1
|
Yin N, Luo J, Wang C, Xiong Y, Sun Y, Yuan E, Zhang H. Comprehensive Evaluation of the Effects of Hot Air Drying Temperature on the Chemical Composition, Flavor Characteristics and Biological Activity of Houttuynia cordata Thunb.. Foods 2025; 14:1962. [PMID: 40509489 PMCID: PMC12154787 DOI: 10.3390/foods14111962] [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: 05/08/2025] [Revised: 05/26/2025] [Accepted: 05/28/2025] [Indexed: 06/16/2025] Open
Abstract
This study systematically investigated the drying kinetics and quality characteristics of Houttuynia cordata Thunb. (HCT) under different processing conditions, evaluating how freeze-drying and hot-air drying (40 °C, 50 °C, 60 °C) affect bioactive compound preservation, antioxidant efficacy, and metabolic profiles to identify the optimal drying method for maximizing its functional benefits. A thin-layer drying model was established to evaluate drying parameters such as effective diffusion coefficient and activation energy. Changes in chemical composition, sensory properties, and antioxidant activity were analyzed using UHPLC-LTQ-Orbitrap-MS, electronic nose/tongue, and HepG2 cell assays. Results showed that the Aghabashlo model was optimal for demonstrating the drying process with the best fit. The 50 °C heating temperature was shown to yield the highest diffusion coefficient. Hot-air drying at 50 °C balanced efficiency and sensory quality, whereas 60 °C significantly altered flavor and metabolite composition. Results of the metabolomic analysis indicated that freeze-drying enhanced the retention of phenolic acids and flavonoids, while hot-air drying led to increased fatty acid metabolites. Freeze-drying preserved the antioxidant activity and natural flavor of HCT. Nevertheless, the metabolic fate of rutin, quercetin, and chlorogenic acid was not significantly affected by the drying method (freeze-drying vs. 50 °C drying). These findings provide a theoretical foundation for improving HCT's therapeutic and sensory qualities through optimized drying techniques.
Collapse
Affiliation(s)
- Ning Yin
- Department of Food Nutrition and Safety, College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330004, China;
| | - Jing Luo
- Department of Pharmaceutics, College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330004, China; (J.L.); (C.W.); (Y.X.)
| | - Chaoping Wang
- Department of Pharmaceutics, College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330004, China; (J.L.); (C.W.); (Y.X.)
| | - Yaokun Xiong
- Department of Pharmaceutics, College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330004, China; (J.L.); (C.W.); (Y.X.)
| | - Yong Sun
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China;
| | - En Yuan
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China;
| | - Hua Zhang
- Department of Food Nutrition and Safety, College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330004, China;
| |
Collapse
|
2
|
Han A, Yermalovich A, Najia MAT, Pearson DS, Fujiwara Y, Bolgov M, Kubaczka C, North TE, Lundin V, Orkin S, Daley GQ. RNA Editors Sculpt the Transcriptome During Terminal Erythropoiesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.03.647020. [PMID: 40236006 PMCID: PMC11996565 DOI: 10.1101/2025.04.03.647020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Selective RNA degradation during terminal erythropoiesis results in a globin-rich transcriptome in mature erythrocytes, but the specific RNA decay pathways remain unknown. We found that deficiency of the terminal uridylyl transferase enzyme Zcchc6 and the 3'-5' exoribonuclease Dis3l2 in mouse models led to fetal and perinatal reticulocytosis, an accumulation of RNA-rich precursors of terminal erythroid cells, suggesting their crucial roles in terminal red cell differentiation. Notably, knockout embryos exhibited persistent high-level expression of Hbb-bh1 globin, the ortholog of human fetal γ- globin. Perturbation of the Zcchc6-Dis3l2 pathway in mice engineered to express the human β-globin locus likewise increased γ -globin levels in fetal erythroid cells, suggesting that globin switching entails post-transcriptional mechanisms of mRNA destabilization in addition to transcriptional down-regulation. We cultured human hematopoietic stem and progenitor cells (HSPCs), performed CRISPR/Cas9-mediated knockout of ZCCHC6 and DIS3L2, and observed accumulation of RNA and elevated γ-globin levels in terminal erythroid cells. Our findings reveal a conserved role for the ZCCHC6/DIS3L2 RNA editors in terminal erythropoiesis and demonstrate a post-transcriptional mechanism for γ- globin gene switching, advancing research into in vitro erythrocyte generation and γ- globin stabilization to ameliorate hemoglobinopathies.
Collapse
|
3
|
Gonzalez-Sanabria N, Contreras GF, Rojas M, Duarte Y, Gonzalez-Nilo FD, Perozo E, Latorre R. The BK channel-NS1619 agonist complex reveals molecular insights on allosteric activation gating. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.27.645783. [PMID: 40235977 PMCID: PMC11996348 DOI: 10.1101/2025.03.27.645783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
BK channels play essential roles in a wealth of physiological functions, including regulating smooth muscle tone and neurotransmitter release. Its dysfunction, often caused by loss-of-function mutations, can lead to severe phenotypes, including ataxia and sensory impairment. Despite the therapeutic potential of BK channel agonists, the molecular mechanisms by which they stabilize the pore's open conformation remain unclear. Using cryo-electron microscopy and molecular dynamic simulations, we identified that NS1619, a synthetic benzimidazolone agonist, first described as a BK opener, binds within a pocket formed by the S6/RCK1 linker and the S4 transmembrane segment. Agonist binding drives a twisting motion in the S6 segment, enabling critical interactions with residues K330, K331, and F223. Our findings clarify the mechanism of NS1619 and suggest that its binding site can accommodate other agonists, highlighting a promising target for therapeutic development. Teaser BK channel activation by NS1619 reveals key binding interactions, offering insights for designing targeted therapeutic agents.
Collapse
|
4
|
Fernández-Castillejo S, Badia J, de la Cruz-Merino L, Martín Garcia-Sáncho A, Carnicero-González F, Palazón-Carrión N, Ríos-Herranz E, de la Cruz-Vicente F, Rueda-Domínguez A, Martínez-Banaclocha N, Gómez-Codina J, Labrador J, Martínez-Madueño F, Amigó N, Salar-Silvestre A, Rodríguez-Abreu D, Gálvez-Carvajal L, Sánchez-Beato M, Provencio-Pulla M, Guirado-Risueño M, Nogales E, Sánchez-Margalet V, Jiménez-Cortegana C, Rodríguez-García G, Cumeras R, Gumà J. Ketone Bodies Are Potential Prognostic Biomarkers in Relapsed/Refractory Diffuse Large B-Cell Lymphoma: Results from the R2-GDP-GOTEL Trial. Cancers (Basel) 2025; 17:532. [PMID: 39941898 PMCID: PMC11817199 DOI: 10.3390/cancers17030532] [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: 12/03/2024] [Revised: 01/10/2025] [Accepted: 01/16/2025] [Indexed: 02/16/2025] Open
Abstract
Background: Patients with relapsed or refractory (R/R) diffuse large B-cell lymphoma (DLBCL) who are ineligible for high-dose chemotherapy have limited treatment options and poor life expectancy. The purpose of this study is to identify a serum metabolomic profile that may be predictive of outcome in patients with R/R-DLBCL. Methods: This study included 69 R/R DLBCL patients from the R2-GDP-GOTEL trial (EudraCT 2014-001620-299). Serum samples were collected at baseline, and the mean length of follow-up was 41 months. Serum metabolites were analyzed by nuclear magnetic resonance (NMR). Metabolites were correlated with treatment response, progression-free survival (PFS), and overall survival (OS). Results: Serum levels of 3-hydroxybutyrate (3OHB) and acetone were significantly (p < 0.001) associated with PFS (3OHB: hazard ratio [HR] 7.7, 95% confidence interval [CI] 2.5-24.1; acetone: HR 9.32, 95% CI 2.75-31.6) and OS (3OHB: HR 9.32, 95% CI 2.75-31.6; acetone: HR 1.92, 95% CI 1.36-2.69). Serum values of 141 µM for 3OHB and 40 µM for acetone were the optimal cutoffs associated with the survival outcomes. Elevated 3OHB levels (>141 μM) were specific to the ABC subtype of DLBCL, while acetone levels were elevated in both types of DLCBL but more pronounced in ABC cases. In a multivariate survival analysis, including the International Prognostic Index (IPI) score and refractoriness status (R/R), 3OHB and acetone remained significant. To aid oncologists employing the R2-GDP regime, we constructed PFS and OS nomograms for R/R-DLBCL risk stratification, incorporating 3OHB levels or acetone levels, IPI score, and refractoriness status. The nomogram with 3OHB and refractoriness status showed a time-dependent AUC of 0.86 for 6-month PFS and 0.84 for 12-month OS. These nomograms provide a comprehensive tool for individualized risk assessment and treatment optimization. Conclusions: The ketone bodies 3OHB and acetone are potential prognostic biomarkers of poor outcome in R/R DLBCL patients treated with the R2-GDP regimen, independently of IPI score and chemorefractoriness status.
Collapse
Affiliation(s)
- Sara Fernández-Castillejo
- Translational, Epidemiological and Clinical Oncological Research Group (GIOTEC), Department of Oncology, Institut d’Investigació Sanitària Pere Virgili (IISPV), 43204 Reus, Tarragona, Spain; (S.F.-C.); (J.B.); (F.M.-M.); (J.G.)
- Institut d’Oncologia de la Catalunya Sud (IOCS), Hospital Universitari Sant Joan de Reus, 43204 Reus, Tarragona, Spain
| | - Joan Badia
- Translational, Epidemiological and Clinical Oncological Research Group (GIOTEC), Department of Oncology, Institut d’Investigació Sanitària Pere Virgili (IISPV), 43204 Reus, Tarragona, Spain; (S.F.-C.); (J.B.); (F.M.-M.); (J.G.)
- Institut d’Oncologia de la Catalunya Sud (IOCS), Hospital Universitari Sant Joan de Reus, 43204 Reus, Tarragona, Spain
| | - Luís de la Cruz-Merino
- Cancer Immunotherapy Group, Oncohematology and Genetics Department, Biomedicine Institute of Seville (IBIS)/CSIC, 41013 Seville, Spain; (L.d.l.C.-M.); (N.P.-C.); (E.N.)
- Department of Clinical Oncology, University Hospital Virgen Macarena and School of Medicine, University of Sevilla, 41013 Sevilla, Spain
| | - Alejandro Martín Garcia-Sáncho
- Department of Hematology, Hospital Universitario de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Universidad de Salamanca, 37007 Salamanca, Spain;
- CIBER de Cáncer (CIBERONC), Institute of Health Carlos III, 28029 Madrid, Spain
| | | | - Natalia Palazón-Carrión
- Cancer Immunotherapy Group, Oncohematology and Genetics Department, Biomedicine Institute of Seville (IBIS)/CSIC, 41013 Seville, Spain; (L.d.l.C.-M.); (N.P.-C.); (E.N.)
- Department of Clinical Oncology, University Hospital Virgen Macarena and School of Medicine, University of Sevilla, 41013 Sevilla, Spain
| | - Eduardo Ríos-Herranz
- Department of Hematology, Hospital Universitario Virgen de Valme, 41014 Sevilla, Spain;
| | - Fátima de la Cruz-Vicente
- Department of Hematology, Hospital Universitario Virgen del Rocío, 41013 Sevilla, Spain; (F.d.l.C.-V.); (G.R.-G.)
| | - Antonio Rueda-Domínguez
- Department of Clinical Oncology. Hospital Universitario Virgen de la Victoria, 29010 Málaga, Spain; (A.R.-D.); (L.G.-C.)
| | - Natividad Martínez-Banaclocha
- Department of Oncology, Dr. Balmis General University Hospital, Alicante Institute for Health and Biomedical Research (ISABIAL), 03010 Alicante, Spain;
| | - José Gómez-Codina
- Department of Clinical Oncology, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain;
| | - Jorge Labrador
- Department of Hematology, Hospital Universitario de Burgos, 09006 Burgos, Spain;
| | - Francisca Martínez-Madueño
- Translational, Epidemiological and Clinical Oncological Research Group (GIOTEC), Department of Oncology, Institut d’Investigació Sanitària Pere Virgili (IISPV), 43204 Reus, Tarragona, Spain; (S.F.-C.); (J.B.); (F.M.-M.); (J.G.)
- Institut d’Oncologia de la Catalunya Sud (IOCS), Hospital Universitari Sant Joan de Reus, 43204 Reus, Tarragona, Spain
- Faculty of Medicine and Health Sciences, Universitat Rovira i Virgili (URV), 43201 Reus, Tarragona, Spain;
| | - Núria Amigó
- Faculty of Medicine and Health Sciences, Universitat Rovira i Virgili (URV), 43201 Reus, Tarragona, Spain;
- Biosfer Teslab, 43206 Reus, Tarragona, Spain
| | | | - Delvys Rodríguez-Abreu
- Department of Clinical Oncology, Hospital Universitario Insular de Gran Canaria, 35016 Las Palmas de Gran Canaria, Las Palmas, Spain;
| | - Laura Gálvez-Carvajal
- Department of Clinical Oncology. Hospital Universitario Virgen de la Victoria, 29010 Málaga, Spain; (A.R.-D.); (L.G.-C.)
| | - Margarita Sánchez-Beato
- Lymphoma Research Group, Department of Medical Oncology, Hospital Universitario Puerta de Hierro-Majadahonda, IDIPHISA, 28222 Majadahonda, Madrid, Spain;
| | - Mariano Provencio-Pulla
- Department of Clinical Oncology, Hospital Universitario Puerta De Hierro-Majadahonda, IDIPHISA, 28222 Majadahonda, Madrid, Spain;
| | - Maria Guirado-Risueño
- Department of Clinical Oncology, Hospital Universitario de Elche, 03203 Elche, Alicante, Spain;
| | - Esteban Nogales
- Cancer Immunotherapy Group, Oncohematology and Genetics Department, Biomedicine Institute of Seville (IBIS)/CSIC, 41013 Seville, Spain; (L.d.l.C.-M.); (N.P.-C.); (E.N.)
- Department of Clinical Oncology, University Hospital Virgen Macarena and School of Medicine, University of Sevilla, 41013 Sevilla, Spain
| | - Víctor Sánchez-Margalet
- Medical Biochemistry and Molecular Biology and Immunology, Hospital Universitario Virgen de la Macarena, 41009 Sevilla, Spain; (V.S.-M.); (C.J.-C.)
| | - Carlos Jiménez-Cortegana
- Medical Biochemistry and Molecular Biology and Immunology, Hospital Universitario Virgen de la Macarena, 41009 Sevilla, Spain; (V.S.-M.); (C.J.-C.)
| | - Guillermo Rodríguez-García
- Department of Hematology, Hospital Universitario Virgen del Rocío, 41013 Sevilla, Spain; (F.d.l.C.-V.); (G.R.-G.)
| | - Raquel Cumeras
- Translational, Epidemiological and Clinical Oncological Research Group (GIOTEC), Department of Oncology, Institut d’Investigació Sanitària Pere Virgili (IISPV), 43204 Reus, Tarragona, Spain; (S.F.-C.); (J.B.); (F.M.-M.); (J.G.)
