, Aljutaily T. Role of γ-Aminobutyric Acid (GABA) as an Inhibitory Neurotransmitter in Diabetes Management: Mechanisms and Therapeutic Implications. Biomolecules 2025; 15:399. [PMID: 40149935 PMCID: PMC11940341 DOI: 10.3390/biom15030399] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/03/2025] [Accepted: 03/07/2025] [Indexed: 03/29/2025] Open
Abstract
GABA (γ-Aminobutyric Acid), a well-established inhibitory neurotransmitter in the central nervous system, has garnered considerable interest for its potential role in diabetes management, particularly due to its presence in pancreatic islets. This review aims to explore the therapeutic role of GABA in diabetes management and its potential mechanisms for antidiabetic effects. Relevant studies were searched across databases such as PubMed and ScienceDirect, applying strict eligibility criteria focused on GABA administration methods and diabetic models. The collective results showed that the administration of GABA in diabetic models resulted in remarkable enhancements in glucose and insulin homeostasis, favorable modifications in lipid profiles, and amelioration of dysfunctions across neural, hepatic, renal, and cardiac systems. The findings from the literature demonstrated that GABAergic signaling within pancreatic tissues can significantly contribute to the stimulation of β cell proliferation through the facilitation of a sustained trans-differentiation process, wherein glucagon-secreting α cells are converted into insulin-secreting β-like cells. In addition, activated GABAergic signaling can trigger the initiation of the PI3K/AKT signaling pathway within pancreatic tissues, leading to improved insulin signaling and maintained glucose homeostasis. GABAergic signaling can further function within hepatic tissues, promoting inhibitory effects on the expression of genes related to gluconeogenesis and lipogenesis. Moreover, GABA may enhance gut microbiota diversity by attenuating gut inflammation, attributable to its anti-inflammatory and immunomodulatory properties. Furthermore, the neuroprotective effects of GABA play a significant role in ameliorating neural disorders associated with diabetes by facilitating a substantial reduction in neuronal apoptosis. In conclusion, GABA emerges as a promising candidate for an antidiabetic agent; however, further research is highly encouraged to develop a rigorously designed framework that comprehensively identifies and optimizes the appropriate dosages and intervention methods for effectively managing and combating diabetes.
Collapse
Affiliation(s)
- Hassan Barakat
- Department of Food Science and Human Nutrition, College of Agriculture and Food, Qassim University, Buraydah 51452, Saudi Arabia;
| | | |
Collapse
|
5
|
Ramezankhani A, Tohidi M, Hadaegh F. Association between the systemic immune-inflammation index and metabolic syndrome and its components: results from the multi-ethnic study of atherosclerosis (MESA). Cardiovasc Diabetol 2025; 24:78. [PMID: 39955525 PMCID: PMC11830208 DOI: 10.1186/s12933-025-02629-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Accepted: 02/04/2025] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND The Systemic Immune-Inflammation Index (SII) is a novel biomarker of systemic inflammation. We explored the association between the SII and metabolic syndrome (MetS) and its components in middle-aged and older adults. METHODS We included 2755 participants (1305 men) aged 45-84 years from the Multi-Ethnic Study of Atherosclerosis (MESA) cohort from examination 5 (2010-2012). Logistic regression was employed to assess the relationship between the SII and MetS, as well as its components. RESULTS A total of 1082 participants (463 men) were diagnosed with MetS. On a continuous scale, the SII was positively associated with MetS (odds ratio (OR): 1.23, 95% confidence interval (CI): 1.05-1.46) and its components including hyperglycemia (1.23: 1.05-1.44) and elevated blood pressure (BP) (1.47: 1.14-1.89). When analyzed on a quartile scale, participants in the quartile 4 of SII had 32% and 63% higher prevalence of hyperglycemia and elevated BP, respectively, compared to those in the quartile 1 (P for trend: 0.021 and < 0.001, respectively). Additionally, we identified 40% higher prevalence of low HDL-C in quartile 2 of the SII compared to quartile 1 (1.40; 1.07-1.83) (P trend = 0.454). In subgroup analysis, general obesity status modified the relationship between SII and abdominal obesity, showing a positive association in obese individuals (1.72: 1.00-2.95) and a negative association (0.80: 0.66-0.97) in non-obese individuals (P for interaction = 0.009). CONCLUSIONS Higher SII scores were associated with an increased likelihood of MetS, hyperglycemia, and high BP among middle-aged and older adults. Longitudinal studies are needed to determine the causal relationships between SII and the development of MetS, as well as to assess the potential role of SII as a screening tool in clinical practice.
