1
|
Jeeva P, Muthusamy A, Kesavan swaminathan J. Deciphering Structural Dynamics of Atherosclerosis Proteins: Insights from Crataegus oxyacantha Phytochemicals that Interceded Functional and Structural Changes in Targeted Atherosclerotic Proteins. ACS OMEGA 2024; 9:48159-48172. [PMID: 39676950 PMCID: PMC11635474 DOI: 10.1021/acsomega.4c04975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 09/18/2024] [Accepted: 09/24/2024] [Indexed: 12/17/2024]
Abstract
Atherosclerosis (ASC) is characterized by foam cell-mediated plaque formation, vascular endothelial inflammation, and lipidosis and is the rudimentary cause of cardiovascular diseases. This is the pre-eminent global factor of mortality. This etiological paradigm is significantly influenced by several proteins, where 23 pivotal proteins involved in ASC were meticulously gleaned on the basis of literature studies. The crux of the present study was aimed to probe the drugability of four active phytochemicals from Crataegus oxyacantha (COC): epicatechin, gallate, tyramine, and vitexin against the selected 23 proteins. The molecular docking analysis was judiciously administered via Glide, the binding free energy was calculated in detail utilizing the prime molecular mechanics-generalized Born surface area (MM-GBSA) module, and a deeper comprehensive investigation of protein-ligand dynamic associations was elucidated through Desmond. Drawing from the upper echelons of our docking results, the molecular dynamics simulation outcomes revealed that the macrophage migration inhibitory factor and prethrombin-1 showed persistent binding nature with gallate. The bioactive compound known as gallate sourced from COC shows the best molecular association with pivotal proteins involved in ASC and has a promising therapeutic potential for drug development endeavors.
Collapse
Affiliation(s)
- Praveen Jeeva
- Department
of Bioinformatics, Bharathidasan University, Tiruchirappalli, Tamilnadu 620024, India
| | - Anusuyadevi Muthusamy
- Department
of Biochemistry, Bharathidasan University, Tiruchirappalli, Tamilnadu 620024, India
| | | |
Collapse
|
2
|
Johnson AR, Rao K, Zhang BB, Mullet S, Goetzman E, Gelhaus S, Tejero J, Shiva S. Myoglobin inhibits breast cancer cell fatty acid oxidation and migration via heme-dependent oxidant production and not fatty acid binding. Free Radic Biol Med 2024; 225:208-220. [PMID: 39368517 DOI: 10.1016/j.freeradbiomed.2024.10.258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 09/17/2024] [Accepted: 10/02/2024] [Indexed: 10/07/2024]
Abstract
The monomeric heme protein myoglobin (Mb) is aberrantly expressed in approximately 40 % of breast tumors. Mb expression is associated with better patient prognosis, yet the molecular mechanisms underlying this effect are unclear. In muscle, Mb's heme moiety confers oxygen storage and delivery. However, prior studies demonstrate that low levels of Mb in cancer cells preclude this function. Several studies propose a fatty acid binding function for Mb via lysine residue K46. Because cancer cells can upregulate fatty acid oxidation (FAO) to fuel cell migration, we tested whether Mb-mediated fatty acid binding modulates FAO and migration. We demonstrate that stable expression of human Mb in MDA-MB-231 breast cancer cells decreases cell migration and FAO. Site-directed mutagenesis of Mb K46 disrupted fatty acid binding but did not improve FAO or migration. Conversely, cells expressing Apo-Mb (with disrupted heme binding) did not show impaired FAO or migration rates, suggesting Mb attenuates FAO and migration via a heme-dependent mechanism rather than through fatty acid binding. Mb's heme-dependent oxidant generation dysregulates migratory gene expression, which is reversed by catalase treatment. Collectively, these data demonstrate that Mb's heme-dependent oxidant production decreases breast cancer cell migration, prompting therapeutic strategies to modulate oxidant production and Mb in tumors.
Collapse
Affiliation(s)
- Aaron R Johnson
- Heart, Lung, Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Krithika Rao
- Heart, Lung, Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Bob B Zhang
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Steven Mullet
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Eric Goetzman
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Stacy Gelhaus
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Jesus Tejero
- Heart, Lung, Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, 15261, USA; Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Sruti Shiva
- Heart, Lung, Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, 15261, USA; Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA, 15261, USA.
| |
Collapse
|
3
|
Madhurantakam S, David BE, Naqvi A, Lee ZJ, Abraham JT, Vankamamidi TS, Prasad S. Advancements in electrochemical immunosensors towards point-of-care detection of cardiac biomarkers. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2024; 16:6615-6633. [PMID: 39114951 DOI: 10.1039/d4ay01049c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/11/2024]
Abstract
Cardiovascular disease remains the leading cause of death worldwide, with mortality rates increasing annually. This underscores the urgent need for accurate diagnostic and monitoring tools. Electrochemical detection has emerged as a promising method for swiftly and precisely measuring specific biomarkers in bodily fluids. This approach is not only cost-effective and efficient compared to traditional clinical methods, but it can also be tailored to detect individual biomarkers, which makes it particularly well-suited for point-of-care (POC) applications. The ability to conduct testing at the point of care is crucial for timely interventions and personalized disease management, empowering healthcare providers to tailor treatment plans based on real-time biomarker data. Thanks to recent advancements in nanomaterials, we've seen significant progress in electrochemical detection, leading to the development of specialized rapid immunoassay systems. These systems utilize specific antibodies to target molecules, expanding the range of detectable biomarkers. This innovation has the potential to revolutionize the diagnosis and treatment of cardiovascular diseases by enhancing detection sensitivity and specificity. Ultimately, these advancements aim to improve patient outcomes by enabling earlier diagnosis, more precise monitoring, and personalized therapeutic interventions, which will contribute to more effective management of cardiovascular health globally.
Collapse
Affiliation(s)
- Sasya Madhurantakam
- Department of Bioengineering, University of Texas at Dallas, 800W Campbell Rd, Richardson, Texas 75080, USA.
| | - Bianca Elizabeth David
- Department of Bioengineering, University of Texas at Dallas, 800W Campbell Rd, Richardson, Texas 75080, USA.
| | - Aliya Naqvi
- Department of Bioengineering, University of Texas at Dallas, 800W Campbell Rd, Richardson, Texas 75080, USA.
| | - Zachary J Lee
- Department of Bioengineering, University of Texas at Dallas, 800W Campbell Rd, Richardson, Texas 75080, USA.
| | - Jacob Thomas Abraham
- Department of Bioengineering, University of Texas at Dallas, 800W Campbell Rd, Richardson, Texas 75080, USA.
| | - Trayi Sai Vankamamidi
- Department of Bioengineering, University of Texas at Dallas, 800W Campbell Rd, Richardson, Texas 75080, USA.
| | - Shalini Prasad
- Department of Bioengineering, University of Texas at Dallas, 800W Campbell Rd, Richardson, Texas 75080, USA.
| |
Collapse
|
4
|
Adepu KK, Anishkin A, Adams SH, Chintapalli SV. A versatile delivery vehicle for cellular oxygen and fuels or metabolic sensor? A review and perspective on the functions of myoglobin. Physiol Rev 2024; 104:1611-1642. [PMID: 38696337 PMCID: PMC11495214 DOI: 10.1152/physrev.00031.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 04/30/2024] [Accepted: 05/01/2024] [Indexed: 05/04/2024] Open
Abstract
A canonical view of the primary physiological function of myoglobin (Mb) is that it is an oxygen (O2) storage protein supporting mitochondrial oxidative phosphorylation, especially as the tissue O2 partial pressure (Po2) drops and Mb off-loads O2. Besides O2 storage/transport, recent findings support functions for Mb in lipid trafficking and sequestration, interacting with cellular glycolytic metabolites such as lactate (LAC) and pyruvate (PYR), and "ectopic" expression in some types of cancer cells and in brown adipose tissue (BAT). Data from Mb knockout (Mb-/-) mice and biochemical models suggest additional metabolic roles for Mb, especially regulation of nitric oxide (NO) pools, modulation of BAT bioenergetics, thermogenesis, and lipid storage phenotypes. From these and other findings in the literature over many decades, Mb's function is not confined to delivering O2 in support of oxidative phosphorylation but may serve as an O2 sensor that modulates intracellular Po2- and NO-responsive molecular signaling pathways. This paradigm reflects a fundamental change in how oxidative metabolism and cell regulation are viewed in Mb-expressing cells such as skeletal muscle, heart, brown adipocytes, and select cancer cells. Here, we review historic and emerging views related to the physiological roles for Mb and present working models illustrating the possible importance of interactions between Mb, gases, and small-molecule metabolites in regulation of cell signaling and bioenergetics.
Collapse
Affiliation(s)
- Kiran Kumar Adepu
- Arkansas Children's Nutrition Center and Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States
| | - Andriy Anishkin
- Department of Biology, University of Maryland, College Park, Maryland, United States
| | - Sean H Adams
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, California, United States
- Center for Alimentary and Metabolic Science, School of Medicine, University of California Davis, Sacramento, California, United States
| | - Sree V Chintapalli
- Arkansas Children's Nutrition Center and Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States
| |
Collapse
|
5
|
Johnson AR, Rao K, Zhang BB, Mullet S, Goetzman E, Gelhaus S, Tejero J, Shiva U. Myoglobin Inhibits Breast Cancer Cell Fatty Acid Oxidation and Migration via Heme-dependent Oxidant Production and Not Fatty Acid Binding. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.30.591659. [PMID: 38746370 PMCID: PMC11092581 DOI: 10.1101/2024.04.30.591659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
The monomeric heme protein myoglobin (Mb), traditionally thought to be expressed exclusively in cardiac and skeletal muscle, is now known to be expressed in approximately 40% of breast tumors. While Mb expression is associated with better patient prognosis, the molecular mechanisms by which Mb limits cancer progression are unclear. In muscle, Mb's predominant function is oxygen storage and delivery, which is dependent on the protein's heme moiety. However, prior studies demonstrate that the low levels of Mb expressed in cancer cells preclude this function. Recent studies propose a novel fatty acid binding function for Mb via a lysine residue (K46) in the heme pocket. Given that cancer cells can upregulate fatty acid oxidation (FAO) to maintain energy production for cytoskeletal remodeling during cell migration, we tested whether Mb-mediated fatty acid binding modulates FAO to decrease breast cancer cell migration. We demonstrate that the stable expression of human Mb in MDA-MB-231 breast cancer cells decreases cell migration and FAO. Site-directed mutagenesis of Mb to disrupt Mb fatty acid binding did not reverse Mb-mediated attenuation of FAO or cell migration in these cells. In contrast, cells expressing Apo-Mb, in which heme incorporation was disrupted, showed a reversal of Mb-mediated attenuation of FAO and cell migration, suggesting that Mb attenuates FAO and migration via a heme-dependent mechanism rather than through fatty acid binding. To this end, we show that Mb's heme-dependent oxidant generation propagates dysregulated gene expression of migratory genes, and this is reversed by catalase treatment. Collectively, these data demonstrate that Mb decreases breast cancer cell migration, and this effect is due to heme-mediated oxidant production rather than fatty acid binding. The implication of these results will be discussed in the context of therapeutic strategies to modulate oxidant production and Mb in tumors. Highlights Myoglobin (Mb) expression in MDA-MB-231 breast cancer cells slows migration.Mb expression decreases mitochondrial respiration and fatty acid oxidation.Mb-dependent fatty acid binding does not regulate cell migration or respiration.Mb-dependent oxidant generation decreases mitochondrial metabolism and migration.Mb-derived oxidants dysregulate migratory gene expression.