- Department of Electrical and Automatic Electronic Engineering, Universitat Rovira i Virgili (URV), 43002 Tarragona, Spain
| | - Josep Gumà
- Translational, Epidemiological and Clinical Oncological Research Group (GIOTEC), Department of Oncology, Institut d’Investigació Sanitària Pere Virgili (IISPV), 43204 Reus, Tarragona, Spain; (S.F.-C.); (J.B.); (F.M.-M.); (J.G.)
- Institut d’Oncologia de la Catalunya Sud (IOCS), Hospital Universitari Sant Joan de Reus, 43204 Reus, Tarragona, Spain
- Faculty of Medicine and Health Sciences, Universitat Rovira i Virgili (URV), 43201 Reus, Tarragona, Spain;
| |
Collapse
|
5
|
Amarjeet, Kumar U, Sodhi M, Kumar D, Vivek P, Niranjan SK, Kataria RS, Kumar S, Sharma M, Tiwari M, Aggarwal RAK, Bharti VK, Iqbal M, Rabgais S, Kumar A, Chanda D, Mukesh M. Characterizing metabolome signature of colostrum, transition and mature milk of indigenous cows (Bos indicus) adapted to high altitude environment of Leh-Ladakh. Food Chem 2025; 464:141767. [PMID: 39515162 DOI: 10.1016/j.foodchem.2024.141767] [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: 06/23/2024] [Revised: 09/26/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024]
Abstract
This study has identified 46 metabolites in colostrum, transition milk and mature milk of unique indigenous high altitude adapted Ladakhi cows using 1D 1H 800 MHz NMR spectroscopy. The multivariate analysis revealed that UDP-galactose, UDP-glucose, citrate, creatine phosphate, myo-inositol, lactose, 2-oxoglutarate, valine, maltose, leucine, dimethylamine, and choline with high VIP scores could differentiate the colostrum, transition and mature milk in separate clusters. Highly enriched metabolites in colostrum such as UDP-galactose, UDP-glucose play crucial roles in cell growth, differentiation, and defense responses. Similarly, the presence of branched chain amino acids in colostrum could be linked to mammary gland development, N-acetylglucosamine, N-acetyl carnitine, choline etc. in high concentration in colostrum l might be helping in growth and development of neonatal calves of Ladakhi cows under hypoxia environment. Overall, this study has helped to characterize the metabolomic signatures of milk/colostrum of Ladakhi cows adapted to high altitude and cold desert of Leh-Ladakh.
Collapse
Affiliation(s)
- Amarjeet
- ICAR-National Bureau of Animal Genetic Resources, Karnal, Haryana 132001, India
| | - Umesh Kumar
- ICAR-National Bureau of Animal Genetic Resources, Karnal, Haryana 132001, India
| | - Monika Sodhi
- ICAR-National Bureau of Animal Genetic Resources, Karnal, Haryana 132001, India
| | - Dinesh Kumar
- ICAR-National Bureau of Animal Genetic Resources, Karnal, Haryana 132001, India
| | - Prince Vivek
- ICAR-National Bureau of Animal Genetic Resources, Karnal, Haryana 132001, India
| | - Saket K Niranjan
- ICAR-National Bureau of Animal Genetic Resources, Karnal, Haryana 132001, India
| | | | - Sudarshan Kumar
- ICAR-National Bureau of Animal Genetic Resources, Karnal, Haryana 132001, India
| | - Manish Sharma
- ICAR-National Bureau of Animal Genetic Resources, Karnal, Haryana 132001, India
| | - Manish Tiwari
- ICAR-National Bureau of Animal Genetic Resources, Karnal, Haryana 132001, India
| | - Rajeev A K Aggarwal
- ICAR-National Bureau of Animal Genetic Resources, Karnal, Haryana 132001, India
| | - Vijay K Bharti
- ICAR-National Bureau of Animal Genetic Resources, Karnal, Haryana 132001, India
| | - M Iqbal
- ICAR-National Bureau of Animal Genetic Resources, Karnal, Haryana 132001, India
| | - Stanzin Rabgais
- ICAR-National Bureau of Animal Genetic Resources, Karnal, Haryana 132001, India
| | - Abhishek Kumar
- ICAR-National Bureau of Animal Genetic Resources, Karnal, Haryana 132001, India
| | - Divya Chanda
- ICAR-National Bureau of Animal Genetic Resources, Karnal, Haryana 132001, India
| | - Manishi Mukesh
- ICAR-National Bureau of Animal Genetic Resources, Karnal, Haryana 132001, India.
| |
Collapse
|
6
|
Chaber P, Andrä-Żmuda S, Śmigiel-Gac N, Zięba M, Dawid K, Martinka Maksymiak M, Adamus G. Enhancing the Potential of PHAs in Tissue Engineering Applications: A Review of Chemical Modification Methods. MATERIALS (BASEL, SWITZERLAND) 2024; 17:5829. [PMID: 39685265 DOI: 10.3390/ma17235829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 11/22/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024]
Abstract
Polyhydroxyalkanoates (PHAs) are a family of polyesters produced by many microbial species. These naturally occurring polymers are widely used in tissue engineering because of their in vivo degradability and excellent biocompatibility. The best studied among them is poly(3-hydroxybutyrate) (PHB) and its copolymer with 3-hydroxyvaleric acid (PHBV). Despite their superior properties, PHB and PHBV suffer from high crystallinity, poor mechanical properties, a slow resorption rate, and inherent hydrophobicity. Not only are PHB and PHBV hydrophobic, but almost all members of the PHA family struggle because of this characteristic. One can overcome the limitations of microbial polyesters by modifying their bulk or surface chemical composition. Therefore, researchers have put much effort into developing methods for the chemical modification of PHAs. This paper explores a rarely addressed topic in review articles-chemical methods for modifying the structure of PHB and PHBV to enhance their suitability as biomaterials for tissue engineering applications. Different chemical strategies for improving the wettability and mechanical properties of PHA scaffolds are discussed in this review. The properties of PHAs that are important for their applications in tissue engineering are also discussed.
Collapse
Affiliation(s)
- Paweł Chaber
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, ul. M. Curie-Skłodowska 34, 41-819 Zabrze, Poland
| | - Silke Andrä-Żmuda
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, ul. M. Curie-Skłodowska 34, 41-819 Zabrze, Poland
| | - Natalia Śmigiel-Gac
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, ul. M. Curie-Skłodowska 34, 41-819 Zabrze, Poland
| | - Magdalena Zięba
- Department of Optoelectronics, Silesian University of Technology, ul. B. Krzywoustego 2, 44-100 Gliwice, Poland
| | - Kamil Dawid
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, ul. M. Curie-Skłodowska 34, 41-819 Zabrze, Poland
| | - Magdalena Martinka Maksymiak
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, ul. M. Curie-Skłodowska 34, 41-819 Zabrze, Poland
| | - Grażyna Adamus
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, ul. M. Curie-Skłodowska 34, 41-819 Zabrze, Poland
| |
Collapse
|
7
|
Samantsidis GR, Smith RC. Exploring new dimensions of immune cell biology in Anopheles gambiae through genetic immunophenotyping. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.22.619690. [PMID: 39484609 PMCID: PMC11526922 DOI: 10.1101/2024.10.22.619690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Mosquito immune cells, or hemocytes, are integral components of the innate immune responses that define vector competence. However, the lack of genetic resources has limited their characterization and our understanding of their functional roles in immune signaling. To overcome these challenges, we engineered transgenic Anopheles gambiae that express fluorescent proteins under the control of candidate hemocyte promoters. Following the characterization of five transgenic constructs through gene expression and microscopy-based approaches, we examine mosquito immune cell populations by leveraging advanced spectral imaging flow cytometry. Our results comprehensively map the composition of mosquito hemocytes, classifying them into twelve distinct populations based on size, granularity, ploidy, phagocytic capacity, and the expression of PPO6, SPARC, and LRIM15 genetic markers. Together, our novel use of morphological properties and genetic markers provides increased resolution into our understanding of mosquito hemocytes, highlighting the complexity and plasticity of these immune cell populations, while providing the foundation for deeper investigations into their roles in immunity and pathogen transmission.
Collapse
Affiliation(s)
| | - Ryan C Smith
- Department of Plant Pathology, Entomology and Microbiology, Iowa State University, Ames, Iowa, USA
| |
Collapse
|
8
|
Calamari ZT, Song A, Cohen E, Akter M, Roy RD, Hallikas O, Christensen MM, Li P, Marangoni P, Jernvall J, Klein OD. Vole genomics links determinate and indeterminate growth of teeth. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.18.572015. [PMID: 38187646 PMCID: PMC10769287 DOI: 10.1101/2023.12.18.572015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Continuously growing teeth are an important innovation in mammalian evolution, yet genetic regulation of continuous growth by stem cells remains incompletely understood. Dental stem cells responsible for tooth crown growth are lost at the onset of tooth root formation. Genetic signaling that initiates this loss is difficult to study with the ever-growing incisor and rooted molars of mice, the most common mammalian dental model species, because signals for root formation overlap with signals that pattern tooth size and shape (i.e., cusp patterns). Different species of voles (Cricetidae, Rodentia, Glires) have evolved rooted and unrooted molars that have similar size and shape, providing alternative models for studying roots. We assembled a de novo genome of Myodes glareolus, a vole with high-crowned, rooted molars, and performed genomic and transcriptomic analyses in a broad phylogenetic context of Glires (rodents and lagomorphs) to assess differential selection and evolution in tooth forming genes. We identified 15 dental genes with changing synteny relationships and six dental genes undergoing positive selection across Glires, two of which were undergoing positive selection in species with unrooted molars, Dspp and Aqp1. Decreased expression of both genes in prairie voles with unrooted molars compared to bank voles supports the presence of positive selection and may underlie differences in root formation. Bulk transcriptomics analyses of embryonic molar development in bank voles also demonstrated conserved patterns of dental gene expression compared to mice, with species-specific variation likely related to developmental timing and morphological differences between mouse and vole molars. Our results support ongoing evolution of dental genes across Glires, revealing the complex evolutionary background of convergent evolution for ever-growing molars.
Collapse
Affiliation(s)
- Zachary T. Calamari
- Baruch College, City University of New York, One Bernard Baruch Way, New York, NY 10010, USA
- The Graduate Center, City University of New York, 365 Fifth Ave, New York, NY 10016, USA
- Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, San Francisco, CA 94158, USA
- Division of Paleontology, American Museum of Natural History, Central Park West at 79th Street, New York, NY, 10024, USA
| | - Andrew Song
- Baruch College, City University of New York, One Bernard Baruch Way, New York, NY 10010, USA
- Cornell University, 616 Thurston Ave, Ithaca, NY 14853, USA
| | - Emily Cohen
- Baruch College, City University of New York, One Bernard Baruch Way, New York, NY 10010, USA
- New York University College of Dentistry, 345 E 34th St, New York, NY 10010
| | - Muspika Akter
- Baruch College, City University of New York, One Bernard Baruch Way, New York, NY 10010, USA
| | - Rishi Das Roy
- Institute of Biotechnology, University of Helsinki, FI-00014 Helsinki, Finland
| | - Outi Hallikas
- Institute of Biotechnology, University of Helsinki, FI-00014 Helsinki, Finland
| | - Mona M. Christensen
- Institute of Biotechnology, University of Helsinki, FI-00014 Helsinki, Finland
| | - Pengyang Li
- Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, San Francisco, CA 94158, USA
- Department of Pediatrics, Cedars-Sinai Guerin Children’s, 8700 Beverly Blvd., Suite 2416, Los Angeles, CA 90048, USA
| | - Pauline Marangoni
- Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, San Francisco, CA 94158, USA
- Department of Pediatrics, Cedars-Sinai Guerin Children’s, 8700 Beverly Blvd., Suite 2416, Los Angeles, CA 90048, USA
| | - Jukka Jernvall
- Institute of Biotechnology, University of Helsinki, FI-00014 Helsinki, Finland
- Department of Geosciences and Geography, University of Helsinki, FI-00014 Helsinki, Finland
| | - Ophir D. Klein
- Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, San Francisco, CA 94158, USA
- Department of Pediatrics, Cedars-Sinai Guerin Children’s, 8700 Beverly Blvd., Suite 2416, Los Angeles, CA 90048, USA
| |
Collapse
|
9
|
Uebanso T, Fukui M, Naito C, Shimohata T, Mawatari K, Takahashi A. SLC16a6, mTORC1, and Autophagy Regulate Ketone Body Excretion in the Intestinal Cells. BIOLOGY 2023; 12:1467. [PMID: 38132294 PMCID: PMC10740559 DOI: 10.3390/biology12121467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 11/21/2023] [Accepted: 11/22/2023] [Indexed: 12/23/2023]
Abstract
Ketone bodies serve several functions in the intestinal epithelium, such as stem cell maintenance, cell proliferation and differentiation, and cancer growth. Nevertheless, there is limited understanding of the mechanisms governing the regulation of intestinal ketone body concentration. In this study, we elucidated the factors responsible for ketone body production and excretion using shRNA-mediated or pharmacological inhibition of specific genes or functions in the intestinal cells. We revealed that a fasting-mimicked culture medium, which excluded glucose, pyruvate, and glutamine, augmented ketone body production and excretion in the Caco2 and HT29 colorectal cells. This effect was attenuated by glucose or glutamine supplementation. On the other hand, the inhibition of the mammalian target of rapamycin complex1 (mTORC1) recovered a fraction of the excreted ketone bodies. In addition, the pharmacological or shbeclin1-mediated inhibition of autophagy suppressed ketone body excretion. The knockdown of basigin, a transmembrane protein responsible for targeting monocarboxylate transporters (MCTs), such as MCT1 and MCT4, suppressed lactic acid and pyruvic acid excretion but increased ketone body excretion. Finally, we found that MCT7 (SLC16a6) knockdown suppressed ketone body excretion. Our findings indicate that the mTORC1-autophagy axis and MCT7 are potential targets to regulate ketone body excretion from the intestinal epithelium.
Collapse
Affiliation(s)
- Takashi Uebanso
- Department of Preventive Environment and Nutrition, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8503, Japan
| | - Moeka Fukui
- Department of Preventive Environment and Nutrition, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8503, Japan
| | - Chisato Naito
- Department of Preventive Environment and Nutrition, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8503, Japan
| | - Takaaki Shimohata
- Department of Preventive Environment and Nutrition, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8503, Japan
- Faculty of Marine Biosciences, Fukui Prefectural University, Fukui 917-0003, Japan
| | - Kazuaki Mawatari
- Department of Preventive Environment and Nutrition, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8503, Japan
| | - Akira Takahashi
- Department of Preventive Environment and Nutrition, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8503, Japan
| |
Collapse
|
10
|
Pervaiz I, Mehta Y, Sherill K, Patel D, Al-Ahmad AJ. Ketone bodies supplementation restores the barrier function, induces a metabolic switch, and elicits beta-hydroxybutyrate diffusion across a monolayer of iPSC-derived brain microvascular endothelial cells. Microvasc Res 2023; 150:104585. [PMID: 37437687 DOI: 10.1016/j.mvr.2023.104585] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 07/05/2023] [Accepted: 07/07/2023] [Indexed: 07/14/2023]
Abstract
Glucose constitutes the main source of energy for the central nervous system (CNS), its entry occurring at the blood-brain barrier (BBB) via the presence of glucose transporter 1 (GLUT1). However, under food intake restrictions, the CNS can utilize ketone bodies (KB) as an alternative source of energy. Notably, the relationship between the BBB and KBs and its effect on their glucose metabolism remains poorly understood. In this study, we investigated the effect of glucose deprivation on the brain endothelium in vitro, and supplementation with KBs using induced pluripotent stem cell (iPSC)-derived brain microvascular endothelial cell-like cells (iBMECs). Glucose-free environment significantly decreased cell metabolic activity and negatively impacted the barrier function. In addition, glucose deprivation did not increase GLUT1 expression but also resulted in a decrease in glucose uptake and glycolysis. Supplementation of glucose-deprived iBMECs monolayers with KB showed no improvement and even worsened upon treatment with acetoacetate. However, under a hypoglycemic condition in the presence of KBs, we noted a slight improvement of the barrier function, with no changes in glucose uptake. Notably, hypoglycemia and/or KB pre-treatment elicited a saturable beta-hydroxybutyrate diffusion across iBMECs monolayers, such diffusion occurred partially via an MCT1-dependent mechanism. Taken together, our study highlights the importance of glucose metabolism and the reliance of the brain endothelium on glucose and glycolysis for its function, such dependence is unlikely to be covered by KBs supplementation. In addition, KB diffusion at the BBB appeared induced by KB pre-treatment and appears to involve an MCT1-dependent mechanism.
