151
|
Ekström A, Villoutreix BO, Halperin J, Renström E, Blom AM, King BC. CD59 double knockout mice express a CD59ba hybrid fusion protein that mediates insulin secretion. FASEB J 2024; 38:e70156. [PMID: 39530539 DOI: 10.1096/fj.202401808r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 09/23/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024]
Abstract
CD59 is a cell-surface inhibitor of the terminal step in the complement cascade. However, in addition to its complement inhibitory function, a non-canonical role of CD59 in pancreatic beta cells has been identified. Two recently discovered intracellular alternative splice forms of CD59, IRIS-1 and IRIS-2, are involved in insulin exocytosis through interactions with SNARE-complex components. In mice, the CD59 gene has undergone duplication and to further explore the role of CD59 in insulin secretion, blood glucose homeostasis was studied in a CD59 double knockout (CD59abKO) mouse model. However, no phenotypic deviation related to insulin secretion or blood glucose homeostasis was observed for the CD59abKO mice. Instead, a CD59ba hybrid transcript formed as a consequence of the mutation induced to generate the model was identified. This hybrid transcript is expressed in pancreatic islets of the CD59abKO mice and is comprised of the remaining exons of the two CD59 genes spliced together. Similar to canonical CD59, the CD59ba hybrid was found to be glycosylated and present on the cell surface when exogenously expressed in INS-1 832/13 cells. Furthermore, INS-1 832/13 cells over-expressing the mouse CD59ba hybrid retained normal insulin secretion following siRNA-mediated knockdown of canonical CD59. Hence, although the CD59ba hybrid has lost the complement inhibitory function, the intracellular insulin secretory function remains. These results provide further information concerning the structural requirements of CD59 in its intracellular role relative to its role as a complement inhibitor. It also highlights the importance of carefully assessing plausible consequences of induced mutations in research models.
Collapse
Affiliation(s)
- A Ekström
- Section for Medical Protein Chemistry, Department of Translational Medicine, Lund university, Malmö, Sweden
| | - B O Villoutreix
- Department of Neuroscience, NeuroDiderot, Inserm U1141, Université Paris Cité, Paris, France
| | - J Halperin
- Division of Haematology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - E Renström
- Section for Islet Pathophysiology, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - A M Blom
- Section for Medical Protein Chemistry, Department of Translational Medicine, Lund university, Malmö, Sweden
| | - B C King
- Section for Medical Protein Chemistry, Department of Translational Medicine, Lund university, Malmö, Sweden
| |
Collapse
|
152
|
Sumalla-Cano S, Eguren-García I, Lasarte-García Á, Prola TA, Martínez-Díaz R, Elío I. Carotenoids Intake and Cardiovascular Prevention: A Systematic Review. Nutrients 2024; 16:3859. [PMID: 39599645 PMCID: PMC11597197 DOI: 10.3390/nu16223859] [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: 09/13/2024] [Revised: 11/07/2024] [Accepted: 11/09/2024] [Indexed: 11/29/2024] Open
Abstract
Background: Cardiovascular diseases (CVDs) encompass a variety of conditions that affect the heart and blood vessels. Carotenoids, a group of fat-soluble organic pigments synthesized by plants, fungi, algae, and some bacteria, may have a beneficial effect in reducing cardiovascular disease (CVD) risk. This study aims to examine and synthesize current research on the relationship between carotenoids and CVDs. Methods: A systematic review was conducted using MEDLINE and the Cochrane Library to identify relevant studies on the efficacy of carotenoid supplementation for CVD prevention. Interventional analytical studies (randomized and non-randomized clinical trials) published in English from January 2011 to February 2024 were included. Results: A total of 38 studies were included in the qualitative analysis. Of these, 17 epidemiological studies assessed the relationship between carotenoids and CVDs, 9 examined the effect of carotenoid supplementation, and 12 evaluated dietary interventions. Conclusions: Elevated serum carotenoid levels are associated with reduced CVD risk factors and inflammatory markers. Increasing the consumption of carotenoid-rich foods appears to be more effective than supplementation, though the specific effects of individual carotenoids on CVD risk remain uncertain.
Collapse
Affiliation(s)
- Sandra Sumalla-Cano
- Research Group on Foods, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, 39011 Santander, Spain; (I.E.-G.); (Á.L.-G.); (R.M.-D.)
- Faculty of Health Sciences, Universidade do Cuanza, Cuito EN250, Bié, Angola
- Faculty of Health Sciences, Universidad de La Romana, La Romana 22000, Dominican Republic
| | - Imanol Eguren-García
- Research Group on Foods, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, 39011 Santander, Spain; (I.E.-G.); (Á.L.-G.); (R.M.-D.)
| | - Álvaro Lasarte-García
- Research Group on Foods, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, 39011 Santander, Spain; (I.E.-G.); (Á.L.-G.); (R.M.-D.)
| | - Thomas A. Prola
- Faculty of Social Sciences and Humanities, European University of the Atlantic, 39011 Santander, Spain;
| | - Raquel Martínez-Díaz
- Research Group on Foods, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, 39011 Santander, Spain; (I.E.-G.); (Á.L.-G.); (R.M.-D.)
- Faculty of Health Sciences, Universidade do Cuanza, Cuito EN250, Bié, Angola
- Faculty of Health Sciences, Universidad de La Romana, La Romana 22000, Dominican Republic
| | - Iñaki Elío
- Research Group on Foods, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, 39011 Santander, Spain; (I.E.-G.); (Á.L.-G.); (R.M.-D.)
- Faculty of Health Sciences, Universidad Internacional Iberoamericana, Arecibo, PR 00613, USA
- Department of Health, Nutrition and Sport, Universidad Internacional Iberoamericana, Campeche 24560, Mexico
| |
Collapse
|
153
|
Kiyuna LA, Krishnamurthy KA, Homan EB, Langelaar-Makkinje M, Gerding A, Bos T, Oosterhuis D, Overduin RJ, Schreuder AB, de Meijer VE, Olinga P, Derks TGJ, van Eunen K, Bakker BM, Oosterveer MH. Precision-cut liver slices as an ex vivo model to assess impaired hepatic glucose production. Commun Biol 2024; 7:1479. [PMID: 39521914 PMCID: PMC11550398 DOI: 10.1038/s42003-024-07070-z] [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: 10/23/2023] [Accepted: 10/14/2024] [Indexed: 11/16/2024] Open
Abstract
Fasting hypoglycemia is a severe and incompletely understood symptom of various inborn errors of metabolism (IEM). Precision-cut liver slices (PCLS) represent a promising model for studying glucose production ex vivo. This study quantified the net glucose production of human and murine PCLS in the presence of different gluconeogenic precursors. Dihydroxyacetone-supplemented slices from the fed mice yielded the highest rate, further stimulated by forskolin and dibutyryl-cAMP. Moreover, using 13C isotope tracing, we assessed the contribution of glycogenolysis and gluconeogenesis to net glucose production over time. Pharmacological inhibition of the glucose 6-phosphate transporter SLC37A4 markedly reduced net glucose production and increased lactate secretion and glycogen storage, while glucose production was completely abolished in PCLS from glycogen storage disease type Ia and Ib patients. In conclusion, this study identifies PCLS as an effective ex vivo model to study hepatic glucose production and opens opportunities for its future application in IEM research and beyond.
Collapse
Affiliation(s)
- Ligia Akemi Kiyuna
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | | | - Esther B Homan
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Miriam Langelaar-Makkinje
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Albert Gerding
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Trijnie Bos
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Dorenda Oosterhuis
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, Groningen, The Netherlands
| | - Ruben J Overduin
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Metabolic Diseases, Beatrix Children's Hospital, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Andrea B Schreuder
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Metabolic Diseases, Beatrix Children's Hospital, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Vincent E de Meijer
- Section of HPB Surgery and Liver Transplantation, Department of Surgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Peter Olinga
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, Groningen, The Netherlands
| | - Terry G J Derks
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Metabolic Diseases, Beatrix Children's Hospital, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Karen van Eunen
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Barbara M Bakker
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| | - Maaike H Oosterveer
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| |
Collapse
|
154
|
Moore D, Morrell JS. Do dietary patterns differ with video game usage in college men? JOURNAL OF AMERICAN COLLEGE HEALTH : J OF ACH 2024; 72:2362-2370. [PMID: 35997702 DOI: 10.1080/07448481.2022.2110381] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 07/23/2022] [Accepted: 07/28/2022] [Indexed: 06/15/2023]
Abstract
Objective: To assess differences in dietary quality among college men reporting high, moderate, and non-video game usage. Participants: College men aged 18-24. Methods: Cross-sectional data were collected between 2012 and 2020. Participants (n = 1259) were categorized according to self-reported video game usage: non-users (NVG), <1 h/day (MVG), and ≥1 h/day (HVG). ANCOVA identified group differences in nutrient intake from 3-day food records. Results: College men reported 30% NVG, 39% MVG, and 31% HVG. Higher saturated fat (30.2 ± 0.4 g and 30.1 ± 0.3 g, vs. 28.5 ± 0.4 g, p < .01, p < .01) and lower fruit/vegetable intake (3.00 ± 0.1 cups and 2.91 ± 0.1 cups, vs. 3.45 ± 0.1 cups, p < .00, p < .00) was observed in HVG and MVG vs. NVG. Higher discretionary calories (750 ± 13 kcals, vs. 686 ± 13 kcals, p < .00) in HVG and sodium (3922 ± 44 mg, vs. 3860 ± 50 mg, p < .02) in MVG were reported vs. NVG. Conclusion: Video game usage was associated with higher saturated fat, sodium, discretionary calories, and lower F/V intake in college men.
Collapse
Affiliation(s)
- Dustin Moore
- Department of Agriculture, Nutrition, and Food Systems, University of New Hampshire, Durham, New Hampshire, USA
| | - Jesse Stabile Morrell
- Department of Agriculture, Nutrition, and Food Systems, University of New Hampshire, Durham, New Hampshire, USA
| |
Collapse
|
155
|
Khalaf MM, Mahmoud HM, Kandeil MA, Mahmoud HA, Salama AA. Fumaric acid protects rats from ciprofloxacin-provoked depression through modulating TLR4, Nrf-2, and p190-rho GTP. Drug Chem Toxicol 2024; 47:897-908. [PMID: 39563658 DOI: 10.1080/01480545.2024.2310641] [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: 01/27/2023] [Revised: 11/17/2023] [Accepted: 11/23/2023] [Indexed: 11/21/2024]
Abstract
Depression is a persistent illness affecting health, behavior, and performance in life. Worldwide morbidity and mortality are caused by depression. The current study intended to explore fumaric acid's potential protective effect against ciprofloxacin-provoked depression in rats and to determine its mechanism of action by studying its antioxidant and anti-inflammatory properties. Five groups of male Wistar albino rats (120 g ± 20) were employed; the first group received physiological saline, the second group received fumaric acid (80 mg/kg/day; orally) for 3 weeks, the third group was administered ciprofloxacin (50 mg/kg/day; orally) for 3 weeks to induce depression, the fourth group received a daily low dose of fumaric acid (40 mg/kg; orally) concurrent with ciprofloxacin and the fifth group received a daily high dose of fumaric acid (80 mg/kg; orally) concurrent with ciprofloxacin for 21 days. Then, behavior tests, oxidative stress indicators, inflammatory biomarkers, neurotransmitters, p190 Rho GTP, and histopathological examination were evaluated. Ciprofloxacin significantly increased oxidative stress biomarkers [malondialdehyde (MDA) as a lipid peroxidation marker and nitric oxide (NO)] and biomarkers of inflammation [Toll-like receptor4 (TLR-4)] and tumor necrosis factor-alpha (TNF-α) with reduction in the activities of the nuclear factor erythroid 2-related factor 2 (Nrf-2) and catalase as well as brain contents of neurotransmitters and P190-RHO GTP. In addition, it causes necrosis of neurons and mild loss of Purkinje cells. Fumaric acid eliminates these effects of ciprofloxacin. Fumaric acid has beneficial effects as an anti-depressant in Wistar albino male rats that received ciprofloxacin.
Collapse
Affiliation(s)
- Marwa M Khalaf
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Heba M Mahmoud
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Mohamed A Kandeil
- Department of Biochemistry, Faculty of Veterinary Medicine, Beni-Suef University, Beni-Suef, Egypt
| | | | - Abeer A Salama
- Department of Pharmacology, National Research Centre, Giza, Egypt
| |
Collapse
|
156
|
Hosseini SM, Tingzhu Y, Zaohong R, Ullah F, Liang A, Hua G, Yang L. Regulatory impacts of PPARGC1A gene expression on milk production and cellular metabolism in buffalo mammary epithelial cells. Anim Biotechnol 2024; 35:2344210. [PMID: 38785376 DOI: 10.1080/10495398.2024.2344210] [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] [Indexed: 05/25/2024]
Abstract
The PPARGC1A gene plays a fundamental role in regulating cellular energy metabolism, including adaptive thermogenesis, mitochondrial biogenesis, adipogenesis, gluconeogenesis, and glucose/fatty acid metabolism. In a previous study, our group investigated seven SNPs in Mediterranean buffalo associated with milk production traits, and the current study builds on this research by exploring the regulatory influences of the PPARGC1A gene in buffalo mammary epithelial cells (BuMECs). Our findings revealed that knockdown of PPARGC1A gene expression significantly affected the growth of BuMECs, including proliferation, cell cycle, and apoptosis. Additionally, we observed downregulated triglyceride secretion after PPARGC1A knockdown. Furthermore, the critical genes related to milk production, including the STATS, BAD, P53, SREBF1, and XDH genes were upregulated after RNAi, while the FABP3 gene, was downregulated. Moreover, Silencing the PPARGC1A gene led to a significant downregulation of β-casein synthesis in BuMECs. Our study provides evidence of the importance of the PPARGC1A gene in regulating cell growth, lipid, and protein metabolism in the buffalo mammary gland. In light of our previous research, the current study underscores the potential of this gene for improving milk production efficiency and overall dairy productivity in buffalo populations.
Collapse
Affiliation(s)
- Seyed Mahdi Hosseini
- National Center for International Research on Animal Genetics, Breeding and Reproduction (NCIRAGBR), College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Ye Tingzhu
- National Center for International Research on Animal Genetics, Breeding and Reproduction (NCIRAGBR), College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Ran Zaohong
- National Center for International Research on Animal Genetics, Breeding and Reproduction (NCIRAGBR), College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Farman Ullah
- National Center for International Research on Animal Genetics, Breeding and Reproduction (NCIRAGBR), College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Aixin Liang
- National Center for International Research on Animal Genetics, Breeding and Reproduction (NCIRAGBR), College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Guohua Hua
- National Center for International Research on Animal Genetics, Breeding and Reproduction (NCIRAGBR), College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Liguo Yang
- National Center for International Research on Animal Genetics, Breeding and Reproduction (NCIRAGBR), College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
157
|
Sauer B, Kueckelhaus J, Lorenz NI, Bozkurt S, Schulte D, Weinem J, Benzarti M, Meiser J, Urban H, Villa G, Harter PN, Münch C, Rieger J, Steinbach JP, Heiland DH, Ronellenfitsch MW. An AMP-activated protein kinase-PGC-1α axis mediates metabolic plasticity in glioblastoma. Clin Transl Med 2024; 14:e70030. [PMID: 39552019 PMCID: PMC11570551 DOI: 10.1002/ctm2.70030] [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: 04/09/2024] [Revised: 09/02/2024] [Accepted: 09/10/2024] [Indexed: 11/19/2024] Open
Abstract
Glioblastoma, the most frequent primary malignant brain tumour in adults, is characterised by profound yet dynamic hypoxia and nutrient depletion. To sustain survival and proliferation, tumour cells are compelled to acquire metabolic plasticity with the induction of adaptive metabolic programs. Here, we interrogated the pathways necessary to enable processing of nutrients other than glucose. We employed genetic approaches (stable/inducible overexpression, CRISPR/Cas9 knockout), pharmacological interventions with a novel inhibitor of AMP-activated protein kinase (AMPK) in glioblastoma cell culture systems and a proteomic approach to investigate mechanisms of metabolic plasticity. Moreover, a spatially resolved multiomic analysis was employed to correlate the gene expression pattern of PGC-1α with the local metabolic and genetic architecture in human glioblastoma tissue sections. A switch from glucose to alternative nutrients triggered an activation of AMPK, which in turn activated PGC-1α-dependent adaptive programs promoting mitochondrial metabolism. This sensor-effector mechanism was essential for metabolic plasticity with both functional AMPK and PGC-1α necessary for survival and growth of cells under nonglucose nutrient sources. In human glioblastoma tissue specimens, PGC-1α-expression correlated with nonhypoxic tumour niches defining a specific metabolic compartment. Our findings reveal a cell-intrinsic nutrient sensing and switching mechanism. The exposure to alternative fuels triggers a starvation signal that subsequently is passed on via AMPK and PGC-1α to induce adaptive programs necessary for broader spectrum nutrient metabolism. The integration of spatially resolved transcriptomic data confirms the relevance of PGC-1α especially in nonhypoxic tumour regions. Thus, the AMPK-PGC-1α axis is a candidate for therapeutic inhibition in glioblastoma. KEY POINTS/HIGHLIGHTS: AMPK activation induces PGC-1α expression in glioblastoma during nutrient scarcity. PGC-1α enables metabolic plasticity by facilitating metabolism of alternative nutrients in glioblastoma. PGC-1α expression is inversely correlated with hypoxic tumour regions in human glioblastomas.
