1
|
Krapoth TC, Henle GS, Avdyli M, Bektić B, Schwarzkopf KM, Bešić L, Zeuzem S, Welsch C, Kraus N, Ortiz C. Wanted: Dead or Alive Cells with Propidium Iodide Staining in Liver Tissue. Int J Mol Sci 2024; 25:13521. [PMID: 39769284 PMCID: PMC11679561 DOI: 10.3390/ijms252413521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 12/11/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
This study demonstrates the effectiveness of propidium iodide as a reliable marker for detecting dead or dying cells in frozen liver tissue sections. By comparing propidium iodide staining with the widely used Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay, both methods showed consistent results in disease models such as alcohol-induced fibrosis and Western diet-induced fatty liver. Additionally, propidium iodide was successfully co-stained with other fluorescent markers, like phalloidin (for actin filaments) and antibodies targeting collagen, enabling detailed spatial analysis of dying cells within tissue. This multiplex approach allows for a deeper understanding of tissue organization and cell death localization, particularly in complex conditions like liver fibrosis. Moreover, our results suggest that propidium iodide staining can be applied beyond current models, offering a more accessible and cost-effective alternative to traditional methods, like TUNEL. Furthermore, its integration with other markers enables simultaneous analysis of immune responses and tissue damage, making it a powerful tool for future studies on liver disease and other inflammatory conditions. This technique has the potential to advance research into disease mechanisms and improve the evaluation of novel therapeutic strategies targeting tissue regeneration and inflammation control.
Collapse
Affiliation(s)
- Tim Christopher Krapoth
- Goethe University, Frankfurt University Hospital, Medical Clinic 1, 60596 Frankfurt, Germany; (T.C.K.); (G.S.H.); (M.A.); (B.B.); (K.M.S.); (S.Z.); (C.W.); (N.K.)
| | - Gina Sophie Henle
- Goethe University, Frankfurt University Hospital, Medical Clinic 1, 60596 Frankfurt, Germany; (T.C.K.); (G.S.H.); (M.A.); (B.B.); (K.M.S.); (S.Z.); (C.W.); (N.K.)
| | - Mihrije Avdyli
- Goethe University, Frankfurt University Hospital, Medical Clinic 1, 60596 Frankfurt, Germany; (T.C.K.); (G.S.H.); (M.A.); (B.B.); (K.M.S.); (S.Z.); (C.W.); (N.K.)
| | - Berina Bektić
- Goethe University, Frankfurt University Hospital, Medical Clinic 1, 60596 Frankfurt, Germany; (T.C.K.); (G.S.H.); (M.A.); (B.B.); (K.M.S.); (S.Z.); (C.W.); (N.K.)
- Department of Genetics and Bioengineering, International Burch University, Francuske Revolucije bb, 71210 Sarajevo, Bosnia and Herzegovina;
| | - Katharina Maria Schwarzkopf
- Goethe University, Frankfurt University Hospital, Medical Clinic 1, 60596 Frankfurt, Germany; (T.C.K.); (G.S.H.); (M.A.); (B.B.); (K.M.S.); (S.Z.); (C.W.); (N.K.)
| | - Larisa Bešić
- Department of Genetics and Bioengineering, International Burch University, Francuske Revolucije bb, 71210 Sarajevo, Bosnia and Herzegovina;
| | - Stefan Zeuzem
- Goethe University, Frankfurt University Hospital, Medical Clinic 1, 60596 Frankfurt, Germany; (T.C.K.); (G.S.H.); (M.A.); (B.B.); (K.M.S.); (S.Z.); (C.W.); (N.K.)
| | - Christoph Welsch
- Goethe University, Frankfurt University Hospital, Medical Clinic 1, 60596 Frankfurt, Germany; (T.C.K.); (G.S.H.); (M.A.); (B.B.); (K.M.S.); (S.Z.); (C.W.); (N.K.)
| | - Nico Kraus
- Goethe University, Frankfurt University Hospital, Medical Clinic 1, 60596 Frankfurt, Germany; (T.C.K.); (G.S.H.); (M.A.); (B.B.); (K.M.S.); (S.Z.); (C.W.); (N.K.)
| | - Cristina Ortiz
- Goethe University, Frankfurt University Hospital, Medical Clinic 1, 60596 Frankfurt, Germany; (T.C.K.); (G.S.H.); (M.A.); (B.B.); (K.M.S.); (S.Z.); (C.W.); (N.K.)
| |
Collapse
|
2
|
Aggarwal H, Gautam J, Gupta SK, Das B, Kumar Y, Jagavelu K, Dikshit M. Improved metabolic stability in iNOS knockout mice with Lactobacillus supplementation. Nutr Res 2024; 132:95-111. [PMID: 39532058 DOI: 10.1016/j.nutres.2024.09.018] [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: 02/21/2024] [Revised: 09/25/2024] [Accepted: 09/25/2024] [Indexed: 11/16/2024]
Abstract
Oxidative and nitrosative stress play pivotal roles in normal physiological processes and the pathogenesis of metabolic disorders. Previous studies from our lab demonstrated insulin resistance (IR), and dyslipidemia in iNOS-/- mice, emphasizing the importance of maintaining optimal redox balance. These mice exhibited altered gut microbiota with decreased Lactobacillus. Therefore, we hypothesized that Lactobacillus supplementation could mitigate metabolic disturbances in iNOS-/- mice. To test this hypothesis, iNOS-/- mice and wild-type (WT) mice were divided into four groups: iNOS-/- with or without Lactobacillus supplementation, WT with or without Lactobacillus supplementation and glucose tolerance, insulin resistance, gluconeogenesis, lipids, gene expression related to glucose and lipid metabolism (qPCR), fecal gut microbiota (16S rRNA sequencing), and serum and caecum metabolomics (LC-MS) were monitored. IR and dyslipidemic iNOS-/- mice exhibited reduced microbial diversity, diminished presence of Lactobacillus, and altered serum metabolites, indicating metabolic dysregulation. Lactobacillus supplementation in iNOS-/- mice effectively reversed glucose intolerance, IR, dyslipidemia, and associated metabolic irregularities compared to WT. These improvements correlated with changes in gene expression related to fatty acid synthesis in liver and adipose tissue, lipid oxidation in liver, and lipid efflux in intestinal tissue as compared to untreated iNOS-/- mice. Despite the positive effects on metabolic markers, Lactobacillus supplementation did not reduce body weight or rectify disrupted energy balance, as evidenced by reduced VCO2 production, heat generation, and metabolic rates in iNOS-/- mice. The results suggest that Lactobacillus supplementation ameliorates metabolic disturbances but did not fully restore disrupted energy balance, highlighting complex interactions between the gut microbiome and metabolism.
Collapse
Affiliation(s)
- Hobby Aggarwal
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India; Non-communicable Diseases Division, Translational Health Science and Technology Institute, Faridabad, Haryana 121001, India
| | - Jyoti Gautam
- Non-communicable Diseases Division, Translational Health Science and Technology Institute, Faridabad, Haryana 121001, India; Department of Pharmacology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Sonu Kumar Gupta
- Non-communicable Diseases Division, Translational Health Science and Technology Institute, Faridabad, Haryana 121001, India
| | - Bhabatosh Das
- Molecular Genetics Laboratory, Infection and Immunology Division, Translational Health Science and Technology Institute, Faridabad, Haryana 121001, India
| | - Yashwant Kumar
- Non-communicable Diseases Division, Translational Health Science and Technology Institute, Faridabad, Haryana 121001, India
| | - Kumaravelu Jagavelu
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India
| | - Madhu Dikshit
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India; Non-communicable Diseases Division, Translational Health Science and Technology Institute, Faridabad, Haryana 121001, India.
| |
Collapse
|
3
|
Hu Q, Luo Y, He H, Chen H, Liao D. Comprehensive analysis of shared risk genes and immunity-metabolisms between non-alcoholic fatty liver disease and atherosclerosis via bulk and single-cell transcriptome analyses. Heliyon 2024; 10:e35453. [PMID: 39165965 PMCID: PMC11334902 DOI: 10.1016/j.heliyon.2024.e35453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 07/26/2024] [Accepted: 07/29/2024] [Indexed: 08/22/2024] Open
Abstract
Objective and design: Considering the clinical link between non-alcoholic fatty liver disease (NAFLD) and atherosclerosis (AS), we performed bioinformatics analysis to uncover their pathogenic interrelationship. Methods and results Data from the U.S. National Health and Nutritional Examination Survey (NHANES) 1999-2018 were included. Among 4851 participants in NHANES, NAFLD was significantly associated with atherosclerotic cardiovascular disease risk (ASCVD risk) (OR = 2.32, 95%CI: 2.04-2.65, P < 0.0001). We conducted WGCNA analysis for NAFLD (GSE130970) and AS (GSE28829) and identified three modules positively related to NAFLD severity and two modules accelerating atherosclerosis plaque progression. 198 key-modules genes were obtained via overlapping these modules. Next, we mined the disease-controlled differentially expressed genes (DEGs) from NAFLD (GSE89632) and AS (GSE100927), respectively. The final common risk genes (ACP5, TP53I3, RPS6KA1, TYMS, TREM2, CA12, and IFI27) were defined by intersecting the upregulated DEGs with 198 genes and validated in new datasets (GSE48452 and GSE43292). Importantly, they showed good diagnostic ability for NAFLD and AS. Immune infiltration analysis showed both illnesses have dysregulated immunity. Analysis of single-cell sequencing datasets NAFLD (GSE179886) and AS (GSE159677) uncovered different abnormal expressions of seven common genes in different immune cells while highlighting metabolic disturbances including upregulation of fatty acid biosynthesis, downregulation of fatty acid degradation and elongation. Conclusion We found 7 shared hub genes with good diagnostic ability and depicted the landscapes of immune and metabolism involved in NAFLD and AS. Our results provided a comprehensive association between them and may contribute to developing potential intervention strategies for targeting both disorders based on these risk factors.
Collapse
Affiliation(s)
- Qian Hu
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
- Key Laboratory of Medical Genetics of Hunan Province, Central South University, Changsha, Hunan, China
| | - Yunfang Luo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, Hunan, China
| | - Hao He
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Hua Chen
- Department of Neurosurgery, the First people's Hospital of Changde City, Changde, Hunan, China
| | - Di Liao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, Hunan, China
| |
Collapse
|
4
|
Chen D, Wang Y, Yang J, Ou W, Lin G, Zeng Z, Lu X, Chen Z, Zou L, Tian Y, Wu A, Keating SE, Yang Q, Lin C, Liang Y. Shenling Baizhu San ameliorates non-alcoholic fatty liver disease in mice by modulating gut microbiota and metabolites. Front Pharmacol 2024; 15:1343755. [PMID: 38720776 PMCID: PMC11076757 DOI: 10.3389/fphar.2024.1343755] [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: 11/24/2023] [Accepted: 03/25/2024] [Indexed: 05/12/2024] Open
Abstract
Purpose: The prevalence of non-alcoholic fatty liver disease (NAFLD) and its related mortality is increasing at an unprecedented rate. Traditional Chinese medicine (TCM) has been shown to offer potential for early prevention and treatment of NAFLD. The new mechanism of "Shenling Baizhu San" (SLBZS) is examined in this study for the prevention and treatment of NAFLD at the preclinical level. Methods: Male C57BL/6J mice were randomly divided into three groups: normal diet (ND), western diet + CCl4 injection (WDC), and SLBZS intervention (WDC + SLBZS). Body weights, energy intake, liver enzymes, pro-inflammatory factors, and steatosis were recorded in detail. Meanwhile, TPH1, 5-HT, HTR2A, and HTR2B were tested using qRT-PCR or ELISA. Dynamic changes in the gut microbiota and metabolites were further detected through the 16S rRNA gene and untargeted metabolomics. Results: SLBZS intervention for 6 weeks could reduce the serum and liver lipid profiles, glucose, and pro-inflammatory factors while improving insulin resistance and liver function indexes in the mice, thus alleviating NAFLD in mice. More importantly, significant changes were found in the intestinal TPH-1, 5-HT, liver 5-HT, and related receptors HTR2A and HTR2B. The 16S rRNA gene analysis suggested that SLBZS was able to modulate the disturbance of gut microbiota, remarkably increasing the relative abundance of probiotics (Bifidobacterium and Parvibacter) and inhibiting the growth of pro-inflammatory bacteria (Erysipelatoclostridium and Lachnoclostridium) in mice with NAFLD. Combined with metabolomics in positive- and negative-ion-mode analyses, approximately 50 common differential metabolites were selected via non-targeted metabolomics detection, which indicated that the targeting effect of SLBZS included lipid metabolites, bile acids (BAs), amino acids (AAs), and tryptophan metabolites. In particular, the lipid metabolites 15-OxEDE, vitamin D3, desoxycortone, and oleoyl ethanol amide were restored by SLBZS. Conclusion: Integrating the above results of multiple omics suggests that SLBZS ameliorates NAFLD via specific gut microbiota, gut-derived 5-HT, and related metabolites to decrease fat accumulation in the liver and inflammatory responses.
Collapse
Affiliation(s)
- Dongliang Chen
- School of Nursing, Jinan University, Guangzhou, Guangdong Province, China
| | - Yuanfei Wang
- School of Nursing, Jinan University, Guangzhou, Guangdong Province, China
| | - Jianmei Yang
- School of Nursing, Jinan University, Guangzhou, Guangdong Province, China
| | - Wanyi Ou
- School of Nursing, Jinan University, Guangzhou, Guangdong Province, China
| | - Guiru Lin
- School of Nursing, Jinan University, Guangzhou, Guangdong Province, China
| | - Ze Zeng
- School of Nursing, Jinan University, Guangzhou, Guangdong Province, China
| | - Xiaomin Lu
- School of Nursing, Jinan University, Guangzhou, Guangdong Province, China
| | - Zumin Chen
- School of Nursing, Jinan University, Guangzhou, Guangdong Province, China
| | - Lili Zou
- School of Medicine, Jinan University, Guangzhou, Guangdong Province, China
| | - Yaling Tian
- School of Medicine, Jinan University, Guangzhou, Guangdong Province, China
| | - Aiping Wu
- School of Nursing, Jinan University, Guangzhou, Guangdong Province, China
| | - Shelley E. Keating
- School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - Qinhe Yang
- School of Chinese Medicine, Jinan University, Guangzhou, Guangdong Province, China
- Health Science Center, Jinan University, Guangzhou, Guangdong Province, China
| | - Chenli Lin
- School of Medicine, Jinan University, Guangzhou, Guangdong Province, China
- Health Science Center, Jinan University, Guangzhou, Guangdong Province, China
| | - Yinji Liang
- School of Nursing, Jinan University, Guangzhou, Guangdong Province, China
- Health Science Center, Jinan University, Guangzhou, Guangdong Province, China
| |
Collapse
|
5
|
Alcober-Boquet L, Kraus N, Huber LS, Vutukuri R, Fuhrmann DC, Stross C, Schaefer L, Scholich K, Zeuzem S, Piiper A, Schulz MH, Trebicka J, Welsch C, Ortiz C. BI-3231, an enzymatic inhibitor of HSD17B13, reduces lipotoxic effects induced by palmitic acid in murine and human hepatocytes. Am J Physiol Cell Physiol 2024; 326:C880-C892. [PMID: 38223924 DOI: 10.1152/ajpcell.00413.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 01/09/2024] [Accepted: 01/11/2024] [Indexed: 01/16/2024]
Abstract
17-β-hydroxysteroid dehydrogenase 13 (HSD17B13), a lipid droplet-associated enzyme, is primarily expressed in the liver and plays an important role in lipid metabolism. Targeted inhibition of enzymatic function is a potential therapeutic strategy for treating steatotic liver disease (SLD). The present study is aimed at investigating the effects of the first selective HSD17B13 inhibitor, BI-3231, in a model of hepatocellular lipotoxicity using human cell lines and primary mouse hepatocytes in vitro. Lipotoxicity was induced with palmitic acid in HepG2 cells and freshly isolated mouse hepatocytes and the cells were coincubated with BI-3231 to assess the protective effects. Under lipotoxic stress, triglyceride (TG) accumulation was significantly decreased in the BI-3231-treated cells compared with that of the control untreated human and mouse hepatocytes. In addition, treatment with BI-3231 led to considerable improvement in hepatocyte proliferation, cell differentiation, and lipid homeostasis. Mechanistically, BI-3231 increased the mitochondrial respiratory function without affecting β-oxidation. BI-3231 inhibited the lipotoxic effects of palmitic acid in hepatocytes, highlighting the potential of targeting HSD17B13 as a specific therapeutic approach in steatotic liver disease.NEW & NOTEWORTHY 17-β-Hydroxysteroid dehydrogenase 13 (HSD17B13) is a lipid droplet protein primarily expressed in the liver hepatocytes. HSD17B13 is associated with the clinical outcome of chronic liver diseases and is therefore a target for the development of drugs. Here, we demonstrate the promising therapeutic effect of BI-3231 as a potent inhibitor of HSD17B13 based on its ability to inhibit triglyceride accumulation in lipid droplets (LDs), restore lipid metabolism and homeostasis, and increase mitochondrial activity in vitro.
Collapse
Affiliation(s)
- Lucia Alcober-Boquet
- Medical Clinic 1, Goethe University Frankfurt, University Hospital, Frankfurt, Germany
| | - Nico Kraus
- Medical Clinic 1, Goethe University Frankfurt, University Hospital, Frankfurt, Germany
| | - Lisa Sophie Huber
- Faculty of Medicine, Institute of Pharmacology and Toxicology, Goethe University Frankfurt, Frankfurt, Germany
| | - Rajkumar Vutukuri
- Faculty of Medicine, Institute of Pharmacology and Toxicology, Goethe University Frankfurt, Frankfurt, Germany
| | - Dominik C Fuhrmann
- Faculty of Medicine, Institute of Biochemistry I, Goethe University Frankfurt, Frankfurt, Germany
| | - Claudia Stross
- Medical Clinic 1, Goethe University Frankfurt, University Hospital, Frankfurt, Germany
| | - Liliana Schaefer
- Faculty of Medicine, Institute of Pharmacology and Toxicology, Goethe University Frankfurt, Frankfurt, Germany
| | - Klaus Scholich
- Faculty of Medicine, Institute of Clinical Pharmacology, Goethe University Frankfurt, Frankfurt, Germany
| | - Stefan Zeuzem
- Medical Clinic 1, Goethe University Frankfurt, University Hospital, Frankfurt, Germany
| | - Albrecht Piiper
- Medical Clinic 1, Goethe University Frankfurt, University Hospital, Frankfurt, Germany
| | - Marcel H Schulz
- Faculty of Medicine, Institute of Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt, Germany
| | - Jonel Trebicka
- Department of Internal Medicine B, University Hospital Münster, Münster, Germany
| | - Christoph Welsch
- Medical Clinic 1, Goethe University Frankfurt, University Hospital, Frankfurt, Germany
| | - Cristina Ortiz
- Medical Clinic 1, Goethe University Frankfurt, University Hospital, Frankfurt, Germany
| |
Collapse
|
6
|
Pandey MK. Uncovering the Lipid Web: Discovering the Multifaceted Roles of Lipids in Human Diseases and Therapeutic Opportunities. Int J Mol Sci 2023; 24:13223. [PMID: 37686028 PMCID: PMC10487860 DOI: 10.3390/ijms241713223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 08/21/2023] [Indexed: 09/10/2023] Open
Abstract
Lipids, characterized by their hydrophobic nature, encompass a wide range of molecules with distinct properties and functions [...].
Collapse
Affiliation(s)
- Manoj Kumar Pandey
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center (CCHMC), 3333 Burnet Avenue, MLC-7016, Suit R1.019A, Cincinnati, OH 45229, USA; or ; Tel.: +1-513-803-1694; Fax: +1-513-636-1321
- Department of Pediatrics, University of Cincinnati College of Medicine, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| |
Collapse
|