151
|
Affiliation(s)
- Navdeep S Chandel
- Northwestern University, Feinberg School of Medicine, Chicago, Illinois 60611, USA
| |
Collapse
|
152
|
Pavel P, Leman G, Hermann M, Ploner C, Eichmann TO, Minzaghi D, Radner FP, Del Frari B, Gruber R, Dubrac S. Peroxisomal Fatty Acid Oxidation and Glycolysis Are Triggered in Mouse Models of Lesional Atopic Dermatitis. JID INNOVATIONS 2021; 1:100033. [PMID: 34909730 PMCID: PMC8659757 DOI: 10.1016/j.xjidi.2021.100033] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 04/09/2021] [Accepted: 04/09/2021] [Indexed: 12/12/2022] Open
Abstract
Alterations of the lipid profile of the stratum corneum have an important role in the pathogenesis of atopic dermatitis (AD) because they contribute to epidermal barrier impairment. However, they have not previously been envisioned as a cellular response to altered metabolic requirements in AD epidermis. In this study, we report that the lipid composition in the epidermis of flaky tail, that is, ft/ft mice mimics that of human lesional AD (ADL) epidermis, both showing a shift toward shorter lipid species. The amounts of C24 and C26 free fatty acids and C24 and C26 ceramides-oxidized exclusively in peroxisomes-were reduced in the epidermis of ft/ft mice despite increased lipid synthesis, similar to that seen in human ADL edpidermis. Increased ACOX1 protein and activity in granular keratinocytes of ft/ft epidermis, altered lipid profile in human epidermal equivalents overexpressing ACOX1, and increased ACOX1 immunostaining in skin biopsies from patients with ADL suggest that peroxisomal β-oxidation significantly contributes to lipid signature in ADL epidermis. Moreover, we show that increased anaerobic glycolysis in ft/ft mouse epidermis is essential for keratinocyte proliferation and adenosine triphosphate synthesis but does not contribute to local inflammation. Thus, this work evidenced a metabolic shift toward enhanced peroxisomal β-oxidation and anaerobic glycolysis in ADL epidermis.
Collapse
Key Words
- AD, atopic dermatitis
- ADL, lesional atopic dermatitis
- ATP, adenosine triphosphate
- Cer, ceramide
- CoA, coenzyme A
- FA, fatty acid
- FFA, free fatty acid
- HEE, human epidermal equivalent
- IMQ, imiquimod
- KC, keratinocyte
- KO, knockout
- LB, lamellar body
- PPAR, peroxisome proliferator–activated receptor
- SC, stratum corneum
- TEWL, transepidermal water loss
- ULCFA, ultra long-chain fatty acid
- VLCFA, very-long-chain fatty acid
Collapse
Affiliation(s)
- Petra Pavel
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Géraldine Leman
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Martin Hermann
- KMT Laboratory, Department of Visceral, Transplant and Thoracic Surgery, Center for Operative Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Christian Ploner
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Deborah Minzaghi
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Franz P.W. Radner
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Barbara Del Frari
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Robert Gruber
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Sandrine Dubrac
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
153
|
Chen D, Zhang Y, Wang W, Chen H, Ling T, Yang R, Wang Y, Duan C, Liu Y, Guo X, Fang L, Liu W, Liu X, Liu J, Otkur W, Qi H, Liu X, Xia T, Liu H, Piao H. Identification and Characterization of Robust Hepatocellular Carcinoma Prognostic Subtypes Based on an Integrative Metabolite-Protein Interaction Network. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2100311. [PMID: 34247449 PMCID: PMC8425875 DOI: 10.1002/advs.202100311] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 05/08/2021] [Indexed: 06/01/2023]
Abstract
Metabolite-protein interactions (MPIs) play key roles in cancer metabolism. However, our current knowledge about MPIs in cancers remains limited due to the complexity of cancer cells. Herein, the authors construct an integrative MPI network and propose a MPI network based hepatocellular carcinoma (HCC) subtyping and mechanism exploration workflow. Based on the expressions of hub proteins on the MPI network, two prognosis-distinctive HCC subtypes are identified. Meanwhile, multiple interdependent features of the poor prognostic subtype are observed, including hypoxia, DNA hypermethylation of metabolic pathways, fatty acid accumulation, immune pathway up-regulation, and exhausted T-cell infiltration. Notably, the immune pathway up-regulation is probably induced by accumulated unsaturated fatty acids which are predicted to interact with multiple immune regulators like SRC and TGFB1. Moreover, based on tumor microenvironment compositions, the poor prognostic subtype is further divided into two sub-populations showing remarkable differences in metabolism. The subtyping shows a strong consistency across multiple HCC cohorts including early-stage HCC. Overall, the authors redefine robust HCC prognosis subtypes and identify potential MPIs linking metabolism to immune regulations, thus promoting understanding and clinical applications about HCC metabolism heterogeneity.
Collapse
Affiliation(s)
- Di Chen
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of SciencesDalian116023China
| | - Yiran Zhang
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of SciencesDalian116023China
- University of Chinese Academy of SciencesBeijing100049China
| | - Wen Wang
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of SciencesDalian116023China
- University of Chinese Academy of SciencesBeijing100049China
| | - Huan Chen
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of SciencesDalian116023China
- University of Chinese Academy of SciencesBeijing100049China
| | - Ting Ling
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of SciencesDalian116023China
- University of Chinese Academy of SciencesBeijing100049China
| | - Renyu Yang
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of SciencesDalian116023China
- University of Chinese Academy of SciencesBeijing100049China
| | - Yawei Wang
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of SciencesDalian116023China
- Department of Thoracic SurgeryCancer Hospital of China Medical UniversityLiaoning Cancer Hospital & InstituteShenyang110042China
| | - Chao Duan
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of SciencesDalian116023China
- Department of Thoracic SurgeryCancer Hospital of China Medical UniversityLiaoning Cancer Hospital & InstituteShenyang110042China
| | - Yu Liu
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of SciencesDalian116023China
- Department of Thoracic SurgeryCancer Hospital of China Medical UniversityLiaoning Cancer Hospital & InstituteShenyang110042China
| | - Xin Guo
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of SciencesDalian116023China
| | - Lei Fang
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of SciencesDalian116023China
| | - Wuguang Liu
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of SciencesDalian116023China
| | - Xiumei Liu
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of SciencesDalian116023China
| | - Jing Liu
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of SciencesDalian116023China
| | - Wuxiyar Otkur
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of SciencesDalian116023China
| | - Huan Qi
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of SciencesDalian116023China
| | - Xiaolong Liu
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of SciencesDalian116023China
| | - Tian Xia
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of SciencesDalian116023China
| | - Hong‐Xu Liu
- Department of Thoracic SurgeryCancer Hospital of China Medical UniversityLiaoning Cancer Hospital & InstituteShenyang110042China
| | - Hai‐long Piao
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of SciencesDalian116023China
- University of Chinese Academy of SciencesBeijing100049China
| |
Collapse
|
154
|
Hypothyroidism Intensifies Both Canonic and the De Novo Pathway of Peroxisomal Biogenesis in Rat Brown Adipocytes in a Time-Dependent Manner. Cells 2021; 10:cells10092248. [PMID: 34571897 PMCID: PMC8472630 DOI: 10.3390/cells10092248] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/18/2021] [Accepted: 08/23/2021] [Indexed: 02/08/2023] Open
Abstract
Despite peroxisomes being important partners of mitochondria by carrying out fatty acid oxidation in brown adipocytes, no clear evidence concerning peroxisome origin and way(s) of biogenesis exists. Herein we used methimazole-induced hypothyroidism for 7, 15, and 21 days to study peroxisomal remodeling and origin in rat brown adipocytes. We found that peroxisomes originated via both canonic, and de novo pathways. Each pathway operates in euthyroid control and over the course of hypothyroidism, in a time-dependent manner. Hypothyroidism increased the peroxisomal number by 1.8-, 3.6- and 5.8-fold on days 7, 15, and 21. Peroxisomal presence, their distribution, and their degree of maturation were heterogeneous in brown adipocytes in a Harlequin-like manner, reflecting differences in their origin. The canonic pathway, through numerous dumbbell-like and “pearls on strings” structures, supported by high levels of Pex11β and Drp1, prevailed on day 7. The de novo pathway of peroxisomal biogenesis started on day 15 and became dominant by day 21. The transition of peroxisomal biogenesis from canonic to the de novo pathway was driven by increased levels of Pex19, PMP70, Pex5S, and Pex26 and characterized by numerous tubular structures. Furthermore, specific peroxisomal origin from mitochondria, regardless of thyroid status, indicates their mutual regulation in rat brown adipocytes.
Collapse
|
155
|
He A, Dean JM, Lodhi IJ. Peroxisomes as cellular adaptors to metabolic and environmental stress. Trends Cell Biol 2021; 31:656-670. [PMID: 33674166 PMCID: PMC8566112 DOI: 10.1016/j.tcb.2021.02.005] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 02/02/2021] [Accepted: 02/04/2021] [Indexed: 02/06/2023]
Abstract
Peroxisomes are involved in multiple metabolic processes, including fatty acid oxidation, ether lipid synthesis, and reactive oxygen species (ROS) metabolism. Recent studies suggest that peroxisomes are critical mediators of cellular responses to various forms of stress, including oxidative stress, hypoxia, starvation, cold exposure, and noise. As dynamic organelles, peroxisomes can modulate their proliferation, morphology, and movement within cells, and engage in crosstalk with other organelles in response to external cues. Although peroxisome-derived hydrogen peroxide has a key role in cellular signaling related to stress, emerging studies suggest that other products of peroxisomal metabolism, such as acetyl-CoA and ether lipids, are also important for metabolic adaptation to stress. Here, we review molecular mechanisms through which peroxisomes regulate metabolic and environmental stress.
Collapse
Affiliation(s)
- Anyuan He
- Division of Endocrinology, Metabolism and Lipid Research, Department of Medicine, Washington University School of Medicine, St Louis, MO 63110, USA.
| | - John M Dean
- Division of Endocrinology, Metabolism and Lipid Research, Department of Medicine, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Irfan J Lodhi
- Division of Endocrinology, Metabolism and Lipid Research, Department of Medicine, Washington University School of Medicine, St Louis, MO 63110, USA.
| |
Collapse
|
156
|
Green CL, Englund DA, Das S, Herrerias MM, Yousefzadeh MJ, Grant RA, Clark J, Pak HH, Liu P, Bai H, Prahlad V, Lamming DW, Chusyd DE. The Second Annual Symposium of the Midwest Aging Consortium: The Future of Aging Research in the Midwestern United States. J Gerontol A Biol Sci Med Sci 2021; 76:2156-2161. [PMID: 34323268 PMCID: PMC8599030 DOI: 10.1093/gerona/glab210] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Indexed: 01/07/2023] Open
Abstract
While the average human life span continues to increase, there is little evidence that this is leading to a contemporaneous increase in "healthy years" experienced by our aging population. Consequently, many scientists focus their research on understanding the process of aging and trialing interventions that can promote healthspan. The 2021 Midwest Aging Consortium consensus statement is to develop and further the understanding of aging and age-related disease using the wealth of expertise across universities in the Midwestern United States. This report summarizes the cutting-edge research covered in a virtual symposium held by a consortium of researchers in the Midwestern United States, spanning topics such as senescence biomarkers, serotonin-induced DNA protection, immune system development, multisystem impacts of aging, neural decline following severe infection, the unique transcriptional impact of calorie restriction of different fat depots, the pivotal role of fasting in calorie restriction, the impact of peroxisome dysfunction, and the influence of early life trauma on health. The symposium speakers presented data from studies conducted in a variety of common laboratory animals as well as less-common species, including Caenorhabditis elegans, Drosophila, mice, rhesus macaques, elephants, and humans. The consensus of the symposium speakers is that this consortium highlights the strength of aging research in the Midwestern United States as well as the benefits of a collaborative and diverse approach to geroscience.
Collapse
Affiliation(s)
- Cara L Green
- Department of Medicine, University of Wisconsin–Madison, Madison, Wisconsin, USA,William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA
| | - Davis A Englund
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota, USA
| | - Srijit Das
- Department of Biology, Aging Mind & Brain Initiative, University of Iowa, Iowa City, Iowa, USA
| | - Mariana M Herrerias
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Matthew J Yousefzadeh
- Department of Biochemistry, Molecular Biology and Biophysics and Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, Minnesota, USA
| | - Rogan A Grant
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Josef Clark
- Department of Medicine, University of Wisconsin–Madison, Madison, Wisconsin, USA,William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA
| | - Heidi H Pak
- Department of Medicine, University of Wisconsin–Madison, Madison, Wisconsin, USA,William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA
| | - Peiduo Liu
- Department of Genetics, Development, and Cell Biology, Iowa State University, Ames, Iowa, USA
| | - Hua Bai
- Department of Genetics, Development, and Cell Biology, Iowa State University, Ames, Iowa, USA
| | - Veena Prahlad
- Department of Biology, Aging Mind & Brain Initiative, University of Iowa, Iowa City, Iowa, USA
| | - Dudley W Lamming
- Department of Medicine, University of Wisconsin–Madison, Madison, Wisconsin, USA,William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA
| | - Daniella E Chusyd
- Department of Epidemiology and Biostatistics, Indiana University-Bloomington, Bloomington, Indiana, USA,Address correspondence to: Daniella E. Chusyd, PhD, School of Public Health, Indiana University-Bloomington, 701 E. Kirkwood Ave., Bloomington, IN 47405-7100, USA. E-mail:
| |
Collapse
|
157
|
Wei X, Maharjan Y, Dorotea D, Dutta RK, Kim D, Kim H, Mu Y, Park C, Park R. Knockdown of PEX16 Induces Autophagic Degradation of Peroxisomes. Int J Mol Sci 2021; 22:ijms22157989. [PMID: 34360754 PMCID: PMC8348608 DOI: 10.3390/ijms22157989] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 07/16/2021] [Accepted: 07/23/2021] [Indexed: 01/16/2023] Open
Abstract
Peroxisome abundance is regulated by homeostasis between the peroxisomal biogenesis and degradation processes. Peroxin 16 (PEX16) is a peroxisomal protein involved in trafficking membrane proteins for de novo peroxisome biogenesis. The present study demonstrates that PEX16 also modulates peroxisome abundance through pexophagic degradation. PEX16 knockdown in human retinal pigment epithelial-1 cells decreased peroxisome abundance and function, represented by reductions in the expression of peroxisome membrane protein ABCD3 and the levels of cholesterol and plasmalogens, respectively. The activation of pexophagy under PEX16 knockdown was shown by (i) abrogated peroxisome loss under PEX16 knockdown in autophagy-deficient ATG5 knockout cell lines, and (ii) increased autophagy flux and co-localization of p62-an autophagy adaptor protein-with ABCD3 in the presence of the autophagy inhibitor chloroquine. However, the levels of cholesterol and plasmalogens did not recover despite the restoration of peroxisome abundance following chloroquine treatment. Thus, PEX16 is indispensable for maintaining peroxisome homeostasis by regulating not only the commonly known biogenesis pathway but also the autophagic degradation of peroxisomes.
Collapse
|
158
|
Meyer MT, Watermann C, Dreyer T, Wagner S, Wittekindt C, Klussmann JP, Ergün S, Baumgart-Vogt E, Karnati S. Differential Expression of Peroxisomal Proteins in Distinct Types of Parotid Gland Tumors. Int J Mol Sci 2021; 22:7872. [PMID: 34360635 PMCID: PMC8345988 DOI: 10.3390/ijms22157872] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/08/2021] [Accepted: 07/19/2021] [Indexed: 11/16/2022] Open
Abstract
Salivary gland cancers are rare but aggressive tumors that have poor prognosis and lack effective cure. Of those, parotid tumors constitute the majority. Functioning as metabolic machinery contributing to cellular redox balance, peroxisomes have emerged as crucial players in tumorigenesis. Studies on murine and human cells have examined the role of peroxisomes in carcinogenesis with conflicting results. These studies either examined the consequences of altered peroxisomal proliferators or compared their expression in healthy and neoplastic tissues. None, however, examined such differences exclusively in human parotid tissue or extended comparison to peroxisomal proteins and their associated gene expressions. Therefore, we examined differences in peroxisomal dynamics in parotid tumors of different morphologies. Using immunofluorescence and quantitative PCR, we compared the expression levels of key peroxisomal enzymes and proliferators in healthy and neoplastic parotid tissue samples. Three parotid tumor subtypes were examined: pleomorphic adenoma, mucoepidermoid carcinoma and acinic cell carcinoma. We observed higher expression of peroxisomal matrix proteins in neoplastic samples with exceptional down regulation of certain enzymes; however, the degree of expression varied between tumor subtypes. Our findings confirm previous experimental results on other organ tissues and suggest peroxisomes as possible therapeutic targets or markers in all or certain subtypes of parotid neoplasms.