Collapse
Affiliation(s)
- Benedikt Sauer
- Dr. Senckenberg Institute of NeurooncologyUniversity HospitalGoethe University FrankfurtFrankfurt am MainGermany
- University Cancer Center Frankfurt (UCT), University HospitalGoethe University FrankfurtFrankfurt am MainGermany
- German Cancer Consortium (DKTK), partner site Frankfurt, a partnership between DKFZ and University Hospital FrankfurtGoethe University FrankfurtFrankfurt am MainGermany
- Frankfurt Cancer Institute (FCI)University HospitalGoethe University FrankfurtFrankfurt am MainGermany
| | - Jan Kueckelhaus
- Microenvironment and Immunology Research LaboratoryMedical CenterUniversity of FreiburgFreiburgGermany
- Department of NeurosurgeryMedical CenterUniversity of FreiburgFreiburgGermany
- Faculty of MedicineUniversity of FreiburgFreiburgGermany
- Faculty of Medicine and Medical CenterComprehensive Cancer Center Freiburg (CCCF)University of FreiburgFreiburgGermany
- German Cancer Consortium (DKTK), partner site Freiburg, a partnership between DKFZ and University Medical Center FreiburgFreiburgGermany
| | - Nadja I. Lorenz
- Dr. Senckenberg Institute of NeurooncologyUniversity HospitalGoethe University FrankfurtFrankfurt am MainGermany
- University Cancer Center Frankfurt (UCT), University HospitalGoethe University FrankfurtFrankfurt am MainGermany
- German Cancer Consortium (DKTK), partner site Frankfurt, a partnership between DKFZ and University Hospital FrankfurtGoethe University FrankfurtFrankfurt am MainGermany
- Frankfurt Cancer Institute (FCI)University HospitalGoethe University FrankfurtFrankfurt am MainGermany
| | - Süleyman Bozkurt
- Institute of Molecular Systems Medicine, Faculty of MedicineGoethe University FrankfurtFrankfurt am MainGermany
| | - Dorothea Schulte
- Institute of Neurology (Edinger Institute)University Hospital, Goethe University FrankfurtFrankfurtGermany
| | - Jan‐Béla Weinem
- Dr. Senckenberg Institute of NeurooncologyUniversity HospitalGoethe University FrankfurtFrankfurt am MainGermany
- University Cancer Center Frankfurt (UCT), University HospitalGoethe University FrankfurtFrankfurt am MainGermany
- German Cancer Consortium (DKTK), partner site Frankfurt, a partnership between DKFZ and University Hospital FrankfurtGoethe University FrankfurtFrankfurt am MainGermany
- Frankfurt Cancer Institute (FCI)University HospitalGoethe University FrankfurtFrankfurt am MainGermany
| | - Mohaned Benzarti
- Cancer Metabolism GroupDepartment of Cancer ResearchLuxembourg Institute of HealthLuxembourgLuxembourg
- Faculty of ScienceTechnology and MedicineUniversity of Luxembourg, 2 avenue de UniversitéEsch‐sur‐AlzetteLuxembourg
| | - Johannes Meiser
- Cancer Metabolism GroupDepartment of Cancer ResearchLuxembourg Institute of HealthLuxembourgLuxembourg
- Faculty of ScienceTechnology and MedicineUniversity of Luxembourg, 2 avenue de UniversitéEsch‐sur‐AlzetteLuxembourg
| | - Hans Urban
- Dr. Senckenberg Institute of NeurooncologyUniversity HospitalGoethe University FrankfurtFrankfurt am MainGermany
- University Cancer Center Frankfurt (UCT), University HospitalGoethe University FrankfurtFrankfurt am MainGermany
- German Cancer Consortium (DKTK), partner site Frankfurt, a partnership between DKFZ and University Hospital FrankfurtGoethe University FrankfurtFrankfurt am MainGermany
- Frankfurt Cancer Institute (FCI)University HospitalGoethe University FrankfurtFrankfurt am MainGermany
| | - Giulia Villa
- Microenvironment and Immunology Research LaboratoryMedical CenterUniversity of FreiburgFreiburgGermany
- Department of NeurosurgeryMedical CenterUniversity of FreiburgFreiburgGermany
- Faculty of MedicineUniversity of FreiburgFreiburgGermany
- Faculty of Medicine and Medical CenterComprehensive Cancer Center Freiburg (CCCF)University of FreiburgFreiburgGermany
- German Cancer Consortium (DKTK), partner site Freiburg, a partnership between DKFZ and University Medical Center FreiburgFreiburgGermany
| | - Patrick N. Harter
- Institute of Neurology (Edinger Institute)University Hospital, Goethe University FrankfurtFrankfurtGermany
- German Cancer Consortium (DKTK), partner site Munich, a partnership between DKFZ and Ludwig‐Maximilians‐Universität (LMU)MunichGermany
- Center for Neuropathology and Prion Research, Faculty of MedicineLudwig‐Maximilians‐Universität (LMU)MunichGermany
| | - Christian Münch
- Frankfurt Cancer Institute (FCI)University HospitalGoethe University FrankfurtFrankfurt am MainGermany
- Institute of Molecular Systems Medicine, Faculty of MedicineGoethe University FrankfurtFrankfurt am MainGermany
- Cardio‐Pulmonary InstituteGoethe University FrankfurtFrankfurt am MainGermany
| | - Johannes Rieger
- Dr. Senckenberg Institute of NeurooncologyUniversity HospitalGoethe University FrankfurtFrankfurt am MainGermany
- Division of Neuro‐OncologyHertie Institute of Clinical Brain ResearchUniversity Hospital TübingenTübingenGermany
| | - Joachim P. Steinbach
- Dr. Senckenberg Institute of NeurooncologyUniversity HospitalGoethe University FrankfurtFrankfurt am MainGermany
- University Cancer Center Frankfurt (UCT), University HospitalGoethe University FrankfurtFrankfurt am MainGermany
- German Cancer Consortium (DKTK), partner site Frankfurt, a partnership between DKFZ and University Hospital FrankfurtGoethe University FrankfurtFrankfurt am MainGermany
- Frankfurt Cancer Institute (FCI)University HospitalGoethe University FrankfurtFrankfurt am MainGermany
| | - Dieter Henrik Heiland
- Microenvironment and Immunology Research LaboratoryMedical CenterUniversity of FreiburgFreiburgGermany
- Department of NeurosurgeryMedical CenterUniversity of FreiburgFreiburgGermany
- Faculty of MedicineUniversity of FreiburgFreiburgGermany
- Faculty of Medicine and Medical CenterComprehensive Cancer Center Freiburg (CCCF)University of FreiburgFreiburgGermany
- German Cancer Consortium (DKTK), partner site Freiburg, a partnership between DKFZ and University Medical Center FreiburgFreiburgGermany
| | - Michael W. Ronellenfitsch
- Dr. Senckenberg Institute of NeurooncologyUniversity HospitalGoethe University FrankfurtFrankfurt am MainGermany
- University Cancer Center Frankfurt (UCT), University HospitalGoethe University FrankfurtFrankfurt am MainGermany
- German Cancer Consortium (DKTK), partner site Frankfurt, a partnership between DKFZ and University Hospital FrankfurtGoethe University FrankfurtFrankfurt am MainGermany
- Frankfurt Cancer Institute (FCI)University HospitalGoethe University FrankfurtFrankfurt am MainGermany
| |
Collapse
|
158
|
Ghafary I, Seoud T, Jorgensen M, Marhaba J, Briggs WM, Jamorabo DS. Inpatient Small Bowel Capsule Endoscopy: Not Associated With Bleeding Site Identification or 30-Day Readmission Prevention. Cureus 2024; 16:e74043. [PMID: 39712853 PMCID: PMC11661884 DOI: 10.7759/cureus.74043] [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] [Accepted: 11/19/2024] [Indexed: 12/24/2024] Open
Abstract
Background The utility of small bowel capsule endoscopy (SBCE) in the inpatient setting is controversial due to retention rates and costs. Aim This study aims to evaluate whether using SBCE significantly improved the identification of potential bleeding sites or reduced the risk of 30-day readmission for overt or occult gastrointestinal bleeding. Methods This was a single-center retrospective cohort study involving inpatients who underwent SBCE at a suburban tertiary care hospital from January 1, 2012, to January 1, 2022, for suspected small bowel bleeding. There was no control group used in this observational study. We used chi-square testing to determine the significance among our categorical variables and t-tests to compare means for our numerical variables. We also did multivariable logistic regression to analyze risk factors for increased hospital stay. All statistical analysis was done in R (R Core Team, 2020, R Foundation for Statistical Computing, Vienna, Austria). Results We identified 514 inpatients who underwent SBCE from January 1, 2012, to January 1, 2022, including 300 (58.4%) men and 214 (41.6%) women. Most (305/514, 59.3%) had no notable findings on SBCE, but 209/514 (40.7%) subsequently underwent endoscopic procedures, and a bleeding site was identified and treated in 168/209 (80.4%). Undergoing a subsequent procedure significantly increased the average number of days between capsule deployment and discharge (9.6 vs. 4.9 days, p < 0.005) without significantly reducing the risk for 30-day readmission (OR 1.33, 95% CI 0.9-1.9, p = 0.2). Among the 209 patients who had a subsequent procedure, identifying and treating a bleeding site did not significantly change readmission rates (OR 1.35, 95% CI 0.6-3.1, p = 0.5) compared to patients who did not have a procedure. Conclusion We did not find that inpatient SBCE significantly affected 30-day readmission rates even if an endoscopic procedure was subsequently done or a potential bleeding site was treated.
Collapse
Affiliation(s)
- Ismail Ghafary
- Gastroenterology and Hepatology, University of Connecticut School of Medicine, Farmington, USA
| | - Talal Seoud
- Gastroenterology and Hepatology, University of Florida, Gainesville, USA
| | - Michael Jorgensen
- Internal Medicine, Stony Brook University Hospital, Stony Brook, USA
| | - Jade Marhaba
- Gastroenterology and Hepatology, Stony Brook University Hospital, Stony Brook, USA
| | | | - Daniel S Jamorabo
- Gastroenterology and Hepatology, Stony Brook Medicine, Stony Brook, USA
| |
Collapse
|
159
|
Lange Y, Steck TL. How active cholesterol coordinates cell cholesterol homeostasis: Test of a hypothesis. Prog Lipid Res 2024; 96:101304. [PMID: 39491591 DOI: 10.1016/j.plipres.2024.101304] [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: 06/27/2024] [Revised: 10/23/2024] [Accepted: 11/01/2024] [Indexed: 11/05/2024]
Abstract
How do cells coordinate the diverse elements that regulate their cholesterol homeostasis? Our model postulates that membrane cholesterol forms simple complexes with bilayer phospholipids. The phospholipids in the plasma membrane are of high affinity; consequently, they are fully complexed with the sterol. This sets the resting level of plasma membrane cholesterol. Cholesterol in excess of the stoichiometric equivalence point of these complexes has high chemical activity; we refer to it as active cholesterol. It equilibrates with the low affinity phospholipids in the intracellular membranes where it serves as a negative feedback signal to a manifold of regulatory proteins that rein in ongoing cholesterol accretion. We tested the model with a review of the literature regarding fourteen homeostatic proteins in enterocytes. It provided strong albeit indirect support for the following hypothesis. Active cholesterol inhibits cholesterol uptake and biosynthesis by suppressing both the expression and the activity of the gene products activated by SREBP-2; namely, HMGCR, LDLR and NPC1L1. It also reduces free cell cholesterol by serving as the substrate for its esterification by ACAT and for the synthesis of side-chain oxysterols, 27-hydroxycholesterol in particular. The oxysterols drive cholesterol depletion by promoting the destruction of HMGCR and stimulating sterol esterification as well as the activation of LXR. The latter fosters the expression of multiple homeostatic proteins, including four transporters for which active cholesterol is the likely substrate. By nulling active cholesterol, the manifold maintains the cellular sterol at its physiologic set point.
Collapse
Affiliation(s)
- Yvonne Lange
- Department of Pathology, Rush University Medical Center, Chicago, IL 60612, United States of America.
| | - Theodore L Steck
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL 60637, United States of America
| |
Collapse
|
160
|
Caldovic L, Ahn JJ, Andricovic J, Balick VM, Brayer M, Chansky PA, Dawson T, Edwards AC, Felsen SE, Ismat K, Jagannathan SV, Mann BT, Medina JA, Morizono T, Morizono M, Salameh S, Vashist N, Williams EC, Zhou Z, Morizono H. Datamining approaches for examining the low prevalence of N-acetylglutamate synthase deficiency and understanding transcriptional regulation of urea cycle genes. J Inherit Metab Dis 2024; 47:1175-1193. [PMID: 37847851 PMCID: PMC11586597 DOI: 10.1002/jimd.12687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 10/09/2023] [Accepted: 10/10/2023] [Indexed: 10/19/2023]
Abstract
Ammonia, which is toxic to the brain, is converted into non-toxic urea, through a pathway of six enzymatically catalyzed steps known as the urea cycle. In this pathway, N-acetylglutamate synthase (NAGS, EC 2.3.1.1) catalyzes the formation of N-acetylglutamate (NAG) from glutamate and acetyl coenzyme A. NAGS deficiency (NAGSD) is the rarest of the urea cycle disorders, yet is unique in that ureagenesis can be restored with the drug N-carbamylglutamate (NCG). We investigated whether the rarity of NAGSD could be due to low sequence variation in the NAGS genomic region, high NAGS tolerance for amino acid replacements, and alternative sources of NAG and NCG in the body. We also evaluated whether the small genomic footprint of the NAGS catalytic domain might play a role. The small number of patients diagnosed with NAGSD could result from the absence of specific disease biomarkers and/or short NAGS catalytic domain. We screened for sequence variants in NAGS regulatory regions in patients suspected of having NAGSD and found a novel NAGS regulatory element in the first intron of the NAGS gene. We applied the same datamining approach to identify regulatory elements in the remaining urea cycle genes. In addition to the known promoters and enhancers of each gene, we identified several novel regulatory elements in their upstream regions and first introns. The identification of cis-regulatory elements of urea cycle genes and their associated transcription factors holds promise for uncovering shared mechanisms governing urea cycle gene expression and potentially leading to new treatments for urea cycle disorders.
Collapse
Affiliation(s)
- Ljubica Caldovic
- Center for Genetic Medicine ResearchChildren's National Research Institute, Children's National HospitalWashingtonDCUSA
- Department of Genomics and Precision Medicine, School of Medicine and Health SciencesThe George Washington UniversityWashingtonDCUSA
| | - Julie J. Ahn
- Department of Anatomy and Cell BiologyThe George Washington University School of Medicine and Health SciencesWashingtonDCUSA
| | - Jacklyn Andricovic
- Department of Anatomy and Cell BiologyThe George Washington University School of Medicine and Health SciencesWashingtonDCUSA
| | - Veronica M. Balick
- Department of Biochemistry and Molecular MedicineThe George Washington University School of Medicine and Health SciencesWashingtonDCUSA
| | - Mallory Brayer
- Department of Biological SciencesThe George Washington UniversityWashingtonDCUSA
| | - Pamela A. Chansky
- The Institute for Biomedical ScienceSchool of Medicine and Health Sciences, George Washington UniversityWashingtonDCUSA
| | - Tyson Dawson
- The Institute for Biomedical ScienceSchool of Medicine and Health Sciences, George Washington UniversityWashingtonDCUSA
- AMPEL BioSolutions LLCCharlottesvilleVirginiaUSA
| | - Alex C. Edwards
- The Institute for Biomedical ScienceSchool of Medicine and Health Sciences, George Washington UniversityWashingtonDCUSA
- Center for Neuroscience ResearchChildren's National Research Institute, Children's National HospitalWashingtonDCUSA
| | - Sara E. Felsen
- The Institute for Biomedical ScienceSchool of Medicine and Health Sciences, George Washington UniversityWashingtonDCUSA
- Center for Neuroscience ResearchChildren's National Research Institute, Children's National HospitalWashingtonDCUSA
| | - Karim Ismat
- Center for Genetic Medicine ResearchChildren's National Research Institute, Children's National HospitalWashingtonDCUSA
- Department of Genomics and Precision Medicine, School of Medicine and Health SciencesThe George Washington UniversityWashingtonDCUSA
| | - Sveta V. Jagannathan
- The Institute for Biomedical ScienceSchool of Medicine and Health Sciences, George Washington UniversityWashingtonDCUSA
| | - Brendan T. Mann
- Department of Microbiology, Immunology, and Tropical MedicineSchool of Medicine and Health Sciences, George Washington UniversityWashingtonDCUSA
| | - Jacob A. Medina
- The Institute for Biomedical ScienceSchool of Medicine and Health Sciences, George Washington UniversityWashingtonDCUSA
| | - Toshio Morizono
- College of Science and EngineeringUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Michio Morizono
- College of Science and EngineeringUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Shatha Salameh
- Department of Pharmacology & PhysiologySchool of Medicine and Health Sciences, George Washington UniversityWashingtonDCUSA
- Sheikh Zayed Institute for Pediatric Surgical InnovationChildren's National HospitalWashingtonDCUSA
| | - Neerja Vashist
- Center for Genetic Medicine ResearchChildren's National Research Institute, Children's National HospitalWashingtonDCUSA
- Department of Genomics and Precision Medicine, School of Medicine and Health SciencesThe George Washington UniversityWashingtonDCUSA
| | - Emily C. Williams
- Department of Anatomy and Cell BiologyThe George Washington University School of Medicine and Health SciencesWashingtonDCUSA
- The George Washington University Cancer Center, School of Medicine and Health SciencesGeorge Washington UniversityWashingtonDCUSA
| | - Zhe Zhou
- Department of Civil and Environmental EngineeringThe George Washington UniversityWashingtonDCUSA
| | - Hiroki Morizono
- Center for Genetic Medicine ResearchChildren's National Research Institute, Children's National HospitalWashingtonDCUSA
- Department of Genomics and Precision Medicine, School of Medicine and Health SciencesThe George Washington UniversityWashingtonDCUSA
| |
Collapse
|
161
|
Ganguly SC, Sangram S, Paul S, Kundu M. Phyto-nanotechnology: A novel beneficial strategy for Alzheimer's disease therapy. Neurochem Int 2024; 180:105868. [PMID: 39326498 DOI: 10.1016/j.neuint.2024.105868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 08/08/2024] [Accepted: 09/24/2024] [Indexed: 09/28/2024]
Abstract
Alzheimer's disease, a neurodegenerative condition, is characterized by the slow and progressive deterioration of the cognitive functions of geriatric patients. It occurs due to exacerbation of neurons in the brain, indicated by loss of memory, mood instability, and even death. The aggregation of amyloid β protein and neurofibrillary tangles-atypical forms of tau protein is the major cause of this disease. Phytoconstituents have been frequently employed in treating Alzheimer's disease. These natural compounds act through different molecular mechanisms to treat the disease. However, their potential in Alzheimer's disease therapy may be limited due to poor blood-brain barrier permeability, off-target effects, low bioavailability, etc. In recent times, nanotechnology has gained attraction to overcome these challenges. This article focuses on the potential phytoconstituents for Alzheimer's disease treatment and the associated limitations. Moreover, it highlights various nanoformulation strategies employed to penetrate the blood-brain barrier effectively, avoid side effects, improve bioavailability, and target specificity in treating Alzheimer's disease. The integration of nanotechnology with plant-derived compounds has the potential to revolutionize the therapeutic landscape for Alzheimer's disease.