Collapse
Affiliation(s)
- Azra Ramezankhani
- Prevention of Metabolic Disorders Research Center, Research Institute for Metabolic and Obesity Disorders, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Floor 3th, Number 24, Yemen Street, Shahid Chamran Highway, P.O. Box: 19395-4763, Tehran, Iran
| | - Maryam Tohidi
- Prevention of Metabolic Disorders Research Center, Research Institute for Metabolic and Obesity Disorders, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Floor 3th, Number 24, Yemen Street, Shahid Chamran Highway, P.O. Box: 19395-4763, Tehran, Iran
| | - Farzad Hadaegh
- Prevention of Metabolic Disorders Research Center, Research Institute for Metabolic and Obesity Disorders, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Floor 3th, Number 24, Yemen Street, Shahid Chamran Highway, P.O. Box: 19395-4763, Tehran, Iran.
| |
Collapse
|
6
|
Ding P, Zhang D, Ling H, Tao T, Gao Y, Wang Y, Zhang H, Wu L, Hang C, Li W. Insulin Resistance Predicts Prognosis in Patients With Subarachnoid Hemorrhage. J Evid Based Med 2024; 17:771-781. [PMID: 39676383 DOI: 10.1111/jebm.12660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 11/27/2024] [Accepted: 11/27/2024] [Indexed: 12/17/2024]
Abstract
OBJECTIVE The objective of this study was to determine whether insulin resistance (IR) could be used as a predictor of poor prognosis at 3 months after subarachnoid hemorrhage (SAH). METHODS The study included patients aged 18 years or older with a confirmed diagnosis of SAH due to ruptured aneurysm from January 2021 to March 2024. Patients with confirmed diabetes mellitus and taking glucose-lowering drugs, or taking lipid-lowering drugs, or SAH not due to ruptured aneurysm, or comorbid systemic diseases were excluded. Patients were classified into good prognosis (modified Rankin scale [MRS] 0-2) and poor prognosis (MRS 3-6). Receiver operating characteristic curve (ROC), least absolute shrinkage and selection operator (LASSO) analysis, and multivariate logistic regression analysis were used to determine the potential of triglyceride-glucose (TyG) index and the triglyceride to high-density lipoprotein cholesterol (TG/HDL) ratio as predictors of poor prognosis. Finally, a prognostic prediction model based on IR was constructed. RESULTS A total of 358 patients were included in this study. Poor prognosis patients had higher age, BMI, hypertension percentage, glucose, triglycerides, TyG index and TG/HDL ratio, and lower HDL. ROC, LASSO, and multivariate logistic regression analysis revealed that age, glucose, TyG index, and TG/HDL ratio had significant potential to predict the prognosis of SAH patients. The prognostic prediction model constructed by combining age, glucose, TyG index, and TG/HDL ratio had high discriminatory power (area under the curve [AUC] = 0.80), satisfactory calibration curves, and good clinical utility. CONCLUSION IR is strongly associated with the prognosis of SAH patients, and the combination of age, glucose, TyG index, and TG/HDL ratio can provide a new direction for future treatment.
Collapse
Affiliation(s)
- Pengfei Ding
- Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Neurosurgical Institute, Nanjing University, Nanjing, China
| | - Dingding Zhang
- Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Neurosurgical Institute, Nanjing University, Nanjing, China
| | - Haiping Ling
- Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Neurosurgical Institute, Nanjing University, Nanjing, China
| | - Tao Tao
- Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Neurosurgical Institute, Nanjing University, Nanjing, China
| | - Yongyue Gao
- Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Neurosurgical Institute, Nanjing University, Nanjing, China
| | - Yunfeng Wang
- Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Neurosurgical Institute, Nanjing University, Nanjing, China
| | - Huasheng Zhang
- Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Neurosurgical Institute, Nanjing University, Nanjing, China
| | - Lingyun Wu
- Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Neurosurgical Institute, Nanjing University, Nanjing, China
| | - Chunhua Hang
- Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Neurosurgical Institute, Nanjing University, Nanjing, China
| | - Wei Li
- Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Neurosurgical Institute, Nanjing University, Nanjing, China
| |
Collapse
|
7
|
Qi L, Groeger M, Sharma A, Goswami I, Chen E, Zhong F, Ram A, Healy K, Hsiao EC, Willenbring H, Stahl A. Adipocyte inflammation is the primary driver of hepatic insulin resistance in a human iPSC-based microphysiological system. Nat Commun 2024; 15:7991. [PMID: 39266553 PMCID: PMC11393072 DOI: 10.1038/s41467-024-52258-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 08/28/2024] [Indexed: 09/14/2024] Open
Abstract
Interactions between adipose tissue, liver and immune system are at the center of metabolic dysfunction-associated steatotic liver disease and type 2 diabetes. To address the need for an accurate in vitro model, we establish an interconnected microphysiological system (MPS) containing white adipocytes, hepatocytes and proinflammatory macrophages derived from isogenic human induced pluripotent stem cells. Using this MPS, we find that increasing the adipocyte-to-hepatocyte ratio moderately affects hepatocyte function, whereas macrophage-induced adipocyte inflammation causes lipid accumulation in hepatocytes and MPS-wide insulin resistance, corresponding to initiation of metabolic dysfunction-associated steatotic liver disease. We also use our MPS to identify and characterize pharmacological intervention strategies for hepatic steatosis and systemic insulin resistance and find that the glucagon-like peptide-1 receptor agonist semaglutide improves hepatocyte function by acting specifically on adipocytes. These results establish our MPS modeling the adipose tissue-liver axis as an alternative to animal models for mechanistic studies or drug discovery in metabolic diseases.