Collapse
Affiliation(s)
- Malin Tordis Meyer
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Giessen, Klinikstrasse 33, Ebene-1, D-35392 Gießen, Germany; (M.T.M.); (C.W.); (S.W.); (C.W.); (J.P.K.)
| | - Christoph Watermann
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Giessen, Klinikstrasse 33, Ebene-1, D-35392 Gießen, Germany; (M.T.M.); (C.W.); (S.W.); (C.W.); (J.P.K.)
| | - Thomas Dreyer
- Institute of Pathology, Justus Liebig University, Langhansstrasse 10, D-35392 Gießen, Germany;
| | - Steffen Wagner
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Giessen, Klinikstrasse 33, Ebene-1, D-35392 Gießen, Germany; (M.T.M.); (C.W.); (S.W.); (C.W.); (J.P.K.)
| | - Claus Wittekindt
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Giessen, Klinikstrasse 33, Ebene-1, D-35392 Gießen, Germany; (M.T.M.); (C.W.); (S.W.); (C.W.); (J.P.K.)
| | - Jens Peter Klussmann
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Giessen, Klinikstrasse 33, Ebene-1, D-35392 Gießen, Germany; (M.T.M.); (C.W.); (S.W.); (C.W.); (J.P.K.)
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical Faculty, University of Cologne, D-50931 Cologne, Germany
| | - Süleyman Ergün
- Institute for Anatomy and Cell Biology, Julius-Maximilians-University Würzburg, Koellikerstrasse 6, D-97070 Würzburg, Germany;
| | - Eveline Baumgart-Vogt
- Institute for Anatomy and Cell Biology II, Medical Cell Biology, Justus Liebig University, D-35385 Gießen, Germany;
| | - Srikanth Karnati
- Institute for Anatomy and Cell Biology, Julius-Maximilians-University Würzburg, Koellikerstrasse 6, D-97070 Würzburg, Germany;
- Institute for Anatomy and Cell Biology II, Medical Cell Biology, Justus Liebig University, D-35385 Gießen, Germany;
| |
Collapse
|
159
|
Boumelhem BB, Pilgrim C, Zwicker VE, Kolanowski JL, Yeo JH, Jolliffe KA, New EJ, Day ML, Assinder SJ, Fraser ST. Intracellular flow cytometric lipid analysis - a multiparametric system to assess distinct lipid classes in live cells. J Cell Sci 2021; 135:269116. [PMID: 34114626 DOI: 10.1242/jcs.258322] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 05/28/2021] [Indexed: 11/20/2022] Open
Abstract
The lipid content of mammalian cells varies greatly between cell type. Current methods for analysing lipid components of cells are technically challenging and destructive. Here, we report a facile, inexpensive method to identify lipid content - intracellular flow cytometric lipid analysis (IFCLA). Distinct lipid classes can be distinguished by Nile Blue fluorescence, Nile Red fluorescence or violet autofluorescence. Nile Blue is fluorescent in the presence of unsaturated fatty acids with a carbon chain length greater than 16. Cis-configured fatty acids induce greater Nile Blue fluorescence than their trans-configured counterparts. In contrast, Nile Red exhibits greatest fluorescence in the presence of cholesterol, cholesteryl esters, some triglycerides and phospholipids. Multiparametric spanning-tree progression analysis for density-normalized events (SPADE) analysis of hepatic cellular lipid distribution, including vitamin A autofluorescence, is presented. This flow cytometric system allows for the rapid, inexpensive and non-destructive identification of lipid content, and highlights the differences in lipid biology between cell types by imaging and flow cytometry.
Collapse
Affiliation(s)
- Badwi B Boumelhem
- Discipline of Physiology, School of Medical Sciences, University of Sydney, Camperdown, NSW, 2050, Australia
| | - Chelsea Pilgrim
- Discipline of Physiology, School of Medical Sciences, University of Sydney, Camperdown, NSW, 2050, Australia
| | - Vincent E Zwicker
- School of Chemistry, University of Sydney, Camperdown, NSW, 2050, Australia
| | - Jacek L Kolanowski
- School of Chemistry, University of Sydney, Camperdown, NSW, 2050, Australia
| | - Jia Hao Yeo
- Discipline of Anatomy and Histology, School of Medical Sciences, University of Sydney, Camperdown, NSW, 2050, Australia
| | - Katrina A Jolliffe
- School of Chemistry, University of Sydney, Camperdown, NSW, 2050, Australia.,University of Sydney Nano Institute, University of Sydney, Camperdown, NSW, 2006, Australia
| | - Elizabeth J New
- School of Chemistry, University of Sydney, Camperdown, NSW, 2050, Australia.,University of Sydney Nano Institute, University of Sydney, Camperdown, NSW, 2006, Australia
| | - Margot L Day
- Discipline of Physiology, School of Medical Sciences, University of Sydney, Camperdown, NSW, 2050, Australia
| | - Stephen J Assinder
- Discipline of Physiology, School of Medical Sciences, University of Sydney, Camperdown, NSW, 2050, Australia.,University of Sydney Nano Institute, University of Sydney, Camperdown, NSW, 2006, Australia
| | - Stuart T Fraser
- Discipline of Physiology, School of Medical Sciences, University of Sydney, Camperdown, NSW, 2050, Australia.,Discipline of Anatomy and Histology, School of Medical Sciences, University of Sydney, Camperdown, NSW, 2050, Australia.,University of Sydney Nano Institute, University of Sydney, Camperdown, NSW, 2006, Australia.,School of Biomedical Engineering, University of Camperdown, NSW, 2006, Australia
| |
Collapse
|
160
|
Blunder S, Pavel P, Minzaghi D, Dubrac S. PPARdelta in Affected Atopic Dermatitis and Psoriasis: A Possible Role in Metabolic Reprograming. Int J Mol Sci 2021; 22:7354. [PMID: 34298981 PMCID: PMC8303290 DOI: 10.3390/ijms22147354] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/06/2021] [Accepted: 07/07/2021] [Indexed: 12/16/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) are nuclear hormone receptors expressed in the skin. Three PPAR isotypes, α (NRC1C1), β or δ (NRC1C2) and γ (NRC1C3), have been identified. After activation through ligand binding, PPARs heterodimerize with the 9-cis-retinoic acid receptor (RXR), another nuclear hormone receptor, to bind to specific PPAR-responsive elements in regulatory regions of target genes mainly involved in organogenesis, cell proliferation, cell differentiation, inflammation and metabolism of lipids or carbohydrates. Endogenous PPAR ligands are fatty acids and fatty acid metabolites. In past years, much emphasis has been given to PPARα and γ in skin diseases. PPARβ/δ is the least studied PPAR family member in the skin despite its key role in several important pathways regulating inflammation, keratinocyte proliferation and differentiation, metabolism and the oxidative stress response. This review focuses on the role of PPARβ/δ in keratinocytes and its involvement in psoriasis and atopic dermatitis. Moreover, the relevance of targeting PPARβ/δ to alleviate skin inflammation is discussed.
Collapse
Affiliation(s)
| | | | | | - Sandrine Dubrac
- Epidermal Biology Laboratory, Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria; (S.B.); (P.P.); (D.M.)
| |
Collapse
|
161
|
Deng Y, Hu S, Luo C, Ouyang Q, Li L, Ma J, Lin Z, Chen J, Liu H, Hu J, Chen G, Shu D, Pan Y, Hu B, He H, Qu H, Wang J. Integrative analysis of histomorphology, transcriptome and whole genome resequencing identified DIO2 gene as a crucial gene for the protuberant knob located on forehead in geese. BMC Genomics 2021; 22:487. [PMID: 34193033 PMCID: PMC8244220 DOI: 10.1186/s12864-021-07822-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 06/17/2021] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND During domestication, remarkable changes in behavior, morphology, physiology and production performance have taken place in farm animals. As one of the most economically important poultry, goose owns a unique appearance characteristic called knob, which is located at the base of the upper bill. However, neither the histomorphology nor the genetic mechanism of the knob phenotype has been revealed in geese. RESULTS In the present study, integrated radiographic, histological, transcriptomic and genomic analyses revealed the histomorphological characteristics and genetic mechanism of goose knob. The knob skin was developed, and radiographic results demonstrated that the knob bone was obviously protuberant and pneumatized. Histologically, there were major differences in structures in both the knob skin and bone between geese owing knob (namely knob-geese) and those devoid of knob (namely non-knob geese). Through transcriptome analysis, 592 and 952 genes differentially expressed in knob skin and bone, and significantly enriched in PPAR and Calcium pathways in knob skin and bone, respectively, which revealed the molecular mechanisms of histomorphological differences of the knob between knob- and non-knob geese. Furthermore, integrated transcriptomic and genomic analysis contributed to the identification of 17 and 21 candidate genes associated with the knob formation in the skin and bone, respectively. Of them, DIO2 gene could play a pivotal role in determining the knob phenotype in geese. Because a non-synonymous mutation (c.642,923 G > A, P265L) changed DIO2 protein secondary structure in knob geese, and Sanger sequencing further showed that the AA genotype was identified in the population of knob geese, and was prevalent in a crossing population which was artificially selected for 10 generations. CONCLUSIONS This study was the first to uncover the knob histomorphological characteristics and genetic mechanism in geese, and DIO2 was identified as the crucial gene associated with the knob phenotype. These data not only expand and enrich our knowledge on the molecular mechanisms underlying the formation of head appendages in both mammalian and avian species, but also have important theoretical and practical significance for goose breeding.
Collapse
Affiliation(s)
- Yan Deng
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Sichuan, 611130, Chengdu, China
| | - Shenqiang Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Sichuan, 611130, Chengdu, China
| | - Chenglong Luo
- The Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangdong, 510640, Guangzhou, China
| | - Qingyuan Ouyang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Sichuan, 611130, Chengdu, China
| | - Li Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Sichuan, 611130, Chengdu, China
| | - Jiaming Ma
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Sichuan, 611130, Chengdu, China
| | - Zhenping Lin
- The Baisha Livestock and Poultry Original Species Research Institute, Guangdong, 515000, Shantou, China
| | - Junpeng Chen
- The Baisha Livestock and Poultry Original Species Research Institute, Guangdong, 515000, Shantou, China
| | - Hehe Liu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Sichuan, 611130, Chengdu, China
| | - Jiwei Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Sichuan, 611130, Chengdu, China
| | - Guohong Chen
- Jiangsu Key Laboratory for Animal Genetic, Breeding and Molecular Design, Yangzhou University, Jiangsu, 225009, Yangzhou, China
| | - Dingming Shu
- The Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangdong, 510640, Guangzhou, China
| | - Yuxuan Pan
- The Baisha Livestock and Poultry Original Species Research Institute, Guangdong, 515000, Shantou, China
| | - Bo Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Sichuan, 611130, Chengdu, China
| | - Hua He
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Sichuan, 611130, Chengdu, China
| | - Hao Qu
- The Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangdong, 510640, Guangzhou, China.
| | - Jiwen Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Sichuan, 611130, Chengdu, China.
| |
Collapse
|
162
|
Yuan ZH, Liu T, Wang H, Xue LX, Wang JJ. Fatty Acids Metabolism: The Bridge Between Ferroptosis and Ionizing Radiation. Front Cell Dev Biol 2021; 9:675617. [PMID: 34249928 PMCID: PMC8264768 DOI: 10.3389/fcell.2021.675617] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 06/04/2021] [Indexed: 12/14/2022] Open
Abstract
Exposure of tumor cells to ionizing radiation (IR) alters the microenvironment, particularly the fatty acid (FA) profile and activity. Moreover, abnormal FA metabolism, either catabolism or anabolism, is essential for synthesizing biological membranes and delivering molecular signals to induce ferroptotic cell death. The current review focuses on the bistable regulation characteristics of FA metabolism and explains how FA catabolism and anabolism pathway crosstalk harmonize different ionizing radiation-regulated ferroptosis responses, resulting in pivotal cell fate decisions. In summary, targeting key molecules involved in lipid metabolism and ferroptosis may amplify the tumor response to IR.
Collapse
Affiliation(s)
- Zhu-hui Yuan
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, China
| | - Tong Liu
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
| | - Hao Wang
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, China
| | - Li-xiang Xue
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
- Biobank, Peking University Third Hospital, Beijing, China
| | - Jun-jie Wang
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, China
| |
Collapse
|
163
|
Dahabieh MS, Huang F, Goncalves C, Flores González RE, Prabhu S, Bolt A, Di Pietro E, Khoury E, Heath J, Xu ZY, Rémy-Sarrazin J, Mann KK, Orthwein A, Boisvert FM, Braverman N, Miller WH, Del Rincón SV. Silencing PEX26 as an unconventional mode to kill drug-resistant cancer cells and forestall drug resistance. Autophagy 2021; 18:540-558. [PMID: 34074205 DOI: 10.1080/15548627.2021.1936932] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Promoting the macroautophagy/autophagy-mediated degradation of specific proteins and organelles can potentially be utilized to induce apoptosis in cancer cells or sensitize tumor cells to therapy. To examine this concept, we enriched for autophagosomes from histone deacetylase inhibitor (HDACi)-sensitive U937 lymphoma cells and isogenic HDACi-resistant cells. Mass spectrometry on autophagosome-enriched fractions revealed that HDACi-resistant cells undergo elevated pexophagy, or autophagy of the peroxisome, an organelle that supports tumor growth. To disturb peroxisome homeostasis, we enhanced pexophagy in HDACi-resistant cells via genetic silencing of peroxisome exportomer complex components (PEX1, PEX6, or PEX26). This consequently sensitized resistant cells to HDACi-mediated apoptosis, which was rescued by inhibiting ATM/ataxia-telangiectasia mutated (ATM serine/threonine kinase), a mediator of pexophagy. We subsequently engineered melanoma cells to stably repress PEX26 using CRISPR interference (CRISPRi). Melanoma cells with repressed PEX26 expression showed evidence of both increased pexophagy and peroxisomal matrix protein import defects versus single guide scrambled (sgSCR) controls. In vivo studies showed that sgPEX26 melanoma xenografts recurred less compared to sgSCR xenografts, following the development of resistance to mitogen-activated protein kinase (MAPK)-targeted therapy. Finally, prognostic analysis of publicly available datasets showed that low expression levels of PEX26, PEX6 and MTOR, were significantly associated with prolonged patient survival in lymphoma, lung cancer and melanoma cohorts. Our work highlighted that drugs designed to disrupt peroxisome homeostasis may serve as unconventional therapies to combat therapy resistance in cancer.Abbreviations: ABCD3/PMP70: ATP binding cassette subfamily D member 3; ACOX1: acyl-CoA oxidase 1; AP: autophagosome; COX: cytochrome c oxidase; CQ: chloroquine; CRISPRi: clustered regularly interspaced short palindromic repeats interference; DLBCL: diffuse large B-cell lymphoma; GO: gene ontology; dCas9: Cas9 endonuclease dead, or dead Cas9; HDACi: histone deacetylase inhibitors; IHC: Immunohistochemistry; LAMP2: lysosomal associated membrane protein 2; LCFAs: long-chain fatty acids; LFQ-MS: label-free quantitation mass spectrometry; LPC: lysophoshatidylcholine; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; MTOR: mechanistic target of rapamycin kinase; PBD: peroxisome biogenesis disorders; PTS1: peroxisomal targeting signal 1; ROS: reactive oxygen species; sgRNA: single guide RNA; VLCFAs: very-long chain fatty acids; Vor: vorinostat; WO: wash-off.