Collapse
Affiliation(s)
| | - Sk Sangram
- Department of Pharmaceutical Chemistry, Calcutta Institute of Pharmaceutical Technology & Allied Health Sciences, West Bengal, India
| | - Sayani Paul
- Department of Pharmaceutical Chemistry, Calcutta Institute of Pharmaceutical Technology & Allied Health Sciences, West Bengal, India; Bengal School of Technology, Hooghly, West Bengal, India
| | - Moumita Kundu
- Department of Pharmaceutical Technology, Brainware University, West Bengal, India; Center for Multidisciplinary Research & Innovations, Brainware University, West Bengal, India.
| |
Collapse
|
162
|
Kato Y, Ariyoshi K, Nohara Y, Matsunaga N, Shimauchi T, Shindo N, Nishimura A, Mi X, Kim SG, Ide T, Kawanishi E, Ojida A, Nakashima N, Mori Y, Nishida M. Inhibition of dynamin-related protein 1-filamin interaction improves systemic glucose metabolism. Br J Pharmacol 2024; 181:4328-4347. [PMID: 38986570 DOI: 10.1111/bph.16487] [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: 09/22/2023] [Revised: 05/05/2024] [Accepted: 05/18/2024] [Indexed: 07/12/2024] Open
Abstract
BACKGROUND AND PURPOSE Maintaining mitochondrial quality is attracting attention as a new strategy to treat diabetes and diabetic complications. We previously reported that mitochondrial hyperfission by forming a protein complex between dynamin-related protein (Drp) 1 and filamin, mediates chronic heart failure and cilnidipine, initially developed as an L/N-type Ca2+ channel blocker, improves heart failure by inhibiting Drp1-filamin protein complex. We investigated whether cilnidipine improves hyperglycaemia of various diabetic mice models. EXPERIMENTAL APPROACH Retrospective analysis focusing on haemoglobin A1c (HbA1c) was performed in hypertensive and hyperglycaemic patients taking cilnidipine and amlodipine. After developing diabetic mice by streptozotocin (STZ) treatment, an osmotic pump including drug was implanted intraperitoneally, followed by weekly measurements of blood glucose levels. Mitochondrial morphology was analysed by electron microscopy. A Ca2+ channel-insensitive cilnidipine derivative (1,4-dihydropyridine [DHP]) was synthesized and its pharmacological effect was evaluated using obese (ob/ob) mice fed with high-fat diet (HFD). KEY RESULTS In patients, cilnidipine was superior to amlodipine in HbA1c lowering effect. Cilnidipine treatment improved systemic hyperglycaemia and mitochondrial morphological abnormalities in STZ-exposed mice, without lowering blood pressure. Cilnidipine failed to improve hyperglycaemia of ob/ob mice, with suppressing insulin secretion. 1,4-DHP improved hyperglycaemia and mitochondria abnormality in ob/ob mice fed HFD. 1,4-DHP and cilnidipine improved basal oxygen consumption rate of HepG2 cells cultured under 25 mM glucose. CONCLUSION AND IMPLICATIONS Inhibition of Drp1-filamin protein complex formation becomes a new strategy for type 2 diabetes treatment.
Collapse
Affiliation(s)
- Yuri Kato
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Kohei Ariyoshi
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Yasunobu Nohara
- Faculty of Advanced Science and Technology, Kumamoto University, Kumamoto, Japan
| | - Naoya Matsunaga
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Tsukasa Shimauchi
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
- Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- National Institute for Physiological Sciences (NIPS), National Institutes of Natural Sciences, Okazaki, Aichi, Japan
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Aichi, Japan
| | - Naoya Shindo
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Akiyuki Nishimura
- National Institute for Physiological Sciences (NIPS), National Institutes of Natural Sciences, Okazaki, Aichi, Japan
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Aichi, Japan
- SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Aichi, Japan
| | - Xinya Mi
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Sang Geon Kim
- College of Pharmacy, Dongguk University-Seoul, Goyang-si, South Korea
| | - Tomomi Ide
- Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Eiji Kawanishi
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Akio Ojida
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Naoki Nakashima
- Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yasuo Mori
- Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Motohiro Nishida
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
- National Institute for Physiological Sciences (NIPS), National Institutes of Natural Sciences, Okazaki, Aichi, Japan
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Aichi, Japan
- SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Aichi, Japan
| |
Collapse
|
163
|
Butrym M, Byvald F, Blanter M, Ringqvist EE, Vasylovska S, Marjomäki V, Lau J, Stone VM, Flodström-Tullberg M. Vemurafenib inhibits the replication of diabetogenic enteroviruses in intestinal epithelial and pancreatic beta cells. Antiviral Res 2024; 231:106021. [PMID: 39419452 DOI: 10.1016/j.antiviral.2024.106021] [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: 08/07/2024] [Revised: 09/27/2024] [Accepted: 10/14/2024] [Indexed: 10/19/2024]
Abstract
Enteroviruses, which infect via the gut, have been implicated in type 1 diabetes (T1D) development. Prolonged faecal shedding of enterovirus has been associated with islet autoimmunity. Additionally, enteroviral proteins and viral RNA have been detected in the pancreatic islets of individuals with recent-onset T1D, implicating their possible role in beta cell destruction. Despite this, no approved antiviral drugs currently exist that specifically target enterovirus infections for utilisation in disease interventions. Drug repurposing allows for the discovery of new clinical uses for existing drugs and can expedite drug discovery. Previously, the cancer drug Vemurafenib demonstrated unprecedented antiviral activity against several enteroviruses. In the present study, we assessed the efficacy of Vemurafenib and an analogue thereof in preventing infection or reducing the replication of enteroviruses associated with T1D. We tested Vemurafenib in intestinal epithelial cells (IECs) and insulin-producing beta cells. Additionally, we established a protocol for infecting human stem cell-derived islets (SC-islets) and used Vemurafenib and its analogue in this model. Our studies revealed that Vemurafenib exhibited strong antiviral properties in IECs and a beta cell line. The antiviral effect was also seen with the Vemurafenib analogue. SC-islets expressed the viral receptors CAR and DAF, with their highest expression in insulin- and glucagon-positive cells, respectively. SC-islets were successfully infected by CVBs and the antiviral activity of Vemurafenib and its analogue was confirmed in most SC-islet batches. In summary, our observations suggest that Vemurafenib and its analogue warrant further exploration as potential antiviral agents for the treatment of enterovirus-induced diseases, including T1D.
Collapse
Affiliation(s)
- Marta Butrym
- Center for Infectious Medicine, Department of Medicine Huddinge / ANA Futura, Karolinska Institutet, Alfred Nobels Allé 8, 141 52, Stockholm, Sweden.
| | - Fabian Byvald
- Center for Infectious Medicine, Department of Medicine Huddinge / ANA Futura, Karolinska Institutet, Alfred Nobels Allé 8, 141 52, Stockholm, Sweden.
| | - Marfa Blanter
- Center for Infectious Medicine, Department of Medicine Huddinge / ANA Futura, Karolinska Institutet, Alfred Nobels Allé 8, 141 52, Stockholm, Sweden.
| | - Emma E Ringqvist
- Center for Infectious Medicine, Department of Medicine Huddinge / ANA Futura, Karolinska Institutet, Alfred Nobels Allé 8, 141 52, Stockholm, Sweden.
| | - Svitlana Vasylovska
- Department of Medical Cell Biology, Uppsala University, BOX 571, 751 23, Uppsala, Sweden.
| | - Varpu Marjomäki
- Department of Biological and Environmental Science, University of Jyväskylä, P.O. Box 35, FI-40014, Jyväskylä, Finland.
| | - Joey Lau
- Department of Medical Cell Biology, Uppsala University, BOX 571, 751 23, Uppsala, Sweden.
| | - Virginia M Stone
- Center for Infectious Medicine, Department of Medicine Huddinge / ANA Futura, Karolinska Institutet, Alfred Nobels Allé 8, 141 52, Stockholm, Sweden.
| | - Malin Flodström-Tullberg
- Center for Infectious Medicine, Department of Medicine Huddinge / ANA Futura, Karolinska Institutet, Alfred Nobels Allé 8, 141 52, Stockholm, Sweden.
| |
Collapse
|
164
|
Ansari P, Khan JT, Chowdhury S, Reberio AD, Kumar S, Seidel V, Abdel-Wahab YHA, Flatt PR. Plant-Based Diets and Phytochemicals in the Management of Diabetes Mellitus and Prevention of Its Complications: A Review. Nutrients 2024; 16:3709. [PMID: 39519546 PMCID: PMC11547802 DOI: 10.3390/nu16213709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 09/27/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Diabetes mellitus (DM) is currently regarded as a global public health crisis for which lifelong treatment with conventional drugs presents limitations in terms of side effects, accessibility, and cost. Type 2 diabetes (T2DM), usually associated with obesity, is characterized by elevated blood glucose levels, hyperlipidemia, chronic inflammation, impaired β-cell function, and insulin resistance. If left untreated or when poorly controlled, DM increases the risk of vascular complications such as hypertension, nephropathy, neuropathy, and retinopathy, which can be severely debilitating or life-threatening. Plant-based foods represent a promising natural approach for the management of T2DM due to the vast array of phytochemicals they contain. Numerous epidemiological studies have highlighted the importance of a diet rich in plant-based foods (vegetables, fruits, spices, and condiments) in the prevention and management of DM. Unlike conventional medications, such natural products are widely accessible, affordable, and generally free from adverse effects. Integrating plant-derived foods into the daily diet not only helps control the hyperglycemia observed in DM but also supports weight management in obese individuals and has broad health benefits. In this review, we provide an overview of the pathogenesis and current therapeutic management of DM, with a particular focus on the promising potential of plant-based foods.
Collapse
Affiliation(s)
- Prawej Ansari
- Comprehensive Diabetes Center, Heersink School of Medicine, University of Alabama, Birmingham (UAB), Birmingham, AL 35233, USA
- School of Pharmacy and Public Health, Department of Pharmacy, Independent University, Bangladesh (IUB), Dhaka 1229, Bangladesh
- Centre for Diabetes Research, School of Biomedical Sciences, Ulster University, Coleraine BT52 1SA, UK; (Y.H.A.A.-W.); (P.R.F.)
| | - Joyeeta T. Khan
- School of Pharmacy and Public Health, Department of Pharmacy, Independent University, Bangladesh (IUB), Dhaka 1229, Bangladesh
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences (UAMS), Little Rock, AR 72205, USA
| | - Suraiya Chowdhury
- School of Pharmacy and Public Health, Department of Pharmacy, Independent University, Bangladesh (IUB), Dhaka 1229, Bangladesh
| | - Alexa D. Reberio
- School of Pharmacy and Public Health, Department of Pharmacy, Independent University, Bangladesh (IUB), Dhaka 1229, Bangladesh
| | - Sandeep Kumar
- Comprehensive Diabetes Center, Heersink School of Medicine, University of Alabama, Birmingham (UAB), Birmingham, AL 35233, USA
| | - Veronique Seidel
- Natural Products Research Laboratory, Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, UK;
| | - Yasser H. A. Abdel-Wahab
- Centre for Diabetes Research, School of Biomedical Sciences, Ulster University, Coleraine BT52 1SA, UK; (Y.H.A.A.-W.); (P.R.F.)
| | - Peter R. Flatt
- Centre for Diabetes Research, School of Biomedical Sciences, Ulster University, Coleraine BT52 1SA, UK; (Y.H.A.A.-W.); (P.R.F.)
| |
Collapse
|
165
|
Wu J, Zhuang W, Lu K, Zhang L, Wang Y, Chai F, Liang XF. Study on the Function of Leptin Nutrient Acquisition and Energy Metabolism of Zebrafish ( Danio rerio). Int J Mol Sci 2024; 25:11647. [PMID: 39519205 PMCID: PMC11546987 DOI: 10.3390/ijms252111647] [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: 10/08/2024] [Revised: 10/22/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
Leptin plays an indispensable role in energy homeostasis, and its involvement in metabolic activities has been extensively explored in fish. We generated mutant lines of leptina (-5 bp) and leptinb (+8 bp) in zebrafish using CRISPR/Cas9 technology to explore the metabolic characteristics of lepa and lepb mutant zebrafish in response to high glucose nutritional stress induced by high levels of carbohydrates. The results were as follows: the body weight and food intake of adult zebrafish of the two mutant species were increased; the visceral fat accumulation, whole-body crude lipid, and crude protein contents of lepb-/- were increased; and the visceral fat accumulation and crude lipid in lepa-/- zebrafish were decreased. The blood glucose levels of the two mutant zebrafish were increased, the mRNA expression levels of glycolytic genes pk and gck were decreased in the two mutant zebrafish, and there were differences between lepa-/- and lepb-/- zebrafish. The expressions of glycogen synthesis and decomposition genes were inhibited and promoted, respectively. The expression of adipose synthesis genes in the liver and muscle was stimulated in lepb-/- zebrafish but suppressed in lepa-/- zebrafish. Lipolysis and oxidation genes were also stimulated in lepa-/- zebrafish livers, while the livers of lepb-/- zebrafish were stimulated but muscle was inhibited. In conclusion, the results indicate that lepa plays a major role in glucose metabolism, which is conducive to promoting glucose utilization and lipogenesis, while lepb mainly promotes lipolysis and oxidation, regulates protein generation, and plays a minor role in glucose metabolism.
Collapse
Affiliation(s)
- Jiaqi Wu
- College of Fisheries, Chinese Perch Research Center, Huazhong Agricultural University, Wuhan 430070, China; (J.W.); (W.Z.); (K.L.); (L.Z.); (Y.W.); (F.C.)
- Engineering Research Center of Green development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan 430070, China
| | - Wuyuan Zhuang
- College of Fisheries, Chinese Perch Research Center, Huazhong Agricultural University, Wuhan 430070, China; (J.W.); (W.Z.); (K.L.); (L.Z.); (Y.W.); (F.C.)
- Engineering Research Center of Green development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan 430070, China
| | - Ke Lu
- College of Fisheries, Chinese Perch Research Center, Huazhong Agricultural University, Wuhan 430070, China; (J.W.); (W.Z.); (K.L.); (L.Z.); (Y.W.); (F.C.)
- Engineering Research Center of Green development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan 430070, China
| | - Lixin Zhang
- College of Fisheries, Chinese Perch Research Center, Huazhong Agricultural University, Wuhan 430070, China; (J.W.); (W.Z.); (K.L.); (L.Z.); (Y.W.); (F.C.)
- Engineering Research Center of Green development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan 430070, China
| | - Yuye Wang
- College of Fisheries, Chinese Perch Research Center, Huazhong Agricultural University, Wuhan 430070, China; (J.W.); (W.Z.); (K.L.); (L.Z.); (Y.W.); (F.C.)
- Engineering Research Center of Green development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan 430070, China
| | - Farui Chai
- College of Fisheries, Chinese Perch Research Center, Huazhong Agricultural University, Wuhan 430070, China; (J.W.); (W.Z.); (K.L.); (L.Z.); (Y.W.); (F.C.)
- Engineering Research Center of Green development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan 430070, China
| | - Xu-Fang Liang
- College of Fisheries, Chinese Perch Research Center, Huazhong Agricultural University, Wuhan 430070, China; (J.W.); (W.Z.); (K.L.); (L.Z.); (Y.W.); (F.C.)
- Engineering Research Center of Green development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan 430070, China
| |
Collapse
|
166
|
Playford RJ. Effects of Chicken Egg Powder, Bovine Colostrum, and Combination Therapy for the Treatment of Gastrointestinal Disorders. Nutrients 2024; 16:3684. [PMID: 39519517 PMCID: PMC11547998 DOI: 10.3390/nu16213684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 10/26/2024] [Accepted: 10/27/2024] [Indexed: 11/16/2024] Open
Abstract
Natural-based products are of interest to the pharmaceutical industry as potential sources of novel medicinal compounds. They are also used by consumers/patients as standalone therapies or as an adjunct to Western medicines. Two natural-based products of interest are chicken egg and bovine colostrum (the milk produced in the first few days following calving). Both products are rich in immunoglobulins, antimicrobial peptides, growth factors, and macro- and micro-nutrients. In vitro, in vivo, and a limited number of clinical studies suggest therapeutic benefits of both components given alone and together. Combination therapy is of particular interest, as preclinical studies suggest synergistic effects on growth, repair, and gut protection, including microbiome-induced damage. This article describes the main constituents of egg and bovine colostrum, studies of their use alone and together for a wide range of conditions, highlights areas requiring further research, and describes novel indications such as GLP-1-associated gut symptoms. While well placed in the food supplement arena, additional high-quality clinical trials are required to establish their benefits in clinical practice.
Collapse
Affiliation(s)
- Raymond John Playford
- School of Medical & Biomedical Sciences, University of West London, St Mary's Road, Ealing, London W5 3TX, UK
| |
Collapse
|
167
|
Wang J, Zhou Y, Liu Y, Sang M, Ding Y, Li T, Wang X, Carvalho V, Ni C, Wang Q, Cai Z, Wang H, Chen Y, Shang Z, Wang D, Qiu S, Sun Z. Association between night blindness history and risk of diabetes in the Chinese population: a multi-center, cross sectional study. BMC Endocr Disord 2024; 24:231. [PMID: 39472978 PMCID: PMC11520579 DOI: 10.1186/s12902-024-01721-2] [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: 04/06/2024] [Accepted: 09/05/2024] [Indexed: 11/02/2024] Open
Abstract
AIMS Night blindness (NB), an important manifestation of VA deficiency, may be associated with the odds of diabetes. The aim of this study was to explore the probable association between NB history and diabetes in Chinese community-dwelling adults. METHODS This multi-center, cross-sectional study enrolled a total of 5664 participants aged 18-82 years from eight sites in China. Information on demographics and medical history was collected using a standardized questionnaire. Diabetes was diagnosed based on the oral glucose tolerance test or a self-reported history. NB history was ascertained by a face-to-face interview with reference to the recommendation by the World Health Organization. Logistic regression analysis was used to evaluate the association between NB history and the odds of diabetes. RESULTS A total of 5049 participants were finally included, with 252 ascertained with NB history and 1076 with diabetes. The mean age of included participants was 52.9 years, and the percentage of participants with NB history was significantly higher in participants with diabetes than those without (7.0% vs. 4.5%). The multivariable adjusted odds ratio for diabetes was 1.41 (95% confidence interval 1.06, 1.89) in participants with NB history compared with those without. Furthermore, mediation analysis showed that obesity, as assessed by waist-height ratio, partially mediated the relationship between NB history and increased odds of diabetes. CONCLUSIONS The results suggest that NB history might be associated with increased odds of diabetes in Chinese community-dwelling adults.
