1
|
Oubraim S, Fauzan M, Studholme K, Gordon C, Glaser ST, Shen RY, Ojima I, Kaczocha M, Haj-Dahmane S. Astrocytic FABP5 mediates retrograde endocannabinoid transport at central synapses. iScience 2025; 28:112342. [PMID: 40292318 PMCID: PMC12033926 DOI: 10.1016/j.isci.2025.112342] [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: 12/11/2024] [Revised: 02/19/2025] [Accepted: 03/31/2025] [Indexed: 04/30/2025] Open
Abstract
Endocannabinoids (eCBs) regulate synaptic function via cannabinoid receptors. While eCB signaling is well understood, the mechanisms underlying eCB synaptic transport are poorly characterized. Using 2-arachidonoylglycerol (2-AG)-mediated depolarization-induced suppression of inhibition (DSI) in the hippocampus as a readout of retrograde eCB signaling, we demonstrate that the deletion of fatty acid binding protein 5 (FABP5) impairs DSI. In FABP5 KO mice, DSI was rescued by re-expressing wild-type FABP5 but not an FABP5 mutant that does not bind 2-AG. Importantly, the deletion of astrocytic FABP5 blunted DSI, which was rescued by its re-expression in the astrocytes of FABP5 KO mice. Neuronal FABP5 was dispensable for 2-AG signaling. DSI was also rescued by expressing a secreted FABP5 variant but not by FABP7, an astrocytic FABP that does not undergo secretion. Our results demonstrate that extracellular FABP5 of astrocytic origin controls 2-AG transport and that FABP5 is adapted to coordinate intracellular and synaptic eCB transport.
Collapse
Affiliation(s)
- Saida Oubraim
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Mohammad Fauzan
- Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, USA
| | - Keith Studholme
- Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Chris Gordon
- Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Sherrye T. Glaser
- Department of Biological Sciences, Kingsborough Community College, Brooklyn, NY, USA
| | - Roh-Yu Shen
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
- University at Buffalo Neuroscience Program, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Iwao Ojima
- Department of Chemistry, Stony Brook University, Stony Brook, NY, USA
- Institute of Chemical Biology and Drug Discovery, Stony Brook University, Stony Brook, NY, USA
| | - Martin Kaczocha
- Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, USA
- Institute of Chemical Biology and Drug Discovery, Stony Brook University, Stony Brook, NY, USA
- Stony Brook University Pain and Analgesia Research Center (SPARC), Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Samir Haj-Dahmane
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
- University at Buffalo Neuroscience Program, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| |
Collapse
|
2
|
Muratoğlu B, Özdemir C, Eylem CC, Reçber T, Nemutlu E, Yet İ, Uçkan-Çetinkaya D. Circadian rhythm and aryl hydrocarbon receptor crosstalk in bone marrow adipose tissue and implications in leukemia. Sci Rep 2025; 15:16387. [PMID: 40350529 PMCID: PMC12066725 DOI: 10.1038/s41598-025-93169-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 03/05/2025] [Indexed: 05/14/2025] Open
Abstract
Leukemic cells modulate the bone marrow microenvironment to enhance their survival. Lipolysis in bone marrow adipose tissue (BMAT) has emerged as a critical factor supporting leukemic cell survival, yet understanding its primary role in leukemia development remains limited. Fanconi anemia (FA), characterized by a predisposition to acute myeloid leukemia (AML) and hypersensitivity to environmental toxins, is a transitional model for studying leukemic transformation. İntegrated multi-omics analyses were conducted on BMAT-derived mesenchymal stem/stromal cells (MSCs) from healthy donors (HD), AML, and FA patients. These analyses revealed intricate interactions among genes, metabolites, and lipids. Particularly noteworthy were the effects observed following the inhibition of aryl hydrocarbon receptor (AhR) signaling by StemRegenin1 (SR1). BMAT-MSCs showed increased expression of epithelial-mesenchymal transition (EMT) genes in FA and AML, suggesting a potential shift towards cancer-associated fibroblasts in the dysregulated marrow microenvironment. Identification of potential circadian rhythm biomarkers (NPAS2, PER2, BHLHE40, PER3, CIART) in BMAT-MSCs indicates a link between related lipid metabolism genes (e.g., PTGS1, PIK3R1) and SR1 treatment, implicating them in lipolysis processes. Dysregulation of circadian rhythm-related genes (CIART, BHLHE40, NPAS2) in AML BMAT-MSCs, along with changes in circulating lipid metabolites like palmitate suggests their role in shaping the leukemia microenvironment. Upregulation of FABP5 and CD36 suggests a novel molecular mechanism involving FABP5 in AhR-mediated circadian regulation and identifies CD36 as a potential partner for FABP5 in BMAT-MSCs. Overall, this study unveils the interplay between AhR signaling, circadian rhythm, and the leukemia microenvironment in BMAT-MSCs, offering new insights into leukemia pathogenesis and therapeutic opportunities.
Collapse
Affiliation(s)
- Bihter Muratoğlu
- Center for Stem Cell Research and Development (PEDI-STEM), Hacettepe University, 06100, Sihhiye, Ankara, Turkey
- Department of Stem Cell Sciences, Institute of Health Sciences, Hacettepe University, 06100, Sihhiye, Ankara, Turkey
| | - Cansu Özdemir
- Center for Stem Cell Research and Development (PEDI-STEM), Hacettepe University, 06100, Sihhiye, Ankara, Turkey.
- Department of Stem Cell Sciences, Institute of Health Sciences, Hacettepe University, 06100, Sihhiye, Ankara, Turkey.
| | - Cemil Can Eylem
- Department of Analytical Chemistry, Faculty of Pharmacy, Hacettepe University, 06100, Sihhiye, Ankara, Turkey
| | - Tuba Reçber
- Department of Analytical Chemistry, Faculty of Pharmacy, Hacettepe University, 06100, Sihhiye, Ankara, Turkey
| | - Emirhan Nemutlu
- Department of Analytical Chemistry, Faculty of Pharmacy, Hacettepe University, 06100, Sihhiye, Ankara, Turkey
| | - İdil Yet
- Department of Bioinformatics, Institute of Health Sciences, Hacettepe University, 06100, Sihhiye, Ankara, Turkey
| | - Duygu Uçkan-Çetinkaya
- Center for Stem Cell Research and Development (PEDI-STEM), Hacettepe University, 06100, Sihhiye, Ankara, Turkey.
- Department of Stem Cell Sciences, Institute of Health Sciences, Hacettepe University, 06100, Sihhiye, Ankara, Turkey.
- Division of Hematology, Department of Pediatrics, Hacettepe University Faculty of Medicine, 06100, Sihhiye, Ankara, Turkey.
| |
Collapse
|
3
|
Zhang Y, Zhang R, Li C, Peng G. Exploring the causal association between fatty acid-binding proteins and anaphylactic shock due to adverse reactions to medications: A two-sample Mendelian randomization study. Medicine (Baltimore) 2025; 104:e42171. [PMID: 40324257 PMCID: PMC12055110 DOI: 10.1097/md.0000000000042171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 03/27/2025] [Accepted: 04/01/2025] [Indexed: 05/07/2025] Open
Abstract
Previous studies have identified a relationship between fatty acid-binding proteins (FABPs) and immune diseases. This study aimed to investigate whether a causal relationship exists between FABPs and anaphylactic shock resulting from adverse drug reactions. Single nucleotide polymorphisms associated with FABPs were utilized as instrumental variables, sourced from the National Human Genome Research Institute-European Bioinformatics Institute Catalog of human genome-wide association studies. Data on anaphylactic shock due to adverse effects of correctly administered drugs were obtained from the FinnGen database, which includes genomic and health data from 500,000 Finnish biobank donors. A two-sample Mendelian randomization analysis was conducted to explore the causality between FABPs and anaphylactic shock due to adverse drug reactions. The analysis revealed a negative causal relationship between FABP5 (odds ratio [OR] = 0.40; 95% confidence interval [CI] = 0.17-0.92; P = .032) and FABP12 (OR = 0.77; 95% CI = 0.63-0.94; P = .009) and anaphylactic shock due to adverse drug reactions. These findings were corroborated by Mendelian randomization-Egger, weighted median, and weighted mode methods. This study provides robust evidence supporting a protective relationship between FABP5 and FABP12 and anaphylactic shock due to adverse drug reactions. Further experimental studies are warranted to elucidate the causal mechanisms and associations between FABP5, FABP12, and anaphylactic shock in the context of adverse drug reactions.
Collapse
Affiliation(s)
- Yu Zhang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
- National Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing, China
| | - Rusheng Zhang
- Department of Cardiology, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Cunyu Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
- National Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing, China
| | - Guoping Peng
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
- National Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing, China
| |
Collapse
|
4
|
Widmer A, Lillegard K, Wood K, Robles M, Fan R, Ye F, Koethe JR, Silver HJ. Consumption of tree nuts as snacks stimulates changes in plasma fatty acid profiles and adipose tissue gene expression in young adults at risk for metabolic syndrome. Clin Nutr 2025; 48:25-34. [PMID: 40117963 DOI: 10.1016/j.clnu.2025.03.002] [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: 10/11/2024] [Revised: 02/10/2025] [Accepted: 03/01/2025] [Indexed: 03/23/2025]
Abstract
BACKGROUND AND AIMS The prevalence of metabolic syndrome has been increasing in young adults, concomitant with the occurrence of increased abdominal adiposity. We previously reported that consuming tree nuts, as replacement for typical high-carbohydrate snacks, reduces visceral fat and waist circumference in young adults who have one or more metabolic syndrome risk factors. We aimed to investigate the effects of tree nuts snack consumption on plasma and adipose tissue fatty acid profiles along with changes in the expression of adipose tissue genes involved in thermogenesis, glycemia, adipocyte signaling, lipolysis, and immunity. METHODS A randomized parallel-arm 16-week intervention trial was conducted in 84 adults aged 22-36 years. Participants in both groups were provided with caloric goals for weight maintenance, daily menus, and pre-portioned snacks at every other week visits with study registered dietitians. Changes in dietary fatty acid intakes, plasma and abdominal subcutaneous adipose tissue (SAT) triglycerides fatty acid profiles using gas-liquid chromatography, and the expression of 241 genes in abdominal SAT were evaluated. RESULTS Consuming tree nuts snacks increased mono- and polyunsaturated fatty acid intakes yielding a 9-fold greater dietary unsaturated to saturated fat ratio. The tree nuts snack group also had significantly greater improvements in plasma 16:1/16:0 ratio; plasma phospholipids oleic and gamma linolenic acid content; plasma diglycerides, triglycerides, and cholesterol esters oleic acid content; and total plasma monounsaturated fatty acids. While abdominal SAT only showed trends for increased oleic acid content and unsaturated to saturated fat ratio, the tree nuts snacks participants had altered expression of 13 genes in abdominal SAT that have roles in nutrient sensing, energy homeostasis, and vulnerability to obesity. CONCLUSIONS Replacing typical high-carbohydrate snacks with tree nuts results in more favorable dietary, plasma, and adipose tissue fatty acid profiles that could aid in preventing the development of excess adiposity and cardiometabolic disease states including metabolic syndrome. CLINICAL TRIAL REGISTRY This trial was registered at clinicaltrials.gov NCT03969264.
Collapse
Affiliation(s)
- Annaliese Widmer
- Vanderbilt University Medical Center, Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, Nashville, TN, USA
| | - Kate Lillegard
- Vanderbilt University Medical Center, Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, Nashville, TN, USA
| | - Kate Wood
- Vanderbilt University Medical Center, Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, Nashville, TN, USA
| | - Michelle Robles
- Vanderbilt University Medical Center, Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, Nashville, TN, USA
| | - Run Fan
- Vanderbilt University Medical Center, Department of Biostatistics, Nashville, TN, USA; Vanderbilt University Medical Center, Department of Epidemiology, Nashville, TN, USA
| | - Fei Ye
- Vanderbilt University Medical Center, Department of Biostatistics, Nashville, TN, USA; Vanderbilt University Medical Center, Department of Epidemiology, Nashville, TN, USA
| | - John R Koethe
- Vanderbilt University Medical Center, Department of Medicine, Division of Infectious Diseases, Nashville, TN, USA; Tennessee Valley Healthcare System, Department of Veterans Affairs, Nashville, TN, USA
| | - Heidi J Silver
- Vanderbilt University Medical Center, Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, Nashville, TN, USA; Tennessee Valley Healthcare System, Department of Veterans Affairs, Nashville, TN, USA.
| |
Collapse
|
5
|
Xu X, Zhan C, Qiao J, Yang Y, Li C, Li P, Ma S. Transcriptomic Analysis of Muscle Satellite Cell Regulation on Intramuscular Preadipocyte Differentiation in Tan Sheep. Int J Mol Sci 2025; 26:3414. [PMID: 40244284 PMCID: PMC11989785 DOI: 10.3390/ijms26073414] [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: 03/07/2025] [Revised: 03/27/2025] [Accepted: 04/02/2025] [Indexed: 04/18/2025] Open
Abstract
Intramuscular fat (IMF) content is a key factor influencing meat properties including tenderness, flavor, and marbling. However, the complex molecular mechanisms regulating IMF deposition, especially the interactions between intramuscular preadipocytes (IMAdCs) and skeletal muscle satellite cells (SMSCs), remain unclear. In this study, a direct co-culture system of sheep IMAdCs and SMSCs was used to elucidate their intercellular interactions. RNA sequencing and bioinformatics analyses were performed under monoculture and co-culture conditions for later stages of differentiation. The obtained results showed that SMSCs significantly inhibited the adipogenic capacity of IMAdCs. This was reflected in the co-culture markedly altered gene expression and observations of lipid droplets in our studies, i.e., the PPARG, ACOX2, PIK3R1, FABP5, FYN, ALDOC, PFKM, PFKL, HADH, and HADHB genes were down-regulated in the co-cultured IMAdCs in association with the inhibition of fat deposition, whereas ACSL3, ACSL4, ATF3, EGR1, and IGF1R within the genes upregulated in co-culture IMAdCs were associated with the promotion of lipid metabolism. In addition, GO, KEGG, and ligand-receptor pairing analyses further elucidated the molecular mechanisms of intercellular communication. These findings emphasize the regulatory role of SMSCs on intramuscular preadipocyte differentiation and lipid metabolism, providing a theoretical framework for targeted molecular strategies to improve sheep meat quality.