Collapse
Affiliation(s)
- Iqra Pervaiz
- Texas Tech University Health Sciences Center - Jerry H. Hodge School of Pharmacy Department of Pharmaceutical Sciences, Amarillo, TX, United States of America; Center for Blood-Brain Barrier Research, Texas Tech University Health Sciences Center - Jerry H. Hodge School of Pharmacy, Amarillo, TX, United States of America.
| | - Yash Mehta
- Texas Tech University Health Sciences Center - Jerry H. Hodge School of Pharmacy Department of Pharmaceutical Sciences, Amarillo, TX, United States of America; Center for Blood-Brain Barrier Research, Texas Tech University Health Sciences Center - Jerry H. Hodge School of Pharmacy, Amarillo, TX, United States of America
| | - Kinzie Sherill
- Texas Tech University Health Sciences Center - Jerry H. Hodge School of Pharmacy Department of Pharmaceutical Sciences, Amarillo, TX, United States of America; Center for Blood-Brain Barrier Research, Texas Tech University Health Sciences Center - Jerry H. Hodge School of Pharmacy, Amarillo, TX, United States of America
| | - Dhavalkumar Patel
- Texas Tech University Health Sciences Center - Jerry H. Hodge School of Pharmacy Department of Pharmaceutical Sciences, Amarillo, TX, United States of America; Center for Blood-Brain Barrier Research, Texas Tech University Health Sciences Center - Jerry H. Hodge School of Pharmacy, Amarillo, TX, United States of America
| | - Abraham J Al-Ahmad
- Texas Tech University Health Sciences Center - Jerry H. Hodge School of Pharmacy Department of Pharmaceutical Sciences, Amarillo, TX, United States of America; Center for Blood-Brain Barrier Research, Texas Tech University Health Sciences Center - Jerry H. Hodge School of Pharmacy, Amarillo, TX, United States of America.
| |
Collapse
|
11
|
Lepiarz-Raba I, Gbadamosi I, Florea R, Paolicelli RC, Jawaid A. Metabolic regulation of microglial phagocytosis: Implications for Alzheimer's disease therapeutics. Transl Neurodegener 2023; 12:48. [PMID: 37908010 PMCID: PMC10617244 DOI: 10.1186/s40035-023-00382-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 10/23/2023] [Indexed: 11/02/2023] Open
Abstract
Microglia, the resident immune cells of the brain, are increasingly implicated in the regulation of brain health and disease. Microglia perform multiple functions in the central nervous system, including surveillance, phagocytosis and release of a variety of soluble factors. Importantly, a majority of their functions are closely related to changes in their metabolism. This natural inter-dependency between core microglial properties and metabolism offers a unique opportunity to modulate microglial activities via nutritional or metabolic interventions. In this review, we examine the existing scientific literature to synthesize the hypothesis that microglial phagocytosis of amyloid beta (Aβ) aggregates in Alzheimer's disease (AD) can be selectively enhanced via metabolic interventions. We first review the basics of microglial metabolism and the effects of common metabolites, such as glucose, lipids, ketone bodies, glutamine, pyruvate and lactate, on microglial inflammatory and phagocytic properties. Next, we examine the evidence for dysregulation of microglial metabolism in AD. This is followed by a review of in vivo studies on metabolic manipulation of microglial functions to ascertain their therapeutic potential in AD. Finally, we discuss the effects of metabolic factors on microglial phagocytosis of healthy synapses, a pathological process that also contributes to the progression of AD. We conclude by enlisting the current challenges that need to be addressed before strategies to harness microglial phagocytosis to clear pathological protein deposits in AD and other neurodegenerative disorders can be widely adopted.
Collapse
Affiliation(s)
- Izabela Lepiarz-Raba
- Laboratory for Translational Research in Neuropsychiatric Disorders (TREND), BRAINCITY: Center of Excellence for Neural Plasticity and Brain Disorders, Nencki Institute of Experimental Biology, Warsaw, Poland.
| | - Ismail Gbadamosi
- Laboratory for Translational Research in Neuropsychiatric Disorders (TREND), BRAINCITY: Center of Excellence for Neural Plasticity and Brain Disorders, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Roberta Florea
- Swiss Federal Institute of Technology (ETH), Zurich, Switzerland
| | | | - Ali Jawaid
- Laboratory for Translational Research in Neuropsychiatric Disorders (TREND), BRAINCITY: Center of Excellence for Neural Plasticity and Brain Disorders, Nencki Institute of Experimental Biology, Warsaw, Poland.
| |
Collapse
|
12
|
Mishima M, Takeda S, Nagane M, Suzuki T, Ogata M, Shima A, Aihara N, Kamiie J, Suzuki R, Mizugaki H, Okamatsu-Ogura Y, Satoh T, Yamashita T. Prebiotic effect of poly-D-3-hydroxybutyrate prevents dyslipidemia in obese mice. FASEB J 2023; 37:e23121. [PMID: 37548278 DOI: 10.1096/fj.202301191r] [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: 06/18/2023] [Revised: 07/16/2023] [Accepted: 07/20/2023] [Indexed: 08/08/2023]
Abstract
Obesity is a global health problem caused by genetic, environmental, and psychological factors and is associated with various health disorders. As such, there is a growing focus on the prevention of obesity and related diseases. The gut microbiota plays a crucial role in these diseases and has become a therapeutic target. Prebiotics, such as poly-d-3-hydroxybutyric acid (PHB), have gained attention for their potential to alter the gut microbiota, promote beneficial bacterial growth, and alleviate obesity. In this study, we examined the prebiotic effects of PHB in obese mice. We found that, in C57BL/6N mice, PHB reduced blood lipid levels. Analysis of the intestinal microflora also revealed an increase in short-chain fatty acid-producing bacteria. When PHB was administered to obese mice, subcutaneous fat and dyslipidemia were reduced, and the number of beneficial bacteria in the intestinal microflora increased. Furthermore, fatty degradation and oxidative stress were suppressed in the liver. PHB regulates gut bacterial changes related to obesity and effectively inhibits dyslipidemia, suggesting that it could be a prebiotic agent for curing various obesity-related diseases. In summary, PHB increases the beneficial gut microbiota, leading to an alleviation of obesity-associated dyslipidemia.
Collapse
Affiliation(s)
- Mayuko Mishima
- School of Veterinary Medicine, Azabu University, Sagamihara, Japan
| | - Shiro Takeda
- School of Veterinary Medicine, Azabu University, Sagamihara, Japan
- Center for Human and Animal Symbiosis Science, Azabu University, Sagamihara, Japan
| | - Masaki Nagane
- School of Veterinary Medicine, Azabu University, Sagamihara, Japan
- Center for Human and Animal Symbiosis Science, Azabu University, Sagamihara, Japan
| | - Takehito Suzuki
- School of Veterinary Medicine, Azabu University, Sagamihara, Japan
- Center for Human and Animal Symbiosis Science, Azabu University, Sagamihara, Japan
| | - Masaya Ogata
- School of Veterinary Medicine, Azabu University, Sagamihara, Japan
| | - Ayaka Shima
- Anicom Specialty Medical Institute Inc., Tokyo, Japan
| | - Naoyuki Aihara
- School of Veterinary Medicine, Azabu University, Sagamihara, Japan
| | - Junichi Kamiie
- School of Veterinary Medicine, Azabu University, Sagamihara, Japan
| | - Rimina Suzuki
- School of Veterinary Medicine, Azabu University, Sagamihara, Japan
| | - Hinano Mizugaki
- School of Veterinary Medicine, Azabu University, Sagamihara, Japan
| | | | - Takumi Satoh
- Department of Antiaging Food Research, School of Bioscience and Biotechnology, Tokyo University of Technology, Hachioji, Japan
| | | |
Collapse
|
13
|
Giha HA. Hidden chronic metabolic acidosis of diabetes type 2 (CMAD): Clues, causes and consequences. Rev Endocr Metab Disord 2023; 24:735-750. [PMID: 37380824 DOI: 10.1007/s11154-023-09816-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/09/2023] [Indexed: 06/30/2023]
Abstract
Interpretation of existing data revealed that chronic metabolic acidosis is a pathognomic feature for type 2 diabetes (T2D), which is described here as "chronic metabolic acidosis of T2D (CMAD)" for the first time. The biochemical clues for the CMAD are summarised in the following; low blood bicarbonate (high anionic gap), low pH of interstitial fluid and urine, and response to acid neutralization, while the causes of extra protons are worked out to be; mitochondrial dysfunction, systemic inflammation, gut microbiota (GM), and diabetic lung. Although, the intracellular pH is largely preserved by the buffer system and ion transporters, a persistent systemic mild acidosis leaves molecular signature in cellular metabolism in diabetics. Reciprocally, there are evidences that CMAD contributes to the initiation and progression of T2D by; reducing insulin production, triggering insulin resistance directly or via altered GM, and inclined oxidative stress. The details about the above clues, causes and consequences of CMAD are obtained by searching literature spanning between 1955 and 2022. Finally, the molecular bases of CMAD are discussed in details by interpretation of an up-to-date data and aid of well constructed diagrams, with a conclusion unravelling that CMAD is a major player in T2D pathophysiology. To this end, the CMAD disclosure offers several therapeutic potentials for prevention, delay or attenuation of T2D and its complications.
Collapse
Affiliation(s)
- Hayder A Giha
- Medical Biochemistry and Molecular Biology, Khartoum, Sudan.
| |
Collapse
|
14
|
García-Velázquez L, Massieu L. The proteomic effects of ketone bodies: implications for proteostasis and brain proteinopathies. Front Mol Neurosci 2023; 16:1214092. [PMID: 37575967 PMCID: PMC10413579 DOI: 10.3389/fnmol.2023.1214092] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 07/07/2023] [Indexed: 08/15/2023] Open
Abstract
A growing body of evidence supports the beneficial effects of the ketone bodies (KBs), acetoacetate and β-hydroxybutyrate (BHB), on diverse physiological processes and diseases. Hence, KBs have been suggested as therapeutic tools for neurodegenerative diseases. KBs are an alternative fuel during fasting and starvation as they can be converted to Ac-CoA to produce ATP. A ketogenic diet (KD), enriched in fats and low in carbohydrates, induces KB production in the liver and favors their use in the brain. BHB is the most abundant KB in the circulation; in addition to its role as energy fuel, it exerts many actions that impact the set of proteins in the cell and tissue. BHB can covalently bind to proteins in lysine residues as a new post-translational modification (PTM) named β-hydroxybutyrylation (Kbhb). Kbhb has been identified in many proteins where Kbhb sites can be critical for binding to other proteins or cofactors. Kbhb is mostly found in proteins involved in chromatin structure, DNA repair, regulation of spliceosome, transcription, and oxidative phosphorylation. Histones are the most studied family of proteins with this PTM, and H3K9bhb is the best studied histone mark. Their target genes are mainly related to cell metabolism, chromatin remodeling and the control of circadian rhythms. The role of Kbhb on physiological processes is poorly known, but it might link KB metabolism to cell signaling and genome regulation. BHB also impacts the proteome by influencing proteostasis. This KB can modulate the Unfolded Protein Response (UPR) and autophagy, two processes involved in the maintenance of protein homeostasis through the clearance of accumulated unfolded and damaged proteins. BHB can support proteostasis and regulate the UPR to promote metabolism adaptation in the liver and prevent cell damage in the brain. Also, BHB stimulates autophagy aiding to the degradation of accumulated proteins. Protein aggregation is common to proteinopathies like Alzheimer's (AD) and Parkinson's (PD) diseases, where the KD and BHB treatment have shown favorable effects. In the present review, the current literature supporting the effects of KBs on proteome conformation and proteostasis is discussed, as well as its possible impact on AD and PD.
Collapse
Affiliation(s)
| | - Lourdes Massieu
- Department of Molecular Neuropathology, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México (UNAM), México City, Mexico
| |
Collapse
|
15
|
Phung NV, Rong F, Xia WY, Fan Y, Li XY, Wang SA, Li FL. Nervonic acid and its sphingolipids: Biological functions and potential food applications. Crit Rev Food Sci Nutr 2023; 64:8766-8785. [PMID: 37114919 DOI: 10.1080/10408398.2023.2203753] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Abstract
Nervonic acid, a 24-carbon fatty acid with only one double bond at the 9th carbon (C24:1n-9), is abundant in the human brain, liver, and kidney. It not only functions in free form but also serves as a critical component of sphingolipids which participate in many biological processes such as cell membrane formation, apoptosis, and neurotransmission. Recent studies show that nervonic acid supplementation is not only beneficial to human health but also can improve the many medical conditions such as neurological diseases, cancers, diabetes, obesity, and their complications. Nervonic acid and its sphingomyelins serve as a special material for myelination in infants and remyelination patients with multiple sclerosis. Besides, the administration of nervonic acid is reported to reduce motor disorder in mice with Parkinson's disease and limit weight gain. Perturbations of nervonic acid and its sphingolipids might lead to the pathogenesis of many diseases and understanding these mechanisms is critical for investigating potential therapeutic approaches for such diseases. However, available studies about this aspect are limited. In this review, relevant findings about functional mechanisms of nervonic acid have been comprehensively and systematically described, focusing on four interconnected functions: cellular structure, signaling, anti-inflammation, lipid mobilization, and their related diseases.