Collapse
Affiliation(s)
- Jinbang Wang
- Department of Endocrinology, Zhongda Hospital, Institute of Diabetes, School of Medicine, Southeast University, Nanjing, China
- Northern Jiangsu People's Hospital, Yangzhou, Jiangsu, 225001, P.R. China
| | - Yunting Zhou
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yu Liu
- Department of Endocrinology, Zhongda Hospital, Institute of Diabetes, School of Medicine, Southeast University, Nanjing, China
| | - Miaomiao Sang
- Department of Endocrinology, Zhongda Hospital, Institute of Diabetes, School of Medicine, Southeast University, Nanjing, China
| | - Yuzhi Ding
- Department of Ophthalmology, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, China
| | - Tingting Li
- Department of Endocrinology, Zhongda Hospital, Institute of Diabetes, School of Medicine, Southeast University, Nanjing, China
| | - Xiaohang Wang
- Institute of Translational Medicine, Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College, Yangzhou University, Yangzhou, 225009, P. R. China
| | - Vladmir Carvalho
- Department of Endocrinology, Zhongda Hospital, Institute of Diabetes, School of Medicine, Southeast University, Nanjing, China
| | - Chengming Ni
- Department of Endocrinology, Zhongda Hospital, Institute of Diabetes, School of Medicine, Southeast University, Nanjing, China
| | - Qianqian Wang
- Department of Endocrinology, Zhongda Hospital, Institute of Diabetes, School of Medicine, Southeast University, Nanjing, China
| | - Zhensheng Cai
- Department of Endocrinology, Zhongda Hospital, Institute of Diabetes, School of Medicine, Southeast University, Nanjing, China
| | - Huan Wang
- Department of Endocrinology, Zhongda Hospital, Institute of Diabetes, School of Medicine, Southeast University, Nanjing, China
| | - Yang Chen
- Department of Endocrinology, Zhongda Hospital, Institute of Diabetes, School of Medicine, Southeast University, Nanjing, China
| | - Zhanjia Shang
- Department of Endocrinology, Zhongda Hospital, Institute of Diabetes, School of Medicine, Southeast University, Nanjing, China
| | - Duolao Wang
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Shanhu Qiu
- Department of General Practice, Zhongda Hospital, Institute of Diabetes, School of Medicine, Southeast University, Nanjing, China.
| | - Zilin Sun
- Department of Endocrinology, Zhongda Hospital, Institute of Diabetes, School of Medicine, Southeast University, Nanjing, China.
| |
Collapse
|
168
|
Esparza D, Lima C, Abuelreich S, Ghaeli I, Hwang J, Oh E, Lenz A, Gu A, Jiang N, Kandeel F, Thurmond DC, Jovanovic-Talisman T. Pancreatic β-cells package double C2-like domain beta protein into extracellular vesicles via tandem C2 domains. Front Endocrinol (Lausanne) 2024; 15:1451279. [PMID: 39497805 PMCID: PMC11532064 DOI: 10.3389/fendo.2024.1451279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 09/10/2024] [Indexed: 11/07/2024] Open
Abstract
Introduction Double C2-like domain beta (DOC2B) is a vesicle priming protein critical for glucose-stimulated insulin secretion in β-cells. Individuals with type 1 diabetes (T1D) have lower levels of DOC2B in their residual functional β-cell mass and platelets, a phenotype also observed in a mouse model of T1D. Thus, DOC2B levels could provide important information on β-cell dys(function). Objective Our objective was to evaluate the DOC2B secretome of β-cells. In addition to soluble extracellular protein, we assessed DOC2B localized within membrane-delimited nanoparticles - extracellular vesicles (EVs). Moreover, in rat clonal β-cells, we probed domains required for DOC2B sorting into EVs. Method Using Single Extracellular VEsicle Nanoscopy, we quantified EVs derived from clonal β-cells (human EndoC-βH1, rat INS-1 832/13, and mouse MIN6); two other cell types known to regulate glucose homeostasis and functionally utilize DOC2B (skeletal muscle rat myotube L6-GLUT4myc and human neuronal-like SH-SY5Y cells); and human islets sourced from individuals with no diabetes (ND). EVs derived from ND human plasma, ND human islets, and cell lines were isolated with either size exclusion chromatography or differential centrifugation. Isolated EVs were comprehensively characterized using dotblots, transmission electron microscopy, nanoparticle tracking analysis, and immunoblotting. Results DOC2B was present within EVs derived from ND human plasma, ND human islets, and INS-1 832/13 β-cells. Compared to neuronal-like SH-SY5Y cells and L6-GLUT4myc myotubes, clonal β-cells (EndoC-βH1, INS-1 832/13, and MIN6) produced significantly more EVs. DOC2B levels in EVs (over whole cell lysates) were higher in INS-1 832/13 β-cells compared to L6-GLUT4myc myotubes; SH-SY5Y neuronal-like cells did not release appreciable DOC2B. Mechanistically, we show that DOC2B was localized to the EV lumen; the tandem C2 domains were sufficient to confer sorting to INS-1 832/13 β-cell EVs. Discussion Clonal β-cells and ND human islets produce abundant EVs. In cell culture, appreciable DOC2B can be packaged into EVs, and a small fraction is excreted as a soluble protein. While DOC2B-laden EVs and soluble protein are present in ND plasma, further studies will be necessary to determine if DOC2B originating from β-cells significantly contributes to the plasma secretome.
Collapse
Affiliation(s)
- Diana Esparza
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute at City of Hope, Duarte, CA, United States
| | - Carinna Lima
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute at City of Hope, Duarte, CA, United States
| | - Sarah Abuelreich
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute at City of Hope, Duarte, CA, United States
| | - Ima Ghaeli
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute at City of Hope, Duarte, CA, United States
| | - Jinhee Hwang
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute at City of Hope, Duarte, CA, United States
| | - Eunjin Oh
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute at City of Hope, Duarte, CA, United States
| | - Ayelet Lenz
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute at City of Hope, Duarte, CA, United States
| | - Angel Gu
- Department of Translational Research and Cellular Therapeutics, Beckman Research Institute at City of Hope, Duarte, CA, United States
| | - Nan Jiang
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute at City of Hope, Duarte, CA, United States
| | - Fouad Kandeel
- Department of Translational Research and Cellular Therapeutics, Beckman Research Institute at City of Hope, Duarte, CA, United States
| | - Debbie C. Thurmond
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute at City of Hope, Duarte, CA, United States
| | - Tijana Jovanovic-Talisman
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute at City of Hope, Duarte, CA, United States
| |
Collapse
|
169
|
Pais R, Conde T, Neves BB, Pinho M, Coelho M, Pereira H, Rodrigues AMC, Domingues P, Gomes AM, Urbatzka R, Domingues R, Melo T. Bioactive Lipids in Dunaliella salina: Implications for Functional Foods and Health. Foods 2024; 13:3321. [PMID: 39456383 PMCID: PMC11507028 DOI: 10.3390/foods13203321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/11/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
Dunaliella salina is a green microalga extensively explored for β-carotene production, while knowledge of its lipid composition is still limited and poorly investigated. Among lipids, polar lipids have been highlighted as bioactive phytochemicals with health-promoting properties. This research aimed to provide an in-depth lipidome profiling of D. salina using liquid and gas chromatography coupled with mass spectrometry. The lipid content was 6.8%, including phospholipids, glycolipids, betaine lipids, sphingolipids, triglycerides, diglycerides, and pigments. Among the total esterified fatty acids, 13.6% were 18:3 omega-3 and 14.7% were 18:1 omega-9. The lipid extract of D. salina showed anti-inflammatory activity by inhibiting cyclooxygenase-2 activity at 100 µg/mL, dose-dependent antioxidant scavenging activity, and antidiabetic activity by inhibiting α-glucosidase activity at 25 and 125 µg/mL. In conclusion, the lipid extract of D. salina has the potential to be used as a functional food ingredient or in the nutraceutical and cosmeceutical industries.
Collapse
Affiliation(s)
- Rita Pais
- CESAM—Centre for Environmental and Marine Studies, Department of Chemistry, University of Aveiro, Santiago University Campus, 3810-193 Aveiro, Portugal; (R.P.); (T.C.); (B.B.N.); (M.P.); (R.D.)
- Mass Spectrometry Centre, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Santiago University Campus, 3810-193 Aveiro, Portugal;
| | - Tiago Conde
- CESAM—Centre for Environmental and Marine Studies, Department of Chemistry, University of Aveiro, Santiago University Campus, 3810-193 Aveiro, Portugal; (R.P.); (T.C.); (B.B.N.); (M.P.); (R.D.)
- Mass Spectrometry Centre, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Santiago University Campus, 3810-193 Aveiro, Portugal;
| | - Bruna B. Neves
- CESAM—Centre for Environmental and Marine Studies, Department of Chemistry, University of Aveiro, Santiago University Campus, 3810-193 Aveiro, Portugal; (R.P.); (T.C.); (B.B.N.); (M.P.); (R.D.)
- Mass Spectrometry Centre, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Santiago University Campus, 3810-193 Aveiro, Portugal;
| | - Marisa Pinho
- CESAM—Centre for Environmental and Marine Studies, Department of Chemistry, University of Aveiro, Santiago University Campus, 3810-193 Aveiro, Portugal; (R.P.); (T.C.); (B.B.N.); (M.P.); (R.D.)
| | - Marta Coelho
- CBQF—Centro de Biotecnologia e Química Fina, Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal; (M.C.); (A.M.G.)
| | - Hugo Pereira
- GreenCoLab—Associação Oceano Verde, University of Algarve, Campus de Gambelas, 8005-139 Faro, Portugal;
| | | | - Pedro Domingues
- Mass Spectrometry Centre, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Santiago University Campus, 3810-193 Aveiro, Portugal;
| | - Ana Maria Gomes
- CBQF—Centro de Biotecnologia e Química Fina, Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal; (M.C.); (A.M.G.)
| | - Ralph Urbatzka
- Biodiscovery for Health Group, CIIMAR/CIMAR, Interdisciplinary Centre of Marine and Environmental Research, Terminal de Cruzeiros do Porto de Leixões, University of Porto, 4450-208 Matosinhos, Portugal;
| | - Rosário Domingues
- CESAM—Centre for Environmental and Marine Studies, Department of Chemistry, University of Aveiro, Santiago University Campus, 3810-193 Aveiro, Portugal; (R.P.); (T.C.); (B.B.N.); (M.P.); (R.D.)
- Mass Spectrometry Centre, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Santiago University Campus, 3810-193 Aveiro, Portugal;
| | - Tânia Melo
- CESAM—Centre for Environmental and Marine Studies, Department of Chemistry, University of Aveiro, Santiago University Campus, 3810-193 Aveiro, Portugal; (R.P.); (T.C.); (B.B.N.); (M.P.); (R.D.)
- Mass Spectrometry Centre, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Santiago University Campus, 3810-193 Aveiro, Portugal;
| |
Collapse
|
170
|
Mutlu B, Sharabi K, Sohn JH, Yuan B, Latorre-Muro P, Qin X, Yook JS, Lin H, Yu D, Camporez JPG, Kajimura S, Shulman GI, Hui S, Kamenecka TM, Griffin PR, Puigserver P. Small molecules targeting selective PCK1 and PGC-1α lysine acetylation cause anti-diabetic action through increased lactate oxidation. Cell Chem Biol 2024; 31:1772-1786.e5. [PMID: 39341205 PMCID: PMC11500315 DOI: 10.1016/j.chembiol.2024.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 06/27/2024] [Accepted: 09/04/2024] [Indexed: 09/30/2024]
Abstract
Small molecules selectively inducing peroxisome proliferator-activated receptor-gamma coactivator (PGC)-1α acetylation and inhibiting glucagon-dependent gluconeogenesis causing anti-diabetic effects have been identified. However, how these small molecules selectively suppress the conversion of gluconeogenic metabolites into glucose without interfering with lipogenesis is unknown. Here, we show that a small molecule SR18292 inhibits hepatic glucose production by increasing lactate and glucose oxidation. SR18292 increases phosphoenolpyruvate carboxykinase 1 (PCK1) acetylation, which reverses its gluconeogenic reaction and favors oxaloacetate (OAA) synthesis from phosphoenolpyruvate. PCK1 reverse catalytic reaction induced by SR18292 supplies OAA to tricarboxylic acid (TCA) cycle and is required for increasing glucose and lactate oxidation and suppressing gluconeogenesis. Acetylation mimetic mutant PCK1 K91Q favors anaplerotic reaction and mimics the metabolic effects of SR18292 in hepatocytes. Liver-specific expression of PCK1 K91Q mutant ameliorates hyperglycemia in obese mice. Thus, SR18292 blocks gluconeogenesis by enhancing gluconeogenic substrate oxidation through PCK1 lysine acetylation, supporting the anti-diabetic effects of these small molecules.
Collapse
Affiliation(s)
- Beste Mutlu
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Kfir Sharabi
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02215, USA; Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Jee Hyung Sohn
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Bo Yuan
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, 655 Huntington Avenue, Boston, MA 02115, USA
| | - Pedro Latorre-Muro
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Xin Qin
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Jin-Seon Yook
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Hua Lin
- Department of Molecular Medicine, The Wertheim UF Scripps Institute for Biomedical Innovation and Technology, University of Florida, Jupiter, FL 33458, USA
| | - Deyang Yu
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02215, USA
| | - João Paulo G Camporez
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06520-8020, USA; Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT 06520-8020, USA
| | - Shingo Kajimura
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA; Howard Hughes Medical Institute, Chevy Chase, MD 020815, USA
| | - Gerald I Shulman
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06520-8020, USA; Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT 06520-8020, USA; Howard Hughes Medical Institute, Chevy Chase, MD 020815, USA
| | - Sheng Hui
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, 655 Huntington Avenue, Boston, MA 02115, USA
| | - Theodore M Kamenecka
- Department of Molecular Medicine, The Wertheim UF Scripps Institute for Biomedical Innovation and Technology, University of Florida, Jupiter, FL 33458, USA
| | - Patrick R Griffin
- Department of Molecular Medicine, The Wertheim UF Scripps Institute for Biomedical Innovation and Technology, University of Florida, Jupiter, FL 33458, USA
| | - Pere Puigserver
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
171
|
Xu R, Zhang L, Pan H, Zhang Y. Retinoid X receptor heterodimers in hepatic function: structural insights and therapeutic potential. Front Pharmacol 2024; 15:1464655. [PMID: 39478961 PMCID: PMC11521896 DOI: 10.3389/fphar.2024.1464655] [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: 07/14/2024] [Accepted: 09/30/2024] [Indexed: 11/02/2024] Open
Abstract
Nuclear receptors (NRs) are key regulators of multiple physiological functions and pathological changes in the liver in response to a variety of extracellular signaling changes. Retinoid X receptor (RXR) is a special member of the NRs, which not only responds to cellular signaling independently, but also regulates multiple signaling pathways by forming heterodimers with various other NR. Therefore, RXR is widely involved in hepatic glucose metabolism, lipid metabolism, cholesterol metabolism and bile acid homeostasis as well as hepatic fibrosis. Specific activation of particular dimers regulating physiological and pathological processes may serve as important pharmacological targets. So here we describe the basic information and structural features of the RXR protein and its heterodimers, focusing on the role of RXR heterodimers in a number of physiological processes and pathological imbalances in the liver, to provide a theoretical basis for RXR as a promising drug target.
Collapse
Affiliation(s)
- Renjie Xu
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Linyue Zhang
- Department of Ultrasound, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hao Pan
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong Zhang
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
172
|
Pareek A, Singhal R, Pareek A, Ghazi T, Kapoor DU, Ratan Y, Singh AK, Jain V, Chuturgoon AA. Retinoic acid in Parkinson's disease: Molecular insights, therapeutic advances, and future prospects. Life Sci 2024; 355:123010. [PMID: 39181315 DOI: 10.1016/j.lfs.2024.123010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 08/17/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
Parkinson's disease (PD) is a common and progressively worsening neurodegenerative disorder characterized by abnormal protein homeostasis and the degeneration of dopaminergic neurons, particularly in the substantia nigra pars compacta. The prevalence of PD has doubled in the past 25 years, now affecting over 8.5 million individuals worldwide, underscoring the need for effective management strategies. While current pharmacological therapies provide symptom relief, they face challenges in treating advanced PD stages. Recent research highlights the therapeutic benefits of retinoic acid (RA) in PD, demonstrating its potential to mitigate neuroinflammation and oxidative stress, regulate brain aging, promote neuronal plasticity, and influence circadian rhythm gene expression and retinoid X receptor heterodimerization. Additionally, RA helps maintain intestinal homeostasis and modulates the enteric nervous system, presenting significant therapeutic potential for managing PD. This review explores RA as a promising alternative to conventional therapies by summarizing the molecular mechanisms underlying its role in PD pathophysiology and presenting up-to-date insights into both preclinical and clinical studies of RA in PD treatment. It also delves into cutting-edge formulations incorporating RA, highlighting ongoing efforts to refine therapeutic strategies by integrating RA into novel treatments. This comprehensive overview aims to advance progress in the field, contribute to the development of effective, targeted treatments for PD, and enhance patient well-being. Further research is essential to fully explore RA's therapeutic potential and validate its efficacy in PD treatment.