Collapse
Affiliation(s)
- Lin Qi
- Department of Nutritional Science and Toxicology, College of Natural Resources, University of California Berkeley, Berkeley, CA, 94720, USA
| | - Marko Groeger
- Division of Transplant Surgery, Department of Surgery, University of California San Francisco, San Francisco, CA, 94143, USA
- Eli and Edythe Broad Center for Regeneration Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Aditi Sharma
- Eli and Edythe Broad Center for Regeneration Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Ishan Goswami
- Department of Bioengineering, College of Engineering, University of California Berkeley, Berkeley, CA, 94720, USA
| | - Erzhen Chen
- Department of Nutritional Science and Toxicology, College of Natural Resources, University of California Berkeley, Berkeley, CA, 94720, USA
| | - Fenmiao Zhong
- Department of Nutritional Science and Toxicology, College of Natural Resources, University of California Berkeley, Berkeley, CA, 94720, USA
| | - Apsara Ram
- Eli and Edythe Broad Center for Regeneration Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Kevin Healy
- Department of Bioengineering, College of Engineering, University of California Berkeley, Berkeley, CA, 94720, USA
- Department of Materials Science and Engineering, College of Engineering, University of California Berkeley, Berkeley, CA, 94720, USA
| | - Edward C Hsiao
- Eli and Edythe Broad Center for Regeneration Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Holger Willenbring
- Division of Transplant Surgery, Department of Surgery, University of California San Francisco, San Francisco, CA, 94143, USA.
- Eli and Edythe Broad Center for Regeneration Medicine, University of California San Francisco, San Francisco, CA, 94143, USA.
- Liver Center, University of California San Francisco, San Francisco, CA, 94143, USA.
| | - Andreas Stahl
- Department of Nutritional Science and Toxicology, College of Natural Resources, University of California Berkeley, Berkeley, CA, 94720, USA.
| |
Collapse
|
8
|
Huang D, Bai F, Hu T, Li J, Wang G, Wu C. Salvia miltiorrhiza bge. f. alba ameliorates type 2 diabetes mellitus-associated non-alcoholic fatty liver disease via the STING pathway. Am J Transl Res 2024; 16:3678-3689. [PMID: 39262750 PMCID: PMC11384384 DOI: 10.62347/xuno9933] [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: 04/07/2024] [Accepted: 06/27/2024] [Indexed: 09/13/2024]
Abstract
OBJECTIVE To elucidate the functional role and underlying mechanism of Salvia miltiorrhiza bge. f. alba (SMBFA) in patients with type 2 diabetes mellitus (T2DM) accompanied by non-alcoholic fatty liver disease (NAFLD). METHODS A retrospective analysis was conducted on 90 patients with T2DM-NAFLD who met the inclusion criteria. The control group was comprised of 45 patients treated with Fenofibrate, while the observation group consisted of 45 patients who received SMBFA in addition to the control treatment. An in vivo mouse model of T2DM-NAFLD was established using a high-fat diet combined with streptozotocin. Serum levels of fasting plasma glucose (FPG), 2-hour postprandial glucose (2h PG), hemoglobin A1c (HbA1c), homeostasis model assessment of insulin resistance (HOMA-IR), total cholesterol (TC), and triglyceride (TG) were measured in both patients and mice using an automated biochemical analyzer. Liver indices and function were also evaluated. ELISA assays were performed to quantify inflammatory cytokine levels. Western blotting was utilized to assess the protein levels related to the stimulator of interferon genes (STING)-interferon regulatory factor 3 (IRF3) pathway. RESULTS After treatment, significant reductions in blood glucose indices, HOMA-IR, lipid metabolism markers, liver function indices, and inflammatory cytokines were observed in both groups of T2DM-NAFLD patients. Notably, the decreases were more pronounced in the observation group compared to the control group. Similarly, in T2DM-NAFLD mouse models, the levels of these parameters were significantly lower in the observation group than in the normal control (NC) group. Additionally, SMBFA suppressed the elevated levels of STING, p-IRF3, and p-TANK-binding kinase 1 in the T2DM-NAFLD mice. CONCLUSION SMBFA exhibits the potential to regulate glucose and lipid metabolism, inhibit insulin resistance, and protect liver function by modulating the STING signaling pathway.