Collapse
|
6
|
Hejlesen R, Kjær-Sørensen K, Fago A, Oxvig C. Generation and validation of a myoglobin knockout zebrafish model. Transgenic Res 2023; 32:537-546. [PMID: 37847464 PMCID: PMC10713697 DOI: 10.1007/s11248-023-00369-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 09/20/2023] [Indexed: 10/18/2023]
Abstract
Previous studies using myoglobin (Mb) knockout mice and knockdown zebrafish have presented conflicting results about in vivo phenotypes resulting from the loss of this conserved and highly expressed protein, and therefore a new well-characterized knockout model is warranted. We here describe the generation of three distinct zebrafish mb knockout lines using the CRISPR/Cas system. None of the three lines exhibited any morphological phenotypes, changes in length, or lethality during embryonic and larval development. The adult homozygous knockout mb(Auzf13.2) zebrafish line were absent of Mb protein, had an almost complete degradation of mb mRNA, and showed no changes in viability, length, or heart size. Furthermore, transcriptomic analysis of adult heart tissue showed that mb knockout did not cause altered expression of other genes. Lastly, no off-targeting was observed in 36 screened loci. In conclusion, we have generated three mb knockout lines with indistinguishable phenotypes during embryonic and larval development and validated one of these lines, mb(Auzf13.2), to have no signs of genetic compensation or off-target effects in the adult heart. These findings suggests that the mb(Auzf13.2) shows promise as a candidate for investigating the biological role of Mb in zebrafish.
Collapse
Affiliation(s)
- Rasmus Hejlesen
- Department of Biology, Zoophysiology, Aarhus University, Aarhus, Denmark
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Kasper Kjær-Sørensen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Angela Fago
- Department of Biology, Zoophysiology, Aarhus University, Aarhus, Denmark.
| | - Claus Oxvig
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
7
|
Aboouf MA, Gorr TA, Hamdy NM, Gassmann M, Thiersch M. Myoglobin in Brown Adipose Tissue: A Multifaceted Player in Thermogenesis. Cells 2023; 12:2240. [PMID: 37759463 PMCID: PMC10526770 DOI: 10.3390/cells12182240] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/01/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Brown adipose tissue (BAT) plays an important role in energy homeostasis by generating heat from chemical energy via uncoupled oxidative phosphorylation. Besides its high mitochondrial content and its exclusive expression of the uncoupling protein 1, another key feature of BAT is the high expression of myoglobin (MB), a heme-containing protein that typically binds oxygen, thereby facilitating the diffusion of the gas from cell membranes to mitochondria of muscle cells. In addition, MB also modulates nitric oxide (NO•) pools and can bind C16 and C18 fatty acids, which indicates a role in lipid metabolism. Recent studies in humans and mice implicated MB present in BAT in the regulation of lipid droplet morphology and fatty acid shuttling and composition, as well as mitochondrial oxidative metabolism. These functions suggest that MB plays an essential role in BAT energy metabolism and thermogenesis. In this review, we will discuss in detail the possible physiological roles played by MB in BAT thermogenesis along with the potential underlying molecular mechanisms and focus on the question of how BAT-MB expression is regulated and, in turn, how this globin regulates mitochondrial, lipid, and NO• metabolism. Finally, we present potential MB-mediated approaches to augment energy metabolism, which ultimately could help tackle different metabolic disorders.
Collapse
Affiliation(s)
- Mostafa A. Aboouf
- Institute of Veterinary Physiology, University of Zurich, 8057 Zurich, Switzerland
- Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, 8057 Zurich, Switzerland
- Department of Biochemistry, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt
| | - Thomas A. Gorr
- Institute of Veterinary Physiology, University of Zurich, 8057 Zurich, Switzerland
| | - Nadia M. Hamdy
- Department of Biochemistry, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt
| | - Max Gassmann
- Institute of Veterinary Physiology, University of Zurich, 8057 Zurich, Switzerland
- Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, 8057 Zurich, Switzerland
| | - Markus Thiersch
- Institute of Veterinary Physiology, University of Zurich, 8057 Zurich, Switzerland
- Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, 8057 Zurich, Switzerland
| |
Collapse
|
8
|
Agriesti F, Landini F, Tamma M, Pacelli C, Mazzoccoli C, Calice G, Ruggieri V, Capitanio G, Mori G, Piccoli C, Capitanio N. Bioenergetic profile and redox tone modulate in vitro osteogenesis of human dental pulp stem cells: new perspectives for bone regeneration and repair. Stem Cell Res Ther 2023; 14:215. [PMID: 37608350 PMCID: PMC10463344 DOI: 10.1186/s13287-023-03447-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 08/10/2023] [Indexed: 08/24/2023] Open
Abstract
BACKGROUND Redox signaling and energy metabolism are known to be involved in controlling the balance between self-renewal and proliferation/differentiation of stem cells. In this study we investigated metabolic and redox changes occurring during in vitro human dental pulp stem cells (hDPSCs) osteoblastic (OB) differentiation and tested on them the impact of the reactive oxygen species (ROS) signaling. METHODS hDPSCs were isolated from dental pulp and subjected to alkaline phosphatase and alizarin red staining, q-RT-PCR, and western blotting analysis of differentiation markers to assess achievement of osteogenic/odontogenic differentiation. Moreover, a combination of metabolic flux analysis and confocal cyto-imaging was used to profile the metabolic phenotype and to evaluate the redox tone of hDPSCs. RESULTS In differentiating hDPSCs we observed the down-regulation of the mitochondrial respiratory chain complexes expression since the early phase of the process, confirmed by metabolic flux analysis, and a reduction of the basal intracellular peroxide level in its later phase. In addition, dampened glycolysis was observed, thereby indicating a lower energy-generating phenotype in differentiating hDPSCs. Treatment with the ROS scavenger Trolox, applied in the early-middle phases of the process, markedly delayed OB differentiation of hDPSCs assessed as ALP activity, Runx2 expression, mineralization capacity, expression of stemness and osteoblast marker genes (Nanog, Lin28, Dspp, Ocn) and activation of ERK1/2. In addition, the antioxidant partly prevented the inhibitory effect on cell metabolism observed following osteogenic induction. CONCLUSIONS Altogether these results provided evidence that redox signaling, likely mediated by peroxide species, influenced the stepwise osteogenic expansion/differentiation of hDPSCs and contributed to shape its accompanying metabolic phenotype changes thus improving their efficiency in bone regeneration and repair.
Collapse
Affiliation(s)
- Francesca Agriesti
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
- Laboratory of Pre-Clinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture, Italy
| | - Francesca Landini
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| | - Mirko Tamma
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| | - Consiglia Pacelli
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| | - Carmela Mazzoccoli
- Laboratory of Pre-Clinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture, Italy
| | - Giovanni Calice
- Laboratory of Pre-Clinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture, Italy
| | - Vitalba Ruggieri
- Laboratory of Pre-Clinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture, Italy
- Clinical Pathology Unit, “Madonna delle Grazie’’ Hospital, Matera, Italy
| | - Giuseppe Capitanio
- Department of Translational Biomedicine and Neuroscience “DiBraiN”, University of Bari “Aldo Moro”, 70124 Bari, Italy
| | - Giorgio Mori
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| | - Claudia Piccoli
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| | - Nazzareno Capitanio
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| |
Collapse
|
9
|
Christen L, Broghammer H, Rapöhn I, Möhlis K, Strehlau C, Ribas‐Latre A, Gebhardt C, Roth L, Krause K, Landgraf K, Körner A, Rohde‐Zimmermann K, Hoffmann A, Klöting N, Ghosh A, Sun W, Dong H, Wolfrum C, Rassaf T, Hendgen‐Cotta UB, Stumvoll M, Blüher M, Heiker JT, Weiner J. Myoglobin-mediated lipid shuttling increases adrenergic activation of brown and white adipocyte metabolism and is as a marker of thermogenic adipocytes in humans. Clin Transl Med 2022; 12:e1108. [PMID: 36480426 PMCID: PMC9731393 DOI: 10.1002/ctm2.1108] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 10/23/2022] [Accepted: 10/25/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Recruitment and activation of brown adipose tissue (BAT) results in increased energy expenditure (EE) via thermogenesis and represents an intriguing therapeutic approach to combat obesity and treat associated diseases. Thermogenesis requires an increased and efficient supply of energy substrates and oxygen to the BAT. The hemoprotein myoglobin (MB) is primarily expressed in heart and skeletal muscle fibres, where it facilitates oxygen storage and flux to the mitochondria during exercise. In the last years, further contributions of MB have been assigned to the scavenging of reactive oxygen species (ROS), the regulation of cellular nitric oxide (NO) levels and also lipid binding. There is a substantial expression of MB in BAT, which is induced during brown adipocyte differentiation and BAT activation. This suggests MB as a previously unrecognized player in BAT contributing to thermogenesis. METHODS AND RESULTS This study analyzed the consequences of MB expression in BAT on mitochondrial function and thermogenesis in vitro and in vivo. Using MB overexpressing, knockdown or knockout adipocytes, we show that expression levels of MB control brown adipocyte mitochondrial respiratory capacity and acute response to adrenergic stimulation, signalling and lipolysis. Overexpression in white adipocytes also increases their metabolic activity. Mutation of lipid interacting residues in MB abolished these beneficial effects of MB. In vivo, whole-body MB knockout resulted in impaired thermoregulation and cold- as well as drug-induced BAT activation in mice. In humans, MB is differentially expressed in subcutaneous (SC) and visceral (VIS) adipose tissue (AT) depots, differentially regulated by the state of obesity and higher expressed in AT samples that exhibit higher thermogenic potential. CONCLUSIONS These data demonstrate for the first time a functional relevance of MBs lipid binding properties and establish MB as an important regulatory element of thermogenic capacity in brown and likely beige adipocytes.