Collapse
Affiliation(s)
- Nghi Van Phung
- Shandong Provincial Key Laboratory of Synthetic Biology, Qingdao C1 Refinery Engineering Research Center, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, China
| | - Fei Rong
- Department of Anesthesiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Wan Yue Xia
- Shandong Provincial Key Laboratory of Synthetic Biology, Qingdao C1 Refinery Engineering Research Center, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, China
| | - Yong Fan
- Shandong Provincial Key Laboratory of Synthetic Biology, Qingdao C1 Refinery Engineering Research Center, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, China
- Qingdao New Energy Shandong Laboratory, Qingdao, China
| | - Xian Yu Li
- China Academy of Chinese Medical Sciences, Beijing, China
| | - Shi An Wang
- Shandong Provincial Key Laboratory of Synthetic Biology, Qingdao C1 Refinery Engineering Research Center, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, China
- Shandong Energy Institute, Qingdao, China
| | - Fu Li Li
- Shandong Provincial Key Laboratory of Synthetic Biology, Qingdao C1 Refinery Engineering Research Center, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, China
- Qingdao New Energy Shandong Laboratory, Qingdao, China
- Shandong Energy Institute, Qingdao, China
| |
Collapse
|
16
|
Gross EC, Putananickal N, Orsini AL, Schoenen J, Fischer D, Soto-Mota A. Defining metabolic migraine with a distinct subgroup of patients with suboptimal inflammatory and metabolic markers. Sci Rep 2023; 13:3787. [PMID: 36882474 PMCID: PMC9992685 DOI: 10.1038/s41598-023-28499-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 01/19/2023] [Indexed: 03/09/2023] Open
Abstract
Emerging evidence suggest migraine is a response to cerebral energy deficiency or oxidative stress in the brain. Beta-hydroxybutyrate (BHB) is likely able to circumvent some of the meta-bolic abnormalities reported in migraine. Exogenous BHB was given to test this assumption and, in this post-hoc analysis, multiple metabolic biomarkers were identified to predict clinical improvements. A randomized clinical trial, involving 41 patients with episodic migraine. Each treatment period was 12 weeks long, followed by eight weeks of washout phase / second run-in phase before entering the corresponding second treatment period. The primary endpoint was the number of migraine days in the last 4 weeks of treatment adjusted for baseline. BHB re-sponders were identified (those with at least a 3-day reduction in migraine days over placebo) and its predictors were evaluated using Akaike's Information Criterion (AIC) stepwise boot-strapped analysis and logistic regression. Responder analysis showed that metabolic markers could identify a "metabolic migraine" subgroup, which responded to BHB with a 5.7 migraine days reduction compared to the placebo. This analysis provides further support for a "metabolic migraine" subtype. Additionally, these analyses identified low-cost and easily accessible biomarkers that could guide recruitment in future research on this subgroup of patients.This study is part of the trial registration: ClinicalTrials.gov: NCT03132233, registered on 27.04.2017, https://clinicaltrials.gov/ct2/show/NCT03132233.
Collapse
Affiliation(s)
- Elena C Gross
- Division of Pediatric Neurology, University Children's Hospital Basel (UKBB), University of Basel, Basel, Switzerland.
| | - Niveditha Putananickal
- Division of Pediatric Neurology, University Children's Hospital Basel (UKBB), University of Basel, Basel, Switzerland
| | - Anna-Lena Orsini
- Division of Pediatric Neurology, University Children's Hospital Basel (UKBB) & Neurology Department, University Hospital Basel (USB), University of Basel, Basel, Switzerland
| | - Jean Schoenen
- Headache Research Unit, Department of Neurology-Citadelle Hospital, University of Liège, Liège, Belgium
| | - Dirk Fischer
- Division of Pediatric Neurology, University Children's Hospital Basel (UKBB), University of Basel, Basel, Switzerland
| | - Adrian Soto-Mota
- Metabolic Diseases Research Unit, National Institute of Medical Sciences and Nutrition Salvador Zubirán (INCMNSZ), Tlalpan, Mexico.,School of Medicine, Tecnologico de Monterrey, Mexico City, Mexico
| |
Collapse
|
17
|
Qiu Y, Hu X, Xu C, Lu C, Cao R, Xie Y, Yang J. Ketogenic diet alleviates renal fibrosis in mice by enhancing fatty acid oxidation through the free fatty acid receptor 3 pathway. Front Nutr 2023; 10:1127845. [PMID: 37032786 PMCID: PMC10081144 DOI: 10.3389/fnut.2023.1127845] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 02/27/2023] [Indexed: 04/11/2023] Open
Abstract
Introduction The ketogenic diet (KD), as a dietary intervention, has gained importance in the treatment of solid organ structural remodeling, but its role in renal fibrosis has not been explored. Methods Male C57BL/6 mice were fed a normal diet or a KD for 6 weeks prior to unilateral ureteral obstruction (UUO), a well-established in vivo model of renal fibrosis in rodents. Seven days after UUO, serum and kidney samples were collected. Serum β-hydroxybutyrate (β-OHB) concentrations and renal fibrosis were assessed. NRK52E cells were treated with TGFβ1, a fibrosis-inducing cytokine, and with or without β-OHB, a ketone body metabolized by KD, to investigate the mechanism underlying renal fibrosis. Results KD significantly enhanced serum β-OHB levels in mice. Histological analysis revealed that KD alleviated structural destruction and fibrosis in obstructed kidneys and reduced the expression of the fibrosis protein markers α-SMA, Col1a1, and Col3a1. Expression of the rate-limiting enzymes involved in fatty acid oxidation (FAO), Cpt1a and Acox1, significantly decreased after UUO and were upregulated by KD. However, the protective effect of KD was abolished by etomoxir (a Cpt1a inhibitor). Besides, our study observed that KD significantly suppressed UUO-induced macrophage infiltration and the expression of IL-6 in the obstructive kidneys. In NRK52E cells, fibrosis-related signaling was increased by TGFβ1 and reduced by β-OHB. β-OHB treatment restored the impaired expression of Cpt1a. The effect of β-OHB was blocked by siRNA targeting free fatty acid receptor 3 (FFAR3), suggesting that β-OHB might function through the FFAR3-dependent pathway. Discussion Our results highlight that KD attenuates UUO-induced renal fibrosis by enhancing FAO via the FFAR3-dependent pathway, which provides a promising dietary therapy for renal fibrosis.
Collapse
|
18
|
Ma N, Guo P, Chen J, Qi Z, Liu C, Shen J, Sun Y, Chen X, Chen GQ, Ma X. Poly-β-hydroxybutyrate alleviated diarrhea and colitis via Lactobacillus johnsonii biofilm-mediated maturation of sulfomucin. SCIENCE CHINA. LIFE SCIENCES 2022:10.1007/s11427-022-2213-6. [PMID: 36580163 DOI: 10.1007/s11427-022-2213-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 10/04/2022] [Indexed: 12/30/2022]
Abstract
Maintainance of sulfomucin is a key end point in the treatment of diarrhea and inflammatory bowel disease (IBD). However, the mechanisms underlying the microbial sense to sulfomucin are poorly understood, and to date, there are no therapies targeting the secretion and maturation of sulfomucin in IBD. Herein, we biosynthesized poly-β-hydroxybutyrate (PHB) and found that PHB could alleviate inflammation caused by diarrhea and colitis by enhancing the differentiation of sulfomucin. Microbiota transplantation and clearance together demonstrate that PHB promoting sulfomucin is mediated by Lactobacillus johnsonii (L. johnsonii). Further studies revealed that PHB provides a favorable niche for L. johnsonii biofilm formation to resist disturbance and support its growth. L. johnsonii-biofilm alleviates colitis by regulating fucose residues to promote goblet cell differentiation and subsequent sulfomucin maturation. Importantly, PHB alleviates colitis by enhancing sulfomucin secretion and maturation in a L. johnsonii-dependent manner. PHB represents a class of guardians, acting as a safe probiotic-biofilm delivery system that significantly promotes probiotic proliferation. Altogether, this study adds weight to the possible role of probiotics and functional materials in the treatment of intestinal inflammation. The application of PHB and biofilm self-coating L. johnsonii carries high translational potential and may be of clinical relevance.
Collapse
Affiliation(s)
- Ning Ma
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Pingting Guo
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Jingyu Chen
- Beijing Laboratory for Food Quality and Safety, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Zengkai Qi
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Chunchen Liu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Jiakun Shen
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Yiwei Sun
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Xiyue Chen
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Guo-Qiang Chen
- Center for Synthetic and Systems Biology, MOE Key Lab for Industrial Biocatalysis, School of Life Sciences and Dept Chemical Engineering, Tsinghua University, Beijing, 100084, China.
| | - Xi Ma
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
19
|
Ley-Ngardigal S, Bertolin G. Approaches to monitor ATP levels in living cells: where do we stand? FEBS J 2022; 289:7940-7969. [PMID: 34437768 DOI: 10.1111/febs.16169] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/30/2021] [Accepted: 08/25/2021] [Indexed: 01/14/2023]
Abstract
ATP is the most universal and essential energy molecule in cells. This is due to its ability to store cellular energy in form of high-energy phosphate bonds, which are extremely stable and readily usable by the cell. This energy is key for a variety of biological functions such as cell growth and division, metabolism, and signaling, and for the turnover of biomolecules. Understanding how ATP is produced and hydrolyzed with a spatiotemporal resolution is necessary to understand its functions both in physiological and in pathological contexts. In this review, first we will describe the organization of the electron transport chain and ATP synthase, the main molecular motor for ATP production in mitochondria. Second, we will review the biochemical assays currently available to estimate ATP quantities in cells, and we will compare their readouts, strengths, and weaknesses. Finally, we will explore the palette of genetically encoded biosensors designed for microscopy-based approaches, and show how their spatiotemporal resolution opened up the possibility to follow ATP levels in living cells.
Collapse
Affiliation(s)
- Seyta Ley-Ngardigal
- CNRS, Univ Rennes, IGDR (Genetics and Development Institute of Rennes), Rennes, France.,LVMH Research Perfumes and Cosmetics, Saint-Jean-de-Braye, France
| | - Giulia Bertolin
- CNRS, Univ Rennes, IGDR (Genetics and Development Institute of Rennes), Rennes, France
| |
Collapse
|
20
|
Dolan LC, Karikachery AR, Thipe VC, Arceneaux BG, Katti KK, Katti KV, Chesne AM. Toxicity Investigations of (R)-3-Hydroxybutyrate Glycerides In Vitro and in Male and Female Rats. Nutrients 2022; 14:nu14204426. [PMID: 36297110 PMCID: PMC9610502 DOI: 10.3390/nu14204426] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/19/2022] [Accepted: 10/19/2022] [Indexed: 11/05/2022] Open
Abstract
TCN006, a formulation of (R)-3-Hydroxybutyrate glycerides, is a promising ingredient for enhancing ketone intake of humans. Ketones have been shown to have beneficial effects on human health. To be used by humans, TCN006 must be determined safe in appropriately designed safety studies. The results of a bacterial reverse mutation assay, an in vitro mammalian micronucleus study, and 14-and 90-day repeat dose toxicity studies in rats are reported herein. In the 14- and 90-day studies, male and female Wistar rats had free access to drinking water containing 0, 75,000, 125,000 or 200,000 ppm TCN006 for 92 and 93 days, respectively. TCN006 tested negative for genotoxicity and the no observed adverse effect level (NOAEL) for toxicity in the 14- and 90-day studies was 200,000 ppm, the highest dose administered. In the longer term study, the mean overall daily intake of TCN006 in the 200,000 ppm groups was 14,027.9 mg/kg bw/day for males and 20,507.0 mg/kg bw/day for females. At this concentration, palatability of water was likely affected, which led to a decrease in water consumption in both males and females compared to respective controls. This had no effect on the health of the animals. Although the rats were administered very high levels of (R)-3-Hydroxybutyrate glycerides, there were no signs of ketoacidosis.
Collapse
Affiliation(s)
- Laurie C. Dolan
- GRAS Associates, LLC., 11810 Grand Park Avenue, Suite 500, North Bethesda, MD 20852, USA
| | - Alice Raphael Karikachery
- Department of Radiology, Institute of Green Nanotechnology, University of Missouri, Columbia, MO 65212, USA
| | - Velaphi C. Thipe
- Department of Radiology, Institute of Green Nanotechnology, University of Missouri, Columbia, MO 65212, USA
| | - Benjamin G. Arceneaux
- GRAS Associates, LLC., 11810 Grand Park Avenue, Suite 500, North Bethesda, MD 20852, USA
| | - Kavita K. Katti
- Department of Radiology, Institute of Green Nanotechnology, University of Missouri, Columbia, MO 65212, USA
| | - Kattesh V. Katti
- Department of Radiology, Institute of Green Nanotechnology, University of Missouri, Columbia, MO 65212, USA
| | - Alton M. Chesne
- Tecton Group, LLC., 370 River Rd., Alexandria, LA 71302, USA
- Correspondence:
| |
Collapse
|
21
|
NADPH and Mitochondrial Quality Control as Targets for a Circadian-Based Fasting and Exercise Therapy for the Treatment of Parkinson's Disease. Cells 2022; 11:cells11152416. [PMID: 35954260 PMCID: PMC9367803 DOI: 10.3390/cells11152416] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/01/2022] [Accepted: 08/01/2022] [Indexed: 02/01/2023] Open
Abstract
Dysfunctional mitochondrial quality control (MQC) is implicated in the pathogenesis of Parkinson's disease (PD). The improper selection of mitochondria for mitophagy increases reactive oxygen species (ROS) levels and lowers ATP levels. The downstream effects include oxidative damage, failure to maintain proteostasis and ion gradients, and decreased NAD+ and NADPH levels, resulting in insufficient energy metabolism and neurotransmitter synthesis. A ketosis-based metabolic therapy that increases the levels of (R)-3-hydroxybutyrate (BHB) may reverse the dysfunctional MQC by partially replacing glucose as an energy source, by stimulating mitophagy, and by decreasing inflammation. Fasting can potentially raise cytoplasmic NADPH levels by increasing the mitochondrial export and cytoplasmic metabolism of ketone body-derived citrate that increases flux through isocitrate dehydrogenase 1 (IDH1). NADPH is an essential cofactor for nitric oxide synthase, and the nitric oxide synthesized can diffuse into the mitochondrial matrix and react with electron transport chain-synthesized superoxide to form peroxynitrite. Excessive superoxide and peroxynitrite production can cause the opening of the mitochondrial permeability transition pore (mPTP) to depolarize the mitochondria and activate PINK1-dependent mitophagy. Both fasting and exercise increase ketogenesis and increase the cellular NAD+/NADH ratio, both of which are beneficial for neuronal metabolism. In addition, both fasting and exercise engage the adaptive cellular stress response signaling pathways that protect neurons against the oxidative and proteotoxic stress implicated in PD. Here, we discuss how intermittent fasting from the evening meal through to the next-day lunch together with morning exercise, when circadian NAD+/NADH is most oxidized, circadian NADP+/NADPH is most reduced, and circadian mitophagy gene expression is high, may slow the progression of PD.
Collapse
|
22
|
Ródenas-González F, Blanco-Gandía MC, Miñarro J, Rodríguez-Arias M. Cognitive profile of male mice exposed to a Ketogenic Diet. Physiol Behav 2022; 254:113883. [PMID: 35716801 DOI: 10.1016/j.physbeh.2022.113883] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 06/09/2022] [Accepted: 06/13/2022] [Indexed: 10/18/2022]
Abstract
In recent years, nutritional interventions for different psychiatric diseases have gained increasing attention, such as the ketogenic diet (KD). This has led to positive effects in neurological disorders such as Parkinson's disease, addiction, autism or epilepsy. The neurobiological mechanisms through which these effects are induced and the effects in cognition still warrant investigation, and considering that other high-fat diets (HFD) can lead to cognitive disturbances that may affect the results achieved, the main aim of the present work was to evaluate the effects of a KD to determine whether it can induce such cognitive effects. A total of 30 OF1 male mice were employed to establish the behavioral profile of mice fed a KD by testing anxiety behavior (Elevated Plus Maze), locomotor activity (Open Field), learning (Hebb Williams Maze), and memory (Passive Avoidance Test). The results revealed that the KD did not affect locomotor activity, memory or hippocampal-dependent learning, as similar results were obtained with mice on a standard diet, albeit with increased anxiety behavior. We conclude that a KD is a promising nutritional approach to apply in research studies, given that it does not cause cognitive alterations.