Collapse
Affiliation(s)
- Donghui Huang
- Department of Endocrinology, The Second Affiliated Hospital of Shandong First Medical University Tai'an 271000, Shandong, China
| | - Fuyan Bai
- Department of Endocrinology, The Second Affiliated Hospital of Shandong First Medical University Tai'an 271000, Shandong, China
| | - Tingting Hu
- Department of Endocrinology, The Second Affiliated Hospital of Shandong First Medical University Tai'an 271000, Shandong, China
| | - Jing Li
- Department of Pediatrics, The Secondary TCM Hospital of Tai'an City Tai'an 271000, Shandong, China
| | - Guoning Wang
- Department of Endocrinology, The Second Affiliated Hospital of Shandong First Medical University Tai'an 271000, Shandong, China
| | - Chengsheng Wu
- Department of Endocrinology, The Second Affiliated Hospital of Shandong First Medical University Tai'an 271000, Shandong, China
| |
Collapse
|
9
|
Guillot A, Tacke F. Liver macrophages revisited: The expanding universe of versatile responses in a spatiotemporal context. Hepatol Commun 2024; 8:e0491. [PMID: 38967563 PMCID: PMC11227356 DOI: 10.1097/hc9.0000000000000491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 05/23/2024] [Indexed: 07/06/2024] Open
Abstract
The liver is a vital organ that continuously adapts to a wide and dynamic diversity of self-antigens and xenobiotics. This involves the active contribution of immune cells, particularly by the liver-resident macrophages, the Kupffer cells (KCs), which exert a variety of central functions in liver homeostasis and disease. As such, KCs interact with their microenvironment to shape the hepatic cellular landscape, control gut-derived signal integration, and modulate metabolism. On injury, the rapid recruitment of bone marrow monocyte-derived macrophages alters this status quo and, when unrestrained, drastically compromises liver homeostasis, immune surveillance, and tissue organization. Several factors determine the functional roles of liver macrophages in these processes, such as their ontogeny, activation/polarization profile and, importantly, spatial distribution within the liver. Loss of tolerance and adaptability of the hepatic immune environment may result in persistent inflammation, hepatic fibrosis, cirrhosis, and a tumorigenic niche promoting liver cancer. In this review, we aim at providing the most recent breakthroughs in our understanding of liver macrophage biology, particularly their diversity and adaptability in the hepatic spatiotemporal context, as well as on potential therapeutic interventions that may hold the key to tackling remaining clinical challenges of varying etiologies in hepatology.
Collapse
|
10
|
Bovi Dos Santos G, de Lima-Vasconcellos TH, Móvio MI, Birbrair A, Del Debbio CB, Kihara AH. New Perspectives in Stem Cell Transplantation and Associated Therapies to Treat Retinal Diseases: From Gene Editing to 3D Bioprinting. Stem Cell Rev Rep 2024; 20:722-737. [PMID: 38319527 DOI: 10.1007/s12015-024-10689-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/29/2024] [Indexed: 02/07/2024]
Abstract
Inherited and non-inherited retinopathies can affect distinct cell types, leading to progressive cell death and visual loss. In the last years, new approaches have indicated exciting opportunities to treat retinopathies. Cell therapy in retinitis pigmentosa, age-related macular disease, and glaucoma have yielded encouraging results in rodents and humans. The first two diseases mainly impact the photoreceptors and the retinal pigmented epithelium, while glaucoma primarily affects the ganglion cell layer. Induced pluripotent stem cells and multipotent stem cells can be differentiated in vitro to obtain specific cell types for use in transplant as well as to assess the impact of candidate molecules aimed at treating retinal degeneration. Moreover, stem cell therapy is presented in combination with newly developed methods, such as gene editing, Müller cells dedifferentiation, sheet & drug delivery, virus-like particles, optogenetics, and 3D bioprinting. This review describes the recent advances in this field, by presenting an updated panel based on cell transplants and related therapies to treat retinopathies.