Collapse
Affiliation(s)
- Lisa Christen
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI‐MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital LeipzigLeipzigGermany
| | - Helen Broghammer
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI‐MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital LeipzigLeipzigGermany
| | - Inka Rapöhn
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI‐MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital LeipzigLeipzigGermany
- Medical Department III ‐ EndocrinologyNephrologyRheumatologyUniversity of Leipzig Medical CenterLeipzigGermany
| | - Kevin Möhlis
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI‐MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital LeipzigLeipzigGermany
| | - Christian Strehlau
- Medical Department III ‐ EndocrinologyNephrologyRheumatologyUniversity of Leipzig Medical CenterLeipzigGermany
| | - Aleix Ribas‐Latre
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI‐MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital LeipzigLeipzigGermany
| | - Claudia Gebhardt
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI‐MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital LeipzigLeipzigGermany
| | - Lisa Roth
- Medical Department III ‐ EndocrinologyNephrologyRheumatologyUniversity of Leipzig Medical CenterLeipzigGermany
| | - Kerstin Krause
- Medical Department III ‐ EndocrinologyNephrologyRheumatologyUniversity of Leipzig Medical CenterLeipzigGermany
| | - Kathrin Landgraf
- Center for Pediatric Research Leipzig (CPL)University Hospital for Children and AdolescentsMedical FacultyUniversity of LeipzigLeipzigGermany
| | - Antje Körner
- Center for Pediatric Research Leipzig (CPL)University Hospital for Children and AdolescentsMedical FacultyUniversity of LeipzigLeipzigGermany
| | - Kerstin Rohde‐Zimmermann
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI‐MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital LeipzigLeipzigGermany
| | - Anne Hoffmann
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI‐MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital LeipzigLeipzigGermany
| | - Nora Klöting
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI‐MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital LeipzigLeipzigGermany
| | - Adhideb Ghosh
- Institute of FoodNutrition and HealthETH ZurichSchwerzenbachSwitzerland
| | - Wenfei Sun
- Institute of FoodNutrition and HealthETH ZurichSchwerzenbachSwitzerland
| | - Hua Dong
- Institute of FoodNutrition and HealthETH ZurichSchwerzenbachSwitzerland
| | - Christian Wolfrum
- Institute of FoodNutrition and HealthETH ZurichSchwerzenbachSwitzerland
| | - Tienush Rassaf
- Department of Cardiology and Vascular MedicineWest German Heart and Vascular CenterMedical FacultyUniversity of Duisburg‐EssenEssenGermany
| | - Ulrike B. Hendgen‐Cotta
- Department of Cardiology and Vascular MedicineWest German Heart and Vascular CenterMedical FacultyUniversity of Duisburg‐EssenEssenGermany
| | - Michael Stumvoll
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI‐MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital LeipzigLeipzigGermany
- Medical Department III ‐ EndocrinologyNephrologyRheumatologyUniversity of Leipzig Medical CenterLeipzigGermany
| | - Matthias Blüher
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI‐MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital LeipzigLeipzigGermany
| | - John T. Heiker
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI‐MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital LeipzigLeipzigGermany
- Institute of Biochemistry, Faculty of Life SciencesUniversity of LeipzigLeipzigGermany
| | - Juliane Weiner
- Medical Department III ‐ EndocrinologyNephrologyRheumatologyUniversity of Leipzig Medical CenterLeipzigGermany
| |
Collapse
|
10
|
Armbruster J, Aboouf MA, Gassmann M, Egert A, Schorle H, Hornung V, Schmidt T, Schmid-Burgk JL, Kristiansen G, Bicker A, Hankeln T, Zhu H, Gorr TA. Myoglobin regulates fatty acid trafficking and lipid metabolism in mammary epithelial cells. PLoS One 2022; 17:e0275725. [PMID: 36223378 PMCID: PMC9555620 DOI: 10.1371/journal.pone.0275725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 09/21/2022] [Indexed: 11/18/2022] Open
Abstract
Myoglobin (MB) is known to bind and deliver oxygen in striated muscles at high expression levels. MB is also expressed at much reduced levels in mammary epithelial cells, where the protein´s function is unclear. In this study, we aim to determine whether MB impacts fatty acid trafficking and facilitates aerobic fatty acid ß-oxidation in mammary epithelial cells. We utilized MB-wildtype versus MB-knockout mice and human breast cancer cells to examine the impact of MB and its oxygenation status on fatty acid metabolism in mouse milk and mammary epithelia. MB deficient cells were generated through CRISPR/Cas9 and TALEN approaches and exposed to various oxygen tensions. Fatty acid profiling of milk and cell extracts were performed along with cell labelling and immunocytochemistry. Our findings show that MB expression in mammary epithelial cells promoted fatty acid oxidation while reducing stearyl-CoA desaturase activity for lipogenesis. In cells and milk product, presence of oxygenated MB significantly elevated indices of limited fatty acid ß-oxidation, i.e., the organelle-bound removal of a C2 moiety from long-chain saturated or monounsaturated fatty acids, thus shifting the composition toward more saturated and shorter fatty acid species. Presence of the globin also increased cytoplasmic fatty acid solubility under normoxia and fatty acid deposition to lipid droplets under severe hypoxia. We conclude that MB can function in mammary epithelia as intracellular O2-dependent shuttle of oxidizable fatty acid substrates. MB's impact on limited oxidation of fatty acids could generate inflammatory mediator lipokines, such as 7-hexadecenoate. Thus, the novel functions of MB in breast epithelia described herein range from controlling fatty acid turnover and homeostasis to influencing inflammatory signalling cascade. Future work is needed to analyse to what extent these novel roles of MB also apply to myocytic cell physiology and malignant cell behaviour, respectively.
Collapse
Affiliation(s)
- Julia Armbruster
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Mostafa A. Aboouf
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
- Department of Biochemistry, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
- Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Max Gassmann
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
- Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Angela Egert
- Institute of Pathology, Department of Developmental Pathology, University Hospital Bonn, Bonn, Germany
| | - Hubert Schorle
- Institute of Pathology, Department of Developmental Pathology, University Hospital Bonn, Bonn, Germany
| | - Veit Hornung
- Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Tobias Schmidt
- Institute of Clinical Chemistry and Clinical Pharmacology, University and University Hospital Bonn, Bonn, Germany
| | - Jonathan L. Schmid-Burgk
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Zurich, Schlieren, Switzerland
| | | | - Anne Bicker
- Institute of Organismic and Molecular Evolution, Molecular and Genome Analysis, Johannes Gutenberg University, Mainz, Germany
| | - Thomas Hankeln
- Institute of Organismic and Molecular Evolution, Molecular and Genome Analysis, Johannes Gutenberg University, Mainz, Germany
| | - Hao Zhu
- Department of Clinical Laboratory Sciences, University of Kansas Medical Center, Kansas City, Kansas, United States of America
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Thomas A. Gorr
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| |
Collapse
|
11
|
Scrima R, Agriesti F, Pacelli C, Piccoli C, Pucci P, Amoresano A, Cela O, Nappi L, Tataranni T, Mori G, Formisano P, Capitanio N. Myoglobin expression by alternative transcript in different mesenchymal stem cells compartments. Stem Cell Res Ther 2022; 13:209. [PMID: 35598009 PMCID: PMC9123686 DOI: 10.1186/s13287-022-02880-6] [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: 03/04/2022] [Accepted: 05/01/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The metabolic phenotype of stem cells is increasingly recognized as a hallmark of their pluripotency with mitochondrial and oxygen-related metabolism playing a not completely defined role in this context. In a previous study, we reported the ectopic expression of myoglobin (MB) in bone marrow-derived hematopoietic stem/progenitor cells. Here, we have extended the analysis to mesenchymal stem cells (MSCs) isolated from different tissues. METHODS MSCs were isolated from human placental membrane, mammary adipose tissue and dental pulp and subjected to RT-PCR, Western blotting and mass spectrometry to investigate the expression of MB. A combination of metabolic flux analysis and cyto-imaging was used to profile the metabolic phenotype and the mitochondria dynamics in the different MSCs. RESULTS As for the hematopoietic stem/progenitor cells, the expression of Mb was largely driven by an alternative transcript with the protein occurring both in the monomer and in the dimer forms as confirmed by mass spectrometry analysis. Comparing the metabolic fluxes between neonatal placental membrane-derived and adult mammary adipose tissue-derived MSCs, we showed a significantly more active bioenergetics profile in the former that correlated with a larger co-localization of myoglobin with the mitochondrial compartment. Differences in the structure of the mitochondrial network as well as in the expression of factors controlling the organelle dynamics were also observed between neonatal and adult mesenchymal stem cells. Finally, the expression of myoglobin was found to be strongly reduced following osteogenic differentiation of dental pulp-derived MSCs, while it was upregulated following reprogramming of human fibroblasts to induce pluripotent stem cells. CONCLUSIONS Ectopic expression of myoglobin in tissues other than muscle raises the question of understanding its function therein. Properties in addition to the canonical oxygen storage/delivery have been uncovered. Finding of Mb expressed via an alternative gene transcript in the context of different stem cells with metabolic phenotypes, its loss during differentiation and recovery in iPSCs suggest a hitherto unappreciated role of Mb in controlling the balance between aerobic metabolism and pluripotency. Understanding how Mb contributes through modulation of the mitochondrial physiology to the stem cell biology paves the way to novel perspectives in regenerative medicine as well as in cancer stem cell therapy.
Collapse
Affiliation(s)
- Rosella Scrima
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy.
| | - Francesca Agriesti
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy.,Laboratory of Pre-Clinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, Rionero in Vulture, PZ, Italy
| | - Consiglia Pacelli
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Claudia Piccoli
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Pietro Pucci
- CEINGE Advanced Biotechnology and Department of Chemical Sciences, University of Napoli Federico II, Naples, Italy
| | - Angela Amoresano
- CEINGE Advanced Biotechnology and Department of Chemical Sciences, University of Napoli Federico II, Naples, Italy
| | - Olga Cela
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Luigi Nappi
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Tiziana Tataranni
- Laboratory of Pre-Clinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, Rionero in Vulture, PZ, Italy
| | - Giorgio Mori
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Pietro Formisano
- Department of Translational Medical Sciences, Federico II University of Naples, Naples, Italy
| | - Nazzareno Capitanio
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy.
| |
Collapse
|
12
|
Zhu Q, Dong Q, Wang X, Xia T, Fu Y, Wang Q, Wu R, Wu T. Palmitic Acid, A Critical Metabolite, Aggravates Cellular Senescence Through Reactive Oxygen Species Generation in Kawasaki Disease. Front Pharmacol 2022; 13:809157. [PMID: 35401162 PMCID: PMC8983937 DOI: 10.3389/fphar.2022.809157] [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: 11/04/2021] [Accepted: 02/17/2022] [Indexed: 11/13/2022] Open
Abstract
Coronary artery lesions (CALs) are severe complications of Kawasaki disease (KD), resulting in stenosis and thrombogenesis. Metabolomic profiling of patients’ plasma could assist in elucidating the pathogenesis of CALs and identifying diagnostic biomarkers, which are imperative for clinical treatment. The metabolic profiles between KD patients with CALs and without CALs (non-coronary artery lesion, or NCAL, group) indicated the most significantly differentially expressed metabolite, palmitic acid (PA), showed the most massive fold change at 9.879. Furthermore, PA was proven to aggravate endothelial cellular senescence by increasing the generation of reactive oxygen species (ROS) in KD, and those two phenotypes were confirmed to be enriched among the differentially expressed genes between KD and normal samples from GEO datasets. Collectively, our findings indicate that cellular senescence may be one of the mechanisms of vascular endothelial damage in KD. PA may be a biomarker and potential therapeutic target for predicting the occurrence of CALs in KD patients. All things considered, our findings confirm that plasma metabolomics was able to identify promising biomarkers and potential pathogenesis mechanisms in KD. To conclude, Palmitic acid could be a novel target in future studies of CALs in patients with KD.