Collapse
Affiliation(s)
- Michael S Dahabieh
- Lady Davis Institute, McGill University, Montréal, Canada.,Department of Experimental Medicine, McGill University, Montréal, Canada
| | - Fan Huang
- Lady Davis Institute, McGill University, Montréal, Canada.,Department of Experimental Medicine, McGill University, Montréal, Canada
| | | | - Raúl Ernesto Flores González
- Lady Davis Institute, McGill University, Montréal, Canada.,Department of Experimental Medicine, McGill University, Montréal, Canada
| | - Sathyen Prabhu
- Lady Davis Institute, McGill University, Montréal, Canada.,Department of Experimental Medicine, McGill University, Montréal, Canada
| | - Alicia Bolt
- Lady Davis Institute, McGill University, Montréal, Canada
| | - Erminia Di Pietro
- Department of Human Genetics and Pediatrics, Research Institute of McGill University Children's Hospital, Montréal, Canada
| | - Elie Khoury
- Lady Davis Institute, McGill University, Montréal, Canada.,Department of Experimental Medicine, McGill University, Montréal, Canada
| | - John Heath
- Lady Davis Institute, McGill University, Montréal, Canada.,Department of Experimental Medicine, McGill University, Montréal, Canada
| | - Zi Yi Xu
- Lady Davis Institute, McGill University, Montréal, Canada
| | | | - Koren K Mann
- Lady Davis Institute, McGill University, Montréal, Canada.,Department of Experimental Medicine, McGill University, Montréal, Canada.,Department of Oncology, McGill University, Montréal, Canada
| | - Alexandre Orthwein
- Lady Davis Institute, McGill University, Montréal, Canada.,Department of Experimental Medicine, McGill University, Montréal, Canada.,Department of Oncology, McGill University, Montréal, Canada
| | | | - Nancy Braverman
- Department of Human Genetics and Pediatrics, Research Institute of McGill University Children's Hospital, Montréal, Canada
| | - Wilson H Miller
- Lady Davis Institute, McGill University, Montréal, Canada.,Department of Experimental Medicine, McGill University, Montréal, Canada.,Department of Oncology, McGill University, Montréal, Canada
| | - Sonia V Del Rincón
- Lady Davis Institute, McGill University, Montréal, Canada.,Department of Experimental Medicine, McGill University, Montréal, Canada.,Department of Oncology, McGill University, Montréal, Canada
| |
Collapse
|
164
|
Levis NA, Kelly PW, Harmon EA, Ehrenreich IM, McKay DJ, Pfennig DW. Transcriptomic bases of a polyphenism. JOURNAL OF EXPERIMENTAL ZOOLOGY PART B-MOLECULAR AND DEVELOPMENTAL EVOLUTION 2021; 336:482-495. [PMID: 34142757 DOI: 10.1002/jez.b.23066] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 03/22/2021] [Accepted: 05/22/2021] [Indexed: 11/06/2022]
Abstract
Polyphenism-in which multiple distinct phenotypes are produced from a single genotype owing to differing environmental conditions-is commonplace, but its molecular bases are poorly understood. Here, we examine the transcriptomic bases of a polyphenism in Mexican spadefoot toads (Spea multiplicata). Depending on their environment, their tadpoles develop into either a default "omnivore" morph or a novel "carnivore" morph. We compared patterns of gene expression among sibships that exhibited high versus low production of carnivores when reared in conditions that induce the carnivore morph versus those that do not. We found that production of the novel carnivore morph actually involved changes in fewer genes than did the maintenance of the default omnivore morph in the inducing environment. However, only body samples showed this pattern; head samples showed the opposite pattern. We also found that changes to lipid metabolism (especially cholesterol biosynthesis) and peroxisome contents and function might be crucial for establishing and maintaining differences between the morphs. Thus, our findings suggest that carnivore phenotype might have originally evolved following the breakdown of robustness mechanisms that maintain the default omnivore phenotype, and that the carnivore morph is developmentally regulated by lipid metabolism and peroxisomal form, function, and/or signaling. This study also serves as a springboard for further exploration into the nature and causes of plasticity in an emerging model system.
Collapse
Affiliation(s)
- Nicholas A Levis
- Department of Biology, University of North Carolina, Chapel Hill, North Carolina, USA.,Current affiliation: Department of Biology, Indiana University, Bloomington, Indiana, USA
| | - Patrick W Kelly
- Department of Biology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Emily A Harmon
- Department of Biology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Ian M Ehrenreich
- Molecular and Computational Biology Section, University of Southern, Los Angeles, California, USA
| | - Daniel J McKay
- Department of Biology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - David W Pfennig
- Department of Biology, University of North Carolina, Chapel Hill, North Carolina, USA
| |
Collapse
|
165
|
Ghaffari MH, Alaedin MT, Sadri H, Hofs I, Koch C, Sauerwein H. Longitudinal changes in fatty acid metabolism and in the mitochondrial protein import system in overconditioned and normal conditioned cows: A transcriptional study using microfluidic quantitative PCR. J Dairy Sci 2021; 104:10338-10354. [PMID: 34147221 DOI: 10.3168/jds.2021-20237] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 05/03/2021] [Indexed: 12/22/2022]
Abstract
This study investigated the effect of body condition around calving on the hepatic mRNA expression of genes involved in fatty acid (FA) metabolism and mitochondrial protein import system of dairy cows during the transition period. Fifteen weeks before their anticipated calving date, 38 multiparous Holstein cows were selected based on their current and previous body condition scores (BCS) and allocated to either a high or a normal BCS group (19 cows each). They received different diets to reach targeted differences in BCS and backfat thickness (BFT) until dry-off. At dry-off, normal BCS (NBCS) cows had a BCS <3.5 and BFT <1.2 cm, and the high BCS (HBCS) cows had a BCS >3.75 and BFT >1.4 cm. The expression of targeted genes in the liver was assayed by reverse-transcription quantitative real-time PCR using microfluidics integrated fluidic circuit chips on a subset of 5 cows from each group. Liver biopsies were collected at d -49, +3, +21, and +84 relative to parturition. The mRNA abundance of 47 genes related to lipid metabolism including carnitine metabolism, FA uptake and transport, lipoprotein export, carnitine metabolism, mitochondrial and proximal FA oxidation, ketogenesis, AMP-activated protein kinase/mammalian target of rapamycin pathway, and mitochondrial protein import system was assessed in liver tissue. The mRNA abundances of FA binding protein (FABP)6 (in both groups), and FABP1 and solute carrier family 22 member 5 (SLC22A5) in HBCS were upregulated (>1.5-fold change, FC) in early lactation (at d +3 and +21 postpartum) compared with antepartum (d -49), indicating promoted FA uptake and intracellular transport in the liver due to the metabolic adaptations of elevated lipo-mobilization after parturition. The upregulation of SLC22A5 and SLC25A20 after parturition was more pronounced in HBCS than in NBCS cows, suggesting a need for increasing the capacity of FA uptake, and FA transport into the hepatocyte. The increased mRNA abundance of carnitine palmitoyltransferase 1A, after parturition and to a greater extent in HBCS (FC = 4.1) versus NBCS (FC = 2.1) indicates a physiological increase in the capacity of long-chain fatty acyl-CoA entry into the liver mitochondria compared with antepartum (ap; d -49 relative to calving). The greater hepatic mRNA abundance of genes encoding enzymes involved in mitochondrial FA oxidation in HBCS than in NBCS points to an increased rate of mitochondrial β-oxidation. The hepatic mRNA abundance of 3-hydroxy-3-methylglutaryl-CoA synthase 2 and 3-hydroxy-3-methylglutaryl-CoA were upregulated after parturition (d +21/d +3 pp) to a greater extent in HBCS than in NBCS cows, indicating that excess acetyl-CoA generated via β-oxidation was increasingly used for ketogenesis. We observed for the first time that the mRNA abundance of genes involved in the translocase of the inner membrane (TIM) complex (TIM22 and TIM23) in the hepatic mitochondrial protein import system were undergoing distinct changes during the transition from late pregnancy to early lactation in dairy cows. Even though sample size in this study was relatively small, the results support that overconditioning around calving may contribute to mitochondrial FA overload and greater ketogenesis at the level of transcription in the liver of early lactation cows.
Collapse
Affiliation(s)
- Morteza H Ghaffari
- Institute of Animal Science, Physiology Unit, University of Bonn, 53115 Bonn, Germany.
| | - Mohamad Taher Alaedin
- Institute of Animal Science, Physiology Unit, University of Bonn, 53115 Bonn, Germany
| | - Hassan Sadri
- Department of Clinical Science, Faculty of Veterinary Medicine, University of Tabriz, 516616471 Tabriz, Iran
| | - Inga Hofs
- Institute of Animal Science, Physiology Unit, University of Bonn, 53115 Bonn, Germany
| | - Christian Koch
- Educational and Research Center for Animal Husbandry, Hofgut Neumuehle, 67728 Muenchweiler an der Alsenz, Germany
| | - Helga Sauerwein
- Institute of Animal Science, Physiology Unit, University of Bonn, 53115 Bonn, Germany
| |
Collapse
|
166
|
Peroxisomal ABC Transporters: An Update. Int J Mol Sci 2021; 22:ijms22116093. [PMID: 34198763 PMCID: PMC8201181 DOI: 10.3390/ijms22116093] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 06/01/2021] [Accepted: 06/03/2021] [Indexed: 12/12/2022] Open
Abstract
ATP-binding cassette (ABC) transporters constitute one of the largest superfamilies of conserved proteins from bacteria to mammals. In humans, three members of this family are expressed in the peroxisomal membrane and belong to the subfamily D: ABCD1 (ALDP), ABCD2 (ALDRP), and ABCD3 (PMP70). These half-transporters must dimerize to form a functional transporter, but they are thought to exist primarily as tetramers. They possess overlapping but specific substrate specificity, allowing the transport of various lipids into the peroxisomal matrix. The defects of ABCD1 and ABCD3 are responsible for two genetic disorders called X-linked adrenoleukodystrophy and congenital bile acid synthesis defect 5, respectively. In addition to their role in peroxisome metabolism, it has recently been proposed that peroxisomal ABC transporters participate in cell signaling and cell control, particularly in cancer. This review presents an overview of the knowledge on the structure, function, and mechanisms involving these proteins and their link to pathologies. We summarize the different in vitro and in vivo models existing across the species to study peroxisomal ABC transporters and the consequences of their defects. Finally, an overview of the known and possible interactome involving these proteins, which reveal putative and unexpected new functions, is shown and discussed.
Collapse
|
167
|
Andrés‐Benito P, Gelpi E, Jové M, Mota‐Martorell N, Obis È, Portero‐Otin M, Povedano M, Pujol A, Pamplona R, Ferrer I. Lipid alterations in human frontal cortex in ALS-FTLD-TDP43 proteinopathy spectrum are partly related to peroxisome impairment. Neuropathol Appl Neurobiol 2021; 47:544-563. [PMID: 33332650 PMCID: PMC8248144 DOI: 10.1111/nan.12681] [Citation(s) in RCA: 13] [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: 08/21/2020] [Revised: 10/26/2020] [Accepted: 12/13/2020] [Indexed: 01/08/2023]
Abstract
AIM Peroxisomes play a key role in lipid metabolism, and peroxisome defects have been associated with neurodegenerative diseases such as X-adrenoleukodystrophy and Alzheimer's disease. This study aims to elucidate the contribution of peroxisomes in lipid alterations of area 8 of the frontal cortex in the spectrum of TDP43-proteinopathies. Cases of frontotemporal lobar degeneration-TDP43 (FTLD-TDP), manifested as sporadic (sFTLD-TDP) or linked to mutations in various genes including expansions of the non-coding region of C9ORF72 (c9FTLD), and of sporadic amyotrophic lateral sclerosis (sALS) as the most common TDP43 proteinopathies, were analysed. METHODS We used transcriptomics and lipidomics methods to define the steady-state levels of gene expression and lipid profiles. RESULTS Our results show alterations in gene expression of some components of peroxisomes and related lipid pathways in frontal cortex area 8 in sALS, sFTLD-TDP and c9FTLD. Additionally, we identify a lipidomic pattern associated with the ALS-FTLD-TDP43 proteinopathy spectrum, notably characterised by down-regulation of ether lipids and acylcarnitine among other lipid species, as well as alterations in the lipidome of each phenotype of TDP43 proteinopathy, which reveals commonalities and disease-dependent differences in lipid composition. CONCLUSION Globally, lipid alterations in the human frontal cortex of the ALS-FTLD-TDP43 proteinopathy spectrum, which involve cell membrane composition and signalling, vulnerability against cellular stress and possible glucose metabolism, are partly related to peroxisome impairment.
Collapse
Affiliation(s)
- Pol Andrés‐Benito
- NeuropathologyBellvitge University Hospital‐Bellvitge Biomedical Research Institute (IDIBELLHospitalet de Llobregat, BarcelonaSpain
- Department of Pathology and Experimental TherapeuticsUniversity of BarcelonaBarcelonaSpain
- CIBERNED (Network Centre of Biomedical Research of Neurodegenerative DiseasesInstitute of Health Carlos IIIMinistry of Economy and CompetitivenessMadridSpain
- International Initiative for Treatment and Research Initiative to Cure ALS (TRICALSUtrechtThe Netherlands
| | - Ellen Gelpi
- Neurological Tissue Bank of the Biobanc‐Hospital Clínic‐Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPSBarcelonaSpain
- Institute of NeurologyMedical University of ViennaViennaAustria
| | - Mariona Jové
- Department of Experimental MedicineUniversity of Lleida ‐ Lleida Biomedical Research Institute (UdL‐IRBLleidaLleidaSpain
| | - Natalia Mota‐Martorell
- Department of Experimental MedicineUniversity of Lleida ‐ Lleida Biomedical Research Institute (UdL‐IRBLleidaLleidaSpain
| | - Èlia Obis
- Department of Experimental MedicineUniversity of Lleida ‐ Lleida Biomedical Research Institute (UdL‐IRBLleidaLleidaSpain
| | - Manuel Portero‐Otin
- Department of Experimental MedicineUniversity of Lleida ‐ Lleida Biomedical Research Institute (UdL‐IRBLleidaLleidaSpain
| | - Mònica Povedano
- International Initiative for Treatment and Research Initiative to Cure ALS (TRICALSUtrechtThe Netherlands
- Functional Unit of Amyotrophic Lateral Sclerosis (UFELAService of NeurologyBellvitge University HospitalHospitalet de LlobregatSpain
| | - Aurora Pujol
- Catalan Institution for Research and Advanced Studies (ICREABarcelonaSpain
- Neurometabolic Diseases LaboratoryBellvitge Biomedical Research InstituteHospital Duran i ReynalsHospitalet de Llobregat, BarcelonaSpain
- Center for Biomedical Research on Rare Diseases (CIBERERInstitute of Health Carlos IIIMadridSpain
| | - Reinald Pamplona
- Department of Experimental MedicineUniversity of Lleida ‐ Lleida Biomedical Research Institute (UdL‐IRBLleidaLleidaSpain
| | - Isidro Ferrer
- NeuropathologyBellvitge University Hospital‐Bellvitge Biomedical Research Institute (IDIBELLHospitalet de Llobregat, BarcelonaSpain
- Department of Pathology and Experimental TherapeuticsUniversity of BarcelonaBarcelonaSpain
- CIBERNED (Network Centre of Biomedical Research of Neurodegenerative DiseasesInstitute of Health Carlos IIIMinistry of Economy and CompetitivenessMadridSpain
- International Initiative for Treatment and Research Initiative to Cure ALS (TRICALSUtrechtThe Netherlands
- Institute of NeurosciencesUniversity of BarcelonaBarcelonaSpain
| |
Collapse
|
168
|
Jiang Y, Li L, Chen X, Liu J, Yuan J, Xie Q, Han H. Three-dimensional ATUM-SEM reconstruction and analysis of hepatic endoplasmic reticulum‒organelle interactions. J Mol Cell Biol 2021; 13:636-645. [PMID: 34048584 PMCID: PMC8648385 DOI: 10.1093/jmcb/mjab032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 02/16/2021] [Accepted: 02/24/2021] [Indexed: 11/24/2022] Open
Abstract
The endoplasmic reticulum (ER) is a contiguous and complicated membrane network in eukaryotic cells, and membrane contact sites (MCSs) between the ER and other organelles perform vital cellular functions, including lipid homeostasis, metabolite exchange, calcium level regulation, and organelle division. Here, we establish a whole pipeline to reconstruct all ER, mitochondria, lipid droplets, lysosomes, peroxisomes, and nuclei by automated tape-collecting ultramicrotome scanning electron microscopy and deep learning techniques, which generates an unprecedented 3D model for mapping liver samples. Furthermore, the morphology of various organelles and the MCSs between the ER and other organelles are systematically analyzed. We found that the ER presents with predominantly flat cisternae and is knitted tightly all throughout the intracellular space and around other organelles. In addition, the ER has a smaller volume-to-membrane surface area ratio than other organelles, which suggests that the ER could be more suited for functions that require a large membrane surface area. Our data also indicate that ER‒mitochondria contacts are particularly abundant, especially for branched mitochondria. Our study provides 3D reconstructions of various organelles in liver samples together with important fundamental information for biochemical and functional studies in the liver.