Collapse
Affiliation(s)
- Francisco Ródenas-González
- Unidad de Investigación Psicobiología de las Drogodependencias, Departamento de Psicobiología, Facultad de Psicología, Universitat de Valencia, Avda. Blasco Ibáñez, 21, Valencia, 46010 Spain
| | - M Carmen Blanco-Gandía
- Departamento de Psicología y Sociología, Facultad de Ciencias Sociales y Humanas, Universidad de Zaragoza, Teruel, Spain
| | - José Miñarro
- Unidad de Investigación Psicobiología de las Drogodependencias, Departamento de Psicobiología, Facultad de Psicología, Universitat de Valencia, Avda. Blasco Ibáñez, 21, Valencia, 46010 Spain
| | - Marta Rodríguez-Arias
- Unidad de Investigación Psicobiología de las Drogodependencias, Departamento de Psicobiología, Facultad de Psicología, Universitat de Valencia, Avda. Blasco Ibáñez, 21, Valencia, 46010 Spain.
| |
Collapse
|
23
|
Abdelhafiz Y, Fernandes JMO, Donati C, Pindo M, Kiron V. Intergenerational Transfer of Persistent Bacterial Communities in Female Nile Tilapia. Front Microbiol 2022; 13:879990. [PMID: 35655994 PMCID: PMC9152445 DOI: 10.3389/fmicb.2022.879990] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 04/11/2022] [Indexed: 11/30/2022] Open
Abstract
Resident microbial communities that can support various host functions play a key role in their development and health. In fishes, microbial symbionts are vertically transferred from the parents to their progeny. Such transfer of microbes in mouthbrooder fish species has not been reported yet. Here, we employed Nile tilapia (Oreochromis niloticus) to investigate the vertical transmission of microbes across generations using a 16S rRNA amplicon sequencing approach, based on the presence of bacteria in different generations. Our analysis revealed that the core microbiome in the buccal cavity and posterior intestine of parents shapes the gut microbiome of the progeny across generations. We speculate that the route of this transmission is via the buccal cavity. The identified core microbiome bacteria, namely Nocardioides, Propionibacterium, and Sphingomonas have been reported to play an essential role in the health and development of offspring. These core microbiome members could have specific functions in fish, similar to mammals.
Collapse
Affiliation(s)
- Yousri Abdelhafiz
- Faculty of Biosciences and Aquaculture, Nord University, Bodø, Norway
| | | | - Claudio Donati
- Unit of Computational Biology, Research and Innovation Centre, Fondazione Edmund Mach, San Michele all'Adige, Italy
| | - Massimo Pindo
- Unit of Computational Biology, Research and Innovation Centre, Fondazione Edmund Mach, San Michele all'Adige, Italy
| | - Viswanath Kiron
- Faculty of Biosciences and Aquaculture, Nord University, Bodø, Norway
| |
Collapse
|
24
|
Amara AAAF. Natural Polymer Types and Applications. BIOMOLECULES FROM NATURAL SOURCES 2022:31-81. [DOI: 10.1002/9781119769620.ch2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
|
25
|
Damasco JA, Huang SY, Perez JVD, Manongdo JAT, Dixon KA, Williams ML, Jacobsen MC, Barbosa R, Canlas GM, Chintalapani G, Melancon AD, Layman RR, Fowlkes NW, Whitley EM, Melancon MP. Bismuth Nanoparticle and Polyhydroxybutyrate Coatings Enhance the Radiopacity of Absorbable Inferior Vena Cava Filters for Fluoroscopy-Guided Placement and Longitudinal Computed Tomography Monitoring in Pigs. ACS Biomater Sci Eng 2022; 8:1676-1685. [PMID: 35343679 PMCID: PMC9045416 DOI: 10.1021/acsbiomaterials.1c01449] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Inferior vena cava filters (IVCFs) constructed with poly-p-dioxanone (PPDO) are promising alternatives to metallic filters and their associated risks and complications. Incorporating high-Z nanoparticles (NPs) improves PPDO IVCFs' radiopacity without adversely affecting their safety or performance. However, increased radiopacity from these studies are insufficient for filter visualization during fluoroscopy-guided PPDO IVCF deployment. This study focuses on the use of bismuth nanoparticles (BiNPs) as radiopacifiers to render sufficient signal intensity for the fluoroscopy-guided deployment and long-term CT monitoring of PPDO IVCFs. The use of polyhydroxybutyate (PHB) as an additional layer to increase the surface adsorption of NPs resulted in a 2-fold increase in BiNP coating (BiNP-PPDO IVCFs, 3.8%; BiNP-PPDO + PHB IVCFs, 6.2%), enabling complete filter visualization during fluoroscopy-guided IVCF deployment and, 1 week later, clot deployment. The biocompatibility, clot-trapping efficacy, and mechanical strength of the control PPDO (load-at-break, 6.23 ± 0.13 kg), BiNP-PPDO (6.10 ± 0.09 kg), and BiNP-PPDO + PHB (6.15 ± 0.13 kg) IVCFs did not differ significantly over a 12-week monitoring period in pigs. These results indicate that BiNP-PPDO + PHB can increase the radiodensity of a novel absorbable IVCF without compromising device strength. Visualizing the device under conventional radiographic imaging is key to allow safe and effective clinical translation of the device.
Collapse
Affiliation(s)
- Jossana A Damasco
- Departments of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - Steven Y Huang
- Departments of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - Joy Vanessa D Perez
- Departments of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| | | | - Katherine A Dixon
- Departments of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - Malea L Williams
- Departments of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - Megan C Jacobsen
- Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - Roland Barbosa
- Covalent Metrology Sunnyvale, Sunnyvale, California 94085, United States
| | - Gino Martin Canlas
- Department of Chemistry, Lamar University, Beaumont, Texas 77710, United States
| | | | - Adam D Melancon
- Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - Rick R Layman
- Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - Natalie W Fowlkes
- Veterinary Medicine and Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - Elizabeth M Whitley
- Veterinary Medicine and Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - Marites P Melancon
- Departments of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
- UT Health Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| |
Collapse
|
26
|
Suchkov S, Seifi Salmi T, Bai CH, Alizargar J, Wu JP. Ketogenic Diet Is Good for Aging-Related Sarcopenic Obesity. ROLE OF OBESITY IN HUMAN HEALTH AND DISEASE 2021. [DOI: 10.5772/intechopen.96028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2025]
Abstract
Sarcopenic obesity is a skeletal muscle weight loss disease. It has happened at an elderly age. A ketogenic diet is a low-carbohydrate (5%), moderate protein (15%), and a higher-fat diet (80%) can help sarcopenic obese patients burn their fat more effectively. It has many benefits for muscle and fat weight loss. A ketogenic diet can be especially useful for losing excess body fat without hunger and for improving type 2 diabetes. That is because of only a few carbohydrates in the diet, the liver converts fat into fatty acids and ketones. Ketone bodies can replace higher ATP energy. This diet forces the human body to burn fat. This is a good way to lose fat weight without restriction.
Collapse
|
27
|
Nanjappa D, Liang Y, Bretherton L, Brown C, Quigg A, Irwin AJ, Finkel ZV. Contrasting transcriptomic responses of a microbial eukaryotic community to oil and dispersant. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 288:117774. [PMID: 34274645 DOI: 10.1016/j.envpol.2021.117774] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 07/05/2021] [Accepted: 07/08/2021] [Indexed: 06/13/2023]
Abstract
Dispersants can aid dispersion and biodegradation of oil in seawater, but the wider ecotoxicological effects of oil and dispersant to the base of marine food webs is unclear. Here we apply a metatranscriptomic approach to identify molecular responses of a natural marine microbial eukaryotic community to oil and chemically dispersed oil. Oil exposure stimulated the upregulation of ketogenesis in the eukaryotic community, which may alleviate carbon- and energy-limitation and reduce oxidative stress. In contrast, a chemically dispersed oil treatment stimulated eukaryotic genes and pathways consistent with nitrogen and oxygen depletion. These results suggest that the addition of dispersant may elevate bacterial biodegradation of crude oil, indirectly increasing competition for nitrogen between prokaryotic and eukaryotic communities as oxygen consumption induces bacterial anaerobic respiration and denitrification. Eukaryotic microbial communities may mitigate some of the negative effects of oil exposure such as reduced photosynthesis and elevated oxidative stress, through ketosis, but the addition of dispersant to the oil fundamentally alters the environmental and ecological conditions and therefore the biochemical response of the eukaryotic community.
Collapse
Affiliation(s)
- Deepak Nanjappa
- Department of Oceanography, Dalhousie University, Halifax, NS, Canada.
| | - Yue Liang
- Department of Oceanography, Dalhousie University, Halifax, NS, Canada
| | - Laura Bretherton
- Department of Oceanography, Dalhousie University, Halifax, NS, Canada
| | - Chris Brown
- Environmental Science Program, Mount Allison University, Sackville, NB, Canada
| | - Antonietta Quigg
- Department of Marine Biology, Texas A&M University at Galveston, Galveston, TX, USA
| | - Andrew J Irwin
- Department of Oceanography, Dalhousie University, Halifax, NS, Canada; Department of Mathematics & Statistics, Dalhousie University, Halifax, NS, Canada
| | - Zoe V Finkel
- Department of Oceanography, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
28
|
Li Z, Zhang S, Zhang Y, Chen J, Wu F, Liu G, Chen GQ. Applications and Mechanism of 3-Hydroxybutyrate (3HB) for Prevention of Colonic Inflammation and Carcinogenesis as a Food Supplement. Mol Nutr Food Res 2021; 65:e2100533. [PMID: 34704372 DOI: 10.1002/mnfr.202100533] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 10/07/2021] [Indexed: 01/19/2023]
Abstract
SCOPE Inflammatory bowel disease and colorectal carcinogenesis (CRC) are common diseases without effective prevention approach. 3-Hydroxybutyrate (3HB) reported to have multiple functions as an oral food supplement. This study observes that 3HB prevents mouse colitis and CRC. METHODS AND RESULTS The sensitivity of wild type (WT) and GPR109a-/- mice to colitis is compared using dextran sulfate sodium salt (DSS)-induced colitis model. Flow cytometry showed that 3HB cellular surface receptor GPR109a that can decrease the percentage of M1 macrophages from 50% of the DSS-induced acute colitis mouse group to 42% DSS+3HB group mediating the inhibitory effect on inflammation. Bone marrow transplantation experiments further demonstrated that the function of 3HB depended on bone marrow cells. Subsequently, the sensitivity of WT and GPR109a-/- mice to CRC is compared using an azoxymethane-DSS-induced CRC mouse model. It is found that the activation of GPR109a inhibited CRC, depended on reduced myeloid-derived suppressor cells accumulation from 27% of the DSS group to 19% of the DSS+3HB group studied using flow cytometry. CONCLUSION It is concluded that 3HB significantly suppresses colonic inflammation and carcinogenesis, promising to benefit colon disease prevention in form of a food supplement.
Collapse
Affiliation(s)
- Zihua Li
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
| | - Shujie Zhang
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Yudian Zhang
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Jin Chen
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Fuqing Wu
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Gang Liu
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
| | - Guo-Qiang Chen
- School of Life Sciences, Tsinghua University, Beijing, 100084, China.,Center for Synthetic and Systems Biology, Tsinghua University, Beijing, 100084, China.,MOE Key Lab of Industrial Biocatalysis, Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
29
|
Thai PN, Miller CV, King MT, Schaefer S, Veech RL, Chiamvimonvat N, Bers DM, Dedkova EN. Ketone Ester D-β-Hydroxybutyrate-(R)-1,3 Butanediol Prevents Decline in Cardiac Function in Type 2 Diabetic Mice. J Am Heart Assoc 2021; 10:e020729. [PMID: 34583524 PMCID: PMC8649133 DOI: 10.1161/jaha.120.020729] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Background Heart failure is responsible for approximately 65% of deaths in patients with type 2 diabetes mellitus. However, existing therapeutics for type 2 diabetes mellitus have limited success on the prevention of diabetic cardiomyopathy. The aim of this study was to determine whether moderate elevation in D‐β‐hydroxybutyrate improves cardiac function in animals with type 2 diabetes mellitus. Methods and Results Type 2 diabetic (db/db) and their corresponding wild‐type mice were fed a control diet or a diet where carbohydrates were equicalorically replaced by D‐β‐hydroxybutyrate‐(R)‐1,3 butanediol monoester (ketone ester [KE]). After 4 weeks, echocardiography demonstrated that a KE diet improved systolic and diastolic function in db/db mice. A KE diet increased expression of mitochondrial succinyl‐CoA:3‐oxoacid‐CoA transferase and restored decreased expression of mitochondrial β‐hydroxybutyrate dehydrogenase, key enzymes in cardiac ketone metabolism. A KE diet significantly enhanced both basal and ADP‐mediated oxygen consumption in cardiac mitochondria from both wild‐type and db/db animals; however, it did not result in the increased mitochondrial respiratory control ratio. Additionally, db/db mice on a KE diet had increased resistance to oxidative and redox stress, with evidence of restoration of decreased expression of thioredoxin and glutathione peroxidase 4 and less permeability transition pore activity in mitochondria. Mitochondrial biogenesis, quality control, and elimination of dysfunctional mitochondria via mitophagy were significantly increased in cardiomyocytes from db/db mice on a KE diet. The increase in mitophagy was correlated with restoration of mitofusin 2 expression, which contributed to improved coupling between cytosolic E3 ubiquitin ligase translocation into mitochondria and microtubule‐associated protein 1 light chain 3–mediated autophagosome formation. Conclusions Moderate elevation in circulating D‐β‐hydroxybutyrate levels via KE supplementation enhances mitochondrial biogenesis, quality control, and oxygen consumption and increases resistance to oxidative/redox stress and mPTP opening, thus resulting in improvement of cardiac function in animals with type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Phung N Thai
- Department of Internal Medicine Cardiovascular Medicine University of California Davis CA
| | | | - M Todd King
- Laboratory of Metabolic Control National Institute on Alcohol Abuse and AlcoholismNational Institutes of Health Rockville MD
| | - Saul Schaefer
- Department of Internal Medicine Cardiovascular Medicine University of California Davis CA.,Department of Veterans Affairs Northern California Health Care System Mather CA
| | - Richard L Veech
- Laboratory of Metabolic Control National Institute on Alcohol Abuse and AlcoholismNational Institutes of Health Rockville MD
| | - Nipavan Chiamvimonvat
- Department of Internal Medicine Cardiovascular Medicine University of California Davis CA.,Department of Veterans Affairs Northern California Health Care System Mather CA
| | - Donald M Bers
- Department of Pharmacology University of California Davis CA
| | - Elena N Dedkova
- Department of Pharmacology University of California Davis CA.,Department of Molecular Biosciences University of California Davis CA
| |
Collapse
|
30
|
Wahba NS, Abdel-Ghany RH, Ghareib SA, Abdel-Aal M, Alsemeh AE, Sabry D. Vitamin D3 potentiates the nephroprotective effects of vildagliptin-metformin combination in a rat model of metabolic syndrome. Fundam Clin Pharmacol 2021; 36:306-323. [PMID: 34453360 DOI: 10.1111/fcp.12721] [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: 05/10/2021] [Accepted: 08/25/2021] [Indexed: 11/30/2022]
Abstract
The current study was conducted to investigate the nephroprotective effects of vildagliptin-metformin combination in an experimental model of fructose/salt-induced metabolic syndrome (MetS). A major aim was to evaluate the potential capacity of vitamin D3 to potentiate the pleiotropic nephroprotective effects of vildagliptin-metformin combination. MetS was induced in adult male Wistar rats by adding fructose (10%) to everyday drinking water and salt (3%) to the diet for 6 weeks. Along with the same concentrations of fructose/salt feeding, MetS rats were then treated orally with either vildagliptin (10 mg/kg/day)-metformin (200 mg/kg/day) combination, vitamin D3 (10 μg/kg/day), or the triple therapy for a further 6 weeks. The incidence of MetS was confirmed 6 weeks after fructose/salt consumption, when the rats exhibited significant weight gain, dyslipidemia, hyperuricemia, insulin resistance, hyperinsulinemia, and impaired glucose tolerance. At the end of the 12-week experimental period, MetS rats displayed significantly deteriorated renal function, enhanced intrarenal oxidative stress and inflammation together with exaggerated renal histopathological damages and interstitial fibrosis. The study has corroborated antioxidant, anti-inflammatory, and antifibrotic effects of vildagliptin-metformin combination, vitamin D3, and the triple collaborative therapy, conferring renoprotection in the setting of MetS. Due attention has been paid to the crucial role of dipeptidyl peptidase-4 inhibition and sirtuin-1/5' adenosine monophosphate-activated protein kinase activation as novel therapeutic targets to optimize renoprotection. The apparent potentiating effect, evoked upon coadministration of vitamin D3 with vildagliptin-metformin combination, may provide a cornerstone for further clinical investigations.