Collapse
Affiliation(s)
- Ashutosh Pareek
- Department of Pharmacy, Banasthali Vidyapith, Banasthali 304022, India.
| | - Runjhun Singhal
- Department of Pharmacy, Banasthali Vidyapith, Banasthali 304022, India
| | - Aaushi Pareek
- Department of Pharmacy, Banasthali Vidyapith, Banasthali 304022, India
| | - Terisha Ghazi
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban 4041, South Africa
| | | | - Yashumati Ratan
- Department of Pharmacy, Banasthali Vidyapith, Banasthali 304022, India
| | - Arun Kumar Singh
- Department of Pharmacy, Vivekananda Global University, Jaipur 303012, India
| | - Vivek Jain
- Department of Pharmaceutical Sciences, Mohanlal Sukhadia University, Udaipur 313001, India
| | - Anil A Chuturgoon
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban 4041, South Africa.
| |
Collapse
|
173
|
Huynh TN, Havrda MC, Zanazzi GJ, Chang CCY, Chang TY. Inhibiting the Cholesterol Storage Enzyme ACAT1/SOAT1 in Myelin Debris-Treated Microglial Cell Lines Activates the Gene Expression of Cholesterol Efflux Transporter ABCA1. Biomolecules 2024; 14:1301. [PMID: 39456234 PMCID: PMC11505751 DOI: 10.3390/biom14101301] [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: 09/17/2024] [Revised: 10/11/2024] [Accepted: 10/12/2024] [Indexed: 10/28/2024] Open
Abstract
Aging is the major risk factor for Alzheimer's disease (AD). In the aged brain, myelin debris accumulates and is cleared by microglia. Phagocytosed myelin debris increases neutral lipid droplet content in microglia. Neutral lipids include cholesteryl esters (CE) and triacylglycerol (TAG). To examine the effects of myelin debris on neutral lipid content in microglia, we added myelin debris to human HMC3 and mouse N9 cells. The results obtained when using 3H-oleate as a precursor in intact cells reveal that myelin debris significantly increases the biosynthesis of CE but not TAG. Mass analyses have shown that myelin debris increases both CE and TAG. The increase in CE biosynthesis was abolished using inhibitors of the cholesterol storage enzyme acyl-CoA:cholesterol acyltransferase 1 (ACAT1/SOAT1). ACAT1 inhibitors are promising drug candidates for AD treatment. In myelin debris-loaded microglia, treatment with two different ACAT1 inhibitors, K604 and F12511, increased the mRNA and protein content of ATP-binding cassette subfamily A1 (ABCA1), a protein that is located at the plasma membrane and which controls cellular cholesterol disposal. The effect of the ACAT1 inhibitor on ABCA1 was abolished by preincubating cells with the liver X receptor (LXR) antagonist GSK2033. We conclude that ACAT1 inhibitors prevent the accumulation of cholesterol and CE in myelin debris-treated microglia by activating ABCA1 gene expression via the LXR pathway.
Collapse
Affiliation(s)
- Thao N. Huynh
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA;
| | - Matthew C. Havrda
- Department of Molecular and System Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA;
| | - George J. Zanazzi
- Department of Pathology and Laboratory Medicine, Dartmouth–Hitchcock Medical Center, Lebanon, NH 03766, USA;
| | - Catherine C. Y. Chang
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA;
| | - Ta Yuan Chang
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA;
| |
Collapse
|
174
|
Abeysekera MV, Ni D, Gilbert L, Hibbert E, Nanan R. Linking the reversal of gestational insulin resistance to postpartum depression. BMC Med 2024; 22:433. [PMID: 39379948 PMCID: PMC11462660 DOI: 10.1186/s12916-024-03659-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 09/26/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND Postpartum depression (PPD) constitutes a significant mental health disorder affecting almost one fifth of pregnancies globally. Despite extensive research, the precise etiological mechanisms underlying PPD remain elusive. However, several risk factors like genetic predisposition, hormonal fluctuations, and stress-related environmental and psychosocial triggers have been found to be implicated in its development. MAIN: Recently, an increased risk of PPD has been reported to be associated with gestational diabetes mellitus (GDM), which is characterized by the disruption of glucose metabolism, primarily attributed to the emergence of insulin resistance (IR). While IR during pregnancy seems to be an evolutionary adaptative mechanism to handle the profound metabolic alterations during pregnancy, its subsequent resolution following delivery necessitates a reconfiguration of the metabolic landscape in both peripheral tissues and the central nervous system (CNS). Considering the pivotal roles of energy metabolism, particularly glucose metabolism, in CNS functions, we propose a novel model that such pronounced changes in IR and the associated glucose metabolism seen postpartum might account for PPD development. This concept is based on the profound influences from insulin and glucose metabolism on brain functions, potentially via modulating neurotransmitter actions of dopamine and serotonin. Their sudden postpartum disruption is likely to be linked to mood changes, as observed in PPD. CONCLUSIONS The detailed pathogenesis of PPD might be multifactorial and still remains to be fully elucidated. Nevertheless, our hypothesis might account in part for an additional etiological factor to PPD development. If our concept is validated, it can provide guidance for future PPD prevention, diagnosis, and intervention.
Collapse
Affiliation(s)
| | - Duan Ni
- Nepean Hospital, Nepean Blue Mountains Local Health District, Sydney, NSW, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Leah Gilbert
- Nepean Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Emily Hibbert
- Nepean Hospital, Nepean Blue Mountains Local Health District, Sydney, NSW, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- Nepean Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Ralph Nanan
- Nepean Hospital, Nepean Blue Mountains Local Health District, Sydney, NSW, Australia.
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia.
- Nepean Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
175
|
Elhassan E, Omolo CA, Gafar MA, Kiruri LW, Ibrahim UH, Ismail EA, Devnarain N, Govender T. Disease-Inspired Design of Biomimetic Tannic Acid-Based Hybrid Nanocarriers for Enhancing the Treatment of Bacterial-Induced Sepsis. Mol Pharm 2024; 21:4924-4946. [PMID: 39214595 DOI: 10.1021/acs.molpharmaceut.4c00048] [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] [Indexed: 09/04/2024]
Abstract
This study explored the development of novel biomimetic tannic acid-based hybrid nanocarriers (HNs) for targeted delivery of ciprofloxacin (CIP-loaded TAH-NPs) against bacterial-induced sepsis. The prepared CIP-loaded TAH-NPs exhibited appropriate physicochemical characteristics and demonstrated biocompatibility and nonhemolytic properties. Computational simulations and microscale thermophoresis studies validated the strong binding affinity of tannic acid (TA) and its nanoformulation to human Toll-like receptor 4, surpassing that of the natural substrate lipopolysaccharide (LPS), suggesting a potential competitive inhibition against LPS-induced inflammatory responses. CIP released from TAH-NPs displayed a sustained release profile over 72 h. The in vitro antibacterial activity studies revealed that CIP-loaded TAH-NPs exhibited enhanced antibacterial efficacy and efflux pump inhibitory activity. Specifically, they showed a 3-fold increase in biofilm eradication activity against MRSA and a 2-fold increase against P. aeruginosa compared to bare CIP. Time-killing assays demonstrated complete bacterial clearance within 8 h of treatment with CIP-loaded TAH-NPs. In vitro DPPH scavenging and anti-inflammatory investigations confirmed the ability of the prepared hybrid nanosystem to neutralize reactive oxygen species (ROS) and modulate LPS-induced inflammatory responses. Collectively, these results suggest that CIP-loaded TAH-NPs may serve as an innovative nanocarrier for the effective and targeted delivery of antibiotics against bacterial-induced sepsis.
Collapse
Affiliation(s)
- Eman Elhassan
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag Durban X54001, South Africa
| | - Calvin A Omolo
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag Durban X54001, South Africa
- Department of Pharmaceutics and Pharmacy Practice, School of Pharmacy and Health Sciences, United States International University-Africa, P.O. Box 14634-00800, Nairobi 00800, Kenya
| | - Mohammed Ali Gafar
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag Durban X54001, South Africa
- Department of Pharmaceutics, Faculty of Pharmacy, University of Khartoum, Khartoum 11111, Sudan
| | - Lucy W Kiruri
- Department of Chemistry, Kenyatta University, P.O. Box 43844, Nairobi 00100, Kenya
| | - Usri H Ibrahim
- Discipline of Human Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban 4300, South Africa
| | - Eman A Ismail
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag Durban X54001, South Africa
| | - Nikita Devnarain
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag Durban X54001, South Africa
| | - Thirumala Govender
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag Durban X54001, South Africa
| |
Collapse
|
176
|
Friedman MI, Sørensen TIA, Taubes G, Lund J, Ludwig DS. Trapped fat: Obesity pathogenesis as an intrinsic disorder in metabolic fuel partitioning. Obes Rev 2024; 25:e13795. [PMID: 38961319 DOI: 10.1111/obr.13795] [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: 12/27/2023] [Revised: 05/24/2024] [Accepted: 06/13/2024] [Indexed: 07/05/2024]
Abstract
Our understanding of the pathophysiology of obesity remains at best incomplete despite a century of research. During this time, two alternative perspectives have helped shape thinking about the etiology of the disorder. The currently prevailing view holds that excessive fat accumulation results because energy intake exceeds energy expenditure, with excessive food consumption being the primary cause of the imbalance. The other perspective attributes the initiating cause of obesity to intrinsic metabolic defects that shift fuel partitioning from pathways for mobilization and oxidation to those for synthesis and storage. The resulting reduction in fuel oxidation and trapping of energy in adipose tissue drives a compensatory increase in energy intake and, under some conditions, a decrease in expenditure. This theory of obesity pathogenesis has historically garnered relatively less attention despite its pedigree. Here, we present an updated comprehensive formulation of the fuel partitioning theory, focused on evidence gathered over the last 80 years from major animal models of obesity showing a redirection of fuel fluxes from oxidation to storage and accumulation of excess body fat with energy intake equal to or even less than that of lean animals. The aim is to inform current discussions about the etiology of obesity and by so doing, help lay new foundations for the design of more efficacious approaches to obesity research, treatment and prevention.
Collapse
Affiliation(s)
| | - Thorkild I A Sørensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
- Department of Public Health, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
- Center for Childhood Health, Copenhagen, Denmark
| | | | - Jens Lund
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - David S Ludwig
- New Balance Foundation Obesity Prevention Center, Boston Children's Hospital, Department of Pediatrics, Harvard Medical School, Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Denmark
| |
Collapse
|
177
|
Bashir I, Wani SM, Jan N, Ali A, Rouf A, Sidiq H, Masood S, Mustafa S. Optimizing ultrasonic parameters for development of vitamin D3-loaded gum arabic nanoemulsions - An approach for vitamin D3 fortification. Int J Biol Macromol 2024; 278:134894. [PMID: 39168215 DOI: 10.1016/j.ijbiomac.2024.134894] [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: 10/24/2023] [Revised: 08/07/2024] [Accepted: 08/18/2024] [Indexed: 08/23/2024]
Abstract
Vitamin D encapsulation can significantly improve its bioavailability, stability, and solubility. Various biopolymers viz. whey protein isolate, carboxymethyl cellulose, alginate and gum arabic were studied for their potential to be used as wall material and gum arabic was selected for encapsulating vitamin D3 as it possesses lesser particle size, apparent viscosity and better stability in terms of zeta potential. Box Behnken design was employed for optimizing the process conditions for developing vitamin D3 nanoemulsion. Box Behnken design was constructed using ultrasonic amplitude, sonication time and vitamin D3/wall material percent as independent factors. The optimum conditions obtained were ultrasonic amplitude (80 %), sonication time (12 min) and vitamin D3/wall material percent (5). The designed nanoemulsion showed a particle size of 20.04 nm, zeta potential of -28.2 mV, and encapsulation efficiency of 71.9 %. Chemical interactions were observed in the developed nanoemulsion as demonstrated by Differential scanning calorimeter thermograms and Fourier transform infrared spectra of the nanoemulsion. The Korsmeyer-Peppas model was the most suitable for describing the release of vitamin D3 from the nanoemulsion. Fabricated nanoemulsion has the potential to be used in food and pharmaceutical industries.
Collapse
Affiliation(s)
- Iqra Bashir
- Division of Food Science and Technology, Sher-e-Kashmir University of Agricultural Sciences and Technology, Kashmir, Jammu and Kashmir 190025, India
| | - Sajad Mohd Wani
- Division of Food Science and Technology, Sher-e-Kashmir University of Agricultural Sciences and Technology, Kashmir, Jammu and Kashmir 190025, India.
| | - Nusrat Jan
- Division of Food Science and Technology, Sher-e-Kashmir University of Agricultural Sciences and Technology, Kashmir, Jammu and Kashmir 190025, India
| | - Asgar Ali
- Centre of Excellence for Postharvest Biotechnology (CEPB), School of Biosciences, University of Nottingham Malaysia, Jalan Broga, 43500 Semenyih, Selangor Darul Ehsan, Malaysia
| | - Abdul Rouf
- Division of Food Science and Technology, Sher-e-Kashmir University of Agricultural Sciences and Technology, Kashmir, Jammu and Kashmir 190025, India
| | - Haamiyah Sidiq
- Division of Food Science and Technology, Sher-e-Kashmir University of Agricultural Sciences and Technology, Kashmir, Jammu and Kashmir 190025, India
| | - Saima Masood
- Division of Basic Science & Humanities, Sher-e-Kashmir University of Agricultural Sciences and Technology, Kashmir, Jammu and Kashmir 190025, India
| | - Sehrish Mustafa
- Division of Food Science and Technology, Sher-e-Kashmir University of Agricultural Sciences and Technology, Kashmir, Jammu and Kashmir 190025, India
| |
Collapse
|
178
|
Liu YH, Liu TT, Niu JQ, Zhang XS, Xu WS, Song S, Wang Z. Characterization of phospholipidome in milk, yogurt and cream, and phospholipid differences related to various dairy processing methods. Food Chem 2024; 454:139733. [PMID: 38805923 DOI: 10.1016/j.foodchem.2024.139733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 04/27/2024] [Accepted: 05/16/2024] [Indexed: 05/30/2024]
Abstract
Milk phospholipids have multiple health benefits, but the deficiency of detailed phospholipid profiles in dairy products brings obstacles to intake calculation and function evaluation of dairy phospholipids. In present study, 306 phospholipid molecular species were identified and quantified among 207 milk, yogurt and cream products using a HILIC-ESI-Q-TOF MS and a HILIC-ESI-QQQ MS. The phospholipid profiles of five mammals' milk show that camel milk contains the most abundant phosphatidylethanolamine, phosphatidylserine and sphingomyelin; cow, yak and goat milk have similar phospholipidomes, while buffalo milk contains abundant phosphatidylinositol. Fewer plasmalogens but more lyso-glycerolphospholipids were found in ultra-high-temperature (UHT) sterilized milk than in pasteurized milk, and higher proportions of lyso-glycerolphospholipid/total phospholipid were observed in both cream and skimmed/semi-skimmed milk than whole milk, indicating that UHT and skimming processes improve glycerolphospholipid degradation and phospholipid nutrition loss. Meanwhile, more diacyl-glycerolphospholipids and less of their degradation products make yogurt a better phospholipid resource than whole milk.
Collapse
Affiliation(s)
- Yue-Han Liu
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| | - Ting-Ting Liu
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| | - Jing-Qi Niu
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| | - Xue-Song Zhang
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| | - Wei-Sheng Xu
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| | - Shuang Song
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing 100050, China; Key Laboratory of Public Nutrition and Health, National Health Commission of the People's Republic of China, Beijing 100000, China.
| | - Zhu Wang
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing 100050, China.
| |
Collapse
|
179
|
Aggarwal K, Singh B, Goel A, Agrawal DK, Bansal S, Kanagala SG, Anamika F, Gupta A, Jain R. Complex dichotomous links of nonalcoholic fatty liver disease and inflammatory bowel disease: exploring risks, mechanisms, and management modalities. Intest Res 2024; 22:414-427. [PMID: 38835139 PMCID: PMC11534450 DOI: 10.5217/ir.2024.00001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 04/04/2024] [Accepted: 04/15/2024] [Indexed: 06/06/2024] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) has been shown to be linked to inflammatory bowel disease (IBD) due to established risk factors such as obesity, age, and type 2 diabetes in numerous studies. However, alternative research suggests that factors related to IBD, such as disease activity, duration, and drug-induced toxicity, can contribute to NAFLD. Recent research findings suggest IBD relapses are correlated with dysbiosis, mucosal damage, and an increase in cytokines. In contrast, remission periods are characterized by reduced metabolic risk factors. There is a dichotomy evident in the associations between NAFLD and IBD during relapses and remissions. This warrants a nuanced understanding of the diverse influences on disease manifestation and progression. It is possible to provide a holistic approach to care for patients with IBD by emphasizing the interdependence between metabolic and inflammatory disorders.
Collapse
Affiliation(s)
- Kanishk Aggarwal
- Department of Medicine, Dayanand Medical College, Ludhiana, India
| | - Bhupinder Singh
- Department of Medicine, Government Medical College Amritsar, Amritsar, India
| | - Abhishek Goel
- Department of Medicine, Cape Fear Valley Medical Center, Fayetteville, NC, USA
| | | | - Sourav Bansal
- Department of Medicine, Government Medical College Amritsar, Amritsar, India
| | | | - Fnu Anamika
- Department of Medicine, University College of Medical Sciences, New Delhi, India
| | | | - Rohit Jain
- Department of Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA, USA
| |
Collapse
|
180
|
Madsen AL, Bonàs-Guarch S, Gheibi S, Prasad R, Vangipurapu J, Ahuja V, Cataldo LR, Dwivedi O, Hatem G, Atla G, Guindo-Martínez M, Jørgensen AM, Jonsson AE, Miguel-Escalada I, Hassan S, Linneberg A, Ahluwalia TS, Drivsholm T, Pedersen O, Sørensen TIA, Astrup A, Witte D, Damm P, Clausen TD, Mathiesen E, Pers TH, Loos RJF, Hakaste L, Fex M, Grarup N, Tuomi T, Laakso M, Mulder H, Ferrer J, Hansen T. Genetic architecture of oral glucose-stimulated insulin release provides biological insights into type 2 diabetes aetiology. Nat Metab 2024; 6:1897-1912. [PMID: 39420167 PMCID: PMC11496110 DOI: 10.1038/s42255-024-01140-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 09/02/2024] [Indexed: 10/19/2024]
Abstract
The genetics of β-cell function (BCF) offer valuable insights into the aetiology of type 2 diabetes (T2D)1,2. Previous studies have expanded the catalogue of BCF genetic associations through candidate gene studies3-7, large-scale genome-wide association studies (GWAS) of fasting BCF8,9 or functional islet studies on T2D risk variants10-14. Nonetheless, GWAS focused on BCF traits derived from oral glucose tolerance test (OGTT) data have been limited in sample size15,16 and have often overlooked the potential for related traits to capture distinct genetic features of insulin-producing β-cells17,18. We reasoned that investigating the genetic basis of multiple BCF estimates could provide a broader understanding of β-cell physiology. Here, we aggregate GWAS data of eight OGTT-based BCF traits from ~26,000 individuals of European descent, identifying 55 independent genetic associations at 44 loci. By examining the effects of BCF genetic signals on related phenotypes, we uncover diverse disease mechanisms whereby genetic regulation of BCF may influence T2D risk. Integrating BCF-GWAS data with pancreatic islet transcriptomic and epigenomic datasets reveals 92 candidate effector genes. Gene silencing in β-cell models highlights ACSL1 and FAM46C as key regulators of insulin secretion. Overall, our findings yield insights into the biology of insulin release and the molecular processes linking BCF to T2D risk, shedding light on the heterogeneity of T2D pathophysiology.