Collapse
Affiliation(s)
- Yi Jiang
- National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China.,School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing 101408, China
| | - Linlin Li
- National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Xi Chen
- National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Jiazheng Liu
- National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China.,School of Future Technology, University of Chinese Academy of Sciences, Beijing 101408, China
| | - Jingbin Yuan
- National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Qiwei Xie
- Data Mining Lab, Beijing University of Technology, Beijing 100124, China
| | - Hua Han
- National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China.,School of Future Technology, University of Chinese Academy of Sciences, Beijing 101408, China.,CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai 200031, China
| |
Collapse
|
169
|
Park JK, Coffey NJ, Limoges A, Le A. The Heterogeneity of Lipid Metabolism in Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1311:39-56. [PMID: 34014533 DOI: 10.1007/978-3-030-65768-0_3] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The study of cancer cell metabolism has traditionally focused on glycolysis and glutaminolysis. However, lipidomic technologies have matured considerably over the last decade and broadened our understanding of how lipid metabolism is relevant to cancer biology [1-3]. Studies now suggest that the reprogramming of cellular lipid metabolism contributes directly to malignant transformation and progression [4, 5]. For example, de novo lipid synthesis can supply proliferating tumor cells with phospholipid components that comprise the plasma and organelle membranes of new daughter cells [6, 7]. Moreover, the upregulation of mitochondrial β-oxidation can support tumor cell energetics and redox homeostasis [8], while lipid-derived messengers can regulate major signaling pathways or coordinate immunosuppressive mechanisms [9-11]. Lipid metabolism has, therefore, become implicated in a variety of oncogenic processes, including metastatic colonization, drug resistance, and cell differentiation [10, 12-16]. However, whether we can safely and effectively modulate the underlying mechanisms of lipid metabolism for cancer therapy is still an open question.
Collapse
Affiliation(s)
- Joshua K Park
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nathan J Coffey
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Aaron Limoges
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Anne Le
- Department of Pathology and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA. .,Department of Chemical and Biomolecular Engineering, Johns Hopkins University Whiting School of Engineering, Baltimore, MD, USA.
| |
Collapse
|
170
|
Kim EH, Ridlo MR, Lee BC, Kim GA. Crosstalk between Peroxisomal Activities and Nrf2 Signaling in Porcine Embryos. Antioxidants (Basel) 2021; 10:antiox10050771. [PMID: 34068072 PMCID: PMC8152488 DOI: 10.3390/antiox10050771] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/06/2021] [Accepted: 05/10/2021] [Indexed: 01/01/2023] Open
Abstract
Melatonin and phytanic acid (PA) are known to be involved in lipid metabolism and β-oxidation, in which peroxisomal activities also significantly participate. In addition, other studies have reported that the nuclear factor-erythroid-derived 2-like 2 (Nrf2 or NFE2L2) signaling pathway mediates lipid metabolism and its subsequent cascades. As these mechanisms are partially involved in porcine oocytes or embryonic development, we hypothesized that the factors governing these mechanisms could be interconnected. Therefore, we aimed to investigate possible crosstalk between peroxisomal activities and Nrf2 signaling in porcine embryos following melatonin and PA treatment. Porcine embryos were cultured for seven days after parthenogenetic activation, and subsequently treated with melatonin and PA, or injected with Pex19-targeted siRNAs. Real-time PCR, immunocytochemistry, and BODIPY staining were used to evaluate peroxisomal activities, Nrf2 signaling, and subsequent lipid metabolism. We found that melatonin/PA treatment enhanced embryonic development, whereas injection with Pex19-targeted siRNAs had the opposite effect. Moreover, melatonin/PA treatment upregulated peroxisomal activities, Nrf2 signaling, lipid metabolism, and mitochondrial membrane potentials, whereas most of these mechanisms were downregulated by Pex19-targeted siRNAs. Therefore, we suggest that there is a connection between the action of melatonin and PA and the Nrf2 signaling pathway and peroxisomal activities, which positively influences porcine embryonic development.
Collapse
Affiliation(s)
- Eui-Hyun Kim
- Department of Theriogenology and Biotechnology, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea; (E.-H.K.); (M.-R.R.); (B.-C.L.)
- Optipharm Inc., Cheongju 28158, Korea
| | - Muhammad-Rosyid Ridlo
- Department of Theriogenology and Biotechnology, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea; (E.-H.K.); (M.-R.R.); (B.-C.L.)
- Department of Bioresources Technology and Veterinary, Vocational College, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia
| | - Byeong-Chun Lee
- Department of Theriogenology and Biotechnology, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea; (E.-H.K.); (M.-R.R.); (B.-C.L.)
| | - Geon A. Kim
- Department of Clinical Pathology, College of Health Science, Eulji University, Uijeongbu 11759, Korea
- Correspondence:
| |
Collapse
|
171
|
Mitochondria and Peroxisome Remodeling across Cytomegalovirus Infection Time Viewed through the Lens of Inter-ViSTA. Cell Rep 2021; 32:107943. [PMID: 32726614 DOI: 10.1016/j.celrep.2020.107943] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 05/20/2020] [Accepted: 06/30/2020] [Indexed: 12/31/2022] Open
Abstract
Nearly all biological processes rely on the finely tuned coordination of protein interactions across cellular space and time. Accordingly, generating protein interactomes has become routine in biological studies, yet interpreting these datasets remains computationally challenging. Here, we introduce Inter-ViSTA (Interaction Visualization in Space and Time Analysis), a web-based platform that quickly builds animated protein interaction networks and automatically synthesizes information on protein abundances, functions, complexes, and subcellular localizations. Using Inter-ViSTA with proteomics and molecular virology, we define virus-host interactions for the human cytomegalovirus (HCMV) anti-apoptotic protein, pUL37x1. We find that spatiotemporal controlled interactions underlie pUL37x1 functions, facilitating the pro-viral remodeling of mitochondria and peroxisomes during infection. Reciprocal isolations, microscopy, and genetic manipulations further characterize these associations, revealing the interplay between pUL37x1 and the MIB complex, which is critical for mitochondrial integrity. At the peroxisome, we show that pUL37x1 activates PEX11β to regulate fission, a key aspect of virus assembly and spread.
Collapse
|
172
|
Spears LD, Adak S, Dong G, Wei X, Spyropoulos G, Zhang Q, Yin L, Feng C, Hu D, Lodhi IJ, Hsu FF, Rajagopal R, Noguchi KK, Halabi CM, Brier L, Bice AR, Lananna BV, Musiek ES, Avraham O, Cavalli V, Holth JK, Holtzman DM, Wozniak DF, Culver JP, Semenkovich CF. Endothelial ether lipids link the vasculature to blood pressure, behavior, and neurodegeneration. J Lipid Res 2021; 62:100079. [PMID: 33894211 PMCID: PMC8144742 DOI: 10.1016/j.jlr.2021.100079] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/05/2021] [Accepted: 04/07/2021] [Indexed: 12/12/2022] Open
Abstract
Vascular disease contributes to neurodegeneration, which is associated with decreased blood pressure in older humans. Plasmalogens, ether phospholipids produced by peroxisomes, are decreased in Alzheimer's disease, Parkinson's disease, and other neurodegenerative disorders. However, the mechanistic links between ether phospholipids, blood pressure, and neurodegeneration are not fully understood. Here, we show that endothelium-derived ether phospholipids affect blood pressure, behavior, and neurodegeneration in mice. In young adult mice, inducible endothelial-specific disruption of PexRAP, a peroxisomal enzyme required for ether lipid synthesis, unexpectedly decreased circulating plasmalogens. PexRAP endothelial knockout (PEKO) mice responded normally to hindlimb ischemia but had lower blood pressure and increased plasma renin activity. In PEKO as compared with control mice, tyrosine hydroxylase was decreased in the locus coeruleus, which maintains blood pressure and arousal. PEKO mice moved less, slept more, and had impaired attention to and recall of environmental events as well as mild spatial memory deficits. In PEKO hippocampus, gliosis was increased, and a plasmalogen associated with memory was decreased. Despite lower blood pressure, PEKO mice had generally normal homotopic functional connectivity by optical neuroimaging of the cerebral cortex. Decreased glycogen synthase kinase-3 phosphorylation, a marker of neurodegeneration, was detected in PEKO cerebral cortex. In a co-culture system, PexRAP knockdown in brain endothelial cells decreased glycogen synthase kinase-3 phosphorylation in co-cultured astrocytes that was rescued by incubation with the ether lipid alkylglycerol. Taken together, our findings suggest that endothelium-derived ether lipids mediate several biological processes and may also confer neuroprotection in mice.
Collapse
Affiliation(s)
- Larry D Spears
- Division of Endocrinology, Metabolism & Lipid Research, Department of Medicine, Washington University, St. Louis, MO, USA
| | - Sangeeta Adak
- Division of Endocrinology, Metabolism & Lipid Research, Department of Medicine, Washington University, St. Louis, MO, USA
| | - Guifang Dong
- Division of Endocrinology, Metabolism & Lipid Research, Department of Medicine, Washington University, St. Louis, MO, USA; Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, China
| | - Xiaochao Wei
- Division of Endocrinology, Metabolism & Lipid Research, Department of Medicine, Washington University, St. Louis, MO, USA
| | | | - Qiang Zhang
- Division of Endocrinology, Metabolism & Lipid Research, Department of Medicine, Washington University, St. Louis, MO, USA
| | - Li Yin
- Division of Endocrinology, Metabolism & Lipid Research, Department of Medicine, Washington University, St. Louis, MO, USA
| | - Chu Feng
- Division of Endocrinology, Metabolism & Lipid Research, Department of Medicine, Washington University, St. Louis, MO, USA
| | - Donghua Hu
- Division of Endocrinology, Metabolism & Lipid Research, Department of Medicine, Washington University, St. Louis, MO, USA
| | - Irfan J Lodhi
- Division of Endocrinology, Metabolism & Lipid Research, Department of Medicine, Washington University, St. Louis, MO, USA
| | - Fong-Fu Hsu
- Division of Endocrinology, Metabolism & Lipid Research, Department of Medicine, Washington University, St. Louis, MO, USA
| | - Rithwick Rajagopal
- Department of Ophthalmology & Visual Sciences, Washington University, St. Louis, MO, USA
| | - Kevin K Noguchi
- Department of Psychiatry, Washington University, St. Louis, MO, USA
| | - Carmen M Halabi
- Department of Pediatrics, Washington University, St. Louis, MO, USA
| | - Lindsey Brier
- Department of Radiology, Washington University, St. Louis, MO, USA
| | - Annie R Bice
- Department of Radiology, Washington University, St. Louis, MO, USA
| | - Brian V Lananna
- Department of Neurology, Washington University, St. Louis, MO, USA
| | - Erik S Musiek
- Department of Neurology, Washington University, St. Louis, MO, USA
| | - Oshri Avraham
- Department of Neuroscience, Washington University, St. Louis, MO, USA
| | - Valeria Cavalli
- Department of Neuroscience, Washington University, St. Louis, MO, USA
| | - Jerrah K Holth
- Department of Neurology, Washington University, St. Louis, MO, USA
| | - David M Holtzman
- Department of Neurology, Washington University, St. Louis, MO, USA
| | - David F Wozniak
- Department of Psychiatry, Washington University, St. Louis, MO, USA
| | - Joseph P Culver
- Department of Radiology, Washington University, St. Louis, MO, USA
| | - Clay F Semenkovich
- Division of Endocrinology, Metabolism & Lipid Research, Department of Medicine, Washington University, St. Louis, MO, USA; Department of Cell Biology & Physiology, Washington University, St. Louis, MO, USA.
| |
Collapse
|
173
|
O'Hearn LA. Genetic variants for personalised management of very low carbohydrate ketogenic diets. BMJ Nutr Prev Health 2021; 4:354. [PMID: 34308146 PMCID: PMC8258056 DOI: 10.1136/bmjnph-2021-000282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 04/07/2021] [Indexed: 11/28/2022] Open
|
174
|
Cahill T, da Silveira WA, Renaud L, Williamson T, Wang H, Chung D, Overton I, Chan SSL, Hardiman G. Induced Torpor as a Countermeasure for Low Dose Radiation Exposure in a Zebrafish Model. Cells 2021; 10:906. [PMID: 33920039 PMCID: PMC8071006 DOI: 10.3390/cells10040906] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 04/07/2021] [Accepted: 04/11/2021] [Indexed: 12/15/2022] Open
Abstract
The development of the Artemis programme with the goal of returning to the moon is spurring technology advances that will eventually take humans to Mars and herald a new era of interplanetary space travel. However, long-term space travel poses unique challenges including exposure to ionising radiation from galactic cosmic rays and potential solar particle events, exposure to microgravity and specific nutritional challenges arising from earth independent exploration. Ionising radiation is one of the major obstacles facing future space travel as it can generate oxidative stress and directly damage cellular structures such as DNA, in turn causing genomic instability, telomere shortening, extracellular-matrix remodelling and persistent inflammation. In the gastrointestinal tract (GIT) this can lead to leaky gut syndrome, perforations and motility issues, which impact GIT functionality and affect nutritional status. While current countermeasures such as shielding from the spacecraft can attenuate harmful biological effects, they produce harmful secondary particles that contribute to radiation exposure. We hypothesised that induction of a torpor-like state would confer a radioprotective effect given the evidence that hibernation extends survival times in irradiated squirrels compared to active controls. To test this hypothesis, a torpor-like state was induced in zebrafish using melatonin treatment and reduced temperature, and radiation exposure was administered twice over the course of 10 days. The protective effects of induced-torpor were assessed via RNA sequencing and qPCR of mRNA extracted from the GIT. Pathway and network analysis were performed on the transcriptomic data to characterise the genomic signatures in radiation, torpor and torpor + radiation groups. Phenotypic analyses revealed that melatonin and reduced temperature successfully induced a torpor-like state in zebrafish as shown by decreased metabolism and activity levels. Genomic analyses indicated that low dose radiation caused DNA damage and oxidative stress triggering a stress response, including steroidal signalling and changes to metabolism, and cell cycle arrest. Torpor attenuated the stress response through an increase in pro-survival signals, reduced oxidative stress via the oxygen effect and detection and removal of misfolded proteins. This proof-of-concept model provides compelling initial evidence for utilizing an induced torpor-like state as a potential countermeasure for radiation exposure.
Collapse
Affiliation(s)
- Thomas Cahill
- School of Biological Sciences & Institute for Global Food Security, Queens University Belfast, Belfast BT9 5DL, UK; (T.C.); (W.A.d.S.); (H.W.)
| | - Willian Abraham da Silveira
- School of Biological Sciences & Institute for Global Food Security, Queens University Belfast, Belfast BT9 5DL, UK; (T.C.); (W.A.d.S.); (H.W.)
| | - Ludivine Renaud
- Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA;
| | - Tucker Williamson
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, SC 29425, USA; (T.W.); (S.S.L.C.)
| | - Hao Wang
- School of Biological Sciences & Institute for Global Food Security, Queens University Belfast, Belfast BT9 5DL, UK; (T.C.); (W.A.d.S.); (H.W.)
| | - Dongjun Chung
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH 43210, USA;
| | - Ian Overton
- Patrick G Johnston Centre for Cancer Research, Queen’s University Belfast, Belfast BT9 7AE, UK;
| | - Sherine S. L. Chan
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, SC 29425, USA; (T.W.); (S.S.L.C.)
| | - Gary Hardiman
- School of Biological Sciences & Institute for Global Food Security, Queens University Belfast, Belfast BT9 5DL, UK; (T.C.); (W.A.d.S.); (H.W.)
- Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA;
| |
Collapse
|
175
|
The Unity of Redox and Structural Remodeling of Brown Adipose Tissue in Hypothyroidism. Antioxidants (Basel) 2021; 10:antiox10040591. [PMID: 33921249 PMCID: PMC8068806 DOI: 10.3390/antiox10040591] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/30/2021] [Accepted: 04/07/2021] [Indexed: 12/20/2022] Open
Abstract
Brown adipose tissue (BAT) is important for maintaining whole-body metabolic and energy homeostasis. However, the effects of hypothyroidism, one of the most common diseases worldwide, which increases the risk of several metabolic disorders, on BAT redox and metabolic homeostasis remain mostly unknown. We aimed to investigate the dynamics of protein expression, enzyme activity, and localization of antioxidant defense (AD) enzymes in rat interscapular BAT upon induction of hypothyroidism by antithyroid drug methimazole for 7, 15, and 21 days. Our results showed an increased protein expression of CuZn- and Mn-superoxide dismutase, catalase, glutamyl-cysteine ligase, thioredoxin, total glutathione content, and activity of catalase and thioredoxin reductase in hypothyroid rats, compared to euthyroid control. Concomitant with the increase in AD, newly established nuclear, mitochondrial, and peroxisomal localization of AD enzymes was found. Hypothyroidism also potentiated associations between mitochondria, peroxisomes, and lipid bodies, creating specific structural-functional units. Moreover, hypothyroidism induced protein expression and nuclear translocation of a master regulator of redox-metabolic homeostasis, nuclear factor erythroid 2-related factor 2 (Nrf2), and an increased amount of 4-hydroxynonenal (4-HNE) protein adducts. The results indicate that spatiotemporal overlap in the remodeling of AD is orchestrated by Nrf2, implicating the role of 4-HNE in this process and suggesting the potential mechanism of redox-structural remodeling during BAT adaptation in hypothyroidism.
Collapse
|
176
|
Xu D, Lü Y, Li Y, Li S, Wang Z, Wang J. Ferroptosis Resistance in Cancer: An Emerging Crisis of New Hope. BIO INTEGRATION 2021. [DOI: 10.15212/bioi-2020-0039] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Ferroptosis, a new mode of nonapoptotic cell death, is increasingly recognized as a new hope in overcoming resistance to chemotherapy in cancer. Both canonical and noncanonical pathways can trigger ferroptosis execution via an iron-dependent lethal lipid peroxidation manner. However,
growing evidence has shown that some cancer cells can survive ferroptotic stress through metabolic remodeling as regards iron metabolism, anti-oxidative systems, and lipid metabolism. In addition to the well-known roles of the XC−/glutathione/glutathione peroxidase 4 (XC‐/GSH/GPX4)
axis in blocking ferroptosis, several recently identified pathways, including the Mevalonate-ferroptosis suppressor protein 1 (MVA-FSP1) axis, the GTP cyclohydrolase 1-Tetrahydrobiopterin (GCH1-BH4) axis, the peroxisome-ether-phospholipid axis, the acyl-CoA synthetase long-chain
family member 3-monounsaturated fatty acids (ACSL3-MUFA) axis, and the Liver kinase B1-AMP-activated protein kinase (LKB1-AMPK) axis, can negatively regulate susceptibility to ferroptosis. Prominin-2, a newly identified ferroptosis-modulating protein, also drives cancer cells to escape from
ferroptosis induction. These findings collectively led to major challenges and opportunities in the development of novel therapies that target the ferroptosis resistance of cancer cells.SignificanceFerroptosis is a lethal consequence of accumulated lipid peroxidation catalyzed
by ferrous iron and oxygen. This unique cell death process appears to involve many diseases, such as neurodegeneration, ischemia/reperfusion injury, kidney disease, and a druggable target in therapy-resistant cancers. There is great expectation of being able to exploit ferroptosis for the
treatment of as yet incurable diseases. However, the state of ferroptosis susceptibility is linked to various regulation pathways. This perspective aims to integrate the current understanding of signaling mechanisms for ferroptotic defenses and facilitates movement toward novel cancer therapeutic
strategies.
Collapse
Affiliation(s)
- Daiyun Xu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518100, China
| | - Yonghui Lü
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518100, China
| | - Yongxiao Li
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518100, China
| | - Shengbin Li
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518100, China
| | - Zhe Wang
- Department of Pathology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518033, China
| | - Junqing Wang
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518100, China
| |
Collapse
|
177
|
Liang X, Yang R, Yin N, Faiola F. Evaluation of the effects of low nanomolar bisphenol A-like compounds' levels on early human embryonic development and lipid metabolism with human embryonic stem cell in vitro differentiation models. JOURNAL OF HAZARDOUS MATERIALS 2021; 407:124387. [PMID: 33172680 DOI: 10.1016/j.jhazmat.2020.124387] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 10/21/2020] [Accepted: 10/23/2020] [Indexed: 06/11/2023]
Abstract
The widely used chemical bisphenol A (BPA) has been associated with several health effects. In recent years, many derivatives were developed to replace BPA although without thorough toxicological evaluation. Here, we employed a human embryoid body (EB)-based in vitro global differentiation and hepatic specification models, followed by RNA-seq analyses, to comprehensively study the potential developmental toxicity of six BPA replacements (BPS, BPF, BPZ, BPB, BPE, and BPAF), as compared to BPA. We found that those bisphenols may disrupt lineage commitment and lipid metabolism during early embryonic development. These effects mostly manifested via the dysregulation of HOX and APO family genes. Moreover, among the seven bisphenols analyzed, BPE seemed to have the mildest effects.
Collapse
Affiliation(s)
- Xiaoxing Liang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Renjun Yang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Nuoya Yin
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Francesco Faiola
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
178
|
Blignaut M, Harries S, Lochner A, Huisamen B. Ataxia Telangiectasia Mutated Protein Kinase: A Potential Master Puppeteer of Oxidative Stress-Induced Metabolic Recycling. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8850708. [PMID: 33868575 PMCID: PMC8032526 DOI: 10.1155/2021/8850708] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 02/15/2021] [Accepted: 02/28/2021] [Indexed: 02/07/2023]
Abstract
Ataxia Telangiectasia Mutated protein kinase (ATM) has recently come to the fore as a regulatory protein fulfilling many roles in the fine balancing act of metabolic homeostasis. Best known for its role as a transducer of DNA damage repair, the activity of ATM in the cytosol is enjoying increasing attention, where it plays a central role in general cellular recycling (macroautophagy) as well as the targeted clearance (selective autophagy) of damaged mitochondria and peroxisomes in response to oxidative stress, independently of the DNA damage response. The importance of ATM activation by oxidative stress has also recently been highlighted in the clearance of protein aggregates, where the expression of a functional ATM construct that cannot be activated by oxidative stress resulted in widespread accumulation of protein aggregates. This review will discuss the role of ATM in general autophagy, mitophagy, and pexophagy as well as aggrephagy and crosstalk between oxidative stress as an activator of ATM and its potential role as a master regulator of these processes.
Collapse
Affiliation(s)
- Marguerite Blignaut
- Centre for Cardio-Metabolic Research in Africa (CARMA), Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, South Africa
| | - Sarah Harries
- Centre for Cardio-Metabolic Research in Africa (CARMA), Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, South Africa
| | - Amanda Lochner
- Centre for Cardio-Metabolic Research in Africa (CARMA), Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, South Africa
| | - Barbara Huisamen
- Centre for Cardio-Metabolic Research in Africa (CARMA), Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, South Africa
| |
Collapse
|
179
|
Manzo T, Prentice BM, Anderson KG, Raman A, Schalck A, Codreanu GS, Nava Lauson CB, Tiberti S, Raimondi A, Jones MA, Reyzer M, Bates BM, Spraggins JM, Patterson NH, McLean JA, Rai K, Tacchetti C, Tucci S, Wargo JA, Rodighiero S, Clise-Dwyer K, Sherrod SD, Kim M, Navin NE, Caprioli RM, Greenberg PD, Draetta G, Nezi L. Accumulation of long-chain fatty acids in the tumor microenvironment drives dysfunction in intrapancreatic CD8+ T cells. J Exp Med 2021; 217:151833. [PMID: 32491160 PMCID: PMC7398173 DOI: 10.1084/jem.20191920] [Citation(s) in RCA: 164] [Impact Index Per Article: 54.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 01/14/2020] [Accepted: 03/19/2020] [Indexed: 12/13/2022] Open
Abstract
CD8+ T cells are master effectors of antitumor immunity, and their presence at tumor sites correlates with favorable outcomes. However, metabolic constraints imposed by the tumor microenvironment (TME) can dampen their ability to control tumor progression. We describe lipid accumulation in the TME areas of pancreatic ductal adenocarcinoma (PDA) populated by CD8+ T cells infiltrating both murine and human tumors. In this lipid-rich but otherwise nutrient-poor TME, access to using lipid metabolism becomes particularly valuable for sustaining cell functions. Here, we found that intrapancreatic CD8+ T cells progressively accumulate specific long-chain fatty acids (LCFAs), which, rather than provide a fuel source, impair their mitochondrial function and trigger major transcriptional reprogramming of pathways involved in lipid metabolism, with the subsequent reduction of fatty acid catabolism. In particular, intrapancreatic CD8+ T cells specifically exhibit down-regulation of the very-long-chain acyl-CoA dehydrogenase (VLCAD) enzyme, which exacerbates accumulation of LCFAs and very-long-chain fatty acids (VLCFAs) that mediate lipotoxicity. Metabolic reprogramming of tumor-specific T cells through enforced expression of ACADVL enabled enhanced intratumoral T cell survival and persistence in an engineered mouse model of PDA, overcoming one of the major hurdles to immunotherapy for PDA.
Collapse
Affiliation(s)
- Teresa Manzo
- Department of Experimental Oncology, IRCCS European Institute of Oncology, Milano, Italy.,Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Boone M Prentice
- Department of Biochemistry, Mass Spectrometry Research Center, Department of Chemistry, Department of Pharmacology and Medicine, Vanderbilt University, Nashville, TN
| | - Kristin G Anderson
- Clinical Research Division and Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA.,Departments of Medicine/Oncology and Immunology, University of Washington School of Medicine, Seattle, WA
| | - Ayush Raman
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Aislyn Schalck
- Department of Genetics and Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Carina B Nava Lauson
- Department of Experimental Oncology, IRCCS European Institute of Oncology, Milano, Italy
| | - Silvia Tiberti
- Department of Experimental Oncology, IRCCS European Institute of Oncology, Milano, Italy
| | - Andrea Raimondi
- Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, San Raffaele Vita-Salute University, Milano, Italy
| | - Marissa A Jones
- Department of Biochemistry, Mass Spectrometry Research Center, Department of Chemistry, Department of Pharmacology and Medicine, Vanderbilt University, Nashville, TN
| | - Michelle Reyzer
- Department of Biochemistry, Mass Spectrometry Research Center, Department of Chemistry, Department of Pharmacology and Medicine, Vanderbilt University, Nashville, TN
| | - Breanna M Bates
- Clinical Research Division and Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA.,Departments of Medicine/Oncology and Immunology, University of Washington School of Medicine, Seattle, WA
| | - Jeffrey M Spraggins
- Department of Biochemistry, Mass Spectrometry Research Center, Department of Chemistry, Department of Pharmacology and Medicine, Vanderbilt University, Nashville, TN
| | - Nathan H Patterson
- Department of Biochemistry, Mass Spectrometry Research Center, Department of Chemistry, Department of Pharmacology and Medicine, Vanderbilt University, Nashville, TN
| | - John A McLean
- Center for Innovative Technology, Vanderbilt University, Nashville, TN
| | - Kunal Rai
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Carlo Tacchetti
- Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, San Raffaele Vita-Salute University, Milano, Italy
| | - Sara Tucci
- Laboratory of Clinical Biochemistry and Metabolism Center for Pediatrics and Adolescent Medicine, University of Freiburg, Freiburg, Germany
| | - Jennifer A Wargo
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX.,Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Simona Rodighiero
- Department of Experimental Oncology, IRCCS European Institute of Oncology, Milano, Italy
| | - Karen Clise-Dwyer
- Department of Stem Cell Transplantation, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Stacy D Sherrod
- Center for Innovative Technology, Vanderbilt University, Nashville, TN
| | - Michael Kim
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Nicholas E Navin
- Department of Genetics and Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Richard M Caprioli
- Department of Biochemistry, Mass Spectrometry Research Center, Department of Chemistry, Department of Pharmacology and Medicine, Vanderbilt University, Nashville, TN
| | - Philip D Greenberg
- Clinical Research Division and Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA.,Departments of Medicine/Oncology and Immunology, University of Washington School of Medicine, Seattle, WA
| | - Giulio Draetta
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Luigi Nezi
- Department of Experimental Oncology, IRCCS European Institute of Oncology, Milano, Italy.,Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
180
|
Xiong Y, Chen Y, Huang X, Yang Z, Zhang J, Yu X, Fang J, Tao J, You K, Cheng Z, Tan S, Xu Y, Yuan F, Liu Y, Zhuang Y, Yang F, Li Y. Icaritin ameliorates hepatic steatosis via promoting fatty acid β-oxidation and insulin sensitivity. Life Sci 2021; 268:119000. [PMID: 33417961 DOI: 10.1016/j.lfs.2020.119000] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 12/18/2020] [Accepted: 12/26/2020] [Indexed: 11/28/2022]
Abstract
AIM This study aimed to reveal the effects of icaritin (ICT) on lipotoxicity induced by palmitate (PA) in hepatic cells and steatosis in high-fat diet (HFD)-fed mice as well as exploring the potential mechanisms. MAIN METHODS Primary mouse hepatocytes and human hepatoma Huh7 cells were used to evaluate ICT effect in vitro. HFD-fed mice were used to evaluate the ICT effect in vivo. RESULTS In vitro study indicated that ICT significantly rescued PA-induced steatosis, mainly through a combination of robust increased mitochondrial respiration, fatty acid oxidation and mildly decreased synthesis of fatty acid. An HFD-fed mouse model with 8 weeks HFD-fed showed metabolic disorders, while ICT application significantly reduced the weight, serum glucose levels, insulin resistance, hepatic steatosis level and adipose contents. In consistent with the observations in cell lines, ICT rescued the HFD-impaired functions and contents of key factors related to fatty acid β-oxidation through elevated expression of peroxisome proliferator-activated receptor α (PPARα). Meanwhile, it also reversed the decreased phosphoryl levels of AKT and glucogen synthase kinase 3 (GSK3β), leading to the improvement of insulin resistance. SIGNIFICANCE ICT administration had a therapeutic effect on PA- or HFD-induced hepatic steatosis and metabolic disorders. It may provide a novel strategy to construct preventive and therapeutic means for hepatic steatosis.
Collapse
Affiliation(s)
- Yue Xiong
- Institute of Public Health, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou 510530, China; University of Chinese Academy of Sciences (UCAS), Beijing 100049, China; Guangdong Provincial Key Laboratory of Biocomputing, GIBH, CAS, Guangzhou 510530, China
| | - Yan Chen
- Institute of Public Health, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou 510530, China; University of Chinese Academy of Sciences (UCAS), Beijing 100049, China; Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, GIBH, CAS, Guangzhou 510530, China; Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou, China
| | - Xinping Huang
- Institute of Public Health, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou 510530, China; University of Chinese Academy of Sciences (UCAS), Beijing 100049, China; Guangdong Provincial Key Laboratory of Biocomputing, GIBH, CAS, Guangzhou 510530, China
| | - Zhen Yang
- Institute of Public Health, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou 510530, China; University of Chinese Academy of Sciences (UCAS), Beijing 100049, China; Guangdong Provincial Key Laboratory of Biocomputing, GIBH, CAS, Guangzhou 510530, China
| | - Jiaye Zhang
- Institute of Public Health, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou 510530, China; University of Chinese Academy of Sciences (UCAS), Beijing 100049, China; Guangdong Provincial Key Laboratory of Biocomputing, GIBH, CAS, Guangzhou 510530, China
| | - Xiaorui Yu
- Institute of Public Health, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou 510530, China; University of Chinese Academy of Sciences (UCAS), Beijing 100049, China; Guangdong Provincial Key Laboratory of Biocomputing, GIBH, CAS, Guangzhou 510530, China
| | - Ji Fang
- Institute of Public Health, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou 510530, China; University of Chinese Academy of Sciences (UCAS), Beijing 100049, China; Guangdong Provincial Key Laboratory of Biocomputing, GIBH, CAS, Guangzhou 510530, China
| | - Jiawang Tao
- Institute of Public Health, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou 510530, China; University of Chinese Academy of Sciences (UCAS), Beijing 100049, China; Guangdong Provincial Key Laboratory of Biocomputing, GIBH, CAS, Guangzhou 510530, China
| | - Kai You
- Institute of Public Health, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou 510530, China; University of Chinese Academy of Sciences (UCAS), Beijing 100049, China; Guangdong Provincial Key Laboratory of Biocomputing, GIBH, CAS, Guangzhou 510530, China
| | - Ziqi Cheng
- Institute of Public Health, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou 510530, China; University of Chinese Academy of Sciences (UCAS), Beijing 100049, China; Guangdong Provincial Key Laboratory of Biocomputing, GIBH, CAS, Guangzhou 510530, China
| | - Shenglin Tan
- Institute of Public Health, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou 510530, China; University of Chinese Academy of Sciences (UCAS), Beijing 100049, China; Guangdong Provincial Key Laboratory of Biocomputing, GIBH, CAS, Guangzhou 510530, China
| | - Yingying Xu
- Institute of Public Health, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou 510530, China; University of Chinese Academy of Sciences (UCAS), Beijing 100049, China; Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, GIBH, CAS, Guangzhou 510530, China; Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou, China
| | - Fang Yuan
- Institute of Public Health, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou 510530, China; University of Chinese Academy of Sciences (UCAS), Beijing 100049, China; Guangdong Provincial Key Laboratory of Biocomputing, GIBH, CAS, Guangzhou 510530, China
| | - Ying Liu
- Institute of Public Health, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou 510530, China; University of Chinese Academy of Sciences (UCAS), Beijing 100049, China; Guangdong Provincial Key Laboratory of Biocomputing, GIBH, CAS, Guangzhou 510530, China
| | - Yuanqi Zhuang
- Institute of Public Health, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou 510530, China; University of Chinese Academy of Sciences (UCAS), Beijing 100049, China; Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, GIBH, CAS, Guangzhou 510530, China; Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou, China
| | - Fan Yang
- Institute of Public Health, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou 510530, China; University of Chinese Academy of Sciences (UCAS), Beijing 100049, China; Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, GIBH, CAS, Guangzhou 510530, China; Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou, China
| | - Yinxiong Li
- Institute of Public Health, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou 510530, China; University of Chinese Academy of Sciences (UCAS), Beijing 100049, China; Guangdong Provincial Key Laboratory of Biocomputing, GIBH, CAS, Guangzhou 510530, China; Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, GIBH, CAS, Guangzhou 510530, China; Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou, China; Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China.
| |
Collapse
|
181
|
Meeks L, De Oliveira Pessoa D, Martinez JA, Limesand KH, Padi M. Integration of metabolomics and transcriptomics reveals convergent pathways driving radiation-induced salivary gland dysfunction. Physiol Genomics 2021; 53:85-98. [PMID: 33522389 PMCID: PMC7988743 DOI: 10.1152/physiolgenomics.00127.2020] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 01/04/2021] [Accepted: 01/22/2021] [Indexed: 11/22/2022] Open
Abstract
Radiation therapy for head and neck cancer causes damage to the surrounding salivary glands, resulting in salivary gland hypofunction and xerostomia. Current treatments do not provide lasting restoration of salivary gland function following radiation; therefore, a new mechanistic understanding of the radiation-induced damage response is necessary for identifying therapeutic targets. The purpose of the present study was to investigate the metabolic phenotype of radiation-induced damage in parotid salivary glands by integrating transcriptomic and metabolomic data. Integrated data were then analyzed to identify significant gene-metabolite interactions. Mice received a single 5 Gy dose of targeted head and neck radiation. Parotid tissue samples were collected 5 days following treatment for RNA sequencing and metabolomics analysis. Altered metabolites and transcripts significantly converged on a specific region in the metabolic reaction network. Both integrative pathway enrichment using rank-based statistics and network analysis highlighted significantly coordinated changes in glutathione metabolism, energy metabolism (TCA cycle and thermogenesis), peroxisomal lipid metabolism, and bile acid production with radiation. Integrated changes observed in energy metabolism suggest that radiation induces a mitochondrial dysfunction phenotype. These findings validated previous pathways involved in the radiation-damage response, such as altered energy metabolism, and identified robust signatures in salivary glands, such as reduced glutathione metabolism, that may be driving salivary gland dysfunction.
Collapse
Affiliation(s)
- Lauren Meeks
- Department of Nutritional Sciences, University of Arizona, Tucson, Arizona
| | | | - Jessica A Martinez
- Department of Nutritional Sciences, University of Arizona, Tucson, Arizona
- University of Arizona Cancer Center, Tucson, Arizona
| | - Kirsten H Limesand
- Department of Nutritional Sciences, University of Arizona, Tucson, Arizona
- University of Arizona Cancer Center, Tucson, Arizona
| | - Megha Padi
- Bioinformatics Shared Resource, Arizona Cancer Center, University of Arizona, Tucson, Arizona
- University of Arizona Cancer Center, Tucson, Arizona
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, Arizona
| |
Collapse
|
182
|
Wang H, Liu H, Dai W, Luo S, Amos CI, Lee JE, Li X, Yue Y, Nan H, Wei Q. Association of genetic variants of TMEM135 and PEX5 in the peroxisome pathway with cutaneous melanoma-specific survival. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:396. [PMID: 33842617 PMCID: PMC8033299 DOI: 10.21037/atm-20-2117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Background Peroxisomes are ubiquitous and dynamic organelles that are involved in the metabolism of reactive oxygen species (ROS) and lipids. However, whether genetic variants in the peroxisome pathway genes are associated with survival in patients with melanoma has not been established. Therefore, our aim was to identify additional genetic variants in the peroxisome pathway that may provide new prognostic biomarkers for cutaneous melanoma (CM). Methods We assessed the associations between 8,397 common single-nucleotide polymorphisms (SNPs) in 88 peroxisome pathway genes and CM disease-specific survival (CMSS) in a two-stage analysis. For the discovery, we extracted the data from a published genome-wide association study from The University of Texas MD Anderson Cancer Center (MDACC). We then replicated the results in another dataset from the Nurse Health Study (NHS)/Health Professionals Follow-up Study (HPFS). Results Overall, 95 (11.1%) patients in the MDACC dataset and 48 (11.7%) patients in the NHS/HPFS dataset died of CM. We found 27 significant SNPs in the peroxisome pathway genes to be associated with CMSS in both datasets after multiple comparison correction using the Bayesian false-discovery probability method. In stepwise Cox proportional hazards regression analysis, with adjustment for other covariates and previously published SNPs in the MDACC dataset, we identified 2 independent SNPs (TMEM135 rs567403 C>G and PEX5 rs7969508 A>G) that predicted CMSS (P=0.003 and 0.031, respectively, in an additive genetic model). The expression quantitative trait loci analysis further revealed that the TMEM135 rs567403 GG and PEX5 rs7969508 GG genotypes were associated with increased and decreased levels of mRNA expression of their genes, respectively. Conclusions Once our findings are replicated by other investigators, these genetic variants may serve as novel biomarkers for the prediction of survival in patients with CM.
Collapse
Affiliation(s)
- Haijiao Wang
- Department of Gynecology Oncology, The First Hospital of Jilin University, Changchun, Jilin, China.,Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA.,Department of Population Health Sciences, Duke University School of Medicine, Durham, NC, USA
| | - Hongliang Liu
- Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA.,Department of Population Health Sciences, Duke University School of Medicine, Durham, NC, USA
| | - Wei Dai
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Sheng Luo
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, USA
| | - Christopher I Amos
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA
| | - Jeffrey E Lee
- Department of Surgical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - Xin Li
- Department of Epidemiology, Richard M. Fairbanks School of Public Health, Indiana University, Indianapolis, IN, USA
| | - Ying Yue
- Department of Gynecology Oncology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Hongmei Nan
- Department of Epidemiology, Richard M. Fairbanks School of Public Health, Indiana University, Indianapolis, IN, USA
| | - Qingyi Wei
- Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA.,Department of Population Health Sciences, Duke University School of Medicine, Durham, NC, USA.,Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
183
|
Tu K, Li J, Mo H, Xian Y, Xu Q, Xiao X. Identification and validation of redox-immune based prognostic signature for hepatocellular carcinoma. Int J Med Sci 2021; 18:2030-2041. [PMID: 33850474 PMCID: PMC8040390 DOI: 10.7150/ijms.56289] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Accepted: 02/24/2021] [Indexed: 01/03/2023] Open
Abstract
The intimate interaction between redox signaling and immunity has been widely revealed. However, the clinical application of relevant therapeutic is unavailable due to the absence of validated markers that stratify patients. Here, we identified novel biomarkers for prognosis prediction in hepatocellular carcinoma (HCC). Prognostic redox-immune-related genes for predicting overall survival (OS) of HCC were identified using datasets from TCGA, LIRI-JP, and GSE14520. LASSO Cox regression was employed to construct the signature model and generate a risk score in the TCGA cohort. The signature contained CDO1, G6PD, LDHA, GPD1L, PPARG, FABP4, CCL20, SPP1, RORC, HDAC1, STC2, HDGF, EPO, and IL18RAP. Patients in the high-risk group had a poor prognosis compared to the low-risk group. Univariate and multivariate Cox regressions identified this signature as an independent factor for predicting OS. Nomogram constructed by multiple clinical parameters showed good performance for predicting OS indicated by the c-index, the calibration curve, and AUC. GSEA showed that oxidoreductase activity and peroxisome-related metabolic pathways were enriched in the low-risk group, while glycolysis activity and hypoxia were higher in the high-risk group. Furthermore, immune profiles analysis showed that the immune score and stromal score were significantly decreased in the high-risk group in the TCGA cohort. There was a considerably lower infiltration of anti-tumor immune cells while a higher proportion of pro-tumor immune cells in silico. Immune markers were distinctly expressed between the subgroups, and redox-sensitive immunoregulatory biomarkers were at higher levels in the high-risk group. Altogether, we identified a redox-immune prognostic signature. A more severe redox perturbation-driven immunosuppressive environment in the high-risk group stratified by the signature may account for poor survival. This may provide a clue to the combined therapy targeting redox and immune in HCC.
Collapse
Affiliation(s)
- Kangsheng Tu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Jin Li
- Department of Shoulder and Elbow Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, China
| | - Huanye Mo
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Yao Xian
- Department of Nutrition, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Qiuran Xu
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou 310014, China
| | - Xuelian Xiao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| |
Collapse
|
184
|
Xu H, Zhong J, Zhuang W, Jiang J, Ma B, He H, Li G, Liao Y, Wang Y. A bifunctional mitochondrial targeting AIE-active fluorescent probe with high sensitivity to hydrogen peroxide and viscosity for fatty liver diagnosis. NEW J CHEM 2021. [DOI: 10.1039/d1nj01712h] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
A mitochondrial targeting AIE-active fluorescent probe with highly sensitive dual-detection of hydrogen peroxide and viscosity for fatty liver diagnosis.
Collapse
Affiliation(s)
- Hong Xu
- National Engineering Research Center for Biomaterials
- Sichuan University
- 29 Wangjiang Road
- Chengdu 610064
- China
| | - Jiehong Zhong
- Alltech Medical Systems
- LLC
- 201 Tianqin Road
- Chengdu 611731
- China
| | - Weihua Zhuang
- Department of Cardiology
- West China Hospital
- Sichuan University
- 37 Guoxue Street
- Chengdu 610041
| | - Jizhou Jiang
- College of Polymer Science and Engineering
- Sichuan University
- 29 Wangjiang Road
- Chengdu 610064
- China
| | - Boxuan Ma
- National Engineering Research Center for Biomaterials
- Sichuan University
- 29 Wangjiang Road
- Chengdu 610064
- China
| | - Haiyang He
- National Engineering Research Center for Biomaterials
- Sichuan University
- 29 Wangjiang Road
- Chengdu 610064
- China
| | - Gaocan Li
- National Engineering Research Center for Biomaterials
- Sichuan University
- 29 Wangjiang Road
- Chengdu 610064
- China
| | - Yanbiao Liao
- Department of Cardiology
- West China Hospital
- Sichuan University
- 37 Guoxue Street
- Chengdu 610041
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials
- Sichuan University
- 29 Wangjiang Road
- Chengdu 610064
- China
| |
Collapse
|
185
|
Torkzaban B, Mohseni Ahooyi T, Duggan M, Amini S, Khalili K. Cross-talk between lipid homeostasis and endoplasmic reticulum stress in neurodegeneration: Insights for HIV-1 associated neurocognitive disorders (HAND). Neurochem Int 2020; 141:104880. [PMID: 33065212 PMCID: PMC8208232 DOI: 10.1016/j.neuint.2020.104880] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 10/06/2020] [Accepted: 10/09/2020] [Indexed: 02/07/2023]
Abstract
The dysregulation of lipid homeostasis is emerging as a hallmark of many CNS diseases. As aberrant protein regulation is suggested to be a shared pathological feature amongst many neurodegenerative conditions, such as Alzheimer's disease (AD), Parkinson's disease (PD), and Huntington's disease (HD), disruptions in neuronal lipid processing may contribute to disease progression in the CNS. Specifically, given the endoplasmic reticulum (ER) dual role in lipid homeostasis as well as protein quality control (PQC) via unfolded protein response (UPR), lipid dysregulation in the CNS may converge on ER functioning and constitute a crucial mechanism underlying aberrant protein aggregation. In the current review, we discuss the diverse roles of lipid species as essential components of the CNS. Moreover, given the importance of both lipid dysregulation and protein aggregation in pathology of CNS diseases, we attempt to assess the potential downstream cross-talk between lipid dysregulation and ER dependent PQC mechanisms, with special focus on HIV-associated neurodegenerative disorders (HAND).
Collapse
Affiliation(s)
- Bahareh Torkzaban
- Department of Neuroscience, Center for Neurovirology, Lewis Katz School of Medicine at Temple University, 3500, N. Broad Street, Philadelphia, PA, USA
| | - Taha Mohseni Ahooyi
- Department of Neuroscience, Center for Neurovirology, Lewis Katz School of Medicine at Temple University, 3500, N. Broad Street, Philadelphia, PA, USA
| | - Michael Duggan
- Department of Neuroscience, Center for Neurovirology, Lewis Katz School of Medicine at Temple University, 3500, N. Broad Street, Philadelphia, PA, USA
| | - Shohreh Amini
- Department of Neuroscience, Center for Neurovirology, Lewis Katz School of Medicine at Temple University, 3500, N. Broad Street, Philadelphia, PA, USA
| | - Kamel Khalili
- Department of Neuroscience, Center for Neurovirology, Lewis Katz School of Medicine at Temple University, 3500, N. Broad Street, Philadelphia, PA, USA.
| |
Collapse
|
186
|
Losko M, Dolicka D, Pydyn N, Jankowska U, Kedracka-Krok S, Kulecka M, Paziewska A, Mikula M, Major P, Winiarski M, Budzynski A, Jura J. Integrative genomics reveal a role for MCPIP1 in adipogenesis and adipocyte metabolism. Cell Mol Life Sci 2020; 77:4899-4919. [PMID: 31893310 PMCID: PMC7658075 DOI: 10.1007/s00018-019-03434-5] [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] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 11/23/2019] [Accepted: 12/18/2019] [Indexed: 12/31/2022]
Abstract
Obesity is considered a serious chronic disease, associated with an increased risk of developing cardiovascular diseases, non-alcoholic fatty liver disease and type 2 diabetes. Monocyte chemoattractant protein-1-induced protein-1 (MCPIP1) is an RNase decreasing stability of transcripts coding for inflammation-related proteins. In addition, MCPIP1 plays an important role in the regulation of adipogenesis in vitro by reducing the expression of key transcription factors, including C/EBPβ. To elucidate the role of MCPIP1 in adipocyte biology, we performed RNA-Seq and proteome analysis in 3T3-L1 adipocytes overexpressing wild-type (WTMCPIP1) and the mutant form of MCPIP1 protein (D141NMCPIP1). Our RNA-Seq analysis followed by confirmatory Q-RT-PCR revealed that elevated MCPIP1 levels in 3T3-L1 adipocytes upregulated transcripts encoding proteins involved in signal transmission and cellular remodeling and downregulated transcripts of factors involved in metabolism. These data are consistent with our proteomic analysis, which showed that MCPIP1 expressing adipocytes exhibit upregulation of proteins involved in cellular organization and movement and decreased levels of proteins involved in lipid and carbohydrate metabolism. Moreover, MCPIP1 adipocytes are characterized by decreased level of insulin receptor, reduced insulin-induced Akt phosphorylation, as well as depleted Glut4 level and impaired glucose uptake. Overexpression of Glut4 in 3T3-L1 cells expressed WTMCPIP1 rescued adipogenesis. Interestingly, we found decreased level of MCPIP1 along with an increase in body mass index in subcutaneous adipose tissue. The presented data show a novel role of MCPIP1 in modulating insulin sensitivity in adipocytes. Overall, our findings demonstrate that MCPIP1 is an important regulator of adipogenesis and adipocyte metabolism.
Collapse
Affiliation(s)
- Magdalena Losko
- Department of General Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Dobrochna Dolicka
- Department of General Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Natalia Pydyn
- Department of General Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Urszula Jankowska
- Proteomics and Mass Spectrometry Laboratory, Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7A, 30-387, Kraków, Poland
| | - Sylwia Kedracka-Krok
- Department of Physical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Maria Kulecka
- Department of Gastroenterology, Hepatology and Clinical Oncology, Medical Center for Postgraduate Education, Marymoncka 99/103, 01-813, Warsaw, Poland
| | - Agnieszka Paziewska
- Department of Gastroenterology, Hepatology and Clinical Oncology, Medical Center for Postgraduate Education, Marymoncka 99/103, 01-813, Warsaw, Poland
| | - Michal Mikula
- Department of Genetics, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Wawelska 15B, 02-034, Warsaw, Poland
| | - Piotr Major
- Second Department of General Surgery, Centre for Research, Training and Innovation in Surgery (CERTAIN Surgery), Jagiellonian University Medical College, Kopernika 21, 31-501, Kraków, Poland
| | - Marek Winiarski
- Second Department of General Surgery, Centre for Research, Training and Innovation in Surgery (CERTAIN Surgery), Jagiellonian University Medical College, Kopernika 21, 31-501, Kraków, Poland
| | - Andrzej Budzynski
- Second Department of General Surgery, Centre for Research, Training and Innovation in Surgery (CERTAIN Surgery), Jagiellonian University Medical College, Kopernika 21, 31-501, Kraków, Poland
| | - Jolanta Jura
- Department of General Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland.
| |
Collapse
|
187
|
Covill-Cooke C, Toncheva VS, Kittler JT. Regulation of peroxisomal trafficking and distribution. Cell Mol Life Sci 2020; 78:1929-1941. [PMID: 33141311 PMCID: PMC7966214 DOI: 10.1007/s00018-020-03687-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 09/02/2020] [Accepted: 10/19/2020] [Indexed: 12/18/2022]
Abstract
Peroxisomes are organelles that perform a wide range of essential metabolic processes. To ensure that peroxisomes are optimally positioned in the cell, they must be transported by both long- and short-range trafficking events in response to cellular needs. Here, we review our current understanding of the mechanisms by which the cytoskeleton and organelle contact sites alter peroxisomal distribution. Though the focus of the review is peroxisomal transport in mammalian cells, findings from flies and fungi are used for comparison and to inform the gaps in our understanding. Attention is given to the apparent overlap in regulatory mechanisms for mitochondrial and peroxisomal trafficking, along with the recently discovered role of the mitochondrial Rho-GTPases, Miro, in peroxisomal dynamics. Moreover, we outline and discuss the known pathological and pharmacological conditions that perturb peroxisomal positioning. We conclude by highlighting several gaps in our current knowledge and suggest future directions that require attention.
Collapse
Affiliation(s)
| | - Viktoriya S Toncheva
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, WC1E 6BT, UK
| | - Josef T Kittler
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, WC1E 6BT, UK.
| |
Collapse
|
188
|
Shen S, Faouzi S, Souquere S, Roy S, Routier E, Libenciuc C, André F, Pierron G, Scoazec JY, Robert C. Melanoma Persister Cells Are Tolerant to BRAF/MEK Inhibitors via ACOX1-Mediated Fatty Acid Oxidation. Cell Rep 2020; 33:108421. [DOI: 10.1016/j.celrep.2020.108421] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 07/10/2020] [Accepted: 11/01/2020] [Indexed: 12/21/2022] Open
|
189
|
Chen Y, Wang X, Yang C, Liu Q, Ran Z, Li X, He C. A mouse model reveals the events and underlying regulatory signals during the gonadotrophin-dependent phase of follicle development. Mol Hum Reprod 2020; 26:920-937. [PMID: 33063120 DOI: 10.1093/molehr/gaaa069] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 09/07/2020] [Indexed: 12/13/2022] Open
Abstract
During folliculogenesis, the gonadotrophin (GTH)-dependent phase begins at the small antral follicle stage and ends with Graafian follicles. In this study, pregnant mare's serum GTH was used to induce GTH-dependent folliculogenesis in mice, following which the developmental events that follicles undergo, as well as the underlying regulatory signals, were investigated at both the morphological and transcriptomic level. GTH-dependent folliculogenesis consisted of three phases: preparation, rapid growth and decelerated growth. In the preparation phase, comprising the first 12 h, granulosa cells completed the preparations for proliferation and differentiation, shifted energy metabolism to glycolysis, and reduced protein synthesis and processing. The rapid growth phase lasted from 12 to 24 h; in this phase, granulosa cells completed their proliferation, and follicles acquired the capacity for estradiol secretion and ovulation. Meanwhile, the decelerating growth phase occurred between 24 and 48 h of GTH-dependent folliculogenesis. In this phase, the proliferation and expansion of the follicular antrum were reduced, energy metabolism was shifted to oxidative phosphorylation, and cell migration and lipid metabolism were enhanced in preparation for luteinization. We also revealed the key signaling pathways that regulate GTH-dependent folliculogenesis and elucidated the activation sequence of these pathways. A comparison of our RNA-sequencing data with that reported for humans suggested that the mechanisms involved in mouse and human folliculogenesis are evolutionarily conserved. In this study, we draw a detailed atlas of GTH-dependent folliculogenesis, thereby laying the foundation for further investigation of the regulatory mechanisms underlying this process.
Collapse
Affiliation(s)
- Yingjun Chen
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan 430070, Hubei, China.,National Center for International Research on Animal Genetics, Breeding and reproduction, Huazhong Agricultural University, Wuhan 430070, Hubei, China.,Department of Hubei Province Engineering Research Center in Buffalo Breeding and Products, Huazhong Agricultural University, Wuhan 430070, Hubei, China.,College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, Hubei, China
| | - Xiaodong Wang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan 430070, Hubei, China.,National Center for International Research on Animal Genetics, Breeding and reproduction, Huazhong Agricultural University, Wuhan 430070, Hubei, China.,Department of Hubei Province Engineering Research Center in Buffalo Breeding and Products, Huazhong Agricultural University, Wuhan 430070, Hubei, China.,College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, Hubei, China
| | - Chan Yang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan 430070, Hubei, China.,National Center for International Research on Animal Genetics, Breeding and reproduction, Huazhong Agricultural University, Wuhan 430070, Hubei, China.,Department of Hubei Province Engineering Research Center in Buffalo Breeding and Products, Huazhong Agricultural University, Wuhan 430070, Hubei, China.,College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, Hubei, China
| | - Qinghua Liu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan 430070, Hubei, China.,National Center for International Research on Animal Genetics, Breeding and reproduction, Huazhong Agricultural University, Wuhan 430070, Hubei, China.,Department of Hubei Province Engineering Research Center in Buffalo Breeding and Products, Huazhong Agricultural University, Wuhan 430070, Hubei, China.,College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, Hubei, China
| | - Zaohong Ran
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan 430070, Hubei, China.,National Center for International Research on Animal Genetics, Breeding and reproduction, Huazhong Agricultural University, Wuhan 430070, Hubei, China.,Department of Hubei Province Engineering Research Center in Buffalo Breeding and Products, Huazhong Agricultural University, Wuhan 430070, Hubei, China.,College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, Hubei, China
| | - Xiang Li
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan 430070, Hubei, China.,National Center for International Research on Animal Genetics, Breeding and reproduction, Huazhong Agricultural University, Wuhan 430070, Hubei, China.,Department of Hubei Province Engineering Research Center in Buffalo Breeding and Products, Huazhong Agricultural University, Wuhan 430070, Hubei, China.,College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, Hubei, China
| | - Changjiu He
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan 430070, Hubei, China.,National Center for International Research on Animal Genetics, Breeding and reproduction, Huazhong Agricultural University, Wuhan 430070, Hubei, China.,Department of Hubei Province Engineering Research Center in Buffalo Breeding and Products, Huazhong Agricultural University, Wuhan 430070, Hubei, China.,College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, Hubei, China
| |
Collapse
|
190
|
Zhou Y, Li P, Wang X, Wu C, Fan N, Liu X, Wu L, Zhang W, Zhang W, Liu Z, Tang B. In situ visualization of peroxisomal viscosity in the liver of mice with non-alcoholic fatty liver disease by near-infrared fluorescence and photoacoustic imaging. Chem Sci 2020; 11:12149-12156. [PMID: 34094429 PMCID: PMC8163019 DOI: 10.1039/d0sc02922j] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Accepted: 10/02/2020] [Indexed: 12/15/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) can gradually develop into hepatic failure, and early diagnosis is crucial to improve treatment efficiency. The occurrence of NAFLD is closely related to lipid metabolism. Peroxisomes act as the first and main site for lipid metabolism in the hepatocytes, so abnormal lipid metabolism might directly affect peroxisomal viscosity. Herein, we developed a new near-infrared fluorescence (NIRF) and photoacoustic (PA) imaging probe (PV-1) for the real-time visualization of peroxisomal viscosity in vivo. This PV-1 encompasses the malononitrile group as the rotor, which emits strong NIRF (at 705 nm) and PA (at 680 nm) signals when rotation is hindered as viscosity increases. Through dual-mode imaging, we discovered distinctly higher viscosity in the liver of NAFLD mice for the first time. We further found the remarkable amelioration of NAFLD upon treatment with N-acetylcysteine (NAC). Therefore, we anticipate that the PV-1 imaging method is promising for the early diagnosis and prognostic evaluation of NAFLD.
Collapse
Affiliation(s)
- Yongqing Zhou
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institutes of Biomedical Sciences, Shandong Normal University Jinan 250014 People's Republic of China
| | - Ping Li
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institutes of Biomedical Sciences, Shandong Normal University Jinan 250014 People's Republic of China
| | - Xin Wang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institutes of Biomedical Sciences, Shandong Normal University Jinan 250014 People's Republic of China
| | - Chuanchen Wu
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institutes of Biomedical Sciences, Shandong Normal University Jinan 250014 People's Republic of China
| | - Nannan Fan
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institutes of Biomedical Sciences, Shandong Normal University Jinan 250014 People's Republic of China
| | - Xiaoning Liu
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institutes of Biomedical Sciences, Shandong Normal University Jinan 250014 People's Republic of China
| | - Lijie Wu
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institutes of Biomedical Sciences, Shandong Normal University Jinan 250014 People's Republic of China
| | - Wei Zhang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institutes of Biomedical Sciences, Shandong Normal University Jinan 250014 People's Republic of China
| | - Wen Zhang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institutes of Biomedical Sciences, Shandong Normal University Jinan 250014 People's Republic of China
| | - Zhenzhen Liu
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institutes of Biomedical Sciences, Shandong Normal University Jinan 250014 People's Republic of China
| | - Bo Tang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institutes of Biomedical Sciences, Shandong Normal University Jinan 250014 People's Republic of China
| |
Collapse
|
191
|
Hyrina A, Jones C, Chen D, Clarkson S, Cochran N, Feucht P, Hoffman G, Lindeman A, Russ C, Sigoillot F, Tsang T, Uehara K, Xie L, Ganem D, Holdorf M. A Genome-wide CRISPR Screen Identifies ZCCHC14 as a Host Factor Required for Hepatitis B Surface Antigen Production. Cell Rep 2020; 29:2970-2978.e6. [PMID: 31801065 DOI: 10.1016/j.celrep.2019.10.113] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 06/23/2019] [Accepted: 10/28/2019] [Indexed: 12/24/2022] Open
Abstract
A hallmark of chronic hepatitis B (CHB) virus infection is the presence of high circulating levels of non-infectious small lipid HBV surface antigen (HBsAg) vesicles. Although rare, sustained HBsAg loss is the idealized endpoint of any CHB therapy. A small molecule, RG7834, has been previously reported to inhibit HBsAg expression by targeting terminal nucleotidyltransferase proteins 4A and 4B (TENT4A and TENT4B). In this study, we describe a genome-wide CRISPR screen to identify other potential host factors required for HBsAg expression and to gain further insights into the mechanism of RG7834. We report more than 60 genes involved in regulating HBsAg and identify additional factors involved in RG7834 activity, including a zinc finger CCHC-type containing 14 (ZCCHC14) protein. We show that ZCCHC14, together with TENT4A/B, stabilizes HBsAg expression through HBV RNA tailing, providing a potential new therapeutic target to achieve functional cure in CHB patients.
Collapse
Affiliation(s)
- Anastasia Hyrina
- Novartis Institutes for BioMedical Research, Emeryville, CA 94608, USA.
| | - Christopher Jones
- Novartis Institutes for BioMedical Research, Emeryville, CA 94608, USA
| | - Darlene Chen
- Novartis Institutes for BioMedical Research, Emeryville, CA 94608, USA
| | - Scott Clarkson
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Nadire Cochran
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Paul Feucht
- Novartis Institutes for BioMedical Research, Emeryville, CA 94608, USA
| | - Gregory Hoffman
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Alicia Lindeman
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Carsten Russ
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | | | - Tiffany Tsang
- Novartis Institutes for BioMedical Research, Emeryville, CA 94608, USA
| | - Kyoko Uehara
- Novartis Institutes for BioMedical Research, Emeryville, CA 94608, USA
| | - Lili Xie
- Novartis Institutes for BioMedical Research, Emeryville, CA 94608, USA
| | - Don Ganem
- Novartis Institutes for BioMedical Research, Emeryville, CA 94608, USA
| | - Meghan Holdorf
- Novartis Institutes for BioMedical Research, Emeryville, CA 94608, USA
| |
Collapse
|
192
|
Dysregulation of metabolic pathways by carnitine palmitoyl-transferase 1 plays a key role in central nervous system disorders: experimental evidence based on animal models. Sci Rep 2020; 10:15583. [PMID: 32973137 PMCID: PMC7519132 DOI: 10.1038/s41598-020-72638-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 08/26/2020] [Indexed: 02/06/2023] Open
Abstract
The etiology of CNS diseases including multiple sclerosis, Parkinson’s disease and amyotrophic lateral sclerosis remains elusive despite decades of research resulting in treatments with only symptomatic effects. In this study, we provide evidence that a metabolic shift from glucose to lipid is a key mechanism in neurodegeneration. We show that, by downregulating the metabolism of lipids through the key molecule carnitine palmitoyl transferase 1 (CPT1), it is possible to reverse or slowdown disease progression in experimental models of autoimmune encephalomyelitis-, SOD1G93A and rotenone models, mimicking these CNS diseases in humans. The effect was seen both when applying a CPT1 blocker or by using a Cpt1a P479L mutant mouse strain. Furthermore, we show that diet, epigenetics, and microbiota are key elements in this metabolic shift. Finally, we present a systemic model for understanding the complex etiology of neurodegeneration and how different regulatory systems are interconnected through a central metabolic pathway that becomes deregulated under specific conditions.
Collapse
|
193
|
Machado-Oliveira G, Ramos C, Marques ARA, Vieira OV. Cell Senescence, Multiple Organelle Dysfunction and Atherosclerosis. Cells 2020; 9:E2146. [PMID: 32977446 PMCID: PMC7598292 DOI: 10.3390/cells9102146] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/19/2020] [Accepted: 09/20/2020] [Indexed: 01/10/2023] Open
Abstract
Atherosclerosis is an age-related disorder associated with long-term exposure to cardiovascular risk factors. The asymptomatic progression of atherosclerotic plaques leads to major cardiovascular diseases (CVD), including acute myocardial infarctions or cerebral ischemic strokes in some cases. Senescence, a biological process associated with progressive structural and functional deterioration of cells, tissues and organs, is intricately linked to age-related diseases. Cell senescence involves coordinated modifications in cellular compartments and has been demonstrated to contribute to different stages of atheroma development. Senescence-based therapeutic strategies are currently being pursued to treat and prevent CVD in humans in the near-future. In addition, distinct experimental settings allowed researchers to unravel potential approaches to regulate anti-apoptotic pathways, facilitate excessive senescent cell clearance and eventually reverse atherogenesis to improve cardiovascular function. However, a deeper knowledge is required to fully understand cellular senescence, to clarify senescence and atherogenesis intertwining, allowing researchers to establish more effective treatments and to reduce the cardiovascular disorders' burden. Here, we present an objective review of the key senescence-related alterations of the major intracellular organelles and analyze the role of relevant cell types for senescence and atherogenesis. In this context, we provide an updated analysis of therapeutic approaches, including clinically relevant experiments using senolytic drugs to counteract atherosclerosis.
Collapse
Affiliation(s)
- Gisela Machado-Oliveira
- CEDOC, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal; (C.R.); (A.R.A.M.)
| | | | | | - Otília V. Vieira
- CEDOC, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal; (C.R.); (A.R.A.M.)
| |
Collapse
|
194
|
Plasticity of ether lipids promotes ferroptosis susceptibility and evasion. Nature 2020; 585:603-608. [PMID: 32939090 DOI: 10.1038/s41586-020-2732-8] [Citation(s) in RCA: 435] [Impact Index Per Article: 108.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 06/24/2020] [Indexed: 12/25/2022]
Abstract
Ferroptosis-an iron-dependent, non-apoptotic cell death process-is involved in various degenerative diseases and represents a targetable susceptibility in certain cancers1. The ferroptosis-susceptible cell state can either pre-exist in cells that arise from certain lineages or be acquired during cell-state transitions2-5. However, precisely how susceptibility to ferroptosis is dynamically regulated remains poorly understood. Here we use genome-wide CRISPR-Cas9 suppressor screens to identify the oxidative organelles peroxisomes as critical contributors to ferroptosis sensitivity in human renal and ovarian carcinoma cells. Using lipidomic profiling we show that peroxisomes contribute to ferroptosis by synthesizing polyunsaturated ether phospholipids (PUFA-ePLs), which act as substrates for lipid peroxidation that, in turn, results in the induction of ferroptosis. Carcinoma cells that are initially sensitive to ferroptosis can switch to a ferroptosis-resistant state in vivo in mice, which is associated with extensive downregulation of PUFA-ePLs. We further find that the pro-ferroptotic role of PUFA-ePLs can be extended beyond neoplastic cells to other cell types, including neurons and cardiomyocytes. Together, our work reveals roles for the peroxisome-ether-phospholipid axis in driving susceptibility to and evasion from ferroptosis, highlights PUFA-ePL as a distinct functional lipid class that is dynamically regulated during cell-state transitions, and suggests multiple regulatory nodes for therapeutic interventions in diseases that involve ferroptosis.
Collapse
|
195
|
Xu L, Yang Y, Chen J. The role of reactive oxygen species in cognitive impairment associated with sleep apnea. Exp Ther Med 2020; 20:4. [PMID: 32934669 PMCID: PMC7471880 DOI: 10.3892/etm.2020.9132] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 02/07/2020] [Indexed: 02/07/2023] Open
Abstract
Obstructive sleep apnea (OSA), a common breathing and sleeping disorder, is associated with a broad range of neurocognitive difficulties. Intermittent hypoxia (IH), one major characteristic of OSA, has been shown to impair learning and memory due to increased levels of reactive oxygen species (ROS). Under normal conditions, ROS are produced in low concentrations and act as signaling molecules in different processes. However, IH treatment leads to elevated ROS production via multiple pathways, including mitochondrial electron transport chain dysfunction and in particular complex I dysfunction, and induces oxidative tissue damage. Moreover, elevated ROS results in the accumulation of unfolded or misfolded proteins in the endoplasmic reticulum (ER) and increased activity of peroxisomes, such as NADPH oxidase, xanthine oxidase and phospholipase A2. Furthermore, oxidative tissue damage has been found in regions of the brains of patients with OSA, including the cortex and hippocampus, which are associated with memory and executive function. Furthermore, increased ROS levels in these regions of the brain induce damage via inflammation, apoptosis, ER stress and neuronal activity disturbance. The present review focuses on the mechanism of excessive ROS production in an OSA model and the relationship between ROS and cognitive impairment.
Collapse
Affiliation(s)
- Linhao Xu
- Department of Cardiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, P.R. China.,Department of Pathology, School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang 310053, P.R. China.,Translational Medicine Research Center, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, P.R. China
| | - Yibo Yang
- College of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai 201424, P.R. China
| | - Jian Chen
- Department of Pathology, School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang 310053, P.R. China
| |
Collapse
|
196
|
Increased Accumulation of Medium-Chain Fatty Acids by Dynamic Degradation of Long-Chain Fatty Acids in Mucor circinelloides. Genes (Basel) 2020; 11:genes11080890. [PMID: 32764225 PMCID: PMC7464202 DOI: 10.3390/genes11080890] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 07/22/2020] [Accepted: 07/27/2020] [Indexed: 12/12/2022] Open
Abstract
Concerns about global warming, fossil-fuel depletion, food security, and human health have promoted metabolic engineers to develop tools/strategies to overproduce microbial functional oils directly from renewable resources. Medium-chain fatty acids (MCFAs, C8–C12) have been shown to be important sources due to their diverse biotechnological importance, providing benefits ranging from functional lipids to uses in bio-fuel production. However, oleaginous microbes do not carry native pathways for the production of MCFAs, and therefore, diverse approaches have been adapted to compensate for the requirements of industrial demand. Mucor circinelloides is a promising organism for lipid production (15–36% cell dry weight; CDW) and the investigation of mechanisms of lipid accumulation; however, it mostly produces long-chain fatty acids (LCFAs). To address this challenge, we genetically modified strain M. circinelloides MU758, first by integrating heterologous acyl-ACP thioesterase (TE) into fatty acid synthase (FAS) complex and subsequently by modifying the β-oxidation pathway by disrupting the acyl-CoA oxidase (ACOX) and/or acyl-CoA thioesterase (ACOT) genes with a preference for medium-chain acyl-CoAs, to elevate the yield of MCFAs. The resultant mutant strains (M-1, M-2, and M-3, respectively) showed a significant increase in lipid production in comparison to the wild-type strain (WT). MCFAs in M-1 (47.45%) was sharply increased compared to the wild type strain (2.25%), and it was further increased in M-2 (60.09%) suggesting a negative role of ACOX in MCFAs production. However, MCFAs in M-3 were much decreased compared to M-1,suggesting a positive role of ACOT in MCFAs production. The M-2 strain showed maximum lipid productivity (~1800 milligram per liter per day or mg/L.d) and MCFAs productivity (~1100 mg/L.d). Taken together, this study elaborates on how the combination of two multidimensional approaches, TE gene over-expression and modification of the β-oxidation pathway via substantial knockout of specific ACOX gene, significantly increased the production of MCFAs. This synergistic approach ultimately offers a novel opportunity for synthetic/industrial biologists to increase the content of MCFAs.
Collapse
|
197
|
Capitão A, Lopes-Marques M, Páscoa I, Ruivo R, Mendiratta N, Fonseca E, Castro LFC, Santos MM. The Echinodermata PPAR: Functional characterization and exploitation by the model lipid homeostasis regulator tributyltin. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 263:114467. [PMID: 32278212 DOI: 10.1016/j.envpol.2020.114467] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 03/23/2020] [Accepted: 03/25/2020] [Indexed: 05/06/2023]
Abstract
The wide ecological relevance of lipid homeostasis modulators in the environment has been increasingly acknowledged. Tributyltin (TBT), for instance, was shown to cause lipid modulation, not only in mammals, but also in fish, molluscs, arthropods and rotifers. In vertebrates, TBT is known to interact with a nuclear receptor heterodimer module, formed by the retinoid X receptor (RXR) and the peroxisome proliferator-activated receptor (PPAR). These modulate the expression of genes involved in lipid homeostasis. In the present work, we isolated for the first time the complete coding region of the Echinodermata (Paracentrotus lividus) gene orthologues of PPAR and RXR and evaluated the ability of a model lipid homeostasis modulator, TBT, to interfere with the lipid metabolism in this species. Our results demonstrate that TBT alters the gonadal fatty acid composition and gene expression patterns: yielding sex-specific responses in fatty acid levels, including the decrease of eicosapentaenoic acid (C20:5 n-3, EPA) in males, and increase of arachidonic acid (20:4n-6, ARA) in females, and upregulation of long-chain acyl-CoA synthetase (acsl), ppar and rxr. Furthermore, an in vitro test using COS-1 cells as host and chimeric receptors with the ligand binding domain (LBD) of P. lividus PPAR and RXR shows that organotins (TBT and TPT (Triphenyltin)) suppressed activity of the heterodimer PPAR/RXR in a concentration-dependent manner. Together, these results suggest that TBT acts as a lipid homeostasis modulator at environmentally relevant concentrations in Echinodermata and highlight a possible conserved mode of action via the PPAR/RXR heterodimer.
Collapse
Affiliation(s)
- Ana Capitão
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto (U.Porto), Matosinhos, Portugal; Faculty of Sciences (FCUP), Department of Biology, University of Porto (U.Porto), Porto, Portugal
| | - Mónica Lopes-Marques
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto (U.Porto), Matosinhos, Portugal
| | - Inês Páscoa
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto (U.Porto), Matosinhos, Portugal
| | - Raquel Ruivo
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto (U.Porto), Matosinhos, Portugal
| | - Nicolau Mendiratta
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto (U.Porto), Matosinhos, Portugal
| | - Elza Fonseca
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto (U.Porto), Matosinhos, Portugal; Faculty of Sciences (FCUP), Department of Biology, University of Porto (U.Porto), Porto, Portugal
| | - L Filipe C Castro
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto (U.Porto), Matosinhos, Portugal; Faculty of Sciences (FCUP), Department of Biology, University of Porto (U.Porto), Porto, Portugal.
| | - Miguel Machado Santos
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto (U.Porto), Matosinhos, Portugal; Faculty of Sciences (FCUP), Department of Biology, University of Porto (U.Porto), Porto, Portugal.
| |
Collapse
|
198
|
A high fat diet with a high C18:0/C16:0 ratio induced worse metabolic and transcriptomic profiles in C57BL/6 mice. Lipids Health Dis 2020; 19:172. [PMID: 32693810 PMCID: PMC7372854 DOI: 10.1186/s12944-020-01346-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 07/09/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Differential effects of individual saturated fatty acids (SFAs), particularly stearic acid (C18:0), relative to the shorter-chain SFAs have drawn interest for more accurate nutritional guidelines. However, specific biologic and pathologic functions that can be assigned to particular SFAs are very limited. The present study was designed to compare changes in metabolic and transcriptomic profiles in mice caused by a high C18:0 diet and high palmitic acid (C16:0) diet. METHODS Male C57BL/6 mice were assigned to a normal fat diet (NFD), a high fat diet with high C18:0/C16:0 ratio (HSF) or an isocaloric high fat diet with a low C18:0/C16:0 ratio (LSF) for 10 weeks. An oral glucose tolerance test, 72-h energy expenditure measurement and CT scan of body fat were done before sacrifice. Fasting glucose and lipids were determined by an autobiochemical analyzer. Blood insulin, tumor necrosis factor-α (TNF-α), and interleukin-6 (IL-6) levels were measured by enzyme-linked immunosorbent assay methods. Free fatty acids (FFAs) profiles in blood and liver were determined by using gas chromatography-mass spectrometry. Microarray analysis was applied to investigate changes in transcriptomic profiles in the liver. Pathway analysis and gene ontology analysis were applied to describe the roles of differentially expressed mRNAs. RESULTS Compared with the NFD group, body weight, body fat ratio, fasting blood glucose, insulin, homeostasis model assessment of insulin resistance (HOMA-IR), triglyceride, IL-6, serum and liver FFAs including total FFAs, C16:0 and C18:0 were increased in both high fat diet groups and were much higher in the HSF group than those in the LSF group. Both HSF and LSF mice exhibited distinguishable long non-coding RNA (lncRNA), microRNA and mRNA expression profiles when compared with those of NFD mice. Additionally, more differentially expressed lncRNAs and mRNAs were observed in the HSF group than in the LSF group. Some biological functions and pathways, other than energy metabolism regulation, were identified as differentially expressed mRNAs between the HSF group and the LSF group. CONCLUSION The high fat diet with a high C18:0/C16:0 ratio induced more severe glucose and lipid metabolic disorders and inflammation and affected expression of more lncRNAs and mRNAs than an isocaloric low C18:0/C16:0 ratio diet in mice. These results provide new insights into the differences in biological functions and related mechanisms, other than glucose and lipid metabolism, between C16:0 and C18:0.
Collapse
|
199
|
Abstract
Peroxisomes are metabolic organelles involved in lipid metabolism and cellular redox balance. Peroxisomal function is central to fatty acid oxidation, ether phospholipid synthesis, bile acid synthesis, and reactive oxygen species homeostasis. Human disorders caused by genetic mutations in peroxisome genes have led to extensive studies on peroxisome biology. Peroxisomal defects are linked to metabolic dysregulation in diverse human diseases, such as neurodegeneration and age-related disorders, revealing the significance of peroxisome metabolism in human health. Cancer is a disease with metabolic aberrations. Despite the critical role of peroxisomes in cell metabolism, the functional effects of peroxisomes in cancer are not as well recognized as those of other metabolic organelles, such as mitochondria. In addition, the significance of peroxisomes in cancer is less appreciated than it is in degenerative diseases. In this review, I summarize the metabolic pathways in peroxisomes and the dysregulation of peroxisome metabolism in cancer. In addition, I discuss the potential of inactivating peroxisomes to target cancer metabolism, which may pave the way for more effective cancer treatment.
Collapse
|
200
|
Wu A, Wojtowicz K, Savary S, Hamon Y, Trombik T. Do ABC transporters regulate plasma membrane organization? Cell Mol Biol Lett 2020; 25:37. [PMID: 32647530 PMCID: PMC7336681 DOI: 10.1186/s11658-020-00224-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 05/05/2020] [Indexed: 12/29/2022] Open
Abstract
The plasma membrane (PM) spatiotemporal organization is one of the major factors controlling cell signaling and whole-cell homeostasis. The PM lipids, including cholesterol, determine the physicochemical properties of the membrane bilayer and thus play a crucial role in all membrane-dependent cellular processes. It is known that lipid content and distribution in the PM are not random, and their transversal and lateral organization is highly controlled. Mainly sphingolipid- and cholesterol-rich lipid nanodomains, historically referred to as rafts, are extremely dynamic “hot spots” of the PM controlling the function of many cell surface proteins and receptors. In the first part of this review, we will focus on the recent advances of PM investigation and the current PM concept. In the second part, we will discuss the importance of several classes of ABC transporters whose substrates are lipids for the PM organization and dynamics. Finally, we will briefly present the significance of lipid ABC transporters for immune responses.
Collapse
Affiliation(s)
- Ambroise Wu
- Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| | | | - Stephane Savary
- Lab. Bio-PeroxIL EA7270, University of Bourgogne Franche-Comté, Dijon, France
| | - Yannick Hamon
- Aix Marseille University, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Tomasz Trombik
- Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| |
Collapse
|