Collapse
Affiliation(s)
- Nehal S Wahba
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Rasha H Abdel-Ghany
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Salah A Ghareib
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Mohamed Abdel-Aal
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Amira E Alsemeh
- Department of Anatomy and Embryology, Faculty of Human Medicine, Zagazig University, Zagazig, Egypt
| | - Dina Sabry
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Cairo University, Cairo, Egypt.,Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Badr University in Cairo, Badr City, Egypt
| |
Collapse
|
31
|
NEFA, BHBa, UREA and Liver Enzyme Variation in the Bloodstream of Weaned Foals up to 18 Months of Age. Animals (Basel) 2021; 11:ani11061746. [PMID: 34208044 PMCID: PMC8230655 DOI: 10.3390/ani11061746] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 06/07/2021] [Accepted: 06/09/2021] [Indexed: 12/01/2022] Open
Abstract
Simple Summary Energy balance assessment of the growing horse requires a complex nutritional evaluation. Some biochemical parameters can be of clinical importance to prevent the onset of metabolic disorders. The adequate nourishment of body tissues in the growing foal may represent an issue in the practice, given the dynamic change of body composition, in view of the potential mobilization of fat from tissue depots and increasing lean mass. As such, the maintenance of adequate body weight and body measures over time (optimal growth curve accomplishment) and the fulfillment of nutrient requirements are cornerstones of individual feeding plans for the expression of the athletic potential of the future sport horse. In this scenario, the metabolic evaluation of growing foals turned out to be a valuable tool to consider the energy distribution within the animal body. In view of those particular conditions, non-esterified fatty acids (NEFA), beta-hydroxybutyric acid (BHBa), UREA and liver enzymes showed to serve as indicators to monitor energy balance and health in growing foals from weaning to 18 months of age. Abstract The pattern of selected metabolites for interpreting homeostasis during the growth of foals can be used as an indicator of energy balance state and liver health. Against this background, the literature on circulating parameters of foals across growth stages is scanty. We hypothesized that circulating metabolites indicating energy distribution such as non-esterified fatty acids (NEFA), β-hydroxy-butyric acid (BHBa), UREA and liver enzyme-like γ-glutamyl-transferase (γ-GT) [interpreted in the light of circulating total bilirubin (TBIL), alanine aminotransferase (ALT) and aspartate aminotransferase (AST)] may be used to monitor the energy balance of growing foals. A total of 12 Anglo-Arab (AA) foals from the same stable were enrolled in this trial. All foals were serially weighed on a digital scale and sampled for total blood at weaning, at 12 and 18 months of age. Feeding and keeping conditions were similar for all the foals involved. Animals appeared healthy and no signs of poor growth performance were pointed out. The peak of circulating NEFA mobilized from body depots was reached at one year of age but markedly dropped at 18 months, when BHBa increased (p < 0.001) alongside with liver enzyme. BHBa and γ-GT levels turned out to positively correlate (p = 0.051). However, at 6, 12 and 18 months, γ-GT dropped in the physiological reference range for the horse, thus showing no prognostic value. ALT and UREA significantly increased (p = 0.008 and p = 0.006, respectively) when NEFA also increased (p = 0.001). Liver enzyme increase could be associated with fat mobilization and ketone bodies production meanwhile amino acid transamination for energy purposes led to the increase of UREA in the bloodstream. However, no prognostic value to liver enzyme could be attributed in this trial.
Collapse
|
32
|
Bird RP, Eskin NAM. The emerging role of phosphorus in human health. ADVANCES IN FOOD AND NUTRITION RESEARCH 2021; 96:27-88. [PMID: 34112356 DOI: 10.1016/bs.afnr.2021.02.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Phosphorus, an essential nutrient, performs vital functions in skeletal and non-skeletal tissues and is pivotal for energy production. The last two decades of research on the physiological importance of phosphorus have provided several novel insights about its dynamic nature as a nutrient performing functions as a phosphate ion. Phosphorous also acts as a signaling molecule and induces complex physiological responses. It is recognized that phosphorus homeostasis is critical for health. The intake of phosphorus by the general population world-wide is almost double the amount required to maintain health. This increase is attributed to the incorporation of phosphate containing food additives in processed foods purchased by consumers. Research findings assessed the impact of excessive phosphorus intake on cells' and organs' responses, and highlighted the potential pathogenic consequences. Research also identified a new class of bioactive phosphates composed of polymers of phosphate molecules varying in chain length. These polymers are involved in metabolic responses including hemostasis, brain and bone health, via complex mechanism(s) with positive or negative health effects, depending on their chain length. It is amazing, that phosphorus, a simple element, is capable of exerting multiple and powerful effects. The role of phosphorus and its polymers in the renal and cardiovascular system as well as on brain health appear to be important and promising future research directions.
Collapse
Affiliation(s)
- Ranjana P Bird
- School of Health Sciences, University of Northern British Columbia, Prince George, BC, Canada.
| | - N A Michael Eskin
- Department of Food and Human Nutritional Sciences, Faculty of Agricultural and Food Sciences, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
33
|
Lv S, Li X, Wang H. The Role of the Effects of Endoplasmic Reticulum Stress on NLRP3 Inflammasome in Diabetes. Front Cell Dev Biol 2021; 9:663528. [PMID: 33937267 PMCID: PMC8079978 DOI: 10.3389/fcell.2021.663528] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 03/15/2021] [Indexed: 12/12/2022] Open
Abstract
Endoplasmic reticulum (ER) is an important organelle for the protein synthesis, modification, folding, assembly, and the transport of new peptide chains. When the folding ability of ER proteins is impaired, the accumulation of unfolded or misfolded proteins in ER leads to endoplasmic reticulum stress (ERS). The nucleotide-binding oligomerization domain-like receptor family, pyrin domain-containing 3 (NLRP3) inflammasome, can induce the maturation and secretion of interleukin-1beta (IL-1β) and IL-18 through activating caspase-1. It is associated with many diseases. Studies have shown that ERS can regulate NLRP3 inflammasome in many diseases including diabetes. However, the mechanism of the effects of ERS on NLRP3 inflammasome in diabetes has not been fully understood. This review summarizes the recent researches about the effects of ERS on NLRP3 inflammasome and the related mechanism in diabetes to provide ideas for the relevant basic research in the future.
Collapse
Affiliation(s)
- Shuangyu Lv
- Bioinformatics Center, School of Basic Medical Sciences, Institute of Biomedical Informatics, Henan University, Kaifeng, China
| | - Xiaotian Li
- Bioinformatics Center, School of Basic Medical Sciences, Institute of Biomedical Informatics, Henan University, Kaifeng, China
| | - Honggang Wang
- Bioinformatics Center, School of Basic Medical Sciences, Institute of Biomedical Informatics, Henan University, Kaifeng, China
| |
Collapse
|
34
|
So V, Klar G, Leitch J, McGillion M, Devereaux PJ, Arellano R, Parlow J, Gilron I. Association between postsurgical pain and heart rate variability: protocol for a scoping review. BMJ Open 2021; 11:e044949. [PMID: 33849852 PMCID: PMC8051399 DOI: 10.1136/bmjopen-2020-044949] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 03/29/2021] [Accepted: 03/30/2021] [Indexed: 11/20/2022] Open
Abstract
INTRODUCTION Surgical interventions can elicit neuroendocrine responses and sympathovagal imbalance, ultimately affecting cardiac autonomic function. Cardiac complications account for 30% of postoperative complications and are the leading cause of morbidity and mortality following non-cardiac surgery. One cardiovascular parameter, heart rate variability (HRV), has been found to be predictive of postoperative morbidity and mortality. HRV is defined as variation in time intervals between heartbeats and is affected by cardiac autonomic balance. Furthermore, altered HRV has been shown to predict cardiovascular events in non-surgical settings. In multiple studies, experimentally induced pain in healthy humans leads to reduced HRV suggesting a causal relationship. In a different studies, chronic pain has been associated with altered HRV, however, in the setting of clinical pain conditions, it remains unclear how much HRV impairment is due to pain itself versus autonomic changes related to analgesia. We aim to review the available evidence describing the association between postsurgical pain and HRV alterations in the early postoperative period. METHODS AND ANALYSIS We will conduct a scoping review of relevant studies using detailed searches of MEDLINE and EMBASE, in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analysis. Included studies will involve participants undergoing non-cardiac surgery and investigate outcomes of (1) measures of pain intensity; (2) measures of HRV and (3) statistical assessment of association between #1 and #2. As secondary review outcomes included studies will also be examined for other cardiovascular events and for their attempts to control for analgesic treatment and presurgical HRV differences among treatment groups in the analysis. This work aims to synthesise available evidence to inform future research questions related to postsurgical pain and cardiac complications. ETHICS AND DISSEMINATION Ethics review and approval is not required for this review. The results will be submitted for publication in peer-reviewed journals.
Collapse
Affiliation(s)
- Vincent So
- Anesthesiology and Perioperative Medicine, Queen's University, Kingston, Ontario, Canada
| | - Gregory Klar
- Anesthesiology and Perioperative Medicine, Queen's University, Kingston, Ontario, Canada
| | - Jordan Leitch
- Anesthesiology and Perioperative Medicine, Queen's University, Kingston, Ontario, Canada
| | - Michael McGillion
- School of Nursing, Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada
| | - P J Devereaux
- Division of Cardiology, Medicine, McMaster University Faculty of Health Sciences, Hamilton, Ontario, Canada
| | - Ramiro Arellano
- Anesthesiology and Perioperative Medicine, Queen's University, Kingston, Ontario, Canada
| | - Joel Parlow
- Anesthesiology and Perioperative Medicine, Queen's University, Kingston, Ontario, Canada
- Departments of Biomedical and Molecular Sciences, Queen's University Faculty of Health Sciences, Kingston, Ontario, Canada
| | - Ian Gilron
- Anesthesiology and Perioperative Medicine, Queen's University, Kingston, Ontario, Canada
- Departments of Biomedical and Molecular Sciences, Centre for Neuroscience Studies, Queen's University Faculty of Health Sciences, Kingston, Ontario, Canada
- School of Policy Studies, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
35
|
Li Y, Zhang X, Ma A, Kang Y. Rational Application of β-Hydroxybutyrate Attenuates Ischemic Stroke by Suppressing Oxidative Stress and Mitochondrial-Dependent Apoptosis via Activation of the Erk/CREB/eNOS Pathway. ACS Chem Neurosci 2021; 12:1219-1227. [PMID: 33739811 DOI: 10.1021/acschemneuro.1c00046] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Stroke is one of the leading causes of disability and death. Increasing evidence indicates that β-hydroxybutyrate (BHB) exerts beneficial effects in treating stroke, but the underlying mechanism remains largely unknown. In this study, we injected different doses of BHB into the lateral ventricle in middle cerebral artery occlusion (MCAO) model rats and neuronal cells were treated with different doses of BHB followed by oxygen-glucose deprivation (OGD). We found that a moderate dose of BHB enhanced mitochondrial complex I respiratory chain complex I activity, reduced oxidative stress, inhibited mitochondrial apoptosis, improved neurological scores, and reduced infarct volume after ischemia. We further showed that the effects of BHB were achieved by upregulating the dedicated BHB transporter SMCT1 and activating the Erk/CREB/eNOS pathway. These results provide us with a foundation for a novel understanding of the neuroprotective effects of BHB in stroke.
Collapse
Affiliation(s)
- Yang Li
- Intensive Care Unit, West China School of Medicine/West China Hospital of Sichuan University, Chengdu, Sichuan 610041, People’s Republic of China
| | - Xuepeng Zhang
- Intensive Care Unit, West China School of Medicine/West China Hospital of Sichuan University, Chengdu, Sichuan 610041, People’s Republic of China
| | - Aijia Ma
- Intensive Care Unit, West China School of Medicine/West China Hospital of Sichuan University, Chengdu, Sichuan 610041, People’s Republic of China
| | - Yan Kang
- Intensive Care Unit, West China School of Medicine/West China Hospital of Sichuan University, Chengdu, Sichuan 610041, People’s Republic of China
| |
Collapse
|
36
|
Semchyshyn H. Is carbonyl/AGE/RAGE stress a hallmark of the brain aging? Pflugers Arch 2021; 473:723-734. [PMID: 33742308 DOI: 10.1007/s00424-021-02529-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 01/21/2021] [Accepted: 01/28/2021] [Indexed: 12/13/2022]
Abstract
Recent studies have linked carbonyl stress to many physiological processes. Increase in the levels of carbonyl compounds, derived from both endogenous and exogenous sources, is believed to accompany normal age-related decline as well as different pathologies. Reactive carbonyl species (RCS) are capable of damaging biomolecules via their involvement in a net of nonspecific reactions. In the advanced stages of RCS metabolism, variety of poorly degraded adducts and crosslinks, collectively named advanced glycoxidation end products (AGEs), arises. They are accumulated in an age-dependent manner in different tissues and organs and can contribute to inflammatory processes. In particular, detrimental effects of the end products are realized via activation of the specific receptor for AGEs (RAGE) and RAGE-dependent inflammatory signaling cascade. Although it is unclear, whether carbonyl stress is causal for age-associated impairments or it results from age- and disease-related cell damages, increased levels of RCS and AGEs are tightly related to inflammaging, and therefore, attenuation of the RAGE signaling is suggested as an effective approach for the treatment of inflammation and age-related disorders. The question raised in this review is whether specific metabolism in the aging brain related to carbonyl/RCS/AGE/RAGE stress.
Collapse
Affiliation(s)
- Halyna Semchyshyn
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, 57 Shevchenko Str, Ivano-Frankivsk, 76018, Ukraine.
| |
Collapse
|
37
|
Did Cyclic Metaphosphates Have a Role in the Origin of Life? ORIGINS LIFE EVOL B 2021; 51:1-60. [PMID: 33721178 DOI: 10.1007/s11084-021-09604-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 01/29/2021] [Indexed: 12/13/2022]
Abstract
How life began still eludes science life, the initial progenote in the context presented herein, being a chemical aggregate of primordial inorganic and organic molecules capable of self-replication and evolution into ever increasingly complex forms and functions.Presented is a hypothesis that a mineral scaffold generated by geological processes and containing polymerized phosphate units was present in primordial seas that provided the initiating factor responsible for the sequestration and organization of primordial life's constituents. Unlike previous hypotheses proposing phosphates as the essential initiating factor, the key phosphate described here is not a polynucleotide or just any condensed phosphate but a large (in the range of at least 1 kilo-phosphate subunits), water soluble, cyclic metaphosphate, which is a closed loop chain of polymerized inorganic phosphate residues containing only phosphate middle groups. The chain forms an intrinsic 4-phosphate helix analogous to its structure in Na Kurrol's salt, and as with DNA, very large metaphosphates may fold into hairpin structures. Using a Holliday-junction-like scrambling mechanism, also analogous to DNA, rings may be manipulated (increased, decreased, exchanged) easily with little to no need for additional energy, the reaction being essentially an isomerization.A literature review is presented describing findings that support the above hypothesis. Reviewed is condensed phosphate inorganic chemistry including its geological origins, biological occurrence, enzymes and their genetics through eukaryotes, polyphosphate functions, circular polynucleotides and the role of the Holliday junction, previous biogenesis hypotheses, and an Eoarchean Era timeline.
Collapse
|
38
|
Mierziak J, Burgberger M, Wojtasik W. 3-Hydroxybutyrate as a Metabolite and a Signal Molecule Regulating Processes of Living Organisms. Biomolecules 2021; 11:biom11030402. [PMID: 33803253 PMCID: PMC8000602 DOI: 10.3390/biom11030402] [Citation(s) in RCA: 109] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/02/2021] [Accepted: 03/03/2021] [Indexed: 02/06/2023] Open
Abstract
3-hydroxybutyrate (3-HB) as a very important metabolite occurs in animals, bacteria and plants. It is well known that in animals, 3-HB is formed as a product of the normal metabolism of fatty acid oxidation and can therefore be used as an energy source in the absence of sufficient blood glucose. In microorganisms, 3-HB mainly serves as a substrate for the synthesis of polyhydroxybutyrate, which is a reserve material. Recent studies show that in plants, 3-HB acts as a regulatory molecule that most likely influences the expression of genes involved in DNA methylation, thereby altering DNA methylation levels. Additionally, in animals, 3-HB is not only an intermediate metabolite, but also an important regulatory molecule that can influence gene expression, lipid metabolism, neuronal function, and overall metabolic rate. Some of these effects are the direct effects of 3-HB itself, while others are indirect effects, regulated by the metabolites into which 3-HB is converted. One of the most important regulatory functions of 3-HB is the inhibition of the activity of histone deacetylases and thus the epigenetic regulation of many genes. Due to the number of functions of this compound, it also shows promising therapeutic properties.
Collapse
|
39
|
Sutton BS, Bermingham SL, Diamantopoulos A, Rosemas SC, Tsintzos SI, Xia Y, Reynolds MR. Economic value of insertable cardiac monitors in unexplained syncope in the United States. Open Heart 2021; 8:e001263. [PMID: 33622962 PMCID: PMC7907887 DOI: 10.1136/openhrt-2020-001263] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 09/17/2020] [Accepted: 01/31/2021] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION Early use of insertable cardiac monitor (ICM) is recommended for patients with unexplained syncope following initial clinical workup, due to its superior ability to establish symptom-rhythm correlation compared with conventional testing (CONV). However, ICMs incur higher upfront costs, and the impact of additional diagnoses and resulting treatment on downstream costs and outcomes is unclear. We aimed to evaluate the cost-effectiveness of ICM compared with CONV for the diagnosis of arrhythmia in patients with unexplained syncope, from a US payer perspective. METHODS A Markov model was developed to estimate lifetime costs and benefits of arrhythmia diagnosis with ICM versus CONV, considering all related diagnostic and arrhythmia-related treatment costs and consequences. Cohort characteristics and costs were informed by original claims database analyses. Risks of mortality, syncopal recurrence, injury due to syncope and quality of life consequences from syncopal events were identified from the literature. RESULTS ICM was less costly and more effective than CONV. Most of the observed US$4532 cost savings were attributed to reduced downstream diagnostic testing. For every 1000 patients, ICM was projected to yield an additional 253 arrhythmia diagnoses and lead to treatment in an additional 168 patients. The ICM strategy resulted in overall improved outcomes (0.30 quality-adjusted life years gained), due to a reduction in syncope recurrence and injury resulting from arrhythmia treatment. The results were robust to changes in the base case parameters but sensitive to the model time horizon, underlying probability of syncope recurrence and prevalence of arrhythmias. CONCLUSIONS Our model projected that early ICM for the diagnosis of unexplained syncope reduced long-term costs, and led to an improvement in overall clinical outcomes by shortening time to arrhythmia treatment. The cost of ICM was outweighed by savings arising from fewer downstream diagnostic episodes, and the increased cost of treatment was counterbalanced by fewer syncope-related event costs.
Collapse
Affiliation(s)
| | | | | | | | - Stelios I Tsintzos
- Health Economics and Reimbursement, Medtronic Europe SA, Tolochenaz, Vaud, Switzerland
| | - Ying Xia
- Medtronic Inc, Minneapolis, Minnesota, USA
| | - Matthew R Reynolds
- Cardiovascular Medicine, Baim Institute for Clinical Research, Boston, Massachusetts, USA
| |
Collapse
|
40
|
Kwak SE, Bae JH, Lee JH, Shin HE, Zhang D, Cho SC, Song W. Effects of exercise-induced beta-hydroxybutyrate on muscle function and cognitive function. Physiol Rep 2021; 9:e14497. [PMID: 33547753 PMCID: PMC7865404 DOI: 10.14814/phy2.14497] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 05/31/2020] [Accepted: 06/01/2020] [Indexed: 12/14/2022] Open
Abstract
Recent studies have shown that exercise improves skeletal muscle and cognitive function by stimulating the secretion of numerous molecules. In particular, previous studies have suggested that exercise-induced beta-hydroxybutyrate (BHB) release might improve skeletal muscle and cognitive function, but to date these studies have been limited to cell and animal models. Therefore, we aimed to determine how an exercise-induced increase in BHB affects skeletal muscle and cognitive function at a cellular level, in an animal model, and in humans. The effects of BHB on skeletal muscle and cognitive function were determined by treating C2C12 and C6 cell lines with BHB, and by measuring the skeletal muscle and serum BHB concentrations in aged mice after endurance or resistance exercise. In addition, serum BHB concentration was measured before and after high-speed band exercise in elderly people, and its relationships with muscle and cognitive function were analyzed. We found that BHB increased cell viability and brain-derived neurotrophic factor expression level in C6 cells, and endurance exercise, but not resistance exercise, increased the muscle BHB concentration in aged mice. Furthermore, the BHB concentration was positively related to skeletal muscle and cognitive function. Exercise did not increase the serum BHB concentration in the elderly people and BHB did not correlate with cognitive function, but after excluding the five people with the highest preexisting serum concentrations of BHB, the BHB concentrations of the remaining participants were increased by exercise, and the concentration showed a tendency toward a positive correlation with cognitive function. Thus, the BHB released by skeletal muscle following endurance exercise may improve muscle and cognitive function in animals and humans.
Collapse
Affiliation(s)
- Seong Eun Kwak
- Institute of Sport ScienceSeoul National UniversitySeoulKorea
| | - Jun Hyun Bae
- Institute of Sport ScienceSeoul National UniversitySeoulKorea
| | - Ji Heun Lee
- Institute of Sport ScienceSeoul National UniversitySeoulKorea
| | - Hyung Eun Shin
- Institute of Sport ScienceSeoul National UniversitySeoulKorea
| | - DiDi Zhang
- Institute of Sport ScienceSeoul National UniversitySeoulKorea
| | | | - Wook Song
- Institute of Sport ScienceSeoul National UniversitySeoulKorea
- Institue on AgingSeoul National UniversitySeoulKorea
| |
Collapse
|
41
|
Abstract
One of the characteristics of the failing human heart is a significant alteration in its energy metabolism. Recently, a ketone body, β-hydroxybutyrate (β-OHB) has been implicated in the failing heart’s energy metabolism as an alternative “fuel source.” Utilization of β-OHB in the failing heart increases, and this serves as a “fuel switch” that has been demonstrated to become an adaptive response to stress during the heart failure progression in both diabetic and non-diabetic patients. In addition to serving as an alternative “fuel,” β-OHB represents a signaling molecule that acts as an endogenous histone deacetylase (HDAC) inhibitor. It can increase histone acetylation or lysine acetylation of other signaling molecules. β-OHB has been shown to decrease the production of reactive oxygen species and activate autophagy. Moreover, β-OHB works as an NLR family pyrin domain-containing protein 3 (Nlrp3) inflammasome inhibitor and reduces Nlrp3-mediated inflammatory responses. It has also been reported that β-OHB plays a role in transcriptional or post-translational regulations of various genes’ expression. Increasing β-OHB levels prior to ischemia/reperfusion injury results in a reduced infarct size in rodents, likely due to the signaling function of β-OHB in addition to its role in providing energy. Sodium-glucose co-transporter-2 (SGLT2) inhibitors have been shown to exert strong beneficial effects on the cardiovascular system. They are also capable of increasing the production of β-OHB, which may partially explain their clinical efficacy. Despite all of the beneficial effects of β-OHB, some studies have shown detrimental effects of long-term exposure to β-OHB. Furthermore, not all means of increasing β-OHB levels in the heart are equally effective in treating heart failure. The best timing and therapeutic strategies for the delivery of β-OHB to treat heart disease are unknown and yet to be determined. In this review, we focus on the crucial role of ketone bodies, particularly β-OHB, as both an energy source and a signaling molecule in the stressed heart and the overall therapeutic potential of this compound for cardiovascular diseases.
Collapse
Affiliation(s)
- Yuxin Chu
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL, United States.,The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, the State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Cheng Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, the State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Min Xie
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
42
|
Wu J, Liu L, Hu H, Gao Z, Lu S. Bioinformatic analysis and experimental identification of blood biomarkers for chronic nonunion. J Orthop Surg Res 2020; 15:208. [PMID: 32503597 PMCID: PMC7275361 DOI: 10.1186/s13018-020-01735-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Accepted: 05/27/2020] [Indexed: 12/13/2022] Open
Abstract
Background Incomplete fracture healing may lead to chronic nonunion; thus, determining fracture healing is the primary issue in the clinical treatment. However, there are no validated early diagnostic biomarkers for assessing chronic nonunion. In this study, bioinformatics analysis combined with an experimental verification strategy was used to identify blood biomarkers for chronic nonunion. Methods First, differentially expressed genes in chronic nonunion were identified by microarray data analysis. Second, Dipsaci Radix (DR), a traditional Chinese medicine for fracture treatment, was used to screen the drug target genes. Third, the drug-disease network was determined, and biomarker genes were obtained. Finally, the potential blood biomarkers were verified by ELISA and qPCR methods. Results Fifty-five patients with open long bone fractures (39 healed and 16 nonunion) were enrolled in this study, and urgent surgical debridement and the severity of soft tissue injury had a significant effect on the prognosis of fracture. After the systems pharmacology analysis, six genes, including QPCT, CA1, LDHB, MMP9, UGCG, and HCAR2, were chosen for experimental validation. We found that all six genes in peripheral blood mononuclear cells (PBMCs) and serum were differentially expressed after injury, and five genes (QPCT, CA1, MMP9, UGCG, and HCAR2) were significantly lower in nonunion patients. Further, CA1, MMP9, and QPCT were markedly increased after DR treatment. Conclusion CA1, MMP9, and QPCT are biomarkers of nonunion patients and DR treatment targets. However, HCAR2 and UGCG are biomarkers of nonunion patients but not DR treatment targets. Therefore, our findings may provide valuable information for nonunion diagnosis and DR treatment. Trial registration ISRCTN, ISRCTN13271153. Registered 05 April 2020—Retrospectively registered.
Collapse
Affiliation(s)
- Jingwei Wu
- Department of Orthopedics, Xuanwu Hospital, Capital Medical University, Beijing, 100053, People's Republic of China
| | - Limin Liu
- Department of Orthopedics, Xuanwu Hospital, Capital Medical University, Beijing, 100053, People's Republic of China.
| | - Huaijian Hu
- Department of Orthopedics, Xuanwu Hospital, Capital Medical University, Beijing, 100053, People's Republic of China
| | - Zhihua Gao
- Department of Orthopedics, Xuanwu Hospital, Capital Medical University, Beijing, 100053, People's Republic of China
| | - Shibao Lu
- Department of Orthopedics, Xuanwu Hospital, Capital Medical University, Beijing, 100053, People's Republic of China.
| |
Collapse
|
43
|
Golonka RM, Xiao X, Abokor AA, Joe B, Vijay-Kumar M. Altered nutrient status reprograms host inflammation and metabolic health via gut microbiota. J Nutr Biochem 2020; 80:108360. [PMID: 32163821 PMCID: PMC7242157 DOI: 10.1016/j.jnutbio.2020.108360] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 02/07/2020] [Accepted: 02/08/2020] [Indexed: 02/07/2023]
Abstract
The metabolism of macro- and micronutrients is a complex and highly regulated biological process. An imbalance in the metabolites and their signaling networks can lead to nonresolving inflammation and consequently to the development of chronic inflammatory-associated diseases. Therefore, identifying the accumulated metabolites and altered pathways during inflammatory disorders would not only serve as "real-time" markers but also help in the development of nutritional therapeutics. In this review, we explore recent research that has delved into elucidating the effects of carbohydrate/calorie restriction, protein malnutrition, lipid emulsions and micronutrient deficiencies on metabolic health and inflammation. Moreover, we describe the integrated stress response in terms of amino acid starvation and lipemia and how this modulates new age diseases such as inflammatory bowel disease and atherosclerosis. Lastly, we explain the latest research on metaflammation and inflammaging. This review focuses on multiple signaling pathways, including, but not limited to, the FGF21-β-hydroxybutryate-NLRP3 axis, the GCN2-eIF2α-ATF4 pathway, the von Hippel-Lindau/hypoxia-inducible transcription factor pathway and the TMAO-PERK-FoxO1 axis. Additionally, throughout the review, we explain how the gut microbiota responds to altered nutrient status and also how antimicrobial peptides generated from nutrient-based signaling pathways can modulate the gut microbiota. Collectively, it must be emphasized that metabolic starvation and inflammation are strongly regulated by both environmental (i.e., nutrition, gut microbiome) and nonenvironmental (i.e., genetics) factors, which can influence the susceptibility to inflammatory disorders.
Collapse
Affiliation(s)
- Rachel M Golonka
- UT Microbiome Consortium, Department of Physiology and Pharmacology, The University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614
| | - Xia Xiao
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Ahmed A Abokor
- UT Microbiome Consortium, Department of Physiology and Pharmacology, The University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614
| | - Bina Joe
- UT Microbiome Consortium, Department of Physiology and Pharmacology, The University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614
| | - Matam Vijay-Kumar
- UT Microbiome Consortium, Department of Physiology and Pharmacology, The University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614.
| |
Collapse
|
44
|
Rezaei H, Rezaie Z, Seifati SM, Ardeshirylajimi A, Basiri A, Taheri M, Omrani MD. Poly-phosphate increases SMC differentiation of mesenchymal stem cells on PLGA-polyurethane nanofibrous scaffold. Cell Tissue Bank 2020; 21:495-505. [PMID: 32388594 PMCID: PMC7223593 DOI: 10.1007/s10561-020-09836-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Accepted: 05/04/2020] [Indexed: 11/30/2022]
Abstract
The use of bioactive scaffolds in tissue engineering has a significant effect on the damaged tissue healing by an increase in speed and quality of the process. Herein, electrospinning was applied to fabricate composite nanofibrous scaffolds by Poly lactic-co-glycolic acid (PLGA) and Polyurethane (PU) with and without poly-phosphate (poly-P). Scaffolds were characterized morphologically by scanning electron microscope (SEM), and their biocompatibility was also investigated by SEM, protein adsorption, cell attachment and survival assays. The applicability of the scaffolds for bladder tissue engineering was also evaluated by culturing mesenchymal stem cells (MSCs) on the scaffolds and their differentiation into smooth muscle cell (SMC) was studied at the gene and protein levels. The results demonstrated that scaffold biocompatibility was increased significantly by loading poly-P. SMC related gene and protein expression level in MSCs cultured on poly-P-loaded scaffold was also increased significantly compared to those cells cultured on empty scaffold. It can be concluded that poly-P hasn’t also increased scaffold biocompatibility, but also SMC differentiation potential of MSCs was also increased while cultured on the poly-P containing scaffold compared to the empty scaffold. Taken together, our study showed that PLGA–PU–poly-P alone and in combination with MSCs has a promising potential for support urinary bladder smooth muscle tissue engineering.
Collapse
Affiliation(s)
| | - Zahra Rezaie
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, No. 23, Shahid Labbafi Nejad Educational Hospital, Amir Ebrahimi St, Pasdaran Ave, Tehran, Iran
| | - Seyed Morteza Seifati
- Medical Biotechnology Research Center, Ashkezar Branch, Islamic Azad University, Ashkezar, Yazd, Iran
| | - Abdolreza Ardeshirylajimi
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Saint Louis, MO, USA
| | - Abbas Basiri
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Taheri
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, No. 23, Shahid Labbafi Nejad Educational Hospital, Amir Ebrahimi St, Pasdaran Ave, Tehran, Iran.
| | - Mir Davood Omrani
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
45
|
Dąbek A, Wojtala M, Pirola L, Balcerczyk A. Modulation of Cellular Biochemistry, Epigenetics and Metabolomics by Ketone Bodies. Implications of the Ketogenic Diet in the Physiology of the Organism and Pathological States. Nutrients 2020; 12:nu12030788. [PMID: 32192146 PMCID: PMC7146425 DOI: 10.3390/nu12030788] [Citation(s) in RCA: 113] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 03/05/2020] [Accepted: 03/11/2020] [Indexed: 12/13/2022] Open
Abstract
Ketone bodies (KBs), comprising β-hydroxybutyrate, acetoacetate and acetone, are a set of fuel molecules serving as an alternative energy source to glucose. KBs are mainly produced by the liver from fatty acids during periods of fasting, and prolonged or intense physical activity. In diabetes, mainly type-1, ketoacidosis is the pathological response to glucose malabsorption. Endogenous production of ketone bodies is promoted by consumption of a ketogenic diet (KD), a diet virtually devoid of carbohydrates. Despite its recently widespread use, the systemic impact of KD is only partially understood, and ranges from physiologically beneficial outcomes in particular circumstances to potentially harmful effects. Here, we firstly review ketone body metabolism and molecular signaling, to then link the understanding of ketone bodies’ biochemistry to controversies regarding their putative or proven medical benefits. We overview the physiological consequences of ketone bodies’ consumption, focusing on (i) KB-induced histone post-translational modifications, particularly β-hydroxybutyrylation and acetylation, which appears to be the core epigenetic mechanisms of activity of β-hydroxybutyrate to modulate inflammation; (ii) inflammatory responses to a KD; (iii) proven benefits of the KD in the context of neuronal disease and cancer; and (iv) consequences of the KD’s application on cardiovascular health and on physical performance.
Collapse
Affiliation(s)
- Arkadiusz Dąbek
- Department of Molecular Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland; (A.D.); (M.W.)
| | - Martyna Wojtala
- Department of Molecular Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland; (A.D.); (M.W.)
| | - Luciano Pirola
- INSERM Unit 1060, CarMeN Laboratory, 165 Chemin du Grand Revoyet - BP12, F-69495 Pierre Bénite CEDEX, France;
| | - Aneta Balcerczyk
- Department of Molecular Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland; (A.D.); (M.W.)
- Correspondence: ; Tel.: +48 42 635 45 10
| |
Collapse
|
46
|
Carroll MS, Ramirez JM, Weese-Mayer DE. Diurnal variation in autonomic regulation among patients with genotyped Rett syndrome. J Med Genet 2020; 57:786-793. [PMID: 32156713 DOI: 10.1136/jmedgenet-2019-106601] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 01/07/2020] [Accepted: 02/05/2020] [Indexed: 01/03/2023]
Abstract
BACKGROUND Rett syndrome is a severe neurological disorder with a range of disabling autonomic and respiratory symptoms and resulting predominantly from variants in the methyl-CpG binding protein 2 gene on the long arm of the X-chromosome. As basic research begins to suggest potential treatments, sensitive measures of the dynamic phenotype are needed to evaluate the results of these research efforts. Here we test the hypothesis that the physiological fingerprint of Rett syndrome in a naturalistic environment differs from that of controls, and differs among genotypes within Rett syndrome. METHODS A comprehensive array of heart rate variability, cardiorespiratory coupling and cardiac repolarisation measures were evaluated from an existing database of overnight and daytime inhome ambulatory recordings in 47 cases and matched controls. RESULTS Differences between girls with Rett syndrome and matched controls were apparent in a range of autonomic measures, and suggest a shift towards sympathetic activation and/or parasympathetic inactivation. Daily temporal trends analysed in the context of circadian rhythms reveal alterations in amplitude and phase of diurnal patterns of autonomic balance. Further analysis by genotype class confirms a graded presentation of the Rett syndrome phenotype such that patients with early truncating mutations were most different from controls, while late truncating and missense mutations were least different from controls. CONCLUSIONS Comprehensive autonomic measures from extensive inhome physiological measurements can detect subtle variations in the phenotype of girls with Rett syndrome, suggesting these techniques are suitable for guiding novel therapies.
Collapse
Affiliation(s)
- Michael Sean Carroll
- Data Analytics and Reporting, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois, USA .,Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.,Division of Autonomic Medicine, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois, USA
| | - Jan-Marino Ramirez
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington, USA.,Department of Neurological Surgery, University of Washington, Seattle, Washington, USA
| | - Debra E Weese-Mayer
- Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.,Division of Autonomic Medicine, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois, USA
| |
Collapse
|
47
|
Morris G, Maes M, Berk M, Carvalho AF, Puri BK. Nutritional ketosis as an intervention to relieve astrogliosis: Possible therapeutic applications in the treatment of neurodegenerative and neuroprogressive disorders. Eur Psychiatry 2020; 63:e8. [PMID: 32093791 PMCID: PMC8057392 DOI: 10.1192/j.eurpsy.2019.13] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Nutritional ketosis, induced via either the classical ketogenic diet or the use of emulsified medium-chain triglycerides, is an established treatment for pharmaceutical resistant epilepsy in children and more recently in adults. In addition, the use of oral ketogenic compounds, fractionated coconut oil, very low carbohydrate intake, or ketone monoester supplementation has been reported to be potentially helpful in mild cognitive impairment, Parkinson’s disease, schizophrenia, bipolar disorder, and autistic spectrum disorder. In these and other neurodegenerative and neuroprogressive disorders, there are detrimental effects of oxidative stress, mitochondrial dysfunction, and neuroinflammation on neuronal function. However, they also adversely impact on neurone–glia interactions, disrupting the role of microglia and astrocytes in central nervous system (CNS) homeostasis. Astrocytes are the main site of CNS fatty acid oxidation; the resulting ketone bodies constitute an important source of oxidative fuel for neurones in an environment of glucose restriction. Importantly, the lactate shuttle between astrocytes and neurones is dependent on glycogenolysis and glycolysis, resulting from the fact that the astrocytic filopodia responsible for lactate release are too narrow to accommodate mitochondria. The entry into the CNS of ketone bodies and fatty acids, as a result of nutritional ketosis, has effects on the astrocytic glutamate–glutamine cycle, glutamate synthase activity, and on the function of vesicular glutamate transporters, EAAT, Na+, K+-ATPase, Kir4.1, aquaporin-4, Cx34 and KATP channels, as well as on astrogliosis. These mechanisms are detailed and it is suggested that they would tend to mitigate the changes seen in many neurodegenerative and neuroprogressive disorders. Hence, it is hypothesized that nutritional ketosis may have therapeutic applications in such disorders.
Collapse
Affiliation(s)
- Gerwyn Morris
- Deakin University, IMPACT Strategic Research Centre, Barwon Health, School of Medicine, Geelong, Victoria, Australia
| | - Michael Maes
- Deakin University, IMPACT Strategic Research Centre, Barwon Health, School of Medicine, Geelong, Victoria, Australia.,Department of Psychiatry, Chulalongkorn University, Faculty of Medicine, Bangkok, Thailand
| | - Michael Berk
- Deakin University, IMPACT Strategic Research Centre, Barwon Health, School of Medicine, Geelong, Victoria, Australia.,Deakin University, CMMR Strategic Research Centre, School of Medicine, Geelong, Victoria, Australia.,Orygen, The National Centre of Excellence in Youth Mental Health, The Department of Psychiatry and the Florey Institute for Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - André F Carvalho
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada.,Centre for Addiction and Mental Health (CAMH), Toronto, Ontario, Canada
| | | |
Collapse
|
48
|
Rojas-Morales P, Pedraza-Chaverri J, Tapia E. Ketone bodies, stress response, and redox homeostasis. Redox Biol 2019; 29:101395. [PMID: 31926621 PMCID: PMC6911969 DOI: 10.1016/j.redox.2019.101395] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 11/16/2019] [Accepted: 11/23/2019] [Indexed: 02/08/2023] Open
Abstract
The ketone body β-hydroxybutyrate is no longer viewed simply as a metabolic intermediate, as it regulates a broad range of physiological processes at cellular and systemic levels. Particularly, β-hydroxybutyrate functions as a stress response molecule and orchestrates an antioxidant defense program to maintain redox homeostasis in response to environmental and metabolic challenges, such as ischemia. This property of β-hydroxybutyrate might be key for the beneficial effect of calorie restriction on stress response and disease processes.
Collapse
Affiliation(s)
- Pedro Rojas-Morales
- Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México, 04510, Mexico
| | - José Pedraza-Chaverri
- Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México, 04510, Mexico
| | - Edilia Tapia
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México, 14080, Mexico.
| |
Collapse
|
49
|
Camberos-Luna L, Massieu L. Therapeutic strategies for ketosis induction and their potential efficacy for the treatment of acute brain injury and neurodegenerative diseases. Neurochem Int 2019; 133:104614. [PMID: 31785349 DOI: 10.1016/j.neuint.2019.104614] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 11/26/2019] [Accepted: 11/26/2019] [Indexed: 12/13/2022]
Abstract
The therapeutic use of ketone bodies (KB) against acute brain injury and neurodegenerative disorders has lately been suggested by many studies. Several mechanisms responsible for the protective action of KB have been described, including metabolic, anti-inflammatory and epigenetic. However, it is still not clear whether a specific mechanism of action can be associated with a particular neurological disorder. Different strategies to induce ketosis including the ketogenic diet (KD), caloric restriction (CR), intermittent fasting (IF), as well as the administration of medium chain triglycerides (MCTs), exogenous ketones or KB derivatives, have been used in animal models of brain injury and in humans. They have shown different degrees of success to prevent neuronal damage, motor alterations and cognitive decline. However, more investigation is needed in order to establish safe protocols for clinical application. Throughout the present review, we describe the different approaches that have been used to elevate blood KB and discuss their effectiveness considering their advantages and limitations, as tested in models of brain injury, neurodegeneration and clinical research. We also describe the mechanisms of action of KB in non-pathologic conditions and in association with their protective effect against neuronal damage in acute neurological disorders and neurodegenerative diseases.
Collapse
Affiliation(s)
- Lucy Camberos-Luna
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, CP 04510, Mexico.
| | - Lourdes Massieu
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, CP 04510, Mexico.
| |
Collapse
|
50
|
McAllister MJ, Waldman HS, Rentería LI, Gonzalez AE, Butawan MB, Bloomer RJ. Acute coffee ingestion with and without medium-chain triglycerides decreases blood oxidative stress markers and increases ketone levels. Can J Physiol Pharmacol 2019; 98:194-200. [PMID: 31689118 DOI: 10.1139/cjpp-2019-0458] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Ingestion of ketone supplements, caffeine, and medium-chain triglycerides (MCTs) may all be effective strategies to increase blood levels of the ketone body beta-hydroxybutyrate (D-BHB). However, acute ingestion of a bolus of lipids may increase oxidative stress (OS). The purpose of the study was to investigate the impact of adding varying amounts of MCTs to coffee on blood levels of D-BHB and markers of OS. Ten college-aged men ingested coffee with 0, 28, and 42 g of MCT in a randomized order. Blood samples were collected pre- as well as 2 and 4 h postprandial and analyzed for D-BHB, total cholesterol (TC), high-density lipoprotein cholesterol (HDL-c), glucose, triglycerides (TAG), insulin, and OS markers: advanced oxidation protein products (AOPP), glutathione (GSH), malondialdehyde (MDA), and hydrogen peroxide (H2O2). All three treatments resulted in a significant increase in D-BHB, HDL-c, and TC as well as a significant decrease in TAG, MDA, H2O2, and insulin. The 42 g treatment was associated with significantly higher levels of AOPP and MDA. Acute ingestion of coffee results in favorable changes to markers of cardiometabolic health that were not impacted by the addition of 28 g of MCT. However, 42 g of MCT caused significantly greater OS.
Collapse
Affiliation(s)
- Matthew J McAllister
- Metabolic and Applied Physiology Laboratory, Department of Health and Human Performance, Texas State University, San Marcos, TX 78666, USA
| | - Hunter S Waldman
- Department of Kinesiology, University of North Alabama, Florence, AL 35632, USA
| | - Liliana I Rentería
- Metabolic and Applied Physiology Laboratory, Department of Health and Human Performance, Texas State University, San Marcos, TX 78666, USA
| | - Andrew E Gonzalez
- Metabolic and Applied Physiology Laboratory, Department of Health and Human Performance, Texas State University, San Marcos, TX 78666, USA
| | - Matthew B Butawan
- School of Health Studies, University of Memphis, Memphis, TN 38152, USA
| | - Richard J Bloomer
- School of Health Studies, University of Memphis, Memphis, TN 38152, USA
| |
Collapse
|