Collapse
Affiliation(s)
- Gabrieli Bovi Dos Santos
- Laboratório de Neurogenética, Universidade Federal do ABC, São Bernardo do Campo, Santo André, SP, Brazil
| | | | - Marília Inês Móvio
- Laboratório de Neurogenética, Universidade Federal do ABC, São Bernardo do Campo, Santo André, SP, Brazil
| | - Alexander Birbrair
- Department of Dermatology, Medical Sciences Center, University of Wisconsin-Madison, Rm 4385, 1300 University Avenue, Madison, WI, 53706, USA
| | - Carolina Beltrame Del Debbio
- Departamento de Biologia Celular e do Desenvolvimento, Instituto de Ciências Biomédicas, Universidade de São Paulo USP, São Paulo, SP, Brazil
| | - Alexandre Hiroaki Kihara
- Laboratório de Neurogenética, Universidade Federal do ABC, São Bernardo do Campo, Santo André, SP, Brazil.
| |
Collapse
|
11
|
Wang D, Ren J, Li J, Li X, Ying J, Jiang T, Wang Z, Pan Z, Guo Q, Li C, Zhang G. Conditioned Media from Deer Antler Stem Cells Effectively Alleviate Type 1 Diabetes Mellitus Possibly via Inhibiting the NF-κB Signaling Pathway. FRONT BIOSCI-LANDMRK 2024; 29:96. [PMID: 38538257 DOI: 10.31083/j.fbl2903096] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 12/01/2023] [Accepted: 12/20/2023] [Indexed: 04/05/2024]
Abstract
BACKGROUND Type 1 diabetes mellitus (T1D) represents a severe threat to human health. Persistent hyperglycemia and dyslipidemia can lead to damaged liver function, while effective interventions for these complications are currently lacking. Deer antler stem cells (AnSCs), a novel type of adult stem cells, significantly reduced liver injury, which was speculated to be achieved through the paracrine pathway. METHODS In this study, AnSC-conditioned medium (AnSC-CM) was used to treat C57BL/6 mice with T1D symptoms induced by streptozotocin (STZ). The therapeutic effects of AnSC-CM on T1D were evaluated, and the underlying mechanism was investigated. RESULTS It was shown that AnSC-CM alleviated the T1D symptom: decreased body weight, increased blood glucose levels and islet lesions, and reduced insulin secretion. Moreover, AnSC-CM treatment improved liver function and mitigated liver injury in T1D mice. Impressively, the therapeutic effects of AnSC-CM on T1D were better than those of bone marrow mesenchymal stem cell-CM (BMSC-CM). The mechanistic study revealed that AnSC-CM significantly downregulated the NF-κB signaling pathway in both pancreatic and liver tissues. CONCLUSIONS Therapeutic effects of AnSC-CM on STZ-induced T1D and liver injury may be achieved through targeting the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Dongxu Wang
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, 130600 Changchun, Jilin, China
| | - Jing Ren
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, 130600 Changchun, Jilin, China
- College of Chinese Medicinal Materials, Jilin Agricultural University, 130118 Changchun, Jilin, China
| | - Jiping Li
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, 130600 Changchun, Jilin, China
| | - Xiuying Li
- Scientific Research center, China-Japan Union Hospital, Jilin University, 130033 Changchun, Jilin, China
| | - Jinchi Ying
- College of Chinese Medicinal Materials, Jilin Agricultural University, 130118 Changchun, Jilin, China
| | - Tiantian Jiang
- College of Chinese Medicinal Materials, Jilin Agricultural University, 130118 Changchun, Jilin, China
| | - Zhen Wang
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, 130600 Changchun, Jilin, China
| | - Zheng Pan
- Scientific Research center, China-Japan Union Hospital, Jilin University, 130033 Changchun, Jilin, China
| | - Qianqian Guo
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, 130600 Changchun, Jilin, China
| | - Chunyi Li
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, 130600 Changchun, Jilin, China
- College of Chinese Medicinal Materials, Jilin Agricultural University, 130118 Changchun, Jilin, China
| | - Guokun Zhang
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, 130600 Changchun, Jilin, China
| |
Collapse
|