Collapse
Affiliation(s)
- Qiongjun Zhu
- Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital, Institute of Cardiovascular Development and Translational Medicine, Wenzhou Medical University, Zhejiang, China
| | - Qianqian Dong
- Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital, Institute of Cardiovascular Development and Translational Medicine, Wenzhou Medical University, Zhejiang, China
| | - Xuliang Wang
- Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital, Institute of Cardiovascular Development and Translational Medicine, Wenzhou Medical University, Zhejiang, China
| | - Tianhe Xia
- Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital, Institute of Cardiovascular Development and Translational Medicine, Wenzhou Medical University, Zhejiang, China
| | - Yu Fu
- Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital, Institute of Cardiovascular Development and Translational Medicine, Wenzhou Medical University, Zhejiang, China
| | - Qiaoyu Wang
- Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital, Institute of Cardiovascular Development and Translational Medicine, Wenzhou Medical University, Zhejiang, China
| | - Rongzhou Wu
- Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital, Institute of Cardiovascular Development and Translational Medicine, Wenzhou Medical University, Zhejiang, China
| | - Tingting Wu
- Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital, Institute of Cardiovascular Development and Translational Medicine, Wenzhou Medical University, Zhejiang, China
| |
Collapse
|
13
|
Melo L, Hagar A, Klaunig J. Gene expression signature of exercise and change of diet on non-alcoholic fatty liver disease in mice. COMPARATIVE EXERCISE PHYSIOLOGY 2022. [DOI: 10.3920/cep210033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Non-alcoholic fatty liver diseases (NAFLD) are particularly prevalent in the general Western adult population, with around one third of the population suffering from the disease. Evidence shows that NAFLD is associated with metabolic syndromes such as obesity, insulin resistance, and hypertension. Currently, the sole therapy for NAFLD involves exercise intervention. Studies showed that, with and without weight loss, exercise interventions produced a significant cutback in intrahepatic lipid content in humans, but better controlled studies that can investigate the cellular and molecular mechanisms are still lacking. In the current study we perform RNA sequencing analysis on liver samples from C57BL/6 mice submitted to aerobic exercise and diet interventions that are human-translatable and determine the genetic expression signature of exercise in the NAFLD onset. We show that aerobic exercise affects genes and pathways related to liver metabolism, muscle contraction and relaxation, immune response and inflammation, and development of liver cancer, counteracting non-alcoholic steatohepatitis and hepatocellular carcinoma development. While genes and pathways implicating immune response are activated by aerobic exercise in all interventions, the most effective intervention in terms of improvement of NASH is the combination of aerobic exercise with change of diet.
Collapse
Affiliation(s)
- L. Melo
- Laboratory of Investigative Toxicology and Pathology, Department of Environmental and Occupational Health, Indiana School of Public Health, 2719E 10th St, Indiana University, Bloomington, IN, 47405, USA
- University of Pittsburgh Medical School, 200 Lothrop St, Pittsburgh, PA 15213, USA
| | - A. Hagar
- History & Philosophy of Science & Medicine Department, Indiana University, 1020 E Kirkwood Ave, Bloomington, IN 47405, USA
- Intelligent Systems Engineering Department, Indiana University, Bloomington, IN, USA
| | - J.E. Klaunig
- Laboratory of Investigative Toxicology and Pathology, Department of Environmental and Occupational Health, Indiana School of Public Health, 2719E 10th St, Indiana University, Bloomington, IN, 47405, USA
| |
Collapse
|
14
|
Garikapati V, Colasante C, Baumgart-Vogt E, Spengler B. Sequential lipidomic, metabolomic, and proteomic analyses of serum, liver, and heart tissue specimens from peroxisomal biogenesis factor 11α knockout mice. Anal Bioanal Chem 2022; 414:2235-2250. [PMID: 35083512 PMCID: PMC8821073 DOI: 10.1007/s00216-021-03860-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 11/25/2021] [Accepted: 12/20/2021] [Indexed: 12/16/2022]
Abstract
Peroxisomes are versatile single membrane-enclosed cytoplasmic organelles, involved in reactive oxygen species (ROS) and lipid metabolism and diverse other metabolic processes. Peroxisomal disorders result from mutations in Pex genes-encoded proteins named peroxins (PEX proteins) and single peroxisomal enzyme deficiencies. The PEX11 protein family (α, β, and γ isoforms) plays an important role in peroxisomal proliferation and fission. However, their specific functions and the metabolic impact caused by their deficiencies have not been precisely characterized. To understand the systemic molecular alterations caused by peroxisomal defects, here we utilized untreated peroxisomal biogenesis factor 11α knockout (Pex11α KO) mouse model and performed serial relative-quantitative lipidomic, metabolomic, and proteomic analyses of serum, liver, and heart tissue homogenates. We demonstrated significant specific changes in the abundances of multiple lipid species, polar metabolites, and proteins and dysregulated metabolic pathways in distinct biological specimens of the Pex11α KO adult mice in comparison to the wild type (WT) controls. Overall, the present study reports comprehensive semi-quantitative molecular omics information of the Pex11α KO mice, which might serve in the future as a reference for a better understanding of the roles of Pex11α and underlying pathophysiological mechanisms of peroxisomal biogenesis disorders.
Collapse
Affiliation(s)
- Vannuruswamy Garikapati
- Institute of Inorganic and Analytical Chemistry, Justus Liebig University Giessen, 35392, Giessen, Germany.,Institute for Anatomy and Cell Biology II, Division of Medical Cell Biology, Justus Liebig University Giessen, 35392, Giessen, Germany
| | - Claudia Colasante
- Institute for Anatomy and Cell Biology II, Division of Medical Cell Biology, Justus Liebig University Giessen, 35392, Giessen, Germany
| | - Eveline Baumgart-Vogt
- Institute for Anatomy and Cell Biology II, Division of Medical Cell Biology, Justus Liebig University Giessen, 35392, Giessen, Germany.
| | - Bernhard Spengler
- Institute of Inorganic and Analytical Chemistry, Justus Liebig University Giessen, 35392, Giessen, Germany.
| |
Collapse
|
15
|
Aboouf MA, Armbruster J, Thiersch M, Gassmann M, Gödecke A, Gnaiger E, Kristiansen G, Bicker A, Hankeln T, Zhu H, Gorr TA. Myoglobin, expressed in brown adipose tissue of mice, regulates the content and activity of mitochondria and lipid droplets. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:159026. [PMID: 34384891 DOI: 10.1016/j.bbalip.2021.159026] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 08/02/2021] [Accepted: 08/04/2021] [Indexed: 12/19/2022]
Abstract
The identification of novel physiological regulators that stimulate energy expenditure through brown adipose tissue (BAT) activity in substrate catalysis is of utmost importance to understand and treat metabolic diseases. Myoglobin (MB), known to store or transport oxygen in heart and skeletal muscles, has recently been found to bind fatty acids with physiological constants in its oxygenated form (i.e., MBO2). Here, we investigated the in vivo effect of MB expression on BAT activity. In particular, we studied mitochondrial function and lipid metabolism as essential determinants of energy expenditure in this tissue. We show in a MB-null (MBko) mouse model that MB expression in BAT impacts on the activity of brown adipocytes in a twofold manner: i) by elevating mitochondrial density plus maximal respiration capacity, and through that, by stimulating BAT oxidative metabolism along with the organelles` uncoupled respiration; and ii) by influencing the free fatty acids pool towards a palmitate-enriched composition and shifting the lipid droplet (LD) equilibrium towards higher counts of smaller droplets. These metabolic changes were accompanied by the up-regulated expression of thermogenesis markers UCP1, CIDEA, CIDEC, PGC1-α and PPAR-α in the BAT of MB wildtype (MBwt) mice. Along with the emergence of the "browning" BAT morphology, MBwt mice exhibited a leaner phenotype when compared to MBko littermates at 20 weeks of age. Our data shed novel insights into MB's role in linking oxygen and lipid-based thermogenic metabolism. The findings suggest potential new strategies of targeting the MB pathway to treat metabolic disorders related to diminishing energy expenditure.
Collapse
Affiliation(s)
- Mostafa A Aboouf
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, 8057 Zurich, Switzerland; Center for Clinical Studies, Vetsuisse Faculty, University of Zurich, 8057 Zurich, Switzerland; Molecular and Translational Biomedicine PhD Program, Life Science Zurich Graduate School, 8057 Zurich, Switzerland; Department of Biochemistry, Faculty of Pharmacy, Ain Shams University, 11566 Cairo, Egypt
| | - Julia Armbruster
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, 8057 Zurich, Switzerland; Center for Clinical Studies, Vetsuisse Faculty, University of Zurich, 8057 Zurich, Switzerland; Molecular and Translational Biomedicine PhD Program, Life Science Zurich Graduate School, 8057 Zurich, Switzerland
| | - Markus Thiersch
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, 8057 Zurich, Switzerland
| | - Max Gassmann
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, 8057 Zurich, Switzerland; Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, 8057 Zurich, Switzerland
| | - Axel Gödecke
- Institute of Cardiovascular Physiology (A.G.), Medical Faculty, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Erich Gnaiger
- Department of Visceral, Transplant and Thoracic Surgery, D. Swarovski Research Laboratory, Medical University Innsbruck, Innrain 66/6, A-6020 Innsbruck, Austria
| | - Glen Kristiansen
- Institute of Pathology, University Hospital Bonn, University of Bonn, D-53127 Bonn, Germany
| | - Anne Bicker
- Institute of Organismic and Molecular Evolution, Molecular Genetics and Genome Analysis, Johannes Gutenberg University, D-55099 Mainz, Germany
| | - Thomas Hankeln
- Institute of Organismic and Molecular Evolution, Molecular Genetics and Genome Analysis, Johannes Gutenberg University, D-55099 Mainz, Germany
| | - Hao Zhu
- Department of Clinical Laboratory Sciences, The University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA; Department of Biochemistry and Molecular Biology, The University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA
| | - Thomas A Gorr
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, 8057 Zurich, Switzerland.