Collapse
Affiliation(s)
- A L Madsen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen (UCPH), Copenhagen, Denmark
| | - S Bonàs-Guarch
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - S Gheibi
- Department of Clinical Sciences, Unit of Molecular Metabolism, Lund University, Malmö, Sweden
| | - R Prasad
- Department of Clinical Sciences, Unit of Genomics, Diabetes and Endocrinology, Lund University, Malmö, Sweden
| | - J Vangipurapu
- Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - V Ahuja
- Institute for Molecular Medicine Finland and Research Program of Clinical and Molecular Medicine, University of Helsinki, Helsinki, Finland
| | - L R Cataldo
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen (UCPH), Copenhagen, Denmark
- Department of Clinical Sciences, Unit of Molecular Metabolism, Lund University, Malmö, Sweden
| | - O Dwivedi
- Institute for Molecular Medicine Finland and Research Program of Clinical and Molecular Medicine, University of Helsinki, Helsinki, Finland
- Folkhalsan Research Centre, Helsinki, Finland
| | - G Hatem
- Department of Clinical Sciences, Unit of Genomics, Diabetes and Endocrinology, Lund University, Malmö, Sweden
| | - G Atla
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - M Guindo-Martínez
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen (UCPH), Copenhagen, Denmark
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - A M Jørgensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen (UCPH), Copenhagen, Denmark
| | - A E Jonsson
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen (UCPH), Copenhagen, Denmark
| | - I Miguel-Escalada
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - S Hassan
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen (UCPH), Copenhagen, Denmark
| | - A Linneberg
- Center for Clinical Research and Prevention, Copenhagen University Hospital-Bispebjerg and Frederiksberg, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health Sciences, UCPH, Copenhagen, Denmark
| | - Tarunveer S Ahluwalia
- Steno Diabetes Center Copenhagen, Herlev, Denmark
- The Bioinformatics Center, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - T Drivsholm
- Center for Clinical Research and Prevention, Copenhagen University Hospital-Bispebjerg and Frederiksberg, Copenhagen, Denmark
- Section of General Practice, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - O Pedersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen (UCPH), Copenhagen, Denmark
| | - T I A Sørensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen (UCPH), Copenhagen, Denmark
- Department of Public Health Sciences (Section of Epidemiology), University of Copenhagen, Copenhagen, Denmark
| | - A Astrup
- Novo Nordisk Fonden, Hellerup, Denmark
| | - D Witte
- Institut for Folkesundhed-Epidemiologi, Aarhus University, Aarhus, Denmark
| | - P Damm
- Center for Pregnant Women with Diabetes and Department of Gynecology, Fertility, and Obstetrics and Department of Clinical Medicine, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - T D Clausen
- Center for Pregnant Women with Diabetes and Department of Gynecology, Fertility, and Obstetrics and Department of Clinical Medicine, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - E Mathiesen
- Center for Pregnant Women with Diabetes, Department of Nephrology and Endocrinology and Department of Clinical Medicine, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - T H Pers
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen (UCPH), Copenhagen, Denmark
| | - R J F Loos
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen (UCPH), Copenhagen, Denmark
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - L Hakaste
- Institute for Molecular Medicine Finland and Research Program of Clinical and Molecular Medicine, University of Helsinki, Helsinki, Finland
- Folkhalsan Research Centre, Helsinki, Finland
| | - M Fex
- Department of Clinical Sciences, Unit of Molecular Metabolism, Lund University, Malmö, Sweden
| | - N Grarup
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen (UCPH), Copenhagen, Denmark
| | - T Tuomi
- Department of Clinical Sciences, Unit of Genomics, Diabetes and Endocrinology, Lund University, Malmö, Sweden
- Institute for Molecular Medicine Finland and Research Program of Clinical and Molecular Medicine, University of Helsinki, Helsinki, Finland
- Folkhalsan Research Centre, Helsinki, Finland
- Helsinki University Hospital, Abdominal Centre / Endocrinology, Helsinki, Finland
| | - M Laakso
- Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - H Mulder
- Department of Clinical Sciences, Unit of Molecular Metabolism, Lund University, Malmö, Sweden
| | - J Ferrer
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain.
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.
| | - T Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen (UCPH), Copenhagen, Denmark.
| |
Collapse
|
181
|
Ferrero E, Masini M, Carli M, Moscato S, Beffy P, Vaglini F, Mattii L, Corti A, Scarselli M, Novelli M, De Tata V. Dopamine-mediated autocrine inhibition of insulin secretion. Mol Cell Endocrinol 2024; 592:112294. [PMID: 38838763 DOI: 10.1016/j.mce.2024.112294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 05/15/2024] [Accepted: 06/01/2024] [Indexed: 06/07/2024]
Abstract
The aim of the present research was to explore the mechanisms underlying the role of dopamine in the regulation of insulin secretion in beta cells. The effect of dopamine on insulin secretion was investigated on INS 832/13 cell line upon glucose and other secretagogues stimulation. Results show that dopamine significantly inhibits insulin secretion stimulated by both glucose and other secretagogues, while it has no effect on the basal secretion. This effect requires the presence of dopamine during incubation with the various secretagogues. Both electron microscopy and immunohistochemistry indicate that in beta cells the D2 dopamine receptor is localized within the insulin granules. Blocking dopamine entry into the insulin granules by inhibiting the VMAT2 transporter with tetrabenazine causes a significant increase in ROS production. Our results confirm that dopamine plays an important role in the regulation of insulin secretion by pancreatic beta cells through a regulated and precise compartmentalization mechanisms.
Collapse
Affiliation(s)
| | | | | | - Stefania Moscato
- Department of Clinical and Experimental Medicine, Italy; Interdepartmental Research Centre "Nutraceuticals and Food for Health", Italy
| | | | | | - Letizia Mattii
- Department of Clinical and Experimental Medicine, Italy; Interdepartmental Research Centre "Nutraceuticals and Food for Health", Italy
| | | | | | | | - Vincenzo De Tata
- Department of Translational Research, Italy; CIME (Interdepartmental Centre of Electron Microscopy), University of Pisa, Pisa, Italy.
| |
Collapse
|
182
|
Nishida T, Ayaori M, Arakawa J, Suenaga Y, Shiotani K, Uto-Kondo H, Komatsu T, Nakaya K, Endo Y, Sasaki M, Ikewaki K. Liver-specific Lxr inhibition represses reverse cholesterol transport in cholesterol-fed mice. Atherosclerosis 2024; 397:117578. [PMID: 38797615 DOI: 10.1016/j.atherosclerosis.2024.117578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 04/26/2024] [Accepted: 05/07/2024] [Indexed: 05/29/2024]
Abstract
BACKGROUND AND AIMS High density lipoprotein (HDL) exerts an anti-atherosclerotic effect via reverse cholesterol transport (RCT). Several phases of RCT are transcriptionally controlled by Liver X receptors (Lxrs). Although macrophage Lxrs reportedly promote RCT, it is still uncertain whether hepatic Lxrs affect RCT in vivo. METHODS To inhibit Lxr-dependent pathways in mouse livers, we performed hepatic overexpression of sulfotransferase family cytosolic 2B member 1 (Sult2b1) using adenoviral vector (Ad-Sult2b1). Ad-Sult2b1 or the control virus was intravenously injected into wild type mice and Lxrα/β double knockout mice, under a normal or high-cholesterol diet. A macrophage RCT assay and an HDL kinetic study were performed. RESULTS Hepatic Sult2b1 overexpression resulted in reduced expression of Lxr-target genes - ATP-binding cassette transporter G5/G8, cholesterol 7α hydroxylase and Lxrα itself - respectively reducing or increasing cholesterol levels in HDL and apolipoprotein B-containing lipoproteins (apoB-L). A macrophage RCT assay revealed that Sult2b1 overexpression inhibited fecal excretion of macrophage-derived 3H-cholesterol only under a high-cholesterol diet. In an HDL kinetic study, Ad-Sult2b1 promoted catabolism/hepatic uptake of HDL-derived cholesterol, thereby reducing fecal excretion. Finally, in Lxrα/β double knockout mice, hepatic Sult2b1 overexpression increased apoB-L levels, but there were no differences in HDL levels or RCT compared to the control, indicating that Sult2b1-mediated effects on HDL/RCT and apoB-L were distinct: the former was Lxr-dependent, but not the latter. CONCLUSIONS Hepatic Lxr inhibition negatively regulates circulating HDL levels and RCT by reducing Lxr-target gene expression.
Collapse
MESH Headings
- Animals
- Liver X Receptors/metabolism
- Liver X Receptors/genetics
- Liver/metabolism
- Mice, Knockout
- Biological Transport
- Mice
- Cholesterol/metabolism
- Macrophages/metabolism
- Mice, Inbred C57BL
- Sulfotransferases/metabolism
- Sulfotransferases/genetics
- ATP Binding Cassette Transporter, Subfamily G, Member 5/metabolism
- ATP Binding Cassette Transporter, Subfamily G, Member 5/genetics
- Cholesterol, Dietary
- ATP Binding Cassette Transporter, Subfamily G, Member 8/metabolism
- ATP Binding Cassette Transporter, Subfamily G, Member 8/genetics
- Male
- Cholesterol, HDL/blood
- Cholesterol, HDL/metabolism
- Lipoproteins, HDL/metabolism
- Lipoproteins
Collapse
Affiliation(s)
- Takafumi Nishida
- Division of Anti-aging and Vascular Medicine, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan.
| | - Makoto Ayaori
- Division of Anti-aging and Vascular Medicine, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan; Tokorozawa Heart Center, Tokorozawa, Japan
| | - Junko Arakawa
- Division of Anti-aging and Vascular Medicine, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Yumiko Suenaga
- Division of Anti-aging and Vascular Medicine, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Kazusa Shiotani
- Division of Anti-aging and Vascular Medicine, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Harumi Uto-Kondo
- Division of Anti-aging and Vascular Medicine, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Tomohiro Komatsu
- Division of Anti-aging and Vascular Medicine, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Kazuhiro Nakaya
- Division of Anti-aging and Vascular Medicine, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Yasuhiro Endo
- Division of Anti-aging and Vascular Medicine, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Makoto Sasaki
- Division of Anti-aging and Vascular Medicine, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Katsunori Ikewaki
- Division of Anti-aging and Vascular Medicine, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan
| |
Collapse
|
183
|
Yang C, Yan P, Wu X, Zhang W, Cui H, Zhang L, Xu Z, Peng S, Tang M, Wang Y, Chen L, Zou Y, Liu Y, Zhang M, Zhao X, Xiao J, Xiao C, Zhang L, Yao Y, Li J, Liu Z, Yang C, Jiang X, Zhang B. Associations of sleep with cardiometabolic risk factors and cardiovascular diseases: An umbrella review of observational and mendelian randomization studies. Sleep Med Rev 2024; 77:101965. [PMID: 39137553 DOI: 10.1016/j.smrv.2024.101965] [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: 09/12/2023] [Revised: 05/09/2024] [Accepted: 05/26/2024] [Indexed: 08/15/2024]
Abstract
Two researchers independently assessed studies published up to February 5, 2023, across PubMed, Web of Science, Embase, and Cochrane Library, to investigate the associations of sleep traits with cardiometabolic risk factors, as well as with cardiovascular diseases. Fourteen systematic reviews consisting of 23 meta-analyses, and 11 Mendelian randomization (MR) studies were included in this study. Short sleep duration was associated with a higher risk of obesity, type 2 diabetes (T2D), hypertension, stroke, and coronary heart disease (CHD) in observational studies, while a causal role was only demonstrated in obesity, hypertension, and CHD by MR. Similarly, long sleep duration showed connections with a higher risk of obesity, T2D, hypertension, stroke, and CHD in observational studies, none was supported by MR analysis. Both observational and MR studies indicated heightened risks of hypertension, stroke, and CHD in relation to insomnia. Napping was linked to elevated risks of T2D and CHD in observational studies, with MR analysis confirming a causal role in T2D. Additionally, snoring was correlated with increased risks of stroke and CHD in both observational and MR studies. This work consolidates existing evidence on a causal relationship between sleep characteristics and cardiometabolic risk factors, as well as cardiovascular diseases.
Collapse
Affiliation(s)
- Chao Yang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, and West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China; School of Public Health, Southwest Medical University, Luzhou, Sichuan, China
| | - Peijing Yan
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, and West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xueyao Wu
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, and West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wenqiang Zhang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, and West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Huijie Cui
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, and West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Li Zhang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, and West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhengxing Xu
- School of Public Health, Southwest Medical University, Luzhou, Sichuan, China
| | - Shanshan Peng
- Department of Occupational and Environmental Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Mingshuang Tang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, and West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yutong Wang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, and West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lin Chen
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, and West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yanqiu Zou
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, and West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yunjie Liu
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, and West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Min Zhang
- Clinical Research Center, Women and Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Xunying Zhao
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, and West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jinyu Xiao
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, and West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chenghan Xiao
- Department of Maternal, Child and Adolescent Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Ling Zhang
- Department of Iatrical Polymer Material and Artificial Apparatus, School of Polymer Science and Engineering, Sichuan University, Chengdu, China
| | - Yuqin Yao
- Department of Occupational and Environmental Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Jiayuan Li
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, and West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhenmi Liu
- Department of Maternal, Child and Adolescent Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Chunxia Yang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, and West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xia Jiang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, and West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China; Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden.
| | - Ben Zhang
- Hainan General Hospital and Hainan Affiliated Hospital, Hainan Medical University, Haikou, China; West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
184
|
Parini P. HDL, reverse cholesterol transport, and atherosclerosis: Unravelling the complexity or adding to the confusion? Atherosclerosis 2024; 397:118562. [PMID: 39137620 DOI: 10.1016/j.atherosclerosis.2024.118562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 07/17/2024] [Accepted: 08/07/2024] [Indexed: 08/15/2024]
Affiliation(s)
- Paolo Parini
- Cardio Metabolic Unit, Dept of Medicine and Dept of Laboratory Medicine, Karolinska Institutet; and Medical Unit Endocrinology, Theme Inflammation and Ageing, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
185
|
Jiao J, Zeng D, Wu Y, Li C, Mo T. Programmable and ultra-efficient Argonaute protein-mediated nucleic acid tests: A review. Int J Biol Macromol 2024; 278:134755. [PMID: 39147338 DOI: 10.1016/j.ijbiomac.2024.134755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 08/08/2024] [Accepted: 08/12/2024] [Indexed: 08/17/2024]
Abstract
With the attributes of high sensitivity, single-base resolution, multiplex detection capability, and programmability upon nucleic acid recognition, Argonaute (Ago)-based biosensing assays are increasingly recognized as one of the most promising tools for precise identification and quantification of target analytes. Employed as highly specific sequence recognition elements of these robust diagnostic methods, Agos are revolutionizing how nucleic acid targets are detected. A systematic and comprehensive summary of this emerging and rapid-advancing technology is necessary to give play to the potential of Ago-based biosensing assays. The structure and function of Agos were briefly overviewed at the beginning of the work, followed by a review of the recent advancements in employing Agos sensing for detecting various targets with a comprehensive analysis such as viruses, tumor biomarkers, pathogens, mycoplasma, and parasite. The significance and benefits of these platforms were then deliberated. In addition, the authors shared subjective viewpoints on the existing challenges and offered relevant guidance for the future progress of Agos assays. Finally, the future research outlook regarding Ago-based sensing in this field was also outlined. As such, this review is expected to offer valuable information and fresh perspectives for a broader group of researchers.
Collapse
Affiliation(s)
- Jinlong Jiao
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Dandan Zeng
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Yafang Wu
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Chentao Li
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Tianlu Mo
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China.
| |
Collapse
|
186
|
Wang Y, Chen H, Wang Y, Zhang H, Weng Q, Liu Y, Xu M. Seasonal changes in vitamin A metabolism-related factors in the oviduct of Chinese brown frog (Rana dybowskii). J Steroid Biochem Mol Biol 2024; 243:106583. [PMID: 38992392 DOI: 10.1016/j.jsbmb.2024.106583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 07/06/2024] [Accepted: 07/09/2024] [Indexed: 07/13/2024]
Abstract
The oviduct of the Chinese brown frog (Rana dybowskii) expands during pre-brumation rather than the breeding period, exhibiting a special physiological feature. Vitamin A is essential for the proper growth and development of many organisms, including the reproductive system such as ovary and oviduct. Vitamin A is metabolized into retinoic acid, which is crucial for oviduct formation. This study examined the relationship between oviducal expansion and vitamin A metabolism. We observed a significant increase in the weight and diameter of the oviduct in Rana dybowskii during pre-brumation. Vitamin A and its active metabolite, retinoic acid, notably increased during pre-brumation. The mRNA levels of retinol binding protein 4 (rbp4) and its receptor stra6 gene, involved in vitamin A transport, were elevated during pre-brumation compared to the breeding period. In the vitamin A metabolic pathway, the mRNA expression level of retinoic acid synthase aldh1a2 decreased significantly during pre-brumation, while the mRNA levels of retinoic acid α receptor (rarα) and the retinoic acid catabolic enzyme cyp26a1 increased significantly during pre-brumation, but not during the breeding period. Immunohistochemical results showed that Rbp4, Stra6, Aldh1a2, Rarα, and Cyp26a1 were expressed in ampulla region of the oviduct. Western blot results indicated that Aldh1a2 expression was lower, while Rbp4, Stra6, RARα, and Cyp26a1 were higher during pre-brumation compared to the breeding period. Transcriptome analyses further identified differential genes in the oviduct and found enrichment of differential genes in the vitamin A metabolism pathway, providing evidences for our study. These results suggest that the vitamin A metabolic pathway is more active during pre-brumation compared to the breeding period, and retinoic acid may regulate pre-brumation oviductal expansion through Rarα-mediated autocrine/paracrine modulation.