Collapse
Affiliation(s)
- Xiaochun Xu
- Collaborative Innovation Center for Food Production and Safety, School of Biological Science & Engineering, North Minzu University, Yinchuan 750021, China; (C.Z.); (J.Q.); (Y.Y.); (C.L.); (P.L.)
| | - Cong Zhan
- Collaborative Innovation Center for Food Production and Safety, School of Biological Science & Engineering, North Minzu University, Yinchuan 750021, China; (C.Z.); (J.Q.); (Y.Y.); (C.L.); (P.L.)
| | - Jiaqi Qiao
- Collaborative Innovation Center for Food Production and Safety, School of Biological Science & Engineering, North Minzu University, Yinchuan 750021, China; (C.Z.); (J.Q.); (Y.Y.); (C.L.); (P.L.)
| | - Yuxuan Yang
- Collaborative Innovation Center for Food Production and Safety, School of Biological Science & Engineering, North Minzu University, Yinchuan 750021, China; (C.Z.); (J.Q.); (Y.Y.); (C.L.); (P.L.)
| | - Changyuan Li
- Collaborative Innovation Center for Food Production and Safety, School of Biological Science & Engineering, North Minzu University, Yinchuan 750021, China; (C.Z.); (J.Q.); (Y.Y.); (C.L.); (P.L.)
| | - Pan Li
- Collaborative Innovation Center for Food Production and Safety, School of Biological Science & Engineering, North Minzu University, Yinchuan 750021, China; (C.Z.); (J.Q.); (Y.Y.); (C.L.); (P.L.)
| | - Sen Ma
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450002, China
| |
Collapse
|
6
|
Jacinto E. Making sense of fat in cancer. Science 2025; 387:1147-1148. [PMID: 40080596 DOI: 10.1126/science.adw1956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/15/2025]
Abstract
A lipid chaperone enables sensing of an essential fatty acid to drive tumor growth.
Collapse
Affiliation(s)
- Estela Jacinto
- Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, USA
| |
Collapse
|
7
|
Blanco‐Pintos T, Regueira‐Iglesias A, Kuz I, Sánchez‐Barco A, Seijas‐Otero N, Chantada‐Vázquez MDP, Balsa‐Castro C, Tomás I. Impact of smoking habit on the subgingival proteome in patients with periodontitis. J Periodontol 2025; 96:217-229. [PMID: 39282712 PMCID: PMC11951952 DOI: 10.1002/jper.24-0062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 07/11/2024] [Accepted: 07/14/2024] [Indexed: 03/29/2025]
Abstract
BACKGROUND Few investigations evaluated smoking's impact on the periodontal proteome. Therefore, this study aimed to analyse the influence of tobacco on the overall periodontal proteome and the differential expression of gingival crevicular fluid (GCF) proteins using sequential window acquisition of all theoretical mass spectra (SWATH-MS). METHODS GCF samples were collected from 40 periodontitis subjects (stages III-IV). These were separated based on smoking status into smokers (17), ex-smokers (10), and non-smokers (13). Samples were analysed using SWATH-MS, and proteins were identified using the UniProt human-specific database. Data are available via ProteomeXchange with the identifier PXD043474. Principal component analysis (PCA) was employed to examine the spectral mass distribution of the proteome. Protein expression was different for a p-value <0.05 and a log2 fold change ≥0.3 (upregulated) or ≤-0.3 (downregulated). RESULTS The distribution of overall proteome did not differ between non-smokers, smokers, and ex-smokers. Considering protein expression, 23 were differentially expressed in smokers vs. non-smokers (16 upregulated and 7 downregulated), 17 in ex-smokers vs. non-smokers (2 upregulated and 15 downregulated), and only 8 in smokers vs. ex-smokers (7 upregulated and 1 downregulated). Smoking increased the expression of proteins related to epithelial hyperkeratinization (keratins type II cytoskeletal 4, type I cytoskeletal 13 and type I cytoskeletal 19, cornulin, and fatty acid-binding protein 5). However, multiple immunoglobulins were underexpressed when comparing smokers and ex-smokers to non-smokers. CONCLUSION Although smoking does not significantly modify the overall GCF proteome associated with periodontitis, it alters the expression of several proteins compared to never-smokers and ex-smokers. PLAIN LANGUAGE SUMMARY Smoking is a critical risk factor for the development and progression of periodontitis. However, evidence of the effect of smoking on the subgingival proteome is scarce. Therefore, this study aimed to determine the impact of smoking on the overall proteome and differential expression of gingival crevicular fluid (GCF) proteins using the sequential window acquisition of all theoretical mass spectra (SWATH-MS) proteomic technique. For this purpose, GCF samples were collected from 40 subjects with periodontitis, of which 17 were smokers, 10 were ex-smokers, and 13 were non-smokers. These samples were analysed by SWATH-MS, and proteins were identified using the UniProt human-specific database. Analysis of the overall proteome showed that its distribution was not significantly different between smokers, ex-smokers, and non-smokers. However, several proteins were found to be differentially expressed according to the smoking status. Smoking can increase the expression of several keratins and proteins related to hyperkeratinization of the epithelium. However, in ex-smokers, these proteins return to similar levels to those of non-smokers. Moreover, smoking may induce a lower expression of proteins related to adaptive immunity, such as immunoglobulins. This immunosuppressive effect may persist in ex-smokers.
Collapse
Affiliation(s)
- Triana Blanco‐Pintos
- Oral Sciences Research Group, Special Needs UnitDepartment of Surgery and Medical‐Surgical SpecialtiesSchool of Medicine and DentistryUniversidade de Santiago de CompostelaHealth Research Institute of Santiago (IDIS)Santiago de CompostelaSpain
| | - Alba Regueira‐Iglesias
- Oral Sciences Research Group, Special Needs UnitDepartment of Surgery and Medical‐Surgical SpecialtiesSchool of Medicine and DentistryUniversidade de Santiago de CompostelaHealth Research Institute of Santiago (IDIS)Santiago de CompostelaSpain
| | - Iryna Kuz
- Oral Sciences Research Group, Special Needs UnitDepartment of Surgery and Medical‐Surgical SpecialtiesSchool of Medicine and DentistryUniversidade de Santiago de CompostelaHealth Research Institute of Santiago (IDIS)Santiago de CompostelaSpain
| | - Alba Sánchez‐Barco
- Oral Sciences Research Group, Special Needs UnitDepartment of Surgery and Medical‐Surgical SpecialtiesSchool of Medicine and DentistryUniversidade de Santiago de CompostelaHealth Research Institute of Santiago (IDIS)Santiago de CompostelaSpain
| | - Noelia Seijas‐Otero
- Oral Sciences Research Group, Special Needs UnitDepartment of Surgery and Medical‐Surgical SpecialtiesSchool of Medicine and DentistryUniversidade de Santiago de CompostelaHealth Research Institute of Santiago (IDIS)Santiago de CompostelaSpain
| | | | - Carlos Balsa‐Castro
- Oral Sciences Research Group, Special Needs UnitDepartment of Surgery and Medical‐Surgical SpecialtiesSchool of Medicine and DentistryUniversidade de Santiago de CompostelaHealth Research Institute of Santiago (IDIS)Santiago de CompostelaSpain
| | - Inmaculada Tomás
- Oral Sciences Research Group, Special Needs UnitDepartment of Surgery and Medical‐Surgical SpecialtiesSchool of Medicine and DentistryUniversidade de Santiago de CompostelaHealth Research Institute of Santiago (IDIS)Santiago de CompostelaSpain
| |
Collapse
|
8
|
Vornoli A, Souid A, Lazzari B, Turri F, Pizzi F, Bramanti E, Campanella B, Trouki C, Raffaelli A, Wójcik M, Della Croce CM, Giorgetti L, Longo V, Capra E, Pozzo L. A Moderate Intake of Beer Improves Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD) in a High-Fat Diet (HFD)-Induced Mouse Model. Molecules 2024; 29:5954. [PMID: 39770043 PMCID: PMC11676803 DOI: 10.3390/molecules29245954] [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: 11/05/2024] [Revised: 12/02/2024] [Accepted: 12/12/2024] [Indexed: 01/04/2025] Open
Abstract
Beer and its components show potential for reducing hepatic steatosis in rodent models through multiple mechanisms. This study aimed to evaluate beer's anti-steatotic effects in a high-fat diet (HFD)-induced mouse model of Metabolic dysfunction-Associated Liver Disease (MASLD) and to explore the underlying mechanisms. In the HFD group, steatosis was confirmed by altered blood parameters, weight gain, elevated liver lipid content, and histological changes. These markers were normalized in the HFD+beer group, reaching levels similar to the control (CTR) group. Protein carbonylation and lipid peroxidation levels were consistent across all groups, suggesting that the model represents an early stage of MASLD without oxidative stress. Transcriptomic and CpG methylation analyses revealed clear distinctions between the CTR and HFD groups. RNA sequencing identified 162 differentially expressed genes (DEGs) between the CTR and HFD groups, primarily related to inflammation and lipid regulation. Beer consumption modified the health of the HFD mice, affecting inflammation but not lipid homeostasis (CTR vs. HFD+beer, DEGs = 43). The CpG methylation analysis indicated that beer lowered methylation, impacting genes linked to lipid accumulation and inflammation. A cecal metabolite analysis suggested that beer improved short-chain fatty acid metabolism (SCFA). In summary, a moderate beer intake may mitigate MASLD by modulating lipid metabolism and SCFA pathways, likely through polyphenol activity.
Collapse
Affiliation(s)
- Andrea Vornoli
- Institute of Agricultural Biology and Biotechnology, National Research Council, Via Moruzzi 1, 56124 Pisa, Italy; (A.V.); (A.S.); (A.R.); (L.G.); (V.L.)
| | - Aymen Souid
- Institute of Agricultural Biology and Biotechnology, National Research Council, Via Moruzzi 1, 56124 Pisa, Italy; (A.V.); (A.S.); (A.R.); (L.G.); (V.L.)
| | - Barbara Lazzari
- Institute of Agricultural Biology and Biotechnology, National Research Council, Via Corti 12, 20133 Milan, Italy;
| | - Federica Turri
- Institute of Agricultural Biology and Biotechnology, National Research Council, Via dell’Università 6, 26900 Lodi, Italy; (F.T.); (F.P.)
| | - Flavia Pizzi
- Institute of Agricultural Biology and Biotechnology, National Research Council, Via dell’Università 6, 26900 Lodi, Italy; (F.T.); (F.P.)
| | - Emilia Bramanti
- Institute of Chemistry of Organometallic Compounds, National Research Council, Via Moruzzi 1, 56124 Pisa, Italy; (E.B.); (B.C.)
| | - Beatrice Campanella
- Institute of Chemistry of Organometallic Compounds, National Research Council, Via Moruzzi 1, 56124 Pisa, Italy; (E.B.); (B.C.)
| | - Cheherazade Trouki
- Institute for Chemical and Physical Processes, National Research Council, Via Moruzzi 1, 56124 Pisa, Italy;
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Andrea Raffaelli
- Institute of Agricultural Biology and Biotechnology, National Research Council, Via Moruzzi 1, 56124 Pisa, Italy; (A.V.); (A.S.); (A.R.); (L.G.); (V.L.)
- Crop Science Research Center, Scuola Superiore Sant’Anna, Piazza Martiri della Libertà 33, 56127 Pisa, Italy
| | - Marta Wójcik
- Sub-Department of Pathophysiology, Department of Preclinical of Veterinary Sciences, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, Akademicka 12, 20-033 Lublin, Poland;
| | - Clara Maria Della Croce
- Institute of Agricultural Biology and Biotechnology, National Research Council, Via Moruzzi 1, 56124 Pisa, Italy; (A.V.); (A.S.); (A.R.); (L.G.); (V.L.)
| | - Lucia Giorgetti
- Institute of Agricultural Biology and Biotechnology, National Research Council, Via Moruzzi 1, 56124 Pisa, Italy; (A.V.); (A.S.); (A.R.); (L.G.); (V.L.)
| | - Vincenzo Longo
- Institute of Agricultural Biology and Biotechnology, National Research Council, Via Moruzzi 1, 56124 Pisa, Italy; (A.V.); (A.S.); (A.R.); (L.G.); (V.L.)
| | - Emanuele Capra
- Institute of Agricultural Biology and Biotechnology, National Research Council, Via dell’Università 6, 26900 Lodi, Italy; (F.T.); (F.P.)
| | - Luisa Pozzo
- Institute of Agricultural Biology and Biotechnology, National Research Council, Via Moruzzi 1, 56124 Pisa, Italy; (A.V.); (A.S.); (A.R.); (L.G.); (V.L.)
| |
Collapse
|
9
|
Phelps DW, Connors AM, Ferrero G, DeWitt JC, Yoder JA. Per- and polyfluoroalkyl substances alter innate immune function: evidence and data gaps. J Immunotoxicol 2024; 21:2343362. [PMID: 38712868 PMCID: PMC11249028 DOI: 10.1080/1547691x.2024.2343362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 04/10/2024] [Indexed: 05/08/2024] Open
Abstract
Per- and polyfluoroalkyl substances (PFASs) are a large class of compounds used in a variety of processes and consumer products. Their unique chemical properties make them ubiquitous and persistent environmental contaminants while also making them economically viable and socially convenient. To date, several reviews have been published to synthesize information regarding the immunotoxic effects of PFASs on the adaptive immune system. However, these reviews often do not include data on the impact of these compounds on innate immunity. Here, current literature is reviewed to identify and incorporate data regarding the effects of PFASs on innate immunity in humans, experimental models, and wildlife. Known mechanisms by which PFASs modulate innate immune function are also reviewed, including disruption of cell signaling, metabolism, and tissue-level effects. For PFASs where innate immune data are available, results are equivocal, raising additional questions about common mechanisms or pathways of toxicity, but highlighting that the innate immune system within several species can be perturbed by exposure to PFASs. Recommendations are provided for future research to inform hazard identification, risk assessment, and risk management practices for PFASs to protect the immune systems of exposed organisms as well as environmental health.