| |
Collapse
|
16
|
Michel L, Helfrich I, Hendgen-Cotta UB, Mincu RI, Korste S, Mrotzek SM, Spomer A, Odersky A, Rischpler C, Herrmann K, Umutlu L, Coman C, Ahrends R, Sickmann A, Löffek S, Livingstone E, Ugurel S, Zimmer L, Gunzer M, Schadendorf D, Totzeck M, Rassaf T. Targeting early stages of cardiotoxicity from anti-PD1 immune checkpoint inhibitor therapy. Eur Heart J 2021; 43:316-329. [PMID: 34389849 DOI: 10.1093/eurheartj/ehab430] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 02/25/2021] [Accepted: 06/25/2021] [Indexed: 12/13/2022] Open
Abstract
AIMS Cardiac immune-related adverse events (irAEs) from immune checkpoint inhibition (ICI) targeting programmed death 1 (PD1) are of growing concern. Once cardiac irAEs become clinically manifest, fatality rates are high. Cardio-oncology aims to prevent detrimental effects before manifestation of severe complications by targeting early pathological changes. We therefore aimed to investigate early consequences of PD1 inhibition for cardiac integrity to prevent the development of overt cardiac disease. METHODS AND RESULTS We investigated cardiac-specific consequences from anti-PD1 therapy in a combined biochemical and in vivo phenotyping approach. Mouse hearts showed broad expression of the ligand PDL1 on cardiac endothelial cells as a main mediator of immune-crosstalk. Using a novel melanoma mouse model, we assessed that anti-PD1 therapy promoted myocardial infiltration with CD4+ and CD8+ T cells, the latter being markedly activated. Left ventricular (LV) function was impaired during pharmacological stress, as shown by pressure-volume catheterization. This was associated with a dysregulated myocardial metabolism, including the proteome and the lipidome. Analogous to the experimental approach, in patients with metastatic melanoma (n = 7) receiving anti-PD1 therapy, LV function in response to stress was impaired under therapy. Finally, we identified that blockade of tumour necrosis factor alpha (TNFα) preserved LV function without attenuating the anti-cancer efficacy of anti-PD1 therapy. CONCLUSIONS Anti-PD1 therapy induces a disruption of cardiac immune homeostasis leading to early impairment of myocardial functional integrity, with potential prognostic effects on the growing number of treated patients. Blockade of TNFα may serve as an approach to prevent the manifestation of ICI-related cardiotoxicity.
Collapse
Affiliation(s)
- Lars Michel
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, Hufelandstraße 55, Essen 45147, Germany
| | - Iris Helfrich
- Department of Dermatology, University Hospital Essen, Hufelandstraße 55, Essen 45147, Germany.,German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Essen 45147, Germany.,Medical Faculty of the Ludwig Maximilian University of Munich, Department of Dermatology and Allergology, Frauenlobstrasse 9-11, Munich 80377, Germany
| | - Ulrike Barbara Hendgen-Cotta
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, Hufelandstraße 55, Essen 45147, Germany
| | - Raluca-Ileana Mincu
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, Hufelandstraße 55, Essen 45147, Germany
| | - Sebastian Korste
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, Hufelandstraße 55, Essen 45147, Germany
| | - Simone Maria Mrotzek
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, Hufelandstraße 55, Essen 45147, Germany
| | - Armin Spomer
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, Hufelandstraße 55, Essen 45147, Germany
| | - Andrea Odersky
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, Hufelandstraße 55, Essen 45147, Germany
| | - Christoph Rischpler
- Department of Nuclear Medicine, University Hospital Essen, Hufelandstraße 55, Essen 45147, Germany
| | - Ken Herrmann
- Department of Nuclear Medicine, University Hospital Essen, Hufelandstraße 55, Essen 45147, Germany
| | - Lale Umutlu
- Department of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Hufelandstraße 55, Essen 45147, Germany
| | - Cristina Coman
- Institute for Analytical Chemistry, Waehringer Straße 38, Vienna A-1090, Austria.,Leibniz Institut für Analytische Wissenschaften-ISAS-e.V., Otto-Hahn-Straße 6b, Dortmund 44227, Germany
| | - Robert Ahrends
- Institute for Analytical Chemistry, Waehringer Straße 38, Vienna A-1090, Austria.,Leibniz Institut für Analytische Wissenschaften-ISAS-e.V., Otto-Hahn-Straße 6b, Dortmund 44227, Germany
| | - Albert Sickmann
- Leibniz Institut für Analytische Wissenschaften-ISAS-e.V., Otto-Hahn-Straße 6b, Dortmund 44227, Germany.,Medizinische Fakultät, Medizinisches Proteom-Center (MPC), Ruhr-Universität Bochum, Bochum 44801, Germany.,Department of Chemistry, College of Physical Sciences, University of Aberdeen, Aberdeen AB243FX, Scotland
| | - Stefanie Löffek
- Department of Dermatology, University Hospital Essen, Hufelandstraße 55, Essen 45147, Germany.,German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Essen 45147, Germany
| | - Elisabeth Livingstone
- Department of Dermatology, University Hospital Essen, Hufelandstraße 55, Essen 45147, Germany.,German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Essen 45147, Germany
| | - Selma Ugurel
- Department of Dermatology, University Hospital Essen, Hufelandstraße 55, Essen 45147, Germany.,German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Essen 45147, Germany
| | - Lisa Zimmer
- Department of Dermatology, University Hospital Essen, Hufelandstraße 55, Essen 45147, Germany.,German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Essen 45147, Germany
| | - Matthias Gunzer
- Leibniz Institut für Analytische Wissenschaften-ISAS-e.V., Otto-Hahn-Straße 6b, Dortmund 44227, Germany.,Institute for Experimental Immunology and Imaging, University Hospital Essen, Hufelandstraße 55, Essen 45147, Germany
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital Essen, Hufelandstraße 55, Essen 45147, Germany.,German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Essen 45147, Germany
| | - Matthias Totzeck
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, Hufelandstraße 55, Essen 45147, Germany
| | - Tienush Rassaf
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, Hufelandstraße 55, Essen 45147, Germany
| |
Collapse
|
17
|
Blackburn ML, Wankhade UD, Ono-Moore KD, Chintapalli SV, Fox R, Rutkowsky JM, Willis BJ, Tolentino T, Lloyd KCK, Adams SH. On the potential role of globins in brown adipose tissue: a novel conceptual model and studies in myoglobin knockout mice. Am J Physiol Endocrinol Metab 2021; 321:E47-E62. [PMID: 33969705 PMCID: PMC8321818 DOI: 10.1152/ajpendo.00662.2020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Myoglobin (Mb) regulates O2 bioavailability in muscle and heart as the partial pressure of O2 (Po2) drops with increased tissue workload. Globin proteins also modulate cellular NO pools, "scavenging" NO at higher Po2 and converting NO2- to NO as Po2 falls. Myoglobin binding of fatty acids may also signal a role in fat metabolism. Interestingly, Mb is expressed in brown adipose tissue (BAT), but its function is unknown. Herein, we present a new conceptual model that proposes links between BAT thermogenic activation, concurrently reduced Po2, and NO pools regulated by deoxy/oxy-globin toggling and xanthine oxidoreductase (XOR). We describe the effect of Mb knockout (Mb-/-) on BAT phenotype [lipid droplets, mitochondrial markers uncoupling protein 1 (UCP1) and cytochrome C oxidase 4 (Cox4), transcriptomics] in male and female mice fed a high-fat diet (HFD, 45% of energy, ∼13 wk), and examine Mb expression during brown adipocyte differentiation. Interscapular BAT weights did not differ by genotype, but there was a higher prevalence of mid-large sized droplets in Mb-/-. COX4 protein expression was significantly reduced in Mb-/- BAT, and a suite of metabolic/NO/stress/hypoxia transcripts were lower. All of these Mb-/--associated differences were most apparent in females. The new conceptual model, and results derived from Mb-/- mice, suggest a role for Mb in BAT metabolic regulation, in part through sexually dimorphic systems and NO signaling. This possibility requires further validation in light of significant mouse-to-mouse variability of BAT Mb mRNA and protein abundances in wild-type mice and lower expression relative to muscle and heart.NEW & NOTEWORTHY Myoglobin confers the distinct red color to muscle and heart, serving as an oxygen-binding protein in oxidative fibers. Less attention has been paid to brown fat, a thermogenic tissue that also expresses myoglobin. In a mouse knockout model lacking myoglobin, brown fat had larger fat droplets and lower markers of mitochondrial oxidative metabolism, especially in females. Gene expression patterns suggest a role for myoglobin as an oxygen/nitric oxide-sensor that regulates cellular metabolic and signaling pathways.
Collapse
Affiliation(s)
- Michael L Blackburn
- Arkansas Children's Nutrition Center, Little Rock, Arkansas
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Umesh D Wankhade
- Arkansas Children's Nutrition Center, Little Rock, Arkansas
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | | | - Sree V Chintapalli
- Arkansas Children's Nutrition Center, Little Rock, Arkansas
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Renee Fox
- Arkansas Children's Nutrition Center, Little Rock, Arkansas
| | - Jennifer M Rutkowsky
- Department of Molecular Biosciences, UC Davis School of Veterinary Medicine, University of California, Davis, California
- Mouse Metabolic Phenotyping Center, University of California, Davis, California
| | - Brandon J Willis
- Mouse Biology Program, University of California, Davis, California
| | - Todd Tolentino
- Mouse Metabolic Phenotyping Center, University of California, Davis, California
- Mouse Biology Program, University of California, Davis, California
| | - K C Kent Lloyd
- Mouse Metabolic Phenotyping Center, University of California, Davis, California
- Mouse Biology Program, University of California, Davis, California
- Department of Surgery, University of California Davis School of Medicine, Sacramento, California
| | - Sean H Adams
- Arkansas Children's Nutrition Center, Little Rock, Arkansas
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
- Department of Surgery, University of California Davis School of Medicine, Sacramento, California
- Center for Alimentary and Metabolic Science, University of California Davis School of Medicine, Sacramento, California
| |
Collapse
|
18
|
Ono-Moore KD, Olfert IM, Rutkowsky JM, Chintapalli SV, Willis BJ, Blackburn ML, Williams DK, O'Reilly J, Tolentino T, Lloyd KCK, Adams SH. Metabolic physiology and skeletal muscle phenotypes in male and female myoglobin knockout mice. Am J Physiol Endocrinol Metab 2021; 321:E63-E79. [PMID: 33969704 PMCID: PMC8321820 DOI: 10.1152/ajpendo.00624.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Myoglobin (Mb) is a regulator of O2 bioavailability in type I muscle and heart, at least when tissue O2 levels drop. Mb also plays a role in regulating cellular nitric oxide (NO) pools. Robust binding of long-chain fatty acids and long-chain acylcarnitines to Mb, and enhanced glucose metabolism in hearts of Mb knockout (KO) mice, suggest additional roles in muscle intermediary metabolism and fuel selection. To evaluate this hypothesis, we measured energy expenditure (EE), respiratory exchange ratio (RER), body weight gain and adiposity, glucose tolerance, and insulin sensitivity in Mb knockout (Mb-/-) and wild-type (WT) mice challenged with a high-fat diet (HFD, 45% of calories). In males (n = 10/genotype) and females (n = 9/genotype) tested at 5-6, 11-12, and 17-18 wk, there were no genotype effects on RER, EE, or food intake. RER and EE during cold (10°C, 72 h), and glucose and insulin tolerance, were not different compared with within-sex WT controls. At ∼18 and ∼19 wk of age, female Mb-/- adiposity was ∼42%-48% higher versus WT females (P = 0.1). Transcriptomics analyses (whole gastrocnemius, soleus) revealed few consistent changes, with the notable exception of a 20% drop in soleus transferrin receptor (Tfrc) mRNA. Capillarity indices were significantly increased in Mb-/-, specifically in Mb-rich soleus and deep gastrocnemius. The results indicate that Mb loss does not have a major impact on whole body glucose homeostasis, EE, RER, or response to a cold challenge in mice. However, the greater adiposity in female Mb-/- mice indicates a sex-specific effect of Mb KO on fat storage and feed efficiency.NEW & NOTEWORTHY The roles of myoglobin remain to be elaborated. We address sexual dimorphism in terms of outcomes in response to the loss of myoglobin in knockout mice and perform, for the first time, a series of comprehensive metabolic studies under conditions in which fat is mobilized (high-fat diet, cold). The results highlight that myoglobin is not necessary and sufficient for maintaining oxidative metabolism and point to alternative roles for this protein in muscle and heart.