Collapse
Affiliation(s)
- Yankun Wang
- College of Biological Sciences and Technology, Beijing Forestry University, Beijing, China
| | - Haohan Chen
- College of Biological Sciences and Technology, Beijing Forestry University, Beijing, China
| | - Yawei Wang
- College of Biological Sciences and Technology, Beijing Forestry University, Beijing, China
| | - Haolin Zhang
- College of Biological Sciences and Technology, Beijing Forestry University, Beijing, China
| | - Qiang Weng
- College of Biological Sciences and Technology, Beijing Forestry University, Beijing, China
| | - Yuning Liu
- College of Biological Sciences and Technology, Beijing Forestry University, Beijing, China.
| | - Meiyu Xu
- College of Biological Sciences and Technology, Beijing Forestry University, Beijing, China.
| |
Collapse
|
187
|
Tattoli I, Mathew AR, Verrienti A, Pallotta L, Severi C, Andreola F, Cavallucci V, Giorgi M, Massimi M, Bencini L, Fidaleo M. The Interplay between Liver and Adipose Tissue in the Onset of Liver Diseases: Exploring the Role of Vitamin Deficiency. Cells 2024; 13:1631. [PMID: 39404394 PMCID: PMC11475612 DOI: 10.3390/cells13191631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 09/24/2024] [Accepted: 09/24/2024] [Indexed: 10/19/2024] Open
Abstract
The deficiency of vitamins, a condition known as "hidden hunger", causes comprehensive pathological states. Research over the years has identified a relationship between liver diseases and hypovitaminosis or defects in vitamin metabolism. The exact mechanisms remain elusive; however, the crucial involvement of specific vitamins in metabolic functions, alongside the reclassification of liver disease as metabolic dysfunction-associated steatotic liver disease (MASLD), has prompted researchers to investigate the potential cause-effect dynamics between vitamin deficiency and liver disease. Moreover, scientists are increasingly investigating how the deficiency of vitamins might disrupt specific organ crosstalk, potentially contributing to liver disease. Although the concept of a dysmetabolic circuit linking adipose tissue and the liver, leading to liver disease, has been discussed, the possible involvement of vitamin deficiency in this axis is a relatively recent area of study, with numerous critical aspects yet to be fully understood. In this review, we examine research from 2019 to July 2024 focusing on the possible link between liver-adipose tissue crosstalk and vitamin deficiency involved in the onset and progression of non-alcoholic fatty liver disease (NAFLD). Studies report that vitamin deficiency can affect the liver-adipose tissue axis, mainly affecting the regulation of systemic energy balance and inflammation.
Collapse
Affiliation(s)
- Ivan Tattoli
- Oncology General Surgery, Azienda Ospedaliero Universitaria Careggi, 50139 Florence, Italy; (I.T.); (L.B.)
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, 00185 Rome, Italy; (A.R.M.); (M.G.)
| | - Aimee Rachel Mathew
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, 00185 Rome, Italy; (A.R.M.); (M.G.)
| | - Antonella Verrienti
- Department of Translational and Precision Medicine, Sapienza University of Rome, 00161 Rome, Italy; (A.V.); (L.P.); (C.S.)
| | - Lucia Pallotta
- Department of Translational and Precision Medicine, Sapienza University of Rome, 00161 Rome, Italy; (A.V.); (L.P.); (C.S.)
| | - Carola Severi
- Department of Translational and Precision Medicine, Sapienza University of Rome, 00161 Rome, Italy; (A.V.); (L.P.); (C.S.)
| | - Fausto Andreola
- Liver Failure Group, Institute for Liver and Digestive Health, Royal Free Hospital, University College London, London NW3 2PF, UK;
| | - Virve Cavallucci
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy;
| | - Mauro Giorgi
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, 00185 Rome, Italy; (A.R.M.); (M.G.)
| | - Mara Massimi
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy;
| | - Lapo Bencini
- Oncology General Surgery, Azienda Ospedaliero Universitaria Careggi, 50139 Florence, Italy; (I.T.); (L.B.)
| | - Marco Fidaleo
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, 00185 Rome, Italy; (A.R.M.); (M.G.)
- Research Center for Nanotechnology for Engineering of Sapienza (CNIS), Sapienza University of Rome, 00185 Rome, Italy
| |
Collapse
|
188
|
Foran D, Antoniades C, Akoumianakis I. Emerging Roles for Sphingolipids in Cardiometabolic Disease: A Rational Therapeutic Target? Nutrients 2024; 16:3296. [PMID: 39408263 PMCID: PMC11478599 DOI: 10.3390/nu16193296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/19/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
Cardiovascular disease is a leading cause of morbidity and mortality. New research elucidates increasingly complex relationships between cardiac and metabolic health, giving rise to new possible therapeutic targets. Sphingolipids are a heterogeneous class of bioactive lipids with critical roles in normal human physiology. They have also been shown to play both protective and deleterious roles in the pathogenesis of cardiovascular disease. Ceramides are implicated in dysregulating insulin signalling, vascular endothelial function, inflammation, oxidative stress, and lipoprotein aggregation, thereby promoting atherosclerosis and vascular disease. Ceramides also advance myocardial disease by enhancing pathological cardiac remodelling and cardiomyocyte death. Glucosylceramides similarly contribute to insulin resistance and vascular inflammation, thus playing a role in atherogenesis and cardiometabolic dysfunction. Sphingosing-1-phosphate, on the other hand, may ameliorate some of the pathological functions of ceramide by protecting endothelial barrier integrity and promoting cell survival. Sphingosine-1-phosphate is, however, implicated in the development of cardiac fibrosis. This review will explore the roles of sphingolipids in vascular, cardiac, and metabolic pathologies and will evaluate the therapeutic potential in targeting sphingolipids with the aim of prevention and reversal of cardiovascular disease in order to improve long-term cardiovascular outcomes.
Collapse
Affiliation(s)
| | | | - Ioannis Akoumianakis
- Cardiovascular Medicine Division, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK; (D.F.); (C.A.)
| |
Collapse
|
189
|
Li H, Murugesan A, Shoaib M, Sheng W, Chen Q. Functionalized metal-organic frameworks with biomolecules for sensing and detection applications of food contaminants. Crit Rev Food Sci Nutr 2024:1-33. [PMID: 39323356 DOI: 10.1080/10408398.2024.2406482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2024]
Abstract
The increasing demand for toxin-free food, driven by the rise in fast food consumption and changing dietary habits, necessitates advanced and efficient detection methods to address the potential risks associated with contaminated food. Nanomaterial-based detection methods have shown significant promise, particularly using metal-organic frameworks (MOFs) combined with biomolecules. This review article provides an overview of recent advancements in using functionalized metal-organic frameworks (FMOFs) with biomolecules to detect various food contaminants, including heavy metals, antibiotics, pesticides, bacteria, mycotoxins and other chemical contaminants. We discuss the fundamental principles of detecting food contaminants, evaluate existing analytical techniques, and explore the development of biomacromolecule-functionalized MOF-based sensors encompassing colorimetric, optical, electrochemical, and portable variants. The review also examines sensing mechanisms, uses FMOFs as signal probes and carriers for capture probes, and assesses sensitivity. Additionally, we explore the opportunities and challenges in producing FMOFs with biomacromolecules for food contaminant assessment. Future directions include improving sensor sensitivity and specificity, developing more cost-effective production methods, and integrating these technologies into real-world food safety monitoring systems. This work aims to pave the way for innovative and reliable solutions to ensure the safety of our food supply.
Collapse
Affiliation(s)
- Huanhuan Li
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, PR China
| | - Arul Murugesan
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, PR China
| | - Muhammad Shoaib
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, PR China
| | - Wei Sheng
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, PR China
| | - Quansheng Chen
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, PR China
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, PR China
| |
Collapse
|
190
|
Falero-Diaz G, Barboza CDA, Kaiser K, Tallman KA, Montoya C, Patel SB, Hutcheson JD, Lassance-Soares RM. The Systemic Effect of Ischemia Training and Its Impact on Bone Marrow-Derived Monocytes. Cells 2024; 13:1602. [PMID: 39404366 PMCID: PMC11475150 DOI: 10.3390/cells13191602] [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: 06/26/2024] [Revised: 08/30/2024] [Accepted: 09/09/2024] [Indexed: 10/19/2024] Open
Abstract
OBJECTIVE Monocytes are innate immune cells that play a central role in inflammation, an essential component during neovascularization. Our recent publication demonstrated that ischemia training by 24 h unilateral occlusion of the femoral artery (FA) can modify bone marrow-derived monocytes (BM-Mono), allowing them to improve collateral remodeling in a mouse model of hindlimb ischemia. Here, we expand on our previous findings, investigating a potential systemic effect of ischemia training and how this training can impact BM-Mono. METHODS AND RESULTS BM-Mono from mice exposed to ischemia training (24 h) or Sham (same surgical procedure without femoral artery occlusion-ischemia training) procedures were used as donors in adoptive transfer experiments where recipients were subjected to hindlimb ischemia. Donor cells were divided corresponding to the limb from which they were isolated (left-limb previously subjected to 24 h ischemia and right-contralateral limb). Recipients who received 24 h ischemic-trained monocytes isolated from either limb had remarkable blood flow recovery compared to recipients with Sham monocytes (monocytes isolated from Sham group-no ischemia training). Since these data suggested a systemic effect of ischemic training, circulating extracellular vesicles (EVs) were investigated as potential players. EVs were isolated from both groups, 24 h-trained and Sham, and the former showed increased expression of histone deacetylase 1 (HDAC1), which is known to downregulate 24-dehydrocholesterol reductase (Dhcr24) gene expression. Since we previously revealed that ischemia training downregulates Dhcr24 in BM-Mono, we incubated EVs from 24 h-trained and Sham groups with wild-type (WT) BM-Mono and demonstrated that WT BM-Mono incubated with 24 h-trained EVs had lower gene expression of Dhcr24 and an HDAC1 inhibitor blunted this effect. Next, we repeated the adoptive transfer experiment using Dhcr24 KO mice as donors of BM-Mono for WT mice subjected to hindlimb ischemia. Recipients who received Dhcr24 KO BM-Mono had greater limb perfusion than those who received WT BM-Mono. Further, we focused on the 24 h-trained monocytes (which previously showed downregulation of Dhcr24 gene expression and higher desmosterol) to test the expression of a few genes downstream of the desmosterol pathway, confirm the Dhcr24 protein level and assess its differentiation in M2-like macrophage phenotype. We found that 24 h-trained BM-Mono had greater expression of key genes in the desmosterol pathway, such as liver X receptors (LXRs) and ATP-binding cassette transporter (ABCA1), and we confirmed low protein expression of Dhcr24. Further, we demonstrated that ischemic-trained BM-Mono polarized towards an anti-inflammatory M2 macrophage phenotype. Finally, we demonstrated that 24 h-trained monocytes adhere less to endothelial cells, and the same pattern was shown by WT BM-Mono treated with Dhcr24 inhibitor. CONCLUSIONS Ischemia training leads to a systemic effect that, at least in part, involves circulating EVs and potential epigenetic modification in BM-Mono. These ischemic-trained BM-Mono demonstrated an anti-inflammatory phenotype towards M2 macrophage differentiation and less ability to adhere to endothelial cells, which is associated with the downregulation of Dhcr24 in those cells. These data together suggest that Dhcr24 might be an important target within monocytes to improve the outcomes of hindlimb ischemia.
Collapse
Affiliation(s)
- Gustavo Falero-Diaz
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (G.F.-D.); (C.M.)
| | - Catarina de A. Barboza
- Department of Medicine, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
| | - Katherine Kaiser
- Department of Biomedical Engineering, Florida International University, Miami, FL 33174, USA; (K.K.); (J.D.H.)
| | - Keri A. Tallman
- Department of Chemistry, Vanderbilt University, Nashville, TN 37235, USA;
| | - Christopher Montoya
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (G.F.-D.); (C.M.)
| | - Shailendra B. Patel
- Department of Internal Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Cincinnati, Cincinnati, OH 45221, USA;
| | - Joshua D. Hutcheson
- Department of Biomedical Engineering, Florida International University, Miami, FL 33174, USA; (K.K.); (J.D.H.)
| | - Roberta M. Lassance-Soares
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (G.F.-D.); (C.M.)
| |
Collapse
|
191
|
Wu J, Li H, Wang Z, Pan M, Wang S. Ratiometric Electrochemical Sensor Applying SWCNHs/T-PEDOT Nanocomposites for Efficient Quantification of Tert-Butylhydroquinone in Foodstuffs. Foods 2024; 13:2996. [PMID: 39335924 PMCID: PMC11431831 DOI: 10.3390/foods13182996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024] Open
Abstract
Tert-butylhydroquinone (TBHQ) is a phenolic substance that is commonly employed to prevent food oxidation. Excessive or improper utilization of this antioxidant can not only impact food quality but may also pose potential risks to human health. In this study, an ultrasensitive, stable, and easily operable ratiometric electrochemical sensor was successfully fabricated by combining the tubular (3,4-ethylenedioxythiophene) (T-PEDOT) with single-wall carbon nanohorns (SWCNHs) for the detection of TBHQ antioxidants in food. The SWCNHs/T-PEDOT nanocomposite fabricated through ultrasound-assisted and template approaches was employed as the modified substrate for the electrode interface. The synergistic effect of SWCNHs and T-PEDOT, which possess excellent electrical conductivity and catalytic properties, enabled the modified electrode to showcase remarkable electrocatalytic performance towards TBHQ, with the redox signal of methylene blue serving as an internal reference. Under optimized conditions, the SWCNHs/T-PEDOT-modified electrode demonstrated good linearity within the TBHQ concentration range of 0.01-200.0 μg mL-1, featuring a low limit of detection (LOD) of 0.005 μg mL-1. The proposed ratiometric electrochemical sensor displayed favorable reproducibility, stability, and anti-interference capacity, thereby offering a promising strategy for monitoring the levels of TBHQ in oil-rich food products.
Collapse
Affiliation(s)
- Jing Wu
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China;
| | - Huilin Li
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science and Technology, Tianjin 300457, China; (H.L.); (Z.W.); (M.P.)
| | - Zhijuan Wang
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science and Technology, Tianjin 300457, China; (H.L.); (Z.W.); (M.P.)
| | - Mingfei Pan
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science and Technology, Tianjin 300457, China; (H.L.); (Z.W.); (M.P.)
| | - Shuo Wang
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China;
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science and Technology, Tianjin 300457, China; (H.L.); (Z.W.); (M.P.)
| |
Collapse
|
192
|
Majeed M, Rather MA. Advancements in vitamin D encapsulation: characterization, wall materials, and fortification applications. NUTRIRE 2024; 49:48. [DOI: 10.1186/s41110-024-00292-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 09/13/2024] [Indexed: 01/06/2025]
|
193
|
Chen T, Lei W, Wang M. Predictive Model of Internal Bleeding in Elderly Aspirin Users Using XGBoost Machine Learning. Risk Manag Healthc Policy 2024; 17:2255-2269. [PMID: 39309118 PMCID: PMC11416773 DOI: 10.2147/rmhp.s478826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 09/15/2024] [Indexed: 09/25/2024] Open
Abstract
Objective This study aimed to develop a predictive model for assessing internal bleeding risk in elderly aspirin users using machine learning. Methods A total of 26,030 elderly aspirin users (aged over 65) were retrospective included in the study. Data on patient demographics, clinical features, underlying diseases, medical history, and laboratory examinations were collected from Affiliated Dongyang Hospital of Wenzhou Medical University. Patients were randomly divided into two groups, with a 7:3 ratio, for model development and internal validation, respectively. Least absolute shrinkage and selection operator (LASSO) regression, extreme gradient boosting (XGBoost), and multivariate logistic regression were employed to develop prediction models. Model performance was evaluated using area under the curve (AUC), calibration curves, decision curve analysis (DCA), clinical impact curve (CIC), and net reduction curve (NRC). Results The XGBoost model exhibited the highest AUC among all models. It consisted of six clinical variables: HGB, PLT, previous bleeding, gastric ulcer, cerebral infarction, and tumor. A visual nomogram was developed based on these six variables. In the training dataset, the model achieved an AUC of 0.842 (95% CI: 0.829-0.855), while in the test dataset, it achieved an AUC of 0.820 (95% CI: 0.800-0.840), demonstrating good discriminatory performance. The calibration curve analysis revealed that the nomogram model closely approximated the ideal curve. Additionally, the DCA curve, CIC, and NRC demonstrated favorable clinical net benefit for the nomogram model. Conclusion This study successfully developed a predictive model to estimate the risk of bleeding in elderly aspirin users. This model can serve as a potential useful tool for clinicians to estimate the risk of bleeding in elderly aspirin users and make informed decisions regarding their treatment and management.