Collapse
Affiliation(s)
- Drake W. Phelps
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC
- Center for Environmental and Health Effects of PFAS, North Carolina State University, Raleigh, NC
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC
| | - Ashley M. Connors
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC
- Center for Environmental and Health Effects of PFAS, North Carolina State University, Raleigh, NC
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC
- Toxicology Program, North Carolina State University, Raleigh, NC
- Genetics and Genomics Academy, North Carolina State University, Raleigh, NC
| | - Giuliano Ferrero
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC
- Center for Environmental and Health Effects of PFAS, North Carolina State University, Raleigh, NC
| | - Jamie C. DeWitt
- Center for Environmental and Health Effects of PFAS, North Carolina State University, Raleigh, NC
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR
| | - Jeffrey A. Yoder
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC
- Center for Environmental and Health Effects of PFAS, North Carolina State University, Raleigh, NC
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC
- Toxicology Program, North Carolina State University, Raleigh, NC
- Genetics and Genomics Academy, North Carolina State University, Raleigh, NC
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC
| |
Collapse
|
10
|
Wei T, Li R, Guo S, Liang C. Stigmasterol exerts antiglioma effects by regulating lipid metabolism. Mol Med Rep 2024; 30:227. [PMID: 39364731 PMCID: PMC11484536 DOI: 10.3892/mmr.2024.13351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 09/18/2024] [Indexed: 10/05/2024] Open
Abstract
Stigmasterol is a sterol compound found in various traditional Chinese medicines; however, its effects on glioma remain unclear. The present study aimed to investigate the effects of stigmasterol on the biological behaviors of glioblastoma (GBM) cells and to explore the underlying mechanisms. In vitro experiments assessed its effects on GBM cell proliferation, apoptosis, cell cycle progression, invasion, migration and vasculogenic mimicry (VM). The potential targets for stigmasterol in treating GBM were identified using databases and Venn diagram analysis, followed by enrichment analysis using R language. A prognostic model related to the target genes of stigmasterol was developed through univariate Cox regression and least absolute shrinkage and selection operator analyses. Stigmasterol was found to suppress the proliferation of GBM cells in a dose‑ and time‑dependent manner, to induce apoptosis, and to inhibit invasion, migration and VM formation. Additionally, 31 potential targets of stigmasterol were identified, linked to lipid metabolism and the G protein‑coupled receptor signaling pathway. Lipid metabolism assays revealed that stigmasterol significantly reduced free fatty acids and total cholesterol levels. Furthermore, two prognosis‑related target genes, fatty acid binding protein 5 and α‑1B adrenergic receptor, were selected, and the prognostic model effectively predicted GBM outcomes. Moreover, molecular docking revealed strong binding affinities between stigmasterol and the target proteins. Overall, these findings suggested that stigmasterol may exert anti‑glioma effects, which could be potentially mediated through the regulation of lipid metabolism.
Collapse
Affiliation(s)
- Ting Wei
- Department of Ophthalmology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Ruichun Li
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Shiwen Guo
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Chen Liang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
11
|
Nakanishi S, Hasegawa T, Maeno K, Motoyama A, Denda M. OBP2A regulates epidermal barrier function and protects against cytotoxic small hydrophobic molecules. iScience 2024; 27:111093. [PMID: 39502293 PMCID: PMC11536036 DOI: 10.1016/j.isci.2024.111093] [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: 04/23/2024] [Revised: 07/14/2024] [Accepted: 09/30/2024] [Indexed: 11/08/2024] Open
Abstract
The skin is constantly exposed to environmental sensory stimuli, which may include harmful volatiles and small hydrophobic molecules. However, the skin's protective mechanism against the latter agents is unclear. Here, we demonstrate that odorant binding protein 2A (OBP2A) protects epidermal keratinocytes against cytotoxic small hydrophobic molecules. OBP2A is mainly expressed in human epidermal keratinocytes. Cellular resistance to cytotoxic aldehyde and lipids was reduced in keratinocytes when OBP2A was silenced. Furthermore, silencing of OBP2A in a three-dimensional epidermal equivalent model resulted in impairment of epidermal barrier function. Inhibition of OBP2A caused disruption of keratinocyte lipid metabolism and induced endoplasmic reticulum stress. OBP2A expression was markedly decreased in the epidermis of atopic dermatitis lesional skin. In addition, interleukin-13 suppressed the expression of OBP2A in keratinocytes. Overall, our findings suggest that OBP2A regulates epidermal barrier function and contributes to protection of the skin against harmful small hydrophobic molecules.
Collapse
Affiliation(s)
| | | | | | - Akira Motoyama
- Shiseido Global Innovation Center, Yokohama 220-0011, Japan
| | - Mitsuhiro Denda
- Institute for Advanced Study of Mathematical Sciences, Meiji University, Nakano-ku, Tokyo 164-8525, Japan
| |
Collapse
|
12
|
Doswell F, Haley JD, Kaczocha M. Proteomic Analysis of Signaling Pathways Modulated by Fatty Acid Binding Protein 5 (FABP5) in Macrophages. J Pharmacol Exp Ther 2024; 391:289-300. [PMID: 38849143 PMCID: PMC11493448 DOI: 10.1124/jpet.123.002006] [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: 11/03/2023] [Revised: 05/20/2024] [Accepted: 05/28/2024] [Indexed: 06/09/2024] Open
Abstract
Although acute inflammation serves essential functions in maintaining tissue homeostasis, chronic inflammation is causally linked to many diseases. Macrophages are a major cell type that orchestrates inflammatory processes. During inflammation, macrophages undergo polarization and activation, thereby mobilizing pro-inflammatory and anti-inflammatory transcriptional programs that regulate ensuing macrophage functions. Fatty acid binding protein 5 (FABP5) is a lipid chaperone highly expressed in macrophages. FABP5 deletion is implicated in driving macrophages toward an anti-inflammatory phenotype, yet signaling pathways regulated by macrophage-FABP5 have not been systematically profiled. We leveraged proteomic and phosphoproteomic approaches to characterize pathways modulated by FABP5 in M1 and M2 polarized bone marrow-derived macrophages (BMDMs). Stable isotope labeling by amino acids-based analysis of M1 and M2 polarized wild-type and FABP5 knockout BMDMs revealed numerous differentially regulated proteins and phosphoproteins. FABP5 deletion impacted downstream pathways associated with inflammation, cytokine production, oxidative stress, and kinase activity. Toll-like receptor 2 (TLR2) emerged as a novel target of FABP5 and pharmacological FABP5 inhibition blunted TLR2-mediated activation of downstream pathways, ascribing a novel role for FABP5 in TLR2 signaling. This study represents a comprehensive characterization of the impact of FABP5 deletion on the proteomic and phosphoproteomic landscape of M1 and M2 polarized BMDMs. Loss of FABP5 altered pathways implicated in inflammatory responses, macrophage function, and TLR2 signaling. This work provides a foundation for future studies seeking to investigate the therapeutic potential of FABP5 inhibition in pathophysiological states resulting from dysregulated inflammatory signaling. SIGNIFICANCE STATEMENT: This research offers a comprehensive analysis of fatty acid binding protein 5 (FABP5) in macrophages during inflammatory response. The authors employed quantitative proteomic and phosphoproteomic approaches to investigate this utilizing bone marrow-derived macrophages that were M1 and M2 polarized using lipopolysaccharide with interferon γ and interleukin-4, respectively. This revealed multiple pathways related to inflammation that were differentially regulated due to the absence of FABP5. These findings underscore the potential therapeutic significance of macrophage-FABP5 as a candidate for addressing inflammatory-related diseases.
Collapse
Affiliation(s)
- Faniya Doswell
- Molecular and Cellular Biology Program, Stony Brook University, Stony Brook, New York (F.D.); Departments of Anesthesiology (F.D., M.K.) and Pathology (J.D.H.) and Biological Mass Spectrometry Facility, (J.D.H.), Renaissance School of Medicine, Stony Brook University, Stony Brook, New York
| | - John D Haley
- Molecular and Cellular Biology Program, Stony Brook University, Stony Brook, New York (F.D.); Departments of Anesthesiology (F.D., M.K.) and Pathology (J.D.H.) and Biological Mass Spectrometry Facility, (J.D.H.), Renaissance School of Medicine, Stony Brook University, Stony Brook, New York
| | - Martin Kaczocha
- Molecular and Cellular Biology Program, Stony Brook University, Stony Brook, New York (F.D.); Departments of Anesthesiology (F.D., M.K.) and Pathology (J.D.H.) and Biological Mass Spectrometry Facility, (J.D.H.), Renaissance School of Medicine, Stony Brook University, Stony Brook, New York
| |
Collapse
|
13
|
Sharma V, Patial V. Insights into the molecular mechanisms of malnutrition-associated steatohepatitis: A review. Liver Int 2024; 44:2156-2173. [PMID: 38775001 DOI: 10.1111/liv.15932] [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: 11/01/2023] [Revised: 03/21/2024] [Accepted: 03/28/2024] [Indexed: 08/10/2024]
Abstract
Malnutrition is a public health epidemic mainly targeting poverty-stricken people, young ones, older people, pregnant women, and individuals with metabolic disorders. Severe malnutrition is linked with several metabolic defects, such as hepatic dysfunction, hypertension, cardiovascular disease, and osteoarthritis. The proper functioning of the liver plays a crucial role in ensuring the supply of nutrients to the body. Consequently, inadequate nutrition can lead to severe periportal hepatic steatosis due to compromised mitochondrial and peroxisome functions. Reduced protein intake disrupts essential metabolic processes like the TCA cycle, oxidative phosphorylation, and β-oxidation, ultimately affecting ATP production. Furthermore, this can trigger a cascade of events, including disturbances in amino acid metabolism, iron metabolism, and gut microbiota, which activate genes involved in de novo lipogenesis, leading to the accumulation of lipids in the liver. The condition, in prolonged cases, progresses to steatohepatitis and liver fibrosis. Limited therapeutic solutions are available; however, few dietary supplements and drugs have demonstrated positive effects on the growth and health of malnourished individuals. These supplements improve parameters such as inflammatory and oxidative status, reduce triglyceride accumulation, enhance insulin sensitivity, and downregulate gene expression in hepatic lipid metabolism. This review elucidates the various mechanisms involved in malnutrition-associated steatohepatitis and provides an overview of the available approaches for treating this condition.
Collapse
Affiliation(s)
- Vinesh Sharma
- Pharmacology and Toxicology Laboratory, Dietetics & Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Vikram Patial
- Pharmacology and Toxicology Laboratory, Dietetics & Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
14
|
Hildebrandt F, Iturritza MU, Zwicker C, Vanneste B, Van Hul N, Semle E, Quin J, Pascini T, Saarenpää S, He M, Andersson ER, Scott CL, Vega-Rodriguez J, Lundeberg J, Ankarklev J. Host-pathogen interactions in the Plasmodium-infected mouse liver at spatial and single-cell resolution. Nat Commun 2024; 15:7105. [PMID: 39160174 PMCID: PMC11333755 DOI: 10.1038/s41467-024-51418-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 08/06/2024] [Indexed: 08/21/2024] Open
Abstract
Upon infecting its vertebrate host, the malaria parasite initially invades the liver where it undergoes massive replication, whilst remaining clinically silent. The coordination of host responses across the complex liver tissue during malaria infection remains unexplored. Here, we perform spatial transcriptomics in combination with single-nuclei RNA sequencing over multiple time points to delineate host-pathogen interactions across Plasmodium berghei-infected liver tissues. Our data reveals significant changes in spatial gene expression in the malaria-infected tissues. These include changes related to lipid metabolism in the proximity to sites of Plasmodium infection, distinct inflammation programs between lobular zones, and regions with enrichment of different inflammatory cells, which we term 'inflammatory hotspots'. We also observe significant upregulation of genes involved in inflammation in the control liver tissues of mice injected with mosquito salivary gland components. However, this response is considerably delayed compared to that observed in P. berghei-infected mice. Our study establishes a benchmark for investigating transcriptome changes during host-parasite interactions in tissues, it provides informative insights regarding in vivo study design linked to infection and offers a useful tool for the discovery and validation of de novo intervention strategies aimed at malaria liver stage infection.
Collapse
Affiliation(s)
- Franziska Hildebrandt
- Molecular Biosciences, the Wenner Gren Institute, Stockholm University, Svante Arrhenius Väg 20C, SE-106 91, Stockholm, Sweden.
| | - Miren Urrutia Iturritza
- Molecular Biosciences, the Wenner Gren Institute, Stockholm University, Svante Arrhenius Väg 20C, SE-106 91, Stockholm, Sweden
| | - Christian Zwicker
- Department of Biomedical Molecular Biology, Faculty of Sciences, Ghent University, Ghent, Belgium
- Laboratory of Myeloid Cell Biology in Tissue Damage and Inflammation, VIB-UGent Center for Inflammation Research, Technologiepark-Zwijnaarde 71, Ghent, 9052, Belgium
| | - Bavo Vanneste
- Department of Biomedical Molecular Biology, Faculty of Sciences, Ghent University, Ghent, Belgium
- Laboratory of Myeloid Cell Biology in Tissue Damage and Inflammation, VIB-UGent Center for Inflammation Research, Technologiepark-Zwijnaarde 71, Ghent, 9052, Belgium
- Laboratory of Myeloid Cell Biology in Tissue Homeostasis and Regeneration, VIB-UGent Center for Inflammation Research, Technologiepark-Zwijnaarde 71, Ghent, 9052, Belgium
| | - Noémi Van Hul
- Department of Cell and Molecular Biology, Karolinska Institutet Stockholm, SE-171 77, Solna, Sweden
| | - Elisa Semle
- Molecular Biosciences, the Wenner Gren Institute, Stockholm University, Svante Arrhenius Väg 20C, SE-106 91, Stockholm, Sweden
| | - Jaclyn Quin
- Molecular Biosciences, the Wenner Gren Institute, Stockholm University, Svante Arrhenius Väg 20C, SE-106 91, Stockholm, Sweden
| | - Tales Pascini
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 12735 Twinbrook Parkway, Rm 2E20A, Rockville, MD, 20852, USA
| | - Sami Saarenpää
- SciLifeLab, Department of Gene Technology, KTH Royal Institute of Technology, Tomtebodavägen 23a, SE-171 65, Solna, Sweden
| | - Mengxiao He
- SciLifeLab, Department of Gene Technology, KTH Royal Institute of Technology, Tomtebodavägen 23a, SE-171 65, Solna, Sweden
| | - Emma R Andersson
- Department of Cell and Molecular Biology, Karolinska Institutet Stockholm, SE-171 77, Solna, Sweden
| | - Charlotte L Scott
- Department of Biomedical Molecular Biology, Faculty of Sciences, Ghent University, Ghent, Belgium
- Laboratory of Myeloid Cell Biology in Tissue Damage and Inflammation, VIB-UGent Center for Inflammation Research, Technologiepark-Zwijnaarde 71, Ghent, 9052, Belgium
| | - Joel Vega-Rodriguez
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 12735 Twinbrook Parkway, Rm 2E20A, Rockville, MD, 20852, USA
| | - Joakim Lundeberg
- SciLifeLab, Department of Gene Technology, KTH Royal Institute of Technology, Tomtebodavägen 23a, SE-171 65, Solna, Sweden
| | - Johan Ankarklev
- Molecular Biosciences, the Wenner Gren Institute, Stockholm University, Svante Arrhenius Väg 20C, SE-106 91, Stockholm, Sweden.
| |
Collapse
|
15
|
Monedeiro F, Ehall B, Tiffner K, Eberl A, Svehlikova E, Prietl B, Pfeifer V, Senekowitsch J, Remm A, Rebane A, Magnes C, Pieber T, Sinner F, Birngruber T. Characterization of Inflammatory Mediators and Metabolome in Interstitial Fluid Collected with Dermal Open Flow Microperfusion before and at the End of Dupilumab Treatment in Atopic Dermatitis. J Proteome Res 2024; 23:3496-3514. [PMID: 38986055 PMCID: PMC11304394 DOI: 10.1021/acs.jproteome.4c00153] [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: 02/29/2024] [Revised: 06/07/2024] [Accepted: 06/20/2024] [Indexed: 07/12/2024]
Abstract
Dupilumab is a monoclonal antibody approved for the treatment of atopic dermatitis (AD); however, its effects on molecular, cellular, and immunological levels remain to be elucidated. In this study, blood and dermal interstitial fluid (ISF) from nonlesional (NL) and lesional (L) skin were collected from eight patients with moderate to severe AD, before (visit 2-v2) and at the end of a 16-week treatment with dupilumab (visit 10-v10). Clinical treatment effect was demonstrated by significantly decreased AD severity scores at the end of treatment. At v10 versus v2, the percentages of CD4+ interleukin-producing cells showed a decreasing trend in ISF L and NL, unbound IL-4 levels in plasma were increased, IL-5 levels in ISF L reduced, and levels of factors involved in anti-inflammatory pathways and re-epithelization increased. At v2, ISF L showed that AD lesions might have altered amino acid pathways and lipid signaling compared to ISF NL. At v10, ISF L exhibited raised levels of long- and very-long-chain fatty acids and lipids compared to v2. Furthermore, dupilumab administration caused reduced expression of miR-155-5p and miR-378a-3p in ISF L. In conclusion, results from the present study provided novel knowledge by linking local immune and metabolic alterations to AD pathogenesis and treatment response.
Collapse
Affiliation(s)
- Fernanda Monedeiro
- HEALTH
− Institute for Biomedical Research and Technologies, Joanneum Research Forschungsgesellschaft mbH, Neue Stiftingtalstraße 2, Graz 8010, Austria
| | - Barbara Ehall
- Division
of Endocrinology and Diabetology, Medical
University of Graz, Neue Stiftingtalstraße 6, Graz 8010, Austria
- BioTechMed, Mozartgasse
12, Graz 8010, Austria
| | - Katrin Tiffner
- HEALTH
− Institute for Biomedical Research and Technologies, Joanneum Research Forschungsgesellschaft mbH, Neue Stiftingtalstraße 2, Graz 8010, Austria
| | - Anita Eberl
- HEALTH
− Institute for Biomedical Research and Technologies, Joanneum Research Forschungsgesellschaft mbH, Neue Stiftingtalstraße 2, Graz 8010, Austria
| | - Eva Svehlikova
- Division
of Endocrinology and Diabetology, Medical
University of Graz, Neue Stiftingtalstraße 6, Graz 8010, Austria
| | - Barbara Prietl
- Division
of Endocrinology and Diabetology, Medical
University of Graz, Neue Stiftingtalstraße 6, Graz 8010, Austria
- Center
for Biomarker Research in Medicine (CBmed) GmbH, Stiftingtalstrasse 5, Graz 8010, Austria
| | - Verena Pfeifer
- Division
of Endocrinology and Diabetology, Medical
University of Graz, Neue Stiftingtalstraße 6, Graz 8010, Austria
- Center
for Biomarker Research in Medicine (CBmed) GmbH, Stiftingtalstrasse 5, Graz 8010, Austria
| | - Julia Senekowitsch
- Division
of Endocrinology and Diabetology, Medical
University of Graz, Neue Stiftingtalstraße 6, Graz 8010, Austria
| | - Anu Remm
- Institute
of Biomedicine and Translational Medicine, University of Tartu, Biomeedikum, Ravila 19, Tartu 50411, Estonia
| | - Ana Rebane
- Institute
of Biomedicine and Translational Medicine, University of Tartu, Biomeedikum, Ravila 19, Tartu 50411, Estonia
| | - Christoph Magnes
- HEALTH
− Institute for Biomedical Research and Technologies, Joanneum Research Forschungsgesellschaft mbH, Neue Stiftingtalstraße 2, Graz 8010, Austria
| | - Thomas Pieber
- HEALTH
− Institute for Biomedical Research and Technologies, Joanneum Research Forschungsgesellschaft mbH, Neue Stiftingtalstraße 2, Graz 8010, Austria
- Division
of Endocrinology and Diabetology, Medical
University of Graz, Neue Stiftingtalstraße 6, Graz 8010, Austria
- Center
for Biomarker Research in Medicine (CBmed) GmbH, Stiftingtalstrasse 5, Graz 8010, Austria
| | - Frank Sinner
- HEALTH
− Institute for Biomedical Research and Technologies, Joanneum Research Forschungsgesellschaft mbH, Neue Stiftingtalstraße 2, Graz 8010, Austria
- Division
of Endocrinology and Diabetology, Medical
University of Graz, Neue Stiftingtalstraße 6, Graz 8010, Austria
| | - Thomas Birngruber
- HEALTH
− Institute for Biomedical Research and Technologies, Joanneum Research Forschungsgesellschaft mbH, Neue Stiftingtalstraße 2, Graz 8010, Austria
| |
Collapse
|
16
|
Chen X, Peng R, Peng D, Liu D, Li R. Helicobacter pylori infection exacerbates metabolic dysfunction-associated steatotic liver disease through lipid metabolic pathways: a transcriptomic study. J Transl Med 2024; 22:701. [PMID: 39075482 PMCID: PMC11288106 DOI: 10.1186/s12967-024-05506-y] [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: 03/31/2024] [Accepted: 07/12/2024] [Indexed: 07/31/2024] Open
Abstract
BACKGROUND The relationship between Helicobacter pylori (H. pylori) infection and metabolic dysfunction-associated steatotic liver disease (MASLD) has attracted increased clinical attention. However, most of those current studies involve cross-sectional studies and meta-analyses, and experimental mechanistic exploration still needs to be improved. This study aimed to investigate the mechanisms by which H. pylori impacts MASLD. METHODS We established two H. pylori-infected (Cag A positive and Cag A negative) mouse models with 16 weeks of chow diet (CD) or high-fat diet (HFD) feeding. Body weight, liver triglyceride, blood glucose, serum biochemical parameters, inflammatory factors, and insulin resistance were measured, and histological analysis of liver tissues was performed. Mouse livers were subjected to transcriptome RNA sequencing analysis. RESULTS Although H. pylori infection could not significantly affect serum inflammatory factor levels and serum biochemical parameters in mice, serum insulin and homeostatic model assessment for insulin resistance levels increased in CD mode. In contrast, H. pylori Cag A + infection significantly aggravated hepatic pathological steatosis induced by HFD and elevated serum inflammatory factors and lipid metabolism parameters. Hepatic transcriptomic analysis in the CD groups revealed 767 differentially expressed genes (DEGs) in the H. pylori Cag A + infected group and 1473 DEGs in the H. pylori Cag A- infected group, and the "nonalcoholic fatty liver disease" pathway was significantly enriched in KEGG analysis. There were 578 DEGs in H. pylori Cag A + infection combined with the HFD feeding group and 820 DEGs in the H. pylori Cag A- infected group. DEGs in the HFD groups were significantly enriched in "fatty acid degradation" and "PPAR pathway." Exploring the effect of different Cag A statuses on mouse liver revealed that fatty acid binding protein 5 was differentially expressed in Cag A- H. pylori. DEG enrichment pathways were concentrated in the "PPAR pathway" and "fatty acid degradation." CONCLUSIONS Clinicians are expected to comprehend the impact of H. pylori on MASLD and better understand and manage MASLD. H. pylori infection may exacerbate the development of MASLD by regulating hepatic lipid metabolism, and the H. pylori virulence factor Cag A plays a vital role in this regulation.
Collapse
Affiliation(s)
- Xingcen Chen
- Department of Gastroenterology, The Second Xiangya Hospital of Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan Province, China
- Research Center of Digestive Diseases, Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan Province, China
- Clinical Research Center for Digestive Diseases in Hunan Province, Changsha, 410011, Hunan Province, China
| | - Ruyi Peng
- Department of Gastroenterology, The Second Xiangya Hospital of Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan Province, China
- Research Center of Digestive Diseases, Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan Province, China
- Clinical Research Center for Digestive Diseases in Hunan Province, Changsha, 410011, Hunan Province, China
| | - Dongzi Peng
- Department of Gastroenterology, The Second Xiangya Hospital of Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan Province, China
- Research Center of Digestive Diseases, Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan Province, China
- Clinical Research Center for Digestive Diseases in Hunan Province, Changsha, 410011, Hunan Province, China
| | - Deliang Liu
- Department of Gastroenterology, The Second Xiangya Hospital of Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan Province, China
- Research Center of Digestive Diseases, Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan Province, China
- Clinical Research Center for Digestive Diseases in Hunan Province, Changsha, 410011, Hunan Province, China
| | - Rong Li
- Department of Gastroenterology, The Second Xiangya Hospital of Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan Province, China.
- Research Center of Digestive Diseases, Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan Province, China.
- Clinical Research Center for Digestive Diseases in Hunan Province, Changsha, 410011, Hunan Province, China.
| |
Collapse
|
17
|
Ohguro H, Watanabe M, Hikage F, Sato T, Nishikiori N, Umetsu A, Higashide M, Ogawa T, Furuhashi M. Fatty Acid-Binding Protein 4-Mediated Regulation Is Pivotally Involved in Retinal Pathophysiology: A Review. Int J Mol Sci 2024; 25:7717. [PMID: 39062961 PMCID: PMC11277531 DOI: 10.3390/ijms25147717] [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: 06/01/2024] [Revised: 07/09/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
Fatty acid-binding proteins (FABPs), a family of lipid chaperone molecules that are involved in intracellular lipid transportation to specific cellular compartments, stimulate lipid-associated responses such as biological signaling, membrane synthesis, transcriptional regulation, and lipid synthesis. Previous studies have shown that FABP4, a member of this family of proteins that are expressed in adipocytes and macrophages, plays pivotal roles in the pathogenesis of various cardiovascular and metabolic diseases, including diabetes mellitus (DM) and hypertension (HT). Since significant increases in the serum levels of FABP4 were detected in those patients, FABP4 has been identified as a crucial biomarker for these systemic diseases. In addition, in the field of ophthalmology, our group found that intraocular levels of FABP4 (ioFABP4) and free fatty acids (ioFFA) were substantially elevated in patients with retinal vascular diseases (RVDs) including proliferative diabetic retinopathy (PDR) and retinal vein occlusion (RVO), for which DM and HT are also recognized as significant risk factors. Recent studies have also revealed that ioFABP4 plays important roles in both retinal physiology and pathogenesis, and the results of these studies have suggested potential molecular targets for retinal diseases that might lead to future new therapeutic strategies.
Collapse
Affiliation(s)
- Hiroshi Ohguro
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (M.W.); (F.H.); (N.N.); (A.U.); (M.H.)
| | - Megumi Watanabe
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (M.W.); (F.H.); (N.N.); (A.U.); (M.H.)
| | - Fumihito Hikage
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (M.W.); (F.H.); (N.N.); (A.U.); (M.H.)
| | - Tatsuya Sato
- Departments of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (T.S.); (T.O.)
- Departments of Cellular Physiology and Signal Transduction, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan
| | - Nami Nishikiori
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (M.W.); (F.H.); (N.N.); (A.U.); (M.H.)
| | - Araya Umetsu
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (M.W.); (F.H.); (N.N.); (A.U.); (M.H.)
| | - Megumi Higashide
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (M.W.); (F.H.); (N.N.); (A.U.); (M.H.)
| | - Toshifumi Ogawa
- Departments of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (T.S.); (T.O.)
- Departments of Cellular Physiology and Signal Transduction, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan
| | - Masato Furuhashi
- Departments of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (T.S.); (T.O.)
| |
Collapse
|
18
|
Zhou Y, Sanchez VB, Xu P, Roule T, Flores-Mendez M, Ciesielski B, Yoo D, Teshome H, Jimenez T, Liu S, Henne M, O’Brien T, He Y, Mesaros C, Akizu N. Altered lipid homeostasis is associated with cerebellar neurodegeneration in SNX14 deficiency. JCI Insight 2024; 9:e168594. [PMID: 38625743 PMCID: PMC11141923 DOI: 10.1172/jci.insight.168594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 04/05/2024] [Indexed: 04/17/2024] Open
Abstract
Dysregulated lipid homeostasis is emerging as a potential cause of neurodegenerative disorders. However, evidence of errors in lipid homeostasis as a pathogenic mechanism of neurodegeneration remains limited. Here, we show that cerebellar neurodegeneration caused by Sorting Nexin 14 (SNX14) deficiency is associated with lipid homeostasis defects. Recent studies indicate that SNX14 is an interorganelle lipid transfer protein that regulates lipid transport, lipid droplet (LD) biogenesis, and fatty acid desaturation, suggesting that human SNX14 deficiency belongs to an expanding class of cerebellar neurodegenerative disorders caused by altered cellular lipid homeostasis. To test this hypothesis, we generated a mouse model that recapitulates human SNX14 deficiency at a genetic and phenotypic level. We demonstrate that cerebellar Purkinje cells (PCs) are selectively vulnerable to SNX14 deficiency while forebrain regions preserve their neuronal content. Ultrastructure and lipidomic studies reveal widespread lipid storage and metabolism defects in SNX14-deficient mice. However, predegenerating SNX14-deficient cerebella show a unique accumulation of acylcarnitines and depletion of triglycerides. Furthermore, defects in LD content and telolysosome enlargement in predegenerating PCs suggest lipotoxicity as a pathogenic mechanism of SNX14 deficiency. Our work shows a selective cerebellar vulnerability to altered lipid homeostasis and provides a mouse model for future therapeutic studies.
Collapse
Affiliation(s)
- Yijing Zhou
- Perelman Center for Cellular and Molecular Therapeutics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pathology and Laboratory Medicine and
| | - Vanessa B. Sanchez
- Perelman Center for Cellular and Molecular Therapeutics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pathology and Laboratory Medicine and
| | - Peining Xu
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Thomas Roule
- Perelman Center for Cellular and Molecular Therapeutics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pathology and Laboratory Medicine and
| | - Marco Flores-Mendez
- Perelman Center for Cellular and Molecular Therapeutics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pathology and Laboratory Medicine and
| | - Brianna Ciesielski
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Donna Yoo
- Perelman Center for Cellular and Molecular Therapeutics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pathology and Laboratory Medicine and
| | - Hiab Teshome
- Perelman Center for Cellular and Molecular Therapeutics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pathology and Laboratory Medicine and
| | - Teresa Jimenez
- Perelman Center for Cellular and Molecular Therapeutics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pathology and Laboratory Medicine and
| | - Shibo Liu
- The Graduate Center of the City University of New York, Advanced Science Research Center, New York, New York, USA
| | - Mike Henne
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Tim O’Brien
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ye He
- The Graduate Center of the City University of New York, Advanced Science Research Center, New York, New York, USA
- Ph.D. Program in Biology, The Graduate Center of the City University of New York, New York, New York, USA
| | - Clementina Mesaros
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Naiara Akizu
- Perelman Center for Cellular and Molecular Therapeutics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pathology and Laboratory Medicine and
| |
Collapse
|
19
|
Iori S, D'Onofrio C, Laham-Karam N, Mushimiyimana I, Lucatello L, Lopparelli RM, Gelain ME, Capolongo F, Pauletto M, Dacasto M, Giantin M. Establishment and characterization of cytochrome P450 1A1 CRISPR/Cas9 Knockout Bovine Foetal Hepatocyte Cell Line (BFH12). Cell Biol Toxicol 2024; 40:18. [PMID: 38528259 PMCID: PMC10963470 DOI: 10.1007/s10565-024-09856-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 03/21/2024] [Indexed: 03/27/2024]
Abstract
The cytochrome P450 1A (CYP1A) subfamily of xenobiotic metabolizing enzymes (XMEs) consists of two different isoforms, namely CYP1A1 and CYP1A2, which are highly conserved among species. These two isoenzymes are involved in the biotransformation of many endogenous compounds as well as in the bioactivation of several xenobiotics into carcinogenic derivatives, thereby increasing the risk of tumour development. Cattle (Bos taurus) are one of the most important food-producing animal species, being a significant source of nutrition worldwide. Despite daily exposure to xenobiotics, data on the contribution of CYP1A to bovine hepatic metabolism are still scarce. The CRISPR/Cas9-mediated knockout (KO) is a useful method for generating in vivo and in vitro models for studying xenobiotic biotransformations. In this study, we applied the ribonucleoprotein (RNP)-complex approach to successfully obtain the KO of CYP1A1 in a bovine foetal hepatocyte cell line (BFH12). After clonal expansion and selection, CYP1A1 excision was confirmed at the DNA, mRNA and protein level. Therefore, RNA-seq analysis revealed significant transcriptomic changes associated with cell cycle regulation, proliferation, and detoxification processes as well as on iron, lipid and mitochondrial homeostasis. Altogether, this study successfully generates a new bovine CYP1A1 KO in vitro model, representing a valuable resource for xenobiotic metabolism studies in this important farm animal species.
Collapse
Affiliation(s)
- Silvia Iori
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale Dell'Università 16, Legnaro, 35020, Padua, Italy
| | - Caterina D'Onofrio
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale Dell'Università 16, Legnaro, 35020, Padua, Italy
| | - Nihay Laham-Karam
- University of Eastern Finland, A.I. Virtanen Institute for Molecular Sciences, Neulaniementie 2, 70211, Kuopio, Finland
| | - Isidore Mushimiyimana
- University of Eastern Finland, A.I. Virtanen Institute for Molecular Sciences, Neulaniementie 2, 70211, Kuopio, Finland
| | - Lorena Lucatello
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale Dell'Università 16, Legnaro, 35020, Padua, Italy
| | - Rosa Maria Lopparelli
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale Dell'Università 16, Legnaro, 35020, Padua, Italy
| | - Maria Elena Gelain
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale Dell'Università 16, Legnaro, 35020, Padua, Italy
| | - Francesca Capolongo
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale Dell'Università 16, Legnaro, 35020, Padua, Italy
| | - Marianna Pauletto
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale Dell'Università 16, Legnaro, 35020, Padua, Italy
| | - Mauro Dacasto
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale Dell'Università 16, Legnaro, 35020, Padua, Italy
| | - Mery Giantin
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale Dell'Università 16, Legnaro, 35020, Padua, Italy.
| |
Collapse
|
20
|
Wu X, Pan X, Zhou Y, Pan J, Kang J, Yu JJJ, Cao Y, Quan C, Gong L, Li Y. Identification of key genes for atherosclerosis in different arterial beds. Sci Rep 2024; 14:6543. [PMID: 38503760 PMCID: PMC10951242 DOI: 10.1038/s41598-024-55575-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 02/25/2024] [Indexed: 03/21/2024] Open
Abstract
Atherosclerosis (AS) is the pathologic basis of various cardiovascular and cerebrovascular events, with a high degree of heterogeneity among different arterial beds. However, mechanistic differences between arterial beds remain unexplored. The aim of this study was to explore key genes and potential mechanistic differences between AS in different arterial beds through bioinformatics analysis. Carotid atherosclerosis (CAS), femoral atherosclerosis (FAS), infrapopliteal atherosclerosis (IPAS), abdominal aortic atherosclerosis (AAS), and AS-specific differentially expressed genes (DEGs) were screened from the GSE100927 and GSE57691 datasets. Immune infiltration analysis was used to identify AS immune cell infiltration differences. Unsupervised cluster analysis of AS samples from different regions based on macrophage polarization gene expression profiles. Weighted gene co-expression network analysis (WGCNA) was performed to identify the most relevant module genes with AS. Hub genes were then screened by LASSO regression, SVM-REF, and single-gene differential analysis, and a nomogram was constructed to predict the risk of AS development. The results showed that differential expression analysis identified 5, 4, 121, and 62 CAS, FAS, IPAS, AAS-specific DEGs, and 42 AS-common DEGs, respectively. Immune infiltration analysis demonstrated that the degree of macrophage and mast cell enrichment differed significantly in different regions of AS. The CAS, FAS, IPAS, and AAS could be distinguished into two different biologically functional and stable molecular clusters based on macrophage polarization gene expression profiles, especially for cardiomyopathy and glycolipid metabolic processes. Hub genes for 6 AS (ADAP2, CSF3R, FABP5, ITGAX, MYOC, and SPP1), 4 IPAS (CLECL1, DIO2, F2RL2, and GUCY1A2), and 3 AAS (RPL21, RPL26, and RPL10A) were obtained based on module gene, gender stratification, machine learning algorithms, and single-gene difference analysis, respectively, and these genes were effective in differentiating between different regions of AS. This study demonstrates that there are similarities and heterogeneities in the pathogenesis of AS between different arterial beds.
Collapse
Affiliation(s)
- Xize Wu
- Nantong Hospital of Traditional Chinese Medicine, Nantong Hospital Affiliated to Nanjing University of Chinese Medicine, No. 41 Jianshe Road, Chongchuan District, Nantong, 226000, Jiangsu, China
- Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, Liaoning, China
| | - Xue Pan
- Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, Liaoning, China
- Dazhou Vocational College of Chinese Medicine, Dazhou, 635000, Sichuan, China
| | - Yi Zhou
- Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, Liaoning, China
| | - Jiaxiang Pan
- The Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, No. 33, Beiling Street, Huanggu District, Shenyang, 110032, Liaoning, China
| | - Jian Kang
- Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, Liaoning, China
| | - J J Jiajia Yu
- Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, Liaoning, China
| | - Yingyue Cao
- Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, Liaoning, China
| | - Chao Quan
- Nantong Hospital of Traditional Chinese Medicine, Nantong Hospital Affiliated to Nanjing University of Chinese Medicine, No. 41 Jianshe Road, Chongchuan District, Nantong, 226000, Jiangsu, China.
| | - Lihong Gong
- Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, Liaoning, China.
- The Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, No. 33, Beiling Street, Huanggu District, Shenyang, 110032, Liaoning, China.
- Liaoning Provincial Key Laboratory of TCM Geriatric Cardio-Cerebrovascular Diseases, Shenyang, 110847, Liaoning, China.
| | - Yue Li
- The Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, No. 33, Beiling Street, Huanggu District, Shenyang, 110032, Liaoning, China.
- Liaoning Provincial Key Laboratory of TCM Geriatric Cardio-Cerebrovascular Diseases, Shenyang, 110847, Liaoning, China.
| |
Collapse
|
21
|
Li Y, Lee W, Zhao ZG, Liu Y, Cui H, Wang HY. Fatty acid binding protein 5 is a novel therapeutic target for hepatocellular carcinoma. World J Clin Oncol 2024; 15:130-144. [PMID: 38292656 PMCID: PMC10823939 DOI: 10.5306/wjco.v15.i1.130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 12/02/2023] [Accepted: 12/25/2023] [Indexed: 01/23/2024] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is an aggressive subtype of liver cancer and is one of the most common cancers with high mortality worldwide. Reprogrammed lipid metabolism plays crucial roles in HCC cancer cell survival, growth, and evolution. Emerging evidence suggests the importance of fatty acid binding proteins (FABPs) in contribution to cancer progression and metastasis; however, how these FABPs are dysregulated in cancer cells, especially in HCC, and the roles of FABPs in cancer progression have not been well defined. AIM To understand the genetic alterations and expression of FABPs and their associated cancer hallmarks and oncogenes in contributing to cancer malignancies. METHODS We used The Cancer Genome Atlas datasets of pan cancer and liver hepatocellular carcinoma (LIHC) as well as patient cohorts with other cancer types in this study. We investigated genetic alterations of FABPs in various cancer types. mRNA expression was used to determine if FABPs are abnormally expressed in tumor tissues compared to non-tumor controls and to investigate whether their expression correlates with patient clinical outcome, enriched cancer hallmarks and oncogenes previously reported for patients with HCC. We determined the protein levels of FABP5 and its correlated genes in two HCC cell lines and assessed the potential of FABP5 inhibition in treating HCC cells. RESULTS We discovered that a gene cluster including five FABP family members (FABP4, FABP5, FABP8, FABP9 and FABP12) is frequently co-amplified in cancer. Amplification, in fact, is the most common genetic alteration for FABPs, leading to overexpression of FABPs. FABP5 showed the greatest differential mRNA expression comparing tumor with non-tumor tissues. High FABP5 expression correlates well with worse patient outcomes (P < 0.05). FABP5 expression highly correlates with enrichment of G2M checkpoint (r = 0.33, P = 1.1e-10), TP53 signaling pathway (r = 0.22, P = 1.7e-5) and many genes in the gene sets such as CDK1 (r = 0.56, P = 0), CDK4 (r = 0.49, P = 0), and TP53 (r = 0.22, P = 1.6e-5). Furthermore, FABP5 also correlates well with two co-expressed oncogenes PLK1 and BIRC5 in pan cancer especially in LIHC patients (r = 0.58, P = 0; r = 0.58, P = 0; respectively). FABP5high Huh7 cells also expressed higher protein levels of p53, BIRC5, CDK1, CDK2, and CDK4 than FABP5low HepG2 cells. FABP5 inhibition more potently inhibited the tumor cell growth in Huh7 cells than in HepG2 cells. CONCLUSION We discovered that FABP5 gene is frequently amplified in cancer, especially in HCC, leading to its significant elevated expression in HCC. Its high expression correlates well with worse patient outcome, enriched cancer hallmarks and oncogenes in HCC. FABP5 inhibition impaired the cell viability of FABP5high Huh7 cells. All these support that FABP5 is a novel therapeutic target for treating FABP5high HCC.
Collapse
Affiliation(s)
- Yan Li
- Department of Gastroenterology, Tianjin Third Central Hospital, Tianjin 300170, China
| | - William Lee
- Biomedical Engineering, Texas A&M University, College Station, TX 77843, United States
| | - Zhen-Gang Zhao
- Department of Gastroenterology, Tianjin Third Central Hospital, Tianjin 300170, China
| | - Yi Liu
- Department of Gastroenterology, Tianjin Third Central Hospital, Tianjin 300170, China
| | - Hao Cui
- Department of Gastroenterology, Tianjin Third Central Hospital, Tianjin 300170, China
| | - Hao-Yu Wang
- Department of Gastroenterology, Tianjin Third Central Hospital, Tianjin 300170, China
| |
Collapse
|
22
|
Wu X, Yang SY, Zhang YH, Fang JZ, Wang S, Xu ZW, Zhang XJ. Prognostic and immunological roles of heat shock protein A4 in lung adenocarcinoma. World J Clin Oncol 2024; 15:45-61. [PMID: 38292659 PMCID: PMC10823936 DOI: 10.5306/wjco.v15.i1.45] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 12/03/2023] [Accepted: 12/29/2023] [Indexed: 01/23/2024] Open
Abstract
BACKGROUND Heat shock protein A4 (HSPA4) belongs to molecular chaperone protein family which plays important roles within variable cellular activities, including cancer initiation and progression. However, the prognostic and immunological significance of HSPA4 in lung adenocarcinoma (LUAD) has not been revealed yet. AIM To explore the prognostic and immunological roles of HSPA4 to identify a novel prognostic biomarker and therapeutic target for LUAD. METHODS We assessed the prognostic and immunological significance of HSPA4 in LUAD using data from The Cancer Genome Atlas database. The association between HSPA4 expression and clinical-pathological features was assessed through Kruskal-Wallis and Wilcoxon signed-rank test. Univariate/multivariate Cox regression analyses and Kaplan-Meier curves were employed to evaluate prognostic factors, including HSPA4, in LUAD. Gene set enrichment analysis (GSEA) was conducted to identify the key signaling pathways associated with HSPA4. The correlation between HSPA4 expression and cancer immune infiltration was evaluated using single-sample gene set enrichment analysis (ssGSEA). RESULTS Overexpressing HSPA4 was significantly related to advanced pathologic TNM stage, advanced pathologic stage, progression disease status of primary therapy outcome and female subgroups with LUAD. In addition, increased HSPA4 expression was found to be related to worse disease-specific survival and overall survival. GSEA analysis indicated a significant correlation between HSPA4 and cell cycle regulation and immune response, particularly through diminishing the function of cytotoxicity cells and CD8 T cells. The ssGSEA algorithm showed a positive correlation between HSPA4 expression and infiltrating levels of Th2 cells, while a negative correlation was observed with cytotoxic cell infiltration levels. CONCLUSION Our findings indicate HSPA4 is related to prognosis and immune cell infiltrates and may act as a novel prognostic biomarker and therapeutic target for LUAD.
Collapse
Affiliation(s)
- Xuan Wu
- Department of Respiratory and Critical Care Medicine, Zhengzhou University People’s Hospital, Zhengzhou 450008, Henan Province, China
| | - Shen-Ying Yang
- Department of Respiratory and Critical Care Medicine, Zhengzhou University People’s Hospital, Zhengzhou 450008, Henan Province, China
| | - Yi-Hua Zhang
- Graduate School, Xinxiang Medical University, Xinxiang 453003, Henan Province, China
| | - Jin-Zhou Fang
- Department of Respiratory and Critical Care Medicine, Zhengzhou University People’s Hospital, Zhengzhou 450008, Henan Province, China
| | - Shuai Wang
- Department of Respiratory and Critical Care Medicine, Zhengzhou University People’s Hospital, Zhengzhou 450008, Henan Province, China
| | - Zhi-Wei Xu
- Department of Respiratory and Critical Care Medicine, Zhengzhou University People’s Hospital, Zhengzhou 450008, Henan Province, China
| | - Xiao-Ju Zhang
- Department of Pulmonary and Critical Care Medicine, Zhengzhou University People’s Hospital, Zhengzhou 450008, Henan Province, China
| |
Collapse
|
23
|
Sullivan MR, White RP, Dashnamoorthy Ravi, Kanetkar N, Fridman IB, Ekenseair A, Evens AM, Konry T. Characterizing influence of rCHOP treatment on diffuse large B-cell lymphoma microenvironment through in vitro microfluidic spheroid model. Cell Death Dis 2024; 15:18. [PMID: 38195589 PMCID: PMC10776622 DOI: 10.1038/s41419-023-06299-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 10/11/2023] [Accepted: 11/13/2023] [Indexed: 01/11/2024]
Abstract
For over two decades, Rituximab and CHOP combination treatment (rCHOP) has remained the standard treatment approach for diffuse large B-cell lymphoma (DLBCL). Despite numerous clinical trials exploring treatment alternatives, few options have shown any promise at further improving patient survival and recovery rates. A wave of new therapeutic approaches have recently been in development with the rise of immunotherapy for cancer, however, the cost of clinical trials is prohibitive of testing all promising approaches. Improved methods of early drug screening are essential for expediting the development of the therapeutic approaches most likely to help patients. Microfluidic devices provide a powerful tool for drug testing with enhanced biological relevance, along with multi-parameter data outputs. Here, we describe a hydrogel spheroid-based microfluidic model for screening lymphoma treatments. We utilized primary patient DLBCL cells in combination with NK cells and rCHOP treatment to determine the biological relevance of this approach. We observed cellular viability in response to treatment, rheological properties, and cell surface marker expression levels correlated well with expected in vivo characteristics. In addition, we explored secretory and transcriptomic changes in response to treatment. Our results showed complex changes in phenotype and transcriptomic response to treatment stimuli, including numerous metabolic and immunogenic changes. These findings support this model as an optimal platform for the comparative screening of novel treatments.
Collapse
Affiliation(s)
- Matthew R Sullivan
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
| | - Rachel P White
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
| | | | - Ninad Kanetkar
- Chemical Engineering Department, Northeastern University, Boston, MA, USA
| | - Ilana Berger Fridman
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
- Avram and Stella Goldstein-Goren Department of Biotechnology and Regenerative Medicine and Stem Cell Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Adam Ekenseair
- Chemical Engineering Department, Northeastern University, Boston, MA, USA
| | | | - Tania Konry
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA.
| |
Collapse
|
24
|
López-Alcalá J, Gordon A, Trávez A, Tercero-Alcázar C, Correa-Sáez A, González-Rellán MJ, Rangel-Zúñiga OA, Rodríguez A, Membrives A, Frühbeck G, Nogueiras R, Calzado MA, Guzmán-Ruiz R, Malagón MM. Localization, traffic and function of Rab34 in adipocyte lipid and endocrine functions. J Biomed Sci 2024; 31:2. [PMID: 38183057 PMCID: PMC10770960 DOI: 10.1186/s12929-023-00990-8] [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: 03/21/2023] [Accepted: 12/20/2023] [Indexed: 01/07/2024] Open
Abstract
BACKGROUND Excessive lipid accumulation in the adipose tissue in obesity alters the endocrine and energy storage functions of adipocytes. Adipocyte lipid droplets represent key organelles coordinating lipid storage and mobilization in these cells. Recently, we identified the small GTPase, Rab34, in the lipid droplet proteome of adipocytes. Herein, we have characterized the distribution, intracellular transport, and potential contribution of this GTPase to adipocyte physiology and its regulation in obesity. METHODS 3T3-L1 and human primary preadipocytes were differentiated in vitro and Rab34 distribution and trafficking were analyzed using markers of cellular compartments. 3T3-L1 adipocytes were transfected with expression vectors and/or Rab34 siRNA and assessed for secretory activity, lipid accumulation and expression of proteins regulating lipid metabolism. Proteomic and protein interaction analyses were employed for the identification of the Rab34 interactome. These studies were combined with functional analysis to unveil the role played by the GTPase in adipocytes, with a focus on the actions conveyed by Rab34 interacting proteins. Finally, Rab34 regulation in response to obesity was also evaluated. RESULTS Our results show that Rab34 localizes at the Golgi apparatus in preadipocytes. During lipid droplet biogenesis, Rab34 translocates from the Golgi to endoplasmic reticulum-related compartments and then reaches the surface of adipocyte lipid droplets. Rab34 exerts distinct functions related to its intracellular location. Thus, at the Golgi, Rab34 regulates cisternae integrity as well as adiponectin trafficking and oligomerization. At the lipid droplets, this GTPase controls lipid accumulation and lipolysis through its interaction with the E1-ubiquitin ligase, UBA1, which induces the ubiquitination and proteasomal degradation of the fatty acid transporter and member of Rab34 interactome, FABP5. Finally, Rab34 levels in the adipose tissue and adipocytes are regulated in response to obesity and related pathogenic insults (i.e., fibrosis). CONCLUSIONS Rab34 plays relevant roles during adipocyte differentiation, including from the regulation of the oligomerization (i.e., biological activity) and secretion of a major adipokine with insulin-sensitizing actions, adiponectin, to lipid storage and mobilization from lipid droplets. Rab34 dysregulation in obesity may contribute to the altered adipokine secretion and lipid metabolism that characterize adipocyte dysfunction in conditions of excess adiposity.
Collapse
Affiliation(s)
- Jaime López-Alcalá
- Department of Cell Biology, Physiology, and Immunology, Maimonides Institute for Biomedical Research of Córdoba (IMIBIC), University of Córdoba (UCO), Reina Sofía University Hospital (HURS), Córdoba, Spain
| | - Ana Gordon
- Department of Cell Biology, Physiology, and Immunology, Maimonides Institute for Biomedical Research of Córdoba (IMIBIC), University of Córdoba (UCO), Reina Sofía University Hospital (HURS), Córdoba, Spain.
| | - Andrés Trávez
- Department of Cell Biology, Physiology, and Immunology, Maimonides Institute for Biomedical Research of Córdoba (IMIBIC), University of Córdoba (UCO), Reina Sofía University Hospital (HURS), Córdoba, Spain
| | - Carmen Tercero-Alcázar
- Department of Cell Biology, Physiology, and Immunology, Maimonides Institute for Biomedical Research of Córdoba (IMIBIC), University of Córdoba (UCO), Reina Sofía University Hospital (HURS), Córdoba, Spain
| | - Alejandro Correa-Sáez
- Department of Cell Biology, Physiology, and Immunology, Maimonides Institute for Biomedical Research of Córdoba (IMIBIC), University of Córdoba (UCO), Reina Sofía University Hospital (HURS), Córdoba, Spain
| | - María Jesús González-Rellán
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), ISCIII, Madrid, Spain
- Department of Physiology, CiMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
| | - Oriol A Rangel-Zúñiga
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), ISCIII, Madrid, Spain
- Lipids and Atherosclerosis Unit, IMIBIC/University of Córdoba (UCO), Reina Sofía University Hospital (HURS), Córdoba, Spain
| | - Amaia Rodríguez
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), ISCIII, Madrid, Spain
- Metabolic Research Laboratory, Department of Endocrinology & Nutrition, Clinic, University of Navarra, IdiSNA, Pamplona, Spain
| | - Antonio Membrives
- Department of Medical-Surgical Specialties, University of Córdoba (UCO), Reina Sofia University Hospital (HURS), Córdoba, Spain
| | - Gema Frühbeck
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), ISCIII, Madrid, Spain
- Metabolic Research Laboratory, Department of Endocrinology & Nutrition, Clinic, University of Navarra, IdiSNA, Pamplona, Spain
| | - Rubén Nogueiras
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), ISCIII, Madrid, Spain
- Department of Physiology, CiMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
| | - Marco A Calzado
- Department of Cell Biology, Physiology, and Immunology, Maimonides Institute for Biomedical Research of Córdoba (IMIBIC), University of Córdoba (UCO), Reina Sofía University Hospital (HURS), Córdoba, Spain
| | - Rocío Guzmán-Ruiz
- Department of Cell Biology, Physiology, and Immunology, Maimonides Institute for Biomedical Research of Córdoba (IMIBIC), University of Córdoba (UCO), Reina Sofía University Hospital (HURS), Córdoba, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), ISCIII, Madrid, Spain
| | - María M Malagón
- Department of Cell Biology, Physiology, and Immunology, Maimonides Institute for Biomedical Research of Córdoba (IMIBIC), University of Córdoba (UCO), Reina Sofía University Hospital (HURS), Córdoba, Spain.
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), ISCIII, Madrid, Spain.
| |
Collapse
|
25
|
Zheng X, Zhou B, Li Y, Zhong H, Huang Z, Gu M. Transcriptome-wide N 6-methyladenosine methylation profile of atherosclerosis in mice. BMC Genomics 2023; 24:774. [PMID: 38097926 PMCID: PMC10720251 DOI: 10.1186/s12864-023-09878-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 12/06/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Atherosclerosis (AS) is a critical pathological event during the progression of cardiovascular diseases. It exhibits fibrofatty lesions on the arterial wall and lacks effective treatment. N6-methyladenosine (m6A) is the most common modification of eukaryotic RNA and plays an important role in regulating the development and progression of cardiovascular diseases. However, the role of m6A modification in AS remains largely unknown. Therefore, in this study, we explored the transcriptome distribution of m6A modification in AS and its potential mechanism. METHODS Methylation Quantification Kit was used to detect the global m6A levels in the aorta of AS mice. Western blot was used to analyze the protein level of methyltransferases. Methylated RNA immunoprecipitation with next-generation sequencing (MeRIP-seq) and RNA sequencing (RNA-seq) were used to obtain the first transcriptome range analysis of the m6A methylene map in the aorta of AS mice, followed by bioinformatics analysis. qRT-PCR and MeRIP-qRT-PCR were used to measure the mRNA and m6A levels in target genes. RESULTS The global m6A and protein levels of methyltransferase METTL3 were significantly increased in the aorta of AS mice. However, the protein level of demethylase ALKBH5 was significantly decreased. Through MeRIP-seq, we obtained m6A methylation maps in AS and control mice. In total, 26,918 m6A peaks associated with 13,744 genes were detected in AS group, whereas 26,157 m6A peaks associated with 13,283 genes were detected in the control group. Peaks mainly appeared in the coding sequence (CDS) regions close to the stop codon with the RRACH motif. Moreover, functional enrichment analysis demonstrated that m6A-containing genes were significantly enriched in AS-relevant pathways. Interestingly, a negative correlation between m6A methylation abundance and gene expression level was found through the integrated analysis of MeRIP-seq and RNA-seq data. Among the m6A-modified genes, a hypo-methylated but up-regulated (hypo-up) gene Fabp5 may be a potential biomarker of AS. CONCLUSIONS Our study provides transcriptome-wide m6A methylation for the first time to determine the association between m6A modification and AS progression. Our study lays a foundation for further exploring the pathogenesis of AS and provides a new direction for the treatment of AS.
Collapse
Affiliation(s)
- Xinbin Zheng
- Clinical Research Center, Hainan Provincial Hospital of Traditional Chinese Medicine, 570203, Haikou, Hainan, P. R. China
- Hainan Clinical Research Center for Preventive Treatment of Diseases, 570203, Haikou, Hainan, P. R. China
- Hospital of Chinese Medicine affiliated by Hainan Medical University, 570203, Haikou, Hainan, P. R. China
| | - Bo Zhou
- Department of Cardiology, Hainan Provincial Hospital of Traditional Chinese Medicine, 570203, Haikou, Hainan, P. R. China
- Hospital of Chinese Medicine affiliated by Hainan Medical University, 570203, Haikou, Hainan, P. R. China
| | - Yuzhen Li
- Clinical Research Center, Hainan Provincial Hospital of Traditional Chinese Medicine, 570203, Haikou, Hainan, P. R. China
- Hospital of Chinese Medicine affiliated by Hainan Medical University, 570203, Haikou, Hainan, P. R. China
| | - Hengren Zhong
- Clinical Research Center, Hainan Provincial Hospital of Traditional Chinese Medicine, 570203, Haikou, Hainan, P. R. China
- Hospital of Chinese Medicine affiliated by Hainan Medical University, 570203, Haikou, Hainan, P. R. China
| | - Zhengxin Huang
- Department of Cardiology, Hainan Provincial Hospital of Traditional Chinese Medicine, 570203, Haikou, Hainan, P. R. China.
- Hospital of Chinese Medicine affiliated by Hainan Medical University, 570203, Haikou, Hainan, P. R. China.
| | - Minhua Gu
- Department of Cardiology, Hainan Provincial Hospital of Traditional Chinese Medicine, 570203, Haikou, Hainan, P. R. China.
- Hospital of Chinese Medicine affiliated by Hainan Medical University, 570203, Haikou, Hainan, P. R. China.
| |
Collapse
|
26
|
Gaffar S, Aathirah AS. Fatty-Acid-Binding Proteins: From Lipid Transporters to Disease Biomarkers. Biomolecules 2023; 13:1753. [PMID: 38136624 PMCID: PMC10741572 DOI: 10.3390/biom13121753] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 10/20/2023] [Accepted: 10/26/2023] [Indexed: 12/24/2023] Open
Abstract
Fatty-acid-binding proteins (FABPs) serve a crucial role in the metabolism and transport of fatty acids and other hydrophobic ligands as an intracellular protein family. They are also recognized as a critical mediator in the inflammatory and ischemic pathways. FABPs are found in a wide range of tissues and organs, allowing them to contribute to various disease/injury developments that have not been widely discussed. We have collected and analyzed research journals that have investigated the role of FABPs in various diseases. Through this review, we discuss the findings on the potential of FABPs as biomarkers for various diseases in different tissues and organs, looking at their expression levels and their roles in related diseases according to available literature data. FABPs have been reported to show significantly increased expression levels in various tissues and organs associated with metabolic and inflammatory diseases. Therefore, FABPs are a promising novel biomarker that needs further development to optimize disease diagnosis and prognosis methods along with previously discovered markers.
Collapse
Affiliation(s)
- Shabarni Gaffar
- Graduate School, Padjadjaran University, Bandung 40132, Indonesia;
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Padjadjaran University, Sumedang 45363, Indonesia
| | | |
Collapse
|
27
|
Kawahata I, Fukunaga K. Pathogenic Impact of Fatty Acid-Binding Proteins in Parkinson's Disease-Potential Biomarkers and Therapeutic Targets. Int J Mol Sci 2023; 24:17037. [PMID: 38069360 PMCID: PMC10707307 DOI: 10.3390/ijms242317037] [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] [Received: 10/30/2023] [Revised: 11/26/2023] [Accepted: 11/30/2023] [Indexed: 12/18/2023] Open
Abstract
Parkinson's disease is a neurodegenerative condition characterized by motor dysfunction resulting from the degeneration of dopamine-producing neurons in the midbrain. This dopamine deficiency gives rise to a spectrum of movement-related symptoms, including tremors, rigidity, and bradykinesia. While the precise etiology of Parkinson's disease remains elusive, genetic mutations, protein aggregation, inflammatory processes, and oxidative stress are believed to contribute to its development. In this context, fatty acid-binding proteins (FABPs) in the central nervous system, FABP3, FABP5, and FABP7, impact α-synuclein aggregation, neurotoxicity, and neuroinflammation. These FABPs accumulate in mitochondria during neurodegeneration, disrupting their membrane potential and homeostasis. In particular, FABP3, abundant in nigrostriatal dopaminergic neurons, is responsible for α-synuclein propagation into neurons and intracellular accumulation, affecting the loss of mesencephalic tyrosine hydroxylase protein, a rate-limiting enzyme of dopamine biosynthesis. This review summarizes the characteristics of FABP family proteins and delves into the pathogenic significance of FABPs in the pathogenesis of Parkinson's disease. Furthermore, it examines potential novel therapeutic targets and early diagnostic biomarkers for Parkinson's disease and related neurodegenerative disorders.
Collapse
Affiliation(s)
- Ichiro Kawahata
- Department of CNS Drug Innovation, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan;
| | - Kohji Fukunaga
- Department of CNS Drug Innovation, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan;
- BRI Pharma Inc., Sendai 982-0804, Japan
| |
Collapse
|
28
|
Glaser ST, Jayanetti K, Oubraim S, Hillowe A, Frank E, Jong J, Wang L, Wang H, Ojima I, Haj-Dahmane S, Kaczocha M. Fatty acid binding proteins are novel modulators of synaptic epoxyeicosatrienoic acid signaling in the brain. Sci Rep 2023; 13:15234. [PMID: 37709856 PMCID: PMC10502087 DOI: 10.1038/s41598-023-42504-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 09/11/2023] [Indexed: 09/16/2023] Open
Abstract
Fatty acid binding proteins (FABPs) govern intracellular lipid transport to cytosolic organelles and nuclear receptors. More recently, FABP5 has emerged as a key regulator of synaptic endocannabinoid signaling, suggesting that FABPs may broadly regulate the signaling of neuroactive lipids in the brain. Herein, we demonstrate that brain-expressed FABPs (FABP3, FABP5, and FABP7) interact with epoxyeicosatrienoic acids (EETs) and the peroxisome proliferator-activated receptor gamma agonist 15-deoxy-Δ12,14-Prostaglandin J2 (15d-PGJ2). Among these lipids, EETs displayed highest affinities for FABP3 and FABP5, and 11,12-EET was identified as the preferred FABP ligand. Similarly, 15d-PGJ2 interacted with FABP3 and FABP5 while binding to FABP7 was markedly lower. Molecular modeling revealed unique binding interactions of the ligands within the FABP binding pockets and highlighted major contributions of van der Waals clashes and acyl chain solvent exposure in dictating FABP affinity and specificity. Functional studies demonstrated that endogenous EETs gate the strength of CA1 hippocampal glutamate synapses and that this function was impaired following FABP inhibition. As such, the present study reveals that FABPs control EET-mediated synaptic gating, thereby expanding the functional roles of this protein family in regulating neuronal lipid signaling.
Collapse
Affiliation(s)
- Sherrye T Glaser
- Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
- Department of Biological Sciences, Kingsborough Community College, Brooklyn, NY, USA
| | - Kalani Jayanetti
- Department of Chemistry, Stony Brook University, Stony Brook, NY, USA
| | - Saida Oubraim
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Andrew Hillowe
- Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Elena Frank
- Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Jason Jong
- Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Liqun Wang
- Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Hehe Wang
- Department of Chemistry, Stony Brook University, Stony Brook, NY, USA
| | - Iwao Ojima
- Department of Chemistry, Stony Brook University, Stony Brook, NY, USA
- Institute of Chemical Biology and Drug Discovery, Stony Brook University, Stony Brook, NY, USA
| | - Samir Haj-Dahmane
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA.
| | - Martin Kaczocha
- Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA.
- Institute of Chemical Biology and Drug Discovery, Stony Brook University, Stony Brook, NY, USA.
- Stony Brook University Pain and Analgesia Research Center (SPARC), Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA.
| |
Collapse
|
29
|
Doswell F, Haley JD, Kaczocha M. Proteomic analysis of signaling pathways modulated by FABP5 in macrophages. RESEARCH SQUARE 2023:rs.3.rs-3332029. [PMID: 37790380 PMCID: PMC10543284 DOI: 10.21203/rs.3.rs-3332029/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Background While acute inflammation serves essential functions in maintaining tissue homeostasis, chronic inflammation is causally linked to many diseases. Macrophages are a major cell-type that orchestrates inflammatory processes. During inflammation, macrophages undergo polarization and activation, thereby mobilizing pro-inflammatory and anti-inflammatory transcriptional programs that regulate ensuing macrophage functions. Fatty acid binding protein 5 (FABP5) is a lipid chaperone that is highly expressed in macrophages. FABP5 deletion is implicated in driving macrophages towards an anti-inflammatory phenotype, yet the signaling pathways regulated by macrophage FABP5 have not been systematically profiled. Herein, we leveraged proteomic and phosphoproteomic approaches to characterize pathways modulated by FABP5 in M1 and M2 polarized bone marrow derived macrophages (BMDMs). Results Stable isotope labeling by amino acids (SILAC) based analysis of M1 and M2 polarized wild-type (WT) and FABP5 knockout (KO) BMDMs revealed numerous differentially regulated proteins and phosphoproteins. FABP5 deletion impacted several downstream pathways associated with inflammation, cytokine production, oxidative stress, and kinase activity. Kinase enrichment analysis based on phosphorylated sites revealed key kinases, including members of the GRK family, that were altered in FABP5 KO BMDMs. Reactive oxygen species (ROS) levels were elevated in M1 polarized KO macrophages, consistent with the differential protein expression profiles. Conclusions This study represents a comprehensive characterization of the impact of FABP5 deletion upon the proteomic and phosphoproteomic landscape of M1 and M2 polarized BMDMs. Loss of FABP5 altered multiple pathways implicated in inflammatory responses and macrophage function. This work provides a foundation for future studies seeking to investigate the therapeutic potential of FABP5 inhibition in pathophysiological states resulting from dysregulated inflammatory signaling.
Collapse
Affiliation(s)
- Faniya Doswell
- Molecular and Cellular Biology Program, Stony Brook University, Stony Brook, NY, USA
- Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - John D Haley
- Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
- Stony Brook Biological Mass Spectrometry Facility, Renaissance School of Medicine, Stony Brook University, Stony Brook, USA
| | - Martin Kaczocha
- Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| |
Collapse
|
30
|
Lu F, Ye M, Hu C, Chen J, Yan L, Gu D, Xu L, Tian Y, Bai J, Tang Q. FABP5 regulates lipid metabolism to facilitate pancreatic neuroendocrine neoplasms progression via FASN mediated Wnt/β-catenin pathway. Cancer Sci 2023; 114:3553-3567. [PMID: 37302809 PMCID: PMC10475765 DOI: 10.1111/cas.15883] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 05/23/2023] [Accepted: 05/28/2023] [Indexed: 06/13/2023] Open
Abstract
Pancreatic neuroendocrine neoplasms (pNENs) are among the most frequently occurring neuroendocrine neoplasms (NENs) and require targeted therapy. High levels of fatty acid binding protein 5 (FABP5) are involved in tumor progression, but its role in pNENs remains unclear. We investigated the mRNA and protein levels of FABP5 in pNEN tissues and cell lines and found them to be upregulated. We evaluated changes in cell proliferation using CCK-8, colony formation, and 5-ethynyl-2'-deoxyuridine assays and examined the effects on cell migration and invasion using transwell assays. We found that knockdown of FABP5 suppressed the proliferation, migration, and invasion of pNEN cell lines, while overexpression of FABP5 had the opposite effect. Co-immunoprecipitation experiments were performed to clarify the interaction between FABP5 and fatty acid synthase (FASN). We further showed that FABP5 regulates the expression of FASN via the ubiquitin proteasome pathway and both proteins facilitate the progression of pNENs. Our study demonstrated that FABP5 acts as an oncogene by promoting lipid droplet deposition and activating the WNT/β-catenin signaling pathway. Moreover, the carcinogenic effects of FABP5 can be reversed by orlistat, providing a novel therapeutic intervention option.
Collapse
Affiliation(s)
- Feiyu Lu
- Department of Geriatric Gastroenterology, Neuroendocrine Tumor Center, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Institute of Neuroendocrine TumorNanjing Medical UniversityNanjingChina
| | - Mujie Ye
- Department of Geriatric Gastroenterology, Neuroendocrine Tumor Center, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Institute of Neuroendocrine TumorNanjing Medical UniversityNanjingChina
| | - Chunhua Hu
- Department of Geriatric Gastroenterology, Neuroendocrine Tumor Center, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Institute of Neuroendocrine TumorNanjing Medical UniversityNanjingChina
| | - Jinhao Chen
- Department of Geriatric Gastroenterology, Neuroendocrine Tumor Center, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Institute of Neuroendocrine TumorNanjing Medical UniversityNanjingChina
| | - Lijun Yan
- Department of Geriatric Gastroenterology, Neuroendocrine Tumor Center, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Institute of Neuroendocrine TumorNanjing Medical UniversityNanjingChina
| | - Danyang Gu
- Department of Geriatric Gastroenterology, Neuroendocrine Tumor Center, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Institute of Neuroendocrine TumorNanjing Medical UniversityNanjingChina
| | - Lin Xu
- Department of Geriatric Gastroenterology, Neuroendocrine Tumor Center, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Institute of Neuroendocrine TumorNanjing Medical UniversityNanjingChina
| | - Ye Tian
- Department of Geriatric Gastroenterology, Neuroendocrine Tumor Center, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Institute of Neuroendocrine TumorNanjing Medical UniversityNanjingChina
| | - Jianan Bai
- Department of Geriatric Gastroenterology, Neuroendocrine Tumor Center, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Institute of Neuroendocrine TumorNanjing Medical UniversityNanjingChina
| | - Qiyun Tang
- Department of Geriatric Gastroenterology, Neuroendocrine Tumor Center, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Institute of Neuroendocrine TumorNanjing Medical UniversityNanjingChina
| |
Collapse
|
31
|
Abstract
Fatty acid-binding proteins (FABPs) are small lipid-binding proteins abundantly expressed in tissues that are highly active in fatty acid (FA) metabolism. Ten mammalian FABPs have been identified, with tissue-specific expression patterns and highly conserved tertiary structures. FABPs were initially studied as intracellular FA transport proteins. Further investigation has demonstrated their participation in lipid metabolism, both directly and via regulation of gene expression, and in signaling within their cells of expression. There is also evidence that they may be secreted and have functional impact via the circulation. It has also been shown that the FABP ligand binding repertoire extends beyond long-chain FAs and that their functional properties also involve participation in systemic metabolism. This article reviews the present understanding of FABP functions and their apparent roles in disease, particularly metabolic and inflammation-related disorders and cancers.
Collapse
Affiliation(s)
- Judith Storch
- Department of Nutritional Sciences and Rutgers Center for Lipid Research, Rutgers University, New Brunswick, New Jersey, United States;
| | - Betina Corsico
- Instituto de Investigaciones Bioquímicas de La Plata, CONICET-UNLP, Facultad de Ciencias Médicas, La Plata, Argentina;
| |
Collapse
|
32
|
Lang H, Noble KV, Barth JL, Rumschlag JA, Jenkins TR, Storm SL, Eckert MA, Dubno JR, Schulte BA. The Stria Vascularis in Mice and Humans Is an Early Site of Age-Related Cochlear Degeneration, Macrophage Dysfunction, and Inflammation. J Neurosci 2023; 43:5057-5075. [PMID: 37268417 PMCID: PMC10324995 DOI: 10.1523/jneurosci.2234-22.2023] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 04/19/2023] [Accepted: 05/25/2023] [Indexed: 06/04/2023] Open
Abstract
Age-related hearing loss, or presbyacusis, is a common degenerative disorder affecting communication and quality of life for millions of older adults. Multiple pathophysiologic manifestations, along with many cellular and molecular alterations, have been linked to presbyacusis; however, the initial events and causal factors have not been clearly established. Comparisons of the transcriptome in the lateral wall (LW) with other cochlear regions in a mouse model (of both sexes) of "normal" age-related hearing loss revealed that early pathophysiological alterations in the stria vascularis (SV) are associated with increased macrophage activation and a molecular signature indicative of inflammaging, a common form of immune dysfunction. Structure-function correlation analyses in mice across the lifespan showed that the age-dependent increase in macrophage activation in the stria vascularis is associated with a decline in auditory sensitivity. High-resolution imaging analysis of macrophage activation in middle-aged and aged mouse and human cochleas, along with transcriptomic analysis of age-dependent changes in mouse cochlear macrophage gene expression, support the hypothesis that aberrant macrophage activity is an important contributor to age-dependent strial dysfunction, cochlear pathology, and hearing loss. Thus, this study highlights the SV as a primary site of age-related cochlear degeneration and aberrant macrophage activity and dysregulation of the immune system as early indicators of age-related cochlear pathology and hearing loss. Importantly, novel new imaging methods described here now provide a means to analyze human temporal bones in a way that had not previously been feasible and thereby represent a significant new tool for otopathological evaluation.SIGNIFICANCE STATEMENT Age-related hearing loss is a common neurodegenerative disorder affecting communication and quality of life. Current interventions (primarily hearing aids and cochlear implants) offer imperfect and often unsuccessful therapeutic outcomes. Identification of early pathology and causal factors is crucial for the development of new treatments and early diagnostic tests. Here, we find that the SV, a nonsensory component of the cochlea, is an early site of structural and functional pathology in mice and humans that is characterized by aberrant immune cell activity. We also establish a new technique for evaluating cochleas from human temporal bones, an important but understudied area of research because of a lack of well-preserved human specimens and difficult tissue preparation and processing approaches.
Collapse
Affiliation(s)
- Hainan Lang
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Kenyaria V Noble
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Jeremy L Barth
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Jeffrey A Rumschlag
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Tyreek R Jenkins
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Shelby L Storm
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Mark A Eckert
- Department of Otolaryngology-Head and Neck Surgery, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Judy R Dubno
- Department of Otolaryngology-Head and Neck Surgery, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Bradley A Schulte
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina 29425
| |
Collapse
|
33
|
Ye M, Hu C, Chen T, Yu P, Chen J, Lu F, Xu L, Zhong Y, Yan L, Kan J, Bai J, Li X, Tian Y, Tang Q. FABP5 suppresses colorectal cancer progression via mTOR-mediated autophagy by decreasing FASN expression. Int J Biol Sci 2023; 19:3115-3127. [PMID: 37416772 PMCID: PMC10321282 DOI: 10.7150/ijbs.85285] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 06/03/2023] [Indexed: 07/08/2023] Open
Abstract
Lipid metabolism plays an important role in the occurrence and development of cancer, in particular, digestive system tumors such as colon cancer. Here, we investigated the role of the fatty acid-binding protein 5 (FABP5) in colorectal cancer (CRC). We observed marked down-regulation of FABP5 in CRC. Data from functional assays revealed inhibitory effects of FABP5 on cell proliferation, colony formation, migration, invasion as well as tumor growth in vivo. In terms of mechanistic insights, FABP5 interacted with fatty acid synthase (FASN) and activated the ubiquitin proteasome pathway, leading to a decrease in FASN expression and lipid accumulation, moreover, suppressing mTOR signaling and facilitating cell autophagy. Orlistat, a FASN inhibitor, exerted anti-cancer effects both in vivo and in vitro. Furthermore, the upstream RNA demethylase ALKBH5 positively regulated FABP5 expression via an m6A-independent mechanism. Overall, our collective findings offer valuable insights into the critical role of the ALKBH5/FABP5/FASN/mTOR axis in tumor progression and uncover a potential mechanism linking lipid metabolism to development of CRC, providing novel therapeutic targets for future interventions.
Collapse
Affiliation(s)
- Mujie Ye
- ✉ Corresponding authors: Qiyun Tang, Department of Geriatric Gastroenterology, Neuroendocrine Tumor Center, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Institute of Neuroendocrine Tumor, Nanjing Medical University, NO.300 Guangzhou Road, Nanjing, China. . Ye Tian, Department of Geriatric Gastroenterology, Neuroendocrine Tumor Center, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Institute of Neuroendocrine Tumor, Nanjing Medical University, NO.300 Guangzhou Road, Nanjing, China. . Mujie Ye, Department of Geriatric Gastroenterology, Neuroendocrine Tumor Center, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Institute of Neuroendocrine Tumor, Nanjing Medical University, NO.300 Guangzhou Road, Nanjing, China.
| | | | | | | | | | | | | | | | | | | | | | | | - Ye Tian
- ✉ Corresponding authors: Qiyun Tang, Department of Geriatric Gastroenterology, Neuroendocrine Tumor Center, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Institute of Neuroendocrine Tumor, Nanjing Medical University, NO.300 Guangzhou Road, Nanjing, China. . Ye Tian, Department of Geriatric Gastroenterology, Neuroendocrine Tumor Center, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Institute of Neuroendocrine Tumor, Nanjing Medical University, NO.300 Guangzhou Road, Nanjing, China. . Mujie Ye, Department of Geriatric Gastroenterology, Neuroendocrine Tumor Center, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Institute of Neuroendocrine Tumor, Nanjing Medical University, NO.300 Guangzhou Road, Nanjing, China.
| | - Qiyun Tang
- ✉ Corresponding authors: Qiyun Tang, Department of Geriatric Gastroenterology, Neuroendocrine Tumor Center, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Institute of Neuroendocrine Tumor, Nanjing Medical University, NO.300 Guangzhou Road, Nanjing, China. . Ye Tian, Department of Geriatric Gastroenterology, Neuroendocrine Tumor Center, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Institute of Neuroendocrine Tumor, Nanjing Medical University, NO.300 Guangzhou Road, Nanjing, China. . Mujie Ye, Department of Geriatric Gastroenterology, Neuroendocrine Tumor Center, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Institute of Neuroendocrine Tumor, Nanjing Medical University, NO.300 Guangzhou Road, Nanjing, China.
| |
Collapse
|
34
|
Ramchatesingh B, Martínez Villarreal A, Arcuri D, Lagacé F, Setah SA, Touma F, Al-Badarin F, Litvinov IV. The Use of Retinoids for the Prevention and Treatment of Skin Cancers: An Updated Review. Int J Mol Sci 2022; 23:ijms232012622. [PMID: 36293471 PMCID: PMC9603842 DOI: 10.3390/ijms232012622] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 10/12/2022] [Accepted: 10/13/2022] [Indexed: 11/21/2022] Open
Abstract
Retinoids are natural and synthetic vitamin A derivatives that are effective for the prevention and the treatment of non-melanoma skin cancers (NMSC). NMSCs constitute a heterogenous group of non-melanocyte-derived skin cancers that impose substantial burdens on patients and healthcare systems. They include entities such as basal cell carcinoma and cutaneous squamous cell carcinoma (collectively called keratinocyte carcinomas), cutaneous lymphomas and Kaposi’s sarcoma among others. The retinoid signaling pathway plays influential roles in skin physiology and pathology. These compounds regulate diverse biological processes within the skin, including proliferation, differentiation, angiogenesis and immune regulation. Collectively, retinoids can suppress skin carcinogenesis. Both topical and systemic retinoids have been investigated in clinical trials as NMSC prophylactics and treatments. Desirable efficacy and tolerability in clinical trials have prompted health regulatory bodies to approve the use of retinoids for NMSC management. Acceptable off-label uses of these compounds as drugs for skin cancers are also described. This review is a comprehensive outline on the biochemistry of retinoids, their activities in the skin, their effects on cancer cells and their adoption in clinical practice.
Collapse
Affiliation(s)
| | | | - Domenico Arcuri
- Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H4A 3J1, Canada
| | - François Lagacé
- Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H4A 3J1, Canada
- Division of Dermatology, McGill University Health Center, Montreal, QC H4A 3J1, Canada
| | - Samy Abu Setah
- Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H4A 3J1, Canada
| | - Fadi Touma
- Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H4A 3J1, Canada
| | - Faris Al-Badarin
- Faculté de Médicine, Université Laval, Québec, QC G1V 0V6, Canada
| | - Ivan V. Litvinov
- Division of Experimental Medicine, McGill University, Montreal, QC H4A 3J1, Canada
- Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H4A 3J1, Canada
- Division of Dermatology, McGill University Health Center, Montreal, QC H4A 3J1, Canada
- Correspondence:
| |
Collapse
|
35
|
Consequences of Autophagy Deletion on the Age-Related Changes in the Epidermal Lipidome of Mice. Int J Mol Sci 2022; 23:ijms231911110. [PMID: 36232414 PMCID: PMC9569666 DOI: 10.3390/ijms231911110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 09/03/2022] [Accepted: 09/18/2022] [Indexed: 12/02/2022] Open
Abstract
Autophagy is a controlled mechanism of intracellular self-digestion with functions in metabolic adaptation to stress, in development, in proteostasis and in maintaining cellular homeostasis in ageing. Deletion of autophagy in epidermal keratinocytes does not prevent the formation of a functional epidermis and the permeability barrier but causes increased susceptibility to damage stress and metabolic alterations and accelerated ageing phenotypes. We here investigated how epidermal autophagy deficiency using Keratin 14 driven Atg7 deletion would affect the lipid composition of the epidermis of young and old mice. Using mass spectrometric lipidomics we found a reduction of age-related accumulation of storage lipids in the epidermis of autophagy-deficient mice, and specific changes in chain length and saturation of fatty acids in several lipid classes. Transcriptomics and immunostaining suggest that these changes are accompanied by changes in expression and localisation of lipid and fatty acid transporter proteins, most notably fatty acid binding protein 5 (FABP5) in autophagy knockouts. Thus, maintaining autophagic activity at an advanced age may be necessary to maintain epidermal lipid homeostasis in mammals.
Collapse
|
36
|
Guo Q, Kawahata I, Cheng A, Jia W, Wang H, Fukunaga K. Fatty Acid-Binding Proteins: Their Roles in Ischemic Stroke and Potential as Drug Targets. Int J Mol Sci 2022; 23:9648. [PMID: 36077044 PMCID: PMC9455833 DOI: 10.3390/ijms23179648] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 08/19/2022] [Accepted: 08/23/2022] [Indexed: 11/17/2022] Open
Abstract
Stroke is among the leading causes of death and disability worldwide. However, despite long-term research yielding numerous candidate neuroprotective drugs, there remains a lack of effective neuroprotective therapies for ischemic stroke patients. Among the factors contributing to this deficiency could be that single-target therapy is insufficient in addressing the complex and extensive mechanistic basis of ischemic brain injury. In this context, lipids serve as an essential component of multiple biological processes and play important roles in the pathogenesis of numerous common neurological diseases. Moreover, in recent years, fatty acid-binding proteins (FABPs), a family of lipid chaperone proteins, have been discovered to be involved in the onset or development of several neurodegenerative diseases, including Alzheimer's and Parkinson's disease. However, comparatively little attention has focused on the roles played by FABPs in ischemic stroke. We have recently demonstrated that neural tissue-associated FABPs are involved in the pathological mechanism of ischemic brain injury in mice. Here, we review the literature published in the past decade that has reported on the associations between FABPs and ischemia and summarize the relevant regulatory mechanisms of FABPs implicated in ischemic injury. We also propose candidate FABPs that could serve as potential therapeutic targets for ischemic stroke.
Collapse
Affiliation(s)
- Qingyun Guo
- Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou 571199, China
- Department of CNS Drug Innovation, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Ichiro Kawahata
- Department of CNS Drug Innovation, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - An Cheng
- Department of CNS Drug Innovation, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Wenbin Jia
- Department of CNS Drug Innovation, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Haoyang Wang
- Department of CNS Drug Innovation, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Kohji Fukunaga
- Department of CNS Drug Innovation, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
- BRI Pharma Incorporated, Sendai 982-0804, Japan
| |
Collapse
|
37
|
Zhao Y, Zhang Q, Tu K, Chen Y, Peng Y, Ni Y, Zhu G, Cheng C, Li Y, Xiao X, Yu C, Lu K, Chen Y, Li C, Tang J, Wang G, Luo W, Zhang W, Che G, Li W, Wang Z, Xie D. Single-Cell Transcriptomics of Immune Cells Reveal Diversity and Exhaustion Signatures in Non-Small-Cell Lung Cancer. Front Immunol 2022; 13:854724. [PMID: 35874785 PMCID: PMC9299430 DOI: 10.3389/fimmu.2022.854724] [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: 01/14/2022] [Accepted: 06/06/2022] [Indexed: 02/05/2023] Open
Abstract
Understanding immune cell phenotypes in the tumor microenvironment (TME) is essential for explaining and predicting progression of non-small cell lung cancer (NSCLC) and its response to immunotherapy. Here we describe the single-cell transcriptomics of CD45+ immune cells from tumors, normal tissues and blood of NSCLC patients. We identified three clusters of immune cells exerting immunosuppressive effects: CD8+ T cells with exhausted phenotype, tumor-associated macrophages (TAMs) with a pro-inflammatory M2 phenotype, and regulatory B cells (B regs) with tumor-promoting characteristics. We identified genes that may be mediating T cell phenotypes, including the transcription factors ONECUT2 and ETV4 in exhausted CD8+ T cells, TIGIT and CTL4 high expression in regulatory T cells. Our results highlight the heterogeneity of CD45+ immune cells in the TME and provide testable hypotheses about the cell types and genes that define the TME.
Collapse
Affiliation(s)
- Ying Zhao
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
| | - Qilin Zhang
- Laboratory of Omics Technology and Bioinformatics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Kailin Tu
- Laboratory of Omics Technology and Bioinformatics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yanmei Chen
- Health Management Center, West China Tianfu Hospital, Sichuan University, Chengdu, China
| | - Yuxuan Peng
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
| | - Yinyun Ni
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
| | - Guonian Zhu
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
| | - Cheng Cheng
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
| | - Yangqian Li
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
| | - Xue Xiao
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
| | - Chunyan Yu
- Laboratory of Omics Technology and Bioinformatics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Keying Lu
- Laboratory of Omics Technology and Bioinformatics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yaxin Chen
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
| | - Chengpin Li
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
| | - Jun Tang
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
| | - Gang Wang
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
| | - Wenxin Luo
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Wengeng Zhang
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
| | - Guowei Che
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Weimin Li
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China.,Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Zhoufeng Wang
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
| | - Dan Xie
- Laboratory of Omics Technology and Bioinformatics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|