Collapse
Affiliation(s)
| | - I Mark Olfert
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Jennifer M Rutkowsky
- Department of Molecular Biosciences, UC Davis School of Veterinary Medicine, University of California, Davis, California
- Mouse Metabolic Phenotyping Center, University of California, Davis, California
| | - Sree V Chintapalli
- Arkansas Children's Nutrition Center, Little Rock, Arkansas
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Brandon J Willis
- Mouse Biology Program, University of California, Davis, California
| | - Michael L Blackburn
- Arkansas Children's Nutrition Center, Little Rock, Arkansas
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - D Keith Williams
- Arkansas Children's Nutrition Center, Little Rock, Arkansas
- Department of Biostatistics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Juliana O'Reilly
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Todd Tolentino
- Mouse Metabolic Phenotyping Center, University of California, Davis, California
- Mouse Biology Program, University of California, Davis, California
| | - K C Kent Lloyd
- Mouse Metabolic Phenotyping Center, University of California, Davis, California
- Mouse Biology Program, University of California, Davis, California
- Department of Surgery, University of California Davis School of Medicine, Sacramento, California
| | - Sean H Adams
- Arkansas Children's Nutrition Center, Little Rock, Arkansas
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
- Department of Surgery, University of California Davis School of Medicine, Sacramento, California
- Center for Alimentary and Metabolic Science, University of California Davis School of Medicine, Sacramento, California
| |
Collapse
|
19
|
Heinen A, Gödecke S, Flögel U, Miklos D, Bottermann K, Spychala A, Gödecke A. 4-hydroxytamoxifen does not deteriorate cardiac function in cardiomyocyte-specific MerCreMer transgenic mice. Basic Res Cardiol 2021; 116:8. [PMID: 33544211 PMCID: PMC7864833 DOI: 10.1007/s00395-020-00841-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 12/28/2020] [Indexed: 01/28/2023]
Abstract
Conditional, cell-type-specific transgenic mouse lines are of high value in cardiovascular research. A standard tool for cardiomyocyte-restricted DNA editing is the αMHC-MerCreMer/loxP system. However, there is an ongoing debate on the occurrence of cardiac side effects caused by unspecific Cre activity or related to tamoxifen/oil overload. Here, we investigated potential adverse effects of DNA editing by the αMHC-MerCreMer/loxP system in combination with a low-dose treatment protocol with the tamoxifen metabolite 4-hydroxytamoxifen (OH-Txf). αMHC-MerCreMer mice received intraperitoneally OH-Txf (20 mg/kg) for 5 or 10 days. These treatment protocols were highly efficient to induce DNA editing in adult mouse hearts. Multi-parametric magnetic resonance imaging revealed neither transient nor permanent effects on cardiac function during or up to 19 days after 5 day OH-Txf treatment. Furthermore, OH-Txf did not affect cardiac phosphocreatine/ATP ratios assessed by in vivo 31P MR spectroscopy, indicating no Cre-mediated side effects on cardiac energy status. No MRI-based indication for the development of cardiac fibrosis was found as mean T1 relaxation time was unchanged. Histological analysis of myocardial collagen III content after OH-Txf confirmed this result. Last, mean T2 relaxation time was not altered after Txf treatment suggesting no pronounced cardiac lipid accumulation or tissue oedema. In additional experiments, cardiac function was assessed for up to 42 days to investigate potential delayed side effects of OH-Txf treatment. Neither 5- nor 10-day treatment resulted in a depression of cardiac function. Efficient cardiomyocyte-restricted DNA editing that is free of unwanted side effects on cardiac function, energetics or fibrosis can be achieved in adult mice when the αMHC-MerCreMer/loxP system is activated by the tamoxifen metabolite OH-Txf.
Collapse
Affiliation(s)
- Andre Heinen
- Institut für Herz- und Kreislaufphysiologie, Medizinische Fakultät und Universitätsklinikum Düsseldorf, Heinrich-Heine-Universität Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Stefanie Gödecke
- Institut für Herz- und Kreislaufphysiologie, Medizinische Fakultät und Universitätsklinikum Düsseldorf, Heinrich-Heine-Universität Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Ulrich Flögel
- Institut für Molekulare Kardiologie, Medizinische Fakultät und Universitätsklinikum Düsseldorf, Heinrich-Heine-Universität Düsseldorf, 40225, Düsseldorf, Germany
| | - Dominika Miklos
- Institut für Herz- und Kreislaufphysiologie, Medizinische Fakultät und Universitätsklinikum Düsseldorf, Heinrich-Heine-Universität Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Katharina Bottermann
- Institut für Herz- und Kreislaufphysiologie, Medizinische Fakultät und Universitätsklinikum Düsseldorf, Heinrich-Heine-Universität Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - André Spychala
- Institut für Herz- und Kreislaufphysiologie, Medizinische Fakultät und Universitätsklinikum Düsseldorf, Heinrich-Heine-Universität Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Axel Gödecke
- Institut für Herz- und Kreislaufphysiologie, Medizinische Fakultät und Universitätsklinikum Düsseldorf, Heinrich-Heine-Universität Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany.
| |
Collapse
|
20
|
Yan J, Song K, Bai Z, Ge RL. WY14643 improves left ventricular myocardial mitochondrial and systolic functions in obese rats under chronic persistent hypoxia via the PPARα pathway. Life Sci 2020; 266:118888. [PMID: 33310031 DOI: 10.1016/j.lfs.2020.118888] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 12/04/2020] [Accepted: 12/04/2020] [Indexed: 02/06/2023]
Abstract
AIMS Peroxisome proliferator-activated receptor (PPAR) α, a key regulator of lipid metabolism, plays a role in maintaining the homeostasis of myocardial energy metabolism. Both hypoxia and obesity inhibit the expression of PPARα in the myocardium. In this study, we verified the inhibitory effects of hypoxia and obesity on PPARα and examined whether WY14643 (4-chloro-6-(2,3-xylidino)-2-pyrimidinylthioacetic acid), an agonist of PPARα, ameliorates myocardial mitochondrial dysfunction and protects cardiac function in obese rats under chronic persistent hypoxia. MAIN METHODS Sprague-Dawley rats were randomly divided into six groups: a control group (normal chow diet, normal oxygen), a high-fat diet (HFD) group (normal oxygen), a chronic persistent hypoxia normal chow diet group, a chronic persistent hypoxia HFD group, a chronic persistent hypoxia HFD group with WY14643 treatment, and a chronic persistent hypoxia HFD group with vehicle treatment. KEY FINDINGS Hypoxia and obesity increased myocardial lipid accumulation, mitochondrial dysfunction, and left ventricular systolic dysfunction. Myocardial lipid metabolism-related genes, including those encoding PPARα, PPARγ coactivator 1α (PGC1α), and carnitine palmitoyl transferase 1α (CPT1α), were downregulated, while acetyl-CoA carboxylase 2 (ACC2) was upregulated under a combination of hypoxia and obesity. WY14643 upregulated PPARα, PGC1α, and CPT1α, and downregulated ACC2. WY14643 alleviated hypoxia- and obesity-induced myocardial lipid accumulation and improved mitochondrial and left ventricular systolic functions. SIGNIFICANCE WY14643 improved myocardial mitochondrial and left ventricular systolic functions in obese rats under chronic persistent hypoxia. Thus, WY14643 possibly exerts its effects by regulating the PPARα pathway and shows potential as a therapeutic target for cardiovascular diseases associated with obesity and hypoxia.
Collapse
Affiliation(s)
- Jun Yan
- Research Center for High Altitude Medicine, Qinghai University Medical College, Xining 810001, PR China; Key Laboratory of Persistent medicine (Qinghai University), Ministry of Education, Xining 810001, PR China; Key Laboratory for Application of Persistent Medicine in Qinghai Province, Xining 810001, PR China; Cardiovascular Medicine Department, Xuzhou Medical University affiliated hospital, Xuzhou 221006, PR China
| | - Kang Song
- Endocrinology Department, Qinghai Provincial People's Hospital, Xining 810000, PR China
| | - Zhenzhong Bai
- Research Center for High Altitude Medicine, Qinghai University Medical College, Xining 810001, PR China; Key Laboratory of Persistent medicine (Qinghai University), Ministry of Education, Xining 810001, PR China; Key Laboratory for Application of Persistent Medicine in Qinghai Province, Xining 810001, PR China
| | - Ri-Li Ge
- Research Center for High Altitude Medicine, Qinghai University Medical College, Xining 810001, PR China; Key Laboratory of Persistent medicine (Qinghai University), Ministry of Education, Xining 810001, PR China; Key Laboratory for Application of Persistent Medicine in Qinghai Province, Xining 810001, PR China.
| |
Collapse
|
21
|
Quesnelle K, Guimaraes DA, Rao K, Singh AB, Wang Y, Hogg N, Shiva S. Myoglobin promotes nitrite-dependent mitochondrial S-nitrosation to mediate cytoprotection after hypoxia/reoxygenation. Nitric Oxide 2020; 104-105:36-43. [PMID: 32891753 DOI: 10.1016/j.niox.2020.08.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 08/15/2020] [Accepted: 08/31/2020] [Indexed: 10/23/2022]
Abstract
It is well established that myoglobin supports mitochondrial respiration through the storage and transport of oxygen as well as through the scavenging of nitric oxide. However, during ischemia/reperfusion (I/R), myoglobin and mitochondria both propagate myocardial injury through the production of oxidants. Nitrite, an endogenous signaling molecule and dietary constituent, mediates potent cardioprotection after I/R and this effect relies on its interaction with both myoglobin and mitochondria. While independent mechanistic studies have demonstrated that nitrite-mediated cardioprotection requires the presence of myoglobin and the post-translational S-nitrosation of critical cysteine residues on mitochondrial complex I, it is unclear whether myoglobin directly catalyzes the S-nitrosation of complex I or whether mitochondrial-dependent nitrite reductase activity contributes to S-nitrosation. Herein, using purified myoglobin and isolated mitochondria, we characterize and directly compare the nitrite reductase activities of mitochondria and myoglobin and assess their contribution to mitochondrial S-nitrosation. We demonstrate that myoglobin is a significantly more efficient nitrite reductase than isolated mitochondria. Further, deoxygenated myoglobin catalyzes the nitrite-dependent S-nitrosation of mitochondrial proteins. This reaction is enhanced in the presence of oxidized (Fe3+) myoglobin and not significantly affected by inhibitors of mitochondrial respiration. Using a Chinese Hamster Ovary cell model stably transfected with human myoglobin, we show that both myoglobin and mitochondrial complex I expression are required for nitrite-dependent attenuation of cell death after anoxia/reoxygenation. These data expand the understanding of myoglobin's role both as a nitrite reductase to a mediator of S-nitrosation and as a regulator of mitochondrial function, and have implications for nitrite-mediated cardioprotection after I/R.