Collapse
Affiliation(s)
- Tenggao Chen
- Department of Colorectal Surgery, Affiliated Dongyang Hospital, Wenzhou Medical University, Dongyang, Zhejiang, 322100, People’s Republic of China
| | - Wanlin Lei
- Department of Biomedical Sciences Laboratory, Affiliated Dongyang Hospital, Wenzhou Medical University, Dongyang, Zhejiang, 322100, People’s Republic of China
| | - Maofeng Wang
- Department of Biomedical Sciences Laboratory, Affiliated Dongyang Hospital, Wenzhou Medical University, Dongyang, Zhejiang, 322100, People’s Republic of China
| |
Collapse
|
194
|
Liu WB, Fermin D, Xu AL, Kopp JB, Xu Q. Single-cell RNA sequencing data locate ALDH1A2-mediated retinoic acid synthetic pathway to glomerular parietal epithelial cells. Exp Biol Med (Maywood) 2024; 249:10167. [PMID: 39360029 PMCID: PMC11444976 DOI: 10.3389/ebm.2024.10167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 08/22/2024] [Indexed: 10/04/2024] Open
Abstract
Aldehyde dehydrogenase 1, family member A2, is a retinoic acid-synthesizing enzyme encoded by Aldh1a2 in mice and ALDH1A2 in humans. This enzyme is indispensable for kidney development, but its role in kidney physiology and pathophysiology remains to be fully defined. In this review, we mined single-cell and single-nucleus RNA sequencing databases of mouse and human kidneys and found that glomerular parietal epithelial cells (PECs) express a full set of genes encoding proteins needed for cellular vitamin A uptake, intracellular transport, and metabolism into retinoic acid. In particular, Aldh1a2/ALDH1A2 mRNAs are selectively enriched in mouse and human PECs. Aldh1a2 expression in PECs is greatly increased in a mouse model of anti-glomerular basement membrane glomerulonephritis and moderately induced in a mouse model of ischemia-reperfusion acute kidney injury. Aldh1a2 expression in PECs is substantially repressed in a chronic kidney disease mouse model combining diabetes, hypertension, and partial nephrectomy and is moderately repressed in mouse models of focal segmental glomerulosclerosis and diabetic nephropathy. Single-nucleus RNA sequencing data show that ALDH1A2 mRNA expression in PECs is diminished in patients with chronic kidney disease associated with diabetes, hypertension and polycystic kidney disease. In addition to data mining, we also performed Spearman's rank correlation coefficient analyses and identified gene transcripts correlated with Aldh1a2/ALDH1A2 transcripts in mouse PECs and PEC subtypes, and in human PECs of healthy subjects and patients with AKI or CKD. Furthermore, we conducted Gene Ontology pathway analyses and identified the biological pathways enriched among these Aldh1a2/ALDH1A2-correlated genes. Our data mining and analyses led us to hypothesize that ALDH1A2-mediated retinoic acid synthesis in PECs plays a yet-undefined role in the kidney and that its dysregulation mediates injury. Conditional, PEC-selective Aldh1a2 knockout, RNA silencing and transgenic mouse models will be useful tools to test this hypothesis. Clinical studies on genetics, epigenetics, expression and functions of ALDH1A2 and other genes needed for retinoic acid biosynthesis and signaling are also warranted.
Collapse
Affiliation(s)
- Wen-Bin Liu
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Damian Fermin
- Department of Internal Medicine, Division of Nephrology, University of Michigan, Ann Arbor, MI, United States
| | - An-Long Xu
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Jeffrey B. Kopp
- Kidney Disease Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Qihe Xu
- Renal Sciences and Integrative Chinese Medicine Laboratory, Department of Inflammation Biology, School of Immunology & Microbial Sciences, Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
| |
Collapse
|
195
|
Raza A, Mushtaq MN, Hassan S, Sharif A, Akhtar B, Akhtar MF. Mitigation of Diabetes Mellitus Using Euphorbia helioscopia Leaf Ethanolic Extract by Modulating GCK, GLUT4, IGF, and G6P Expressions in Streptozotocin-Induced Diabetic Rats. J Diabetes Res 2024; 2024:5497320. [PMID: 39329045 PMCID: PMC11424858 DOI: 10.1155/2024/5497320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 06/27/2024] [Accepted: 08/12/2024] [Indexed: 09/28/2024] Open
Abstract
Diabetes mellitus is a metabolic disorder. Synthetic antidiabetics are the commonly used treatment options associated with complications. The objective of this study was to explore the antioxidative and antidiabetic potential of Euphorbia helioscopia whole plant ethanolic extract using in vitro and in vivo models. For that purpose, the antioxidative potential was explored by using 2,2-diphenyl-1-picrylhydrazyl analysis. In vitro antidiabetic potential of the extract was evaluated using amylase inhibitory analysis. In vivo antidiabetic activity of the extract was assessed in diabetic rats using streptozotocin/nicotinamide (60 mg/kg/120 mg/kg) as an inducing agent. Metformin was used as standard. The results indicated the presence of significant quantities of phenolic 82.18 ± 1.28 mgg-1 gallic acid equivalent (GAE) and flavonoid 66.55±1.22 mgg-1 quercetin equivalent (QE) contents in the extract. Quantitation of phytoconstituents exhibited the presence of sinapic acid, myricetin, and quercetin using HPLC analysis. The extract inhibited α-amylase by 84.71%, and an antiglycemic potential of 50.34% was assessed in the OGTT assay. Biochemical analysis demonstrated a reduction in urea, creatinine, cholesterol, low-density lipoprotein, and alkaline phosphatase (p < 0.001) as compared to diabetic control rats at the dose of 500 mg/kg. An upregulation in the expressions of glucokinase, glucose transporter 4, peroxisome proliferator-activated receptor γ, and insulin-like growth factor was observed in treated rats in contrast to G6P expression, which was downregulated upon treatment. In conclusion, this study provided evidence of the antioxidative and antidiabetic potential of E. helioscopia whole plant ethanolic extract through in vitro and in vivo analysis and emphasized its promising role as a natural alternative.
Collapse
Affiliation(s)
- Ahmed Raza
- Faculty of PharmacyThe University of Lahore, Lahore 54000, Pakistan
| | | | - Sadia Hassan
- Department of Biomedical Engineering and SciencesSchool of Mechanical and Manufacturing EngineeringNational University of Science and Technology, Islamabad 24090, Pakistan
| | - Ali Sharif
- Department of PharmacologyFaculty of Pharmaceutical and Allied Health SciencesLahore College for Women University, Lahore 54000, Pakistan
| | - Bushra Akhtar
- Department of PharmacyUniversity of Agriculture, Faisalabad 38000, Pakistan
| | - Muhammad Furqan Akhtar
- Riphah Institute of Pharmaceutical SciencesRiphah International UniversityLahore Campus, Lahore 54000, Pakistan
| |
Collapse
|
196
|
Kositanurit W, Siritaweechai N, Varachotisate P, Burana C, Sukswai N, Surintrspanont J, Siriviriyakul P, Kaikaew K, Werawatganon D. Genistein mitigates diet-induced obesity and metabolic dysfunctions in gonadectomized mice with some sex-differential effects. Front Endocrinol (Lausanne) 2024; 15:1392866. [PMID: 39351533 PMCID: PMC11439649 DOI: 10.3389/fendo.2024.1392866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 08/26/2024] [Indexed: 10/04/2024] Open
Abstract
Background Obesity is associated with insulin resistance (IR) and metabolic dysfunction-associated steatotic liver disease (MASLD). Genistein, an isoflavone, is a promising natural compound for preventing and treating obesity and metabolic dysfunctions. We aimed to investigate the sex-specific protective effects of genistein on obesity, IR, and MASLD in a murine model of sex hormone deprivation with diet-induced obesity (DIO), mimicking postmenopausal women or aging men with metabolic syndrome. Methods Gonadectomized and sham-operated C57BL/6NJcl mice were fed a high-fat high-sucrose diet for 4 weeks to induce obesity (7 mice per group). In gonadectomized mice, genistein (16 mg/kg/day) or vehicle (7.5% dimethyl sulfoxide) was orally administered for 45 days. We assessed glucose homeostasis parameters, hepatic histopathology, and hepatic gene expression to investigate the effects of gonadectomy and genistein treatment. Results Gonadectomy exacerbated adiposity in both sexes. Ovariectomy diminished the protective effects of female gonadal hormones on the homeostatic model assessment for insulin resistance (HOMA-IR), serum alanine transaminase levels, hepatic steatosis score, and the expression of hepatic genes associated with MASLD progression and IR, such as Fasn, Srebf1, Saa1, Cd36, Col1a1, Pck1, and Ppargc1a. Genistein treatment in gonadectomized mice significantly reduced body weight gain and the hepatic steatosis score in both sexes. However, genistein treatment significantly attenuated HOMA-IR and the expression of the hepatic genes only in female mice. Conclusion Genistein treatment mitigates DIO-related MASLD in both male and female gonadectomized mice. Regarding hepatic gene expression associated with MASLD and IR, the beneficial effect of genistein was significantly evident only in female mice. This study suggests a potential alternative application of genistein in individuals with obesity and sex hormone deprivation, yet pending clinical trials.
Collapse
Affiliation(s)
- Weerapat Kositanurit
- Department of Physiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Natakorn Siritaweechai
- Department of Physiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Pachara Varachotisate
- Department of Physiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Chuti Burana
- Department of Physiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Narittee Sukswai
- Precision Pathology of Neoplasia Research Group, Department of Pathology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Jerasit Surintrspanont
- Precision Pathology of Neoplasia Research Group, Department of Pathology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Department of Pathology, King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, Thailand
| | - Prasong Siriviriyakul
- Department of Physiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Alternative and Complementary Medicine for Gastrointestinal and Liver Diseases, Chulalongkorn University, Bangkok, Thailand
| | - Kasiphak Kaikaew
- Department of Physiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Alternative and Complementary Medicine for Gastrointestinal and Liver Diseases, Chulalongkorn University, Bangkok, Thailand
| | - Duangporn Werawatganon
- Department of Physiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Alternative and Complementary Medicine for Gastrointestinal and Liver Diseases, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
197
|
Cabral RP, Ribeiro APD, Monte MG, Fujimori ASS, Tonon CR, Ferreira NF, Zanatti SG, Minicucci MF, Zornoff LAM, Paiva SARD, Polegato BF. Pera orange juice ( Citrus sinensis L. Osbeck) alters lipid metabolism and attenuates oxidative stress in the heart and liver of rats treated with doxorubicin. Heliyon 2024; 10:e36834. [PMID: 39263053 PMCID: PMC11388782 DOI: 10.1016/j.heliyon.2024.e36834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 08/14/2024] [Accepted: 08/22/2024] [Indexed: 09/13/2024] Open
Abstract
Background Doxorubicin (DOX) is a highly effective chemotherapy drug widely used to treat cancer, but its use is limited due to multisystemic toxicity. Lipid metabolism is also affected by doxorubicin. Orange juice can reduce dyslipidemia in other clinical situations and has already been shown to attenuate cardiotoxicity. Our aim is to evaluate the effects of Pera orange juice (Citrus sinensis L. Osbeck) on mitigating lipid metabolism imbalance, metabolic pathways, and DOX induced cytotoxic effects in the heart and liver. Methods Twenty-four male Wistar rats were allocated into 3 groups: Control (C); DOX (D); and DOX plus Pera orange juice (DOJ). DOJ received orange juice for 4 weeks, while C and D received water. At the end of each week, D and DOJ groups received 4 mg/kg/week DOX, intraperitoneal. At the end of 4 weeks animals were submitted to echocardiography and euthanasia. Results Animals treated with DOX decreased water intake and lost weight over time. At echocardiography, DOX treated rats presented morphologic alterations in the heart. DOX increased aspartate aminotransferase (AST), alanine aminotransferase (ALT), total cholesterol, high density lipoprotein (HDL), low-density lipoprotein (LDL), and triglycerides. It also reduced superoxide dismutase (SOD) activity, increased protein carbonylation in the heart and dihydroethidium (DHE) expression in the liver, decreased glucose transporter type 4 (GLUT4) and the nuclear receptor peroxisome proliferator-activated receptor γ (PPARγ1) in the heart, and reduced carnitine palmitoyltransferase I (CPT1) in the liver. Conclusion DOX caused dyslipidemia, liver and cardiac toxicity by increasing oxidative stress, and altered energy metabolic parameters in both organs. Despite not improving changes in left ventricular morphology, orange juice did attenuate oxidative stress and mitigate the metabolic effects of DOX.
Collapse
|
198
|
Sha J, Yi Q, Jiang X, Wang Z, Cao H, Jiang S. Pacing strategies in marathons: A systematic review. Heliyon 2024; 10:e36760. [PMID: 39281580 PMCID: PMC11400961 DOI: 10.1016/j.heliyon.2024.e36760] [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: 06/02/2024] [Revised: 08/21/2024] [Accepted: 08/21/2024] [Indexed: 09/18/2024] Open
Abstract
Background The pacing strategy embodies the tactical behavior of athletes in distributing their energy across different segments of a race; therefore, a quantitative analysis of pacing strategies in marathon races could deepen the understanding of both pacing behavior and physical capacity in marathon athletics. Objective The objective of this systematic review was to synthesize and characterize pacing strategies in marathon road races by exploring the categories of pacing strategies and the factors that influence these strategies during marathon events. Methods Preferred Reporting Items for Systematic Reviews guidelines were followed for systematic searches, appraisals, and syntheses of literature on this topic. Electronic databases such as Science Direct, SPORTDiscuss, PubMed, and Web of Science were searched up to July 2024. Records were eligible if they included pace performance measurements during competition, without experimental intervention that may influence their pace, in healthy, adult athletes at any level. Results A total of 39 studies were included in the review. Twenty-nine were observational studies, and 10 were experimental (randomized controlled trials). The assessment of article quality revealed an overall median NOS score of 8 (range 5-9). The included studies examined the pacing profiles of master athletes and finishers in half-marathon (n = 7, plus numbers compared to full marathon), full-marathon (n = 21), and ultramarathon (n = 11) road races. Considering that some studies refer to multiple pacing strategies, in general, 5 studies (∼13 %) reported even pacing, 3 (∼8 %) reported parabolic pacing, 7 (∼18 %) reported negative pacing, and 30 (∼77 %) reported positive pacing during marathon competitions. Gender, age, performance, pack, and physiological and psychological factors influence pacing strategies. Conclusion This study synthesized pacing performance in marathons and highlighted the significance of examining pacing strategies in these events, offering valuable insights for coaches and athletes. Several key findings were highlighted: (1) pacing profiles and pacing ranges were identified as the primary indicators of pacing strategies; (2) the pacing strategy was found to be dynamic, with the most substantial effects attributed to gender and distance; and (3) three distinct types of pacing strategies for marathons were classified: positive, negative, and even pacing. These findings advance the understanding of marathon pacing strategies by shedding light on the factors that influence athletes' pacing decisions and behaviors. Additionally, these findings offer practical benefits, aiding athletes in making well-informed tactical choices and developing effective pace plans to enhance marathon performance. However, due to the complex nature of marathon racing, further research is required to explore additional factors that might impact pacing strategies. A better grasp of optimal pacing strategies will foster progress in this area and serve as a basis for future research and advancements.
Collapse
Affiliation(s)
- Jungong Sha
- School of Physical Education, Shanghai University of Sport, Shanghai, China
| | - Qing Yi
- College of Physical Education, Dalian University, Dalian, China
| | - Xin Jiang
- College of Physical Education, Dalian University, Dalian, China
| | - Zhengwei Wang
- Department of physical education, Dalian Jiaotong University, Dalian, China
| | - Houwen Cao
- School of Kinesiology and Health Promotion, Dalian University of Technology, Dalian, China
| | - Shan Jiang
- Department of Sports Science and Physical Education, The Chinese University of Hong Kong, Shatin, Hong Kong
| |
Collapse
|
199
|
Chung Y, Kim JS. Association between health-related behaviors and obstructive sleep apnea among Korean adults. Sci Rep 2024; 14:21244. [PMID: 39261636 PMCID: PMC11390712 DOI: 10.1038/s41598-024-72401-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 09/06/2024] [Indexed: 09/13/2024] Open
Abstract
We aimed to investigate the association between health-related behaviors and obstructive sleep apnea (OSA) among Korean adults. A cross-sectional design using national open data was employed. Data from 8,096 adults aged 40 years and above who participated in the Korea National Health and Nutrition Examination Survey between 2019 and 2021 were analyzed. The participants' OSA risk level was assessed using the STOP-Bang questionnaire. A logistic regression analysis was performed to investigate the association between health-related behaviors and high risk for OSA. The association between health-related behaviors and OSA risk remained significant for former smokers (OR = 1.643) and high-risk drinking (OR = 1.365), after adjusting for variables that showed significant differences in general and metabolic characteristics. Implementing lifestyle modifications is crucial for mitigating the health and societal impact of OSA. Understanding and addressing modifiable risk factors, including high-risk drinking and smoking, should be prioritized in nursing intervention. Nursing interventions are critical for preventing and managing OSA among Korean adults. Prioritizing high-risk behaviors through cessation programs and education is essential. Vulnerability of individuals living alone must be addressed through community outreach and support services. Emphasizing routine screenings for pre-hypertension and pre-diabetes, promoting balanced nutrition, and encouraging physical activity are crucial.
Collapse
Affiliation(s)
- Yoongi Chung
- Department of Nursing, Chung-Ang University, 84, Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea
| | - Ji-Su Kim
- Department of Nursing, Chung-Ang University, 84, Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea.
| |
Collapse
|
200
|
Shekho D, Mishra R, Kamal R, Bhatia R, Awasthi A. Breaking Barriers in Alzheimer's Disease: the Role of Advanced Drug Delivery Systems. AAPS PharmSciTech 2024; 25:207. [PMID: 39237748 DOI: 10.1208/s12249-024-02923-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 08/18/2024] [Indexed: 09/07/2024] Open
Abstract
Alzheimer's disease (AD), characterized by cognitive impairment, brain plaques, and tangles, is a global health concern affecting millions. It involves the build-up of amyloid-β (Aβ) and tau proteins, the formation of neuritic plaques and neurofibrillary tangles, cholinergic system dysfunction, genetic variations, and mitochondrial dysfunction. Various signaling pathways and metabolic processes are implicated in AD, along with numerous biomarkers used for diagnosis, risk assessment, and research. Despite these, there is no cure or effective treatment for AD. It is critically important to address this immediately to develop novel drug delivery systems (NDDS) capable of targeting the brain and delivering therapeutic agents to modulate the pathological processes of AD. This review summarizes AD, its pathogenesis, related signaling pathways, biomarkers, conventional treatments, the need for NDDS, and their application in AD treatment. It also covers preclinical, clinical, and ongoing trials, patents, and marketed AD formulations.
Collapse
Affiliation(s)
- Devank Shekho
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Ritika Mishra
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Raj Kamal
- Department of Quality Assurance, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Rohit Bhatia
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Ankit Awasthi
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, 142001, Punjab, India.
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India.
| |
Collapse
|