Collapse
Affiliation(s)
- Kelly Quesnelle
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Danielle A Guimaraes
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Krithika Rao
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | | | - Yinna Wang
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Neil Hogg
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Sruti Shiva
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, 15261, USA; Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA, 15261, USA.
| |
Collapse
|
22
|
Wang PY, Ma J, Li J, Starost MF, Wolfgang MJ, Singh K, Pirooznia M, Kang JG, Hwang PM. Reducing Fatty Acid Oxidation Improves Cancer-free Survival in a Mouse Model of Li-Fraumeni Syndrome. Cancer Prev Res (Phila) 2020; 14:31-40. [PMID: 32958587 DOI: 10.1158/1940-6207.capr-20-0368] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/11/2020] [Accepted: 09/15/2020] [Indexed: 11/16/2022]
Abstract
Germline mutations of TP53, which cause the cancer predisposition disorder Li-Fraumeni syndrome (LFS), can increase mitochondrial activity as well as fatty acid β-oxidation (FAO) in mice. Increased fatty acid metabolism can promote cancer malignancy, but its specific contribution to tumorigenesis in LFS remains unclear. To investigate this, we crossed LFS mice carrying the p53 R172H knock-in mutation (p53172H/H , homolog of the human TP53 R175H LFS mutation) with myoglobin-knockout (MB-/- ) mice known to have decreased FAO. MB-/- p53172H/H double-mutant mice also showed mildly reduced FAO in thymus, a common site of T lymphoma development in LFS mice, in association with an approximately 40% improvement in cancer-free survival time. RNA sequencing profiling revealed that the p53 R172H mutation promotes mitochondrial metabolism and ribosome biogenesis, both of which are suppressed by the disruption of MB. The activation of ribosomal protein S6, involved in protein translation and implicated in cancer promotion, was also inhibited in the absence of MB. To further confirm the role of FAO in lymphomagenesis, mitochondrial FAO enzyme, carnitine palmitoyltransferase 2 (CPT2), was specifically disrupted in T cells of p53172H/H mice using a Cre-loxP-mediated strategy. The heterozygous knockout of CPT2 resulted in thymus FAO haploinsufficiency and an approximately 30% improvement in survival time, paralleling the antiproliferative signaling observed with MB disruption. Thus, this study demonstrates that moderating FAO in LFS can suppress tumorigenesis and improve cancer-free survival with potential implications for cancer prevention. PREVENTION RELEVANCE: Mildly inhibiting the increased fatty acid oxidation observed in a mouse model of Li-Fraumeni syndrome, a cancer predisposition disorder caused by inherited mutations of TP53, dampens aberrant pro-tumorigenic cell signaling and improves the survival time of these mice, thereby revealing a potential strategy for cancer prevention in patients.
Collapse
Affiliation(s)
- Ping-Yuan Wang
- Cardiovascular Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland
| | - Jin Ma
- Cardiovascular Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland
| | - Jie Li
- Cardiovascular Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland
| | | | - Michael J Wolfgang
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Komudi Singh
- Bioinformatics and Computational Biology, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland
| | - Mehdi Pirooznia
- Bioinformatics and Computational Biology, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland
| | - Ju-Gyeong Kang
- Cardiovascular Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland
| | - Paul M Hwang
- Cardiovascular Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland.
| |
Collapse
|
23
|
Lessons from the post-genomic era: Globin diversity beyond oxygen binding and transport. Redox Biol 2020; 37:101687. [PMID: 32863222 PMCID: PMC7475203 DOI: 10.1016/j.redox.2020.101687] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 08/11/2020] [Accepted: 08/11/2020] [Indexed: 12/16/2022] Open
Abstract
Vertebrate hemoglobin (Hb) and myoglobin (Mb) were among the first proteins whose structures and sequences were determined over 50 years ago. In the subsequent pregenomic period, numerous related proteins came to light in plants, invertebrates and bacteria, that shared the myoglobin fold, a signature sequence motif characteristic of a 3-on-3 α-helical sandwich. Concomitantly, eukaryote and bacterial globins with a truncated 2-on-2 α-helical fold were discovered. Genomic information over the last 20 years has dramatically expanded the list of known globins, demonstrating their existence in a limited number of archaeal genomes, a majority of bacterial genomes and an overwhelming majority of eukaryote genomes. In vertebrates, 6 additional globin types were identified, namely neuroglobin (Ngb), cytoglobin (Cygb), globin E (GbE), globin X (GbX), globin Y (GbY) and androglobin (Adgb). Furthermore, functions beyond the familiar oxygen transport and storage have been discovered within the vertebrate globin family, including NO metabolism, peroxidase activity, scavenging of free radicals, and signaling functions. The extension of the knowledge on globin functions suggests that the original roles of bacterial globins must have been enzymatic, involved in defense against NO toxicity, and perhaps also as sensors of O2, regulating taxis away or towards high O2 concentrations. In this review, we aimed to discuss the evolution and remarkable functional diversity of vertebrate globins with particular focus on the variety of non-canonical expression sites of mammalian globins and their according impressive variability of atypical functions.
Collapse
|
24
|
Basmaeil Y, Rashid MA, Khatlani T, AlShabibi M, Bahattab E, Abdullah ML, Abomaray F, Kalionis B, Massoudi S, Abumaree M. Preconditioning of Human Decidua Basalis Mesenchymal Stem/Stromal Cells with Glucose Increased Their Engraftment and Anti-diabetic Properties. Tissue Eng Regen Med 2020; 17:209-222. [PMID: 32077075 PMCID: PMC7105536 DOI: 10.1007/s13770-020-00239-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 11/10/2019] [Accepted: 01/06/2020] [Indexed: 12/19/2022] Open
Abstract
Background: Mesenchymal stem/stromal cells (MSCs) from the decidua basalis (DBMSCs) of the human placenta have important functions that make them potential candidates for cellular therapy. Previously, we showed that DBMSC functions do not change significantly in a high oxidative stress environment, which was induced by hydrogen peroxide (H2O2) and immune cells. Here, we studied the consequences of glucose, another oxidative stress inducer, on the phenotypic and functional changes in DBMSCs. Methods: DBMSCs were exposed to a high level of glucose, and its effect on DBMSC phenotypic and functional properties was determined. DBMSC expression of oxidative stress and immune molecules after exposure to glucose were also identified. Results: Conditioning of DBMSCs with glucose improved their adhesion and invasion. Glucose also increased DBMSC expression of genes with survival, proliferation, migration, invasion, anti-inflammatory, anti-chemoattractant and antimicrobial properties. In addition, DBMSC expression of B7H4, an inhibitor of T cell proliferation was also enhanced by glucose. Interestingly, glucose modulated DBMSC expression of genes involved in insulin secretion and prevention of diabetes. Conclusion: These data show the potentially beneficial effects of glucose on DBMSC functions. Preconditioning of DBMSCs with glucose may therefore be a rational strategy for increasing their therapeutic potential by enhancing their engraftment efficiency. In addition, glucose may program DBMSCs into insulin producing cells with ability to counteract inflammation and infection associated with diabetes. However, future in vitro and in vivo studies are essential to investigate the findings of this study further. Electronic supplementary material The online version of this article (10.1007/s13770-020-00239-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yasser Basmaeil
- Stem Cells and Regenerative Medicine Department, King Abdullah International Medical Research Center, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Mail Code 1515, P.O. Box 22490, Riyadh, 11426, Kingdom of Saudi Arabia.
| | - Manar Al Rashid
- Stem Cells and Regenerative Medicine Department, King Abdullah International Medical Research Center, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Mail Code 1515, P.O. Box 22490, Riyadh, 11426, Kingdom of Saudi Arabia
| | - Tanvir Khatlani
- Stem Cells and Regenerative Medicine Department, King Abdullah International Medical Research Center, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Mail Code 1515, P.O. Box 22490, Riyadh, 11426, Kingdom of Saudi Arabia
| | - Manal AlShabibi
- National Center for Stem Cell Technology, Life Sciences and Environment Research Institute, King Abdulaziz City for Science and Technology, P.O Box 6086, Riyadh, 11442, Kingdom of Saudi Arabia
| | - Eman Bahattab
- National Center for Stem Cell Technology, Life Sciences and Environment Research Institute, King Abdulaziz City for Science and Technology, P.O Box 6086, Riyadh, 11442, Kingdom of Saudi Arabia
| | - Meshan L Abdullah
- Experimental Medicine, King Abdullah International Medical Research Center MNG-HA, Ali Al Arini, Ar Rimayah, Riyadh, 11481, Kingdom of Saudi Arabia
| | - Fawaz Abomaray
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, 14186, Stockholm, Sweden
| | - Bill Kalionis
- Department of Maternal-Fetal Medicine, Pregnancy Research Centre and University of Melbourne, Parkville, VIC, 3010, Australia.,Department of Obstetrics and Gynaecology, Royal Women's Hospital, 20 Flemington Rd, Parkville, VIC, 3052, Australia
| | - Safia Massoudi
- Department of Forensic Biology, College of Forensic Sciences, Naif Arab University for Security Sciences, Khurais Rd, Ar Rimayah, Riyadh, 14812, Kingdom of Saudi Arabia
| | - Mohammad Abumaree
- Stem Cells and Regenerative Medicine Department, King Abdullah International Medical Research Center, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Mail Code 1515, P.O. Box 22490, Riyadh, 11426, Kingdom of Saudi Arabia.,College of Science and Health Professions, King Saud Bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Mail Code 3124, P.O. Box 3660, Riyadh, 11481, Kingdom of Saudi Arabia
| |
Collapse
|
25
|
Hochman-Mendez C, Pereira de Campos DB, Pinto RS, Mendes BJDS, Rocha GM, Monnerat G, Weissmuller G, Sampaio LC, Carvalho AB, Taylor DA, de Carvalho ACC. Tissue-engineered human embryonic stem cell-containing cardiac patches: evaluating recellularization of decellularized matrix. J Tissue Eng 2020; 11:2041731420921482. [PMID: 32742631 PMCID: PMC7375712 DOI: 10.1177/2041731420921482] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 03/27/2020] [Indexed: 02/05/2023] Open
Abstract
Decellularized cardiac extracellular matrix scaffolds with preserved composition and architecture can be used in tissue engineering to reproduce the complex cardiac extracellular matrix. However, evaluating the extent of cardiomyocyte repopulation of decellularized cardiac extracellular matrix scaffolds after recellularization attempts is challenging. Here, we describe a unique combination of biochemical, biomechanical, histological, and physiological parameters for quantifying recellularization efficiency of tissue-engineered cardiac patches compared with native cardiac tissue. Human embryonic stem cell-derived cardiomyocytes were seeded into rat heart atrial and ventricular decellularized cardiac extracellular matrix patches. Confocal and atomic force microscopy showed cell integration within the extracellular matrix basement membrane that was accompanied by restoration of native cardiac tissue passive mechanical properties. Multi-electrode array and immunostaining (connexin 43) were used to determine synchronous field potentials with electrical coupling. Myoglobin content (~60%) and sarcomere length measurement (>45% vs 2D culture) were used to evaluate cardiomyocyte maturation of integrated cells. The combination of these techniques allowed us to demonstrate that as cellularization efficiency improves, cardiomyocytes mature and synchronize electrical activity, and tissue mechanical/biochemical properties improve toward those of native tissue.
Collapse
Affiliation(s)
- Camila Hochman-Mendez
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Regenerative Medicine Research, Texas Heart Institute, Houston, TX, USA
| | - Dilza Balteiro Pereira de Campos
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Regenerative Medicine Research, Texas Heart Institute, Houston, TX, USA
| | - Rafael Serafim Pinto
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Gustavo Miranda Rocha
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gustavo Monnerat
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gilberto Weissmuller
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luiz C Sampaio
- Regenerative Medicine Research, Texas Heart Institute, Houston, TX, USA
| | - Adriana Bastos Carvalho
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- National Institute of Science and Technology for Regenerative Medicine, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Doris A Taylor
- Regenerative Medicine Research, Texas Heart Institute, Houston, TX, USA
| | - Antonio Carlos Campos de Carvalho
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- National Institute of Science and Technology for Regenerative Medicine, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
26
|
Bicker A, Nauth T, Gerst D, Aboouf MA, Fandrey J, Kristiansen G, Gorr TA, Hankeln T. The role of myoglobin in epithelial cancers: Insights from transcriptomics. Int J Mol Med 2019; 45:385-400. [PMID: 31894249 PMCID: PMC6984796 DOI: 10.3892/ijmm.2019.4433] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 11/08/2019] [Indexed: 12/25/2022] Open
Abstract
The muscle-associated respiratory protein myoglobin (MB) is expressed in multiple types of cancer, including breast and prostate tumors. In Kaplan-Meier analyses of the two tumor types, MB positivity is associated with favorable prognoses. Despite its well-characterized function in myocytes, the role of MB in cancer remains unclear. To study the impact of endogenous MB expression, small interfering RNA MB-knockdown cells were engineered using breast, prostate and colon cancer cell lines (MDA-MB468, LNCaP, DLD-1), and their transcriptomes were investigated using RNA-Seq at different oxygen levels. In MB-positive cells, increased expression of glycolytic genes was observed, which was possibly mediated by a higher activity of hypoxia-inducible factor 1α. In addition, the results of the gene set enrichment analysis suggested that MB contributed to fatty acid transport and turnover. MB-positive, wild-type-p53 LNCaP cells also exhibited increased expression of p53 target genes involved in cell cycle checkpoint control and prevention of cell migration. MB-positive cells expressing mutant p53 exhibited upregulation of genes associated with prolonged cancer cell viability and motility. Therefore, it was hypothesized that these transcriptomic differences may result from MB-mediated generation of nitric oxide or reactive oxygen species, thus employing established enzymatic activities of the globin. In summary, the transcriptome comparisons identified potential molecular functions of MB in carcinogenesis by highlighting the interaction of MB with key metabolic and regulatory processes.
Collapse
Affiliation(s)
- Anne Bicker
- Institute of Organismic and Molecular Evolution, Molecular Genetics and Genome Analysis, Johannes Gutenberg University, D‑55099 Mainz, Germany
| | - Theresa Nauth
- Institute of Organismic and Molecular Evolution, Molecular Genetics and Genome Analysis, Johannes Gutenberg University, D‑55099 Mainz, Germany
| | - Daniela Gerst
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, CH‑8057 Zurich, Switzerland
| | - Mostafa Ahmed Aboouf
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, CH‑8057 Zurich, Switzerland
| | - Joachim Fandrey
- Institute of Physiology, University of Duisburg‑Essen, D‑45147 Essen, Germany
| | - Glen Kristiansen
- Institute of Pathology, Center for Integrated Oncology, University Hospital Bonn, University of Bonn, D‑53127 Bonn, Germany
| | - Thomas Alexander Gorr
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, CH‑8057 Zurich, Switzerland
| | - Thomas Hankeln
- Institute of Organismic and Molecular Evolution, Molecular Genetics and Genome Analysis, Johannes Gutenberg University, D‑55099 Mainz, Germany
| |
Collapse
|
27
|
Catalase-Like Antioxidant Activity is Unaltered in Hypochlorous Acid Oxidized Horse Heart Myoglobin. Antioxidants (Basel) 2019; 8:antiox8090414. [PMID: 31540488 PMCID: PMC6770884 DOI: 10.3390/antiox8090414] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 08/27/2019] [Accepted: 09/16/2019] [Indexed: 01/01/2023] Open
Abstract
Activated neutrophils release myeloperoxidase that produces the potent oxidant hypochlorous acid (HOCl). Exposure of the oxygen transport protein horse heart myoglobin (hhMb) to HOCl inhibits Iron III (Fe(III))-heme reduction by cytochrome b5 to oxygen-binding Iron II (Fe(II))Mb. Pathological concentrations of HOCl yielded myoglobin oxidation products of increased electrophoretic mobility and markedly different UV/Vis absorbance. Mass analysis indicated HOCl caused successive mass increases of 16 a.m.u., consistent serial addition of molecular oxygen to the protein. By contrast, parallel analysis of protein chlorination by quantitative mass spectrometry revealed a comparatively minor increase in the 3-chlorotyrosine/tyrosine ratio. Pre-treatment of hhMb with HOCl affected the peroxidase reaction between the hemoprotein and H2O2 as judged by a HOCl dose-dependent decrease in spin-trapped tyrosyl radical detected by electron paramagnetic resonance (EPR) spectroscopy and the rate constant of 2,2'-azino-bis(3-ethylbenzothiazoline-6-sulphonic acid (ABTS) oxidation. By contrast, Mb catalase-like antioxidant activity remained unchanged under the same conditions. Notably, HOCl-modification of Mb decreased the rate of ferric-to-ferrous Mb reduction by a cytochrome b5 reductase system. Taken together, these data indicate oxidizing HOCl promotes Mb oxidation but not chlorination and that oxidized Mb shows altered Mb peroxidase-like activity and diminished rates of one-electron reduction by cytochrome b5 reductase, possibly affecting oxygen storage and transport however, Mb-catalase-like antioxidant activity remains unchanged.
Collapse
|
28
|
Plöhn S, Hose M, Schlüter A, Michel L, Diaz-Cano S, Hendgen-Cotta UB, Banga JP, Bechrakis NE, Hansen W, Eckstein A, Berchner-Pfannschmidt U. Fingolimod Improves the Outcome of Experimental Graves' Disease and Associated Orbitopathy by Modulating the Autoimmune Response to the Thyroid-Stimulating Hormone Receptor. Thyroid 2019; 29:1286-1301. [PMID: 31237525 DOI: 10.1089/thy.2018.0754] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Graves' disease (GD) and Graves' orbitopathy are associated with stimulating thyrotropin receptor (TSHR) autoantibodies and autoreactive T cells. Recent in vitro studies suggested that sphingosine-1-phosphate (S1P) signaling is involved in the pathogenesis of orbitopathy. In this study, we explored the immune modulatory potential of S1P receptor antagonist fingolimod in a murine model for GD. Fingolimod was orally administered preventively during disease onset or therapeutically after disease onset. Administration of fingolimod during disease onset completely prevented the formation of TSHR-stimulating autoantibodies. Intervention after disease onset rarely reduced TSHR-stimulating autoantibodies and blocking autoantibodies were induced in some animals. Consequently, autoimmune hyperthyroidism characterized by elevated serum thyroxin levels, hyperplastic thyroid morphology accompanied by T cell infiltration, weight gain, enhanced body temperature, and tachycardia did not manifest preventively and showed milder manifestation in therapeutically treated animals. Importantly, examination of orbital tissue showed significant amelioration of orbitopathy manifestations through reduction of T cell infiltration, adipogenesis, and hyaluronan deposition. Autoimmune hyperthyroidism and orbitopathy were accompanied by changes in peripheral and splenic T cell proportions with high CD3+, CD4+, and CD8+ T cells. Activated T cells CD4+CD25+ were elevated whereas regulatory T cells CD4+Foxp3+ cells remained unchanged in spleens. Fingolimod decreased elevated T cell levels and increased CD4+CD25+Foxp3+ regulatory T cell populations. Analysis of total disease outcome revealed that treatment during disease onset protected animals against autoimmune hyperthyroidism and orbitopathy. Of note, therapeutic intervention after disease onset suppressed disease in half of the animals and in the other half disease remained at mild stages. The results of this study support a clinical trial to investigate the immunologic and clinical benefits of early treatment with S1P-based drugs in GD.
Collapse
Affiliation(s)
- Svenja Plöhn
- Molecular Ophthalmology, Department of Ophthalmology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Matthias Hose
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Anke Schlüter
- Department of Oto-Rhino-Laryngology, Head and Neck Surgery, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Lars Michel
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Salvador Diaz-Cano
- Department of Histopathology, King's College Hospital, King's College, London, United Kingdom
| | - Ulrike B Hendgen-Cotta
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Jasvinder Paul Banga
- Molecular Ophthalmology, Department of Ophthalmology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Nikolaos E Bechrakis
- Department of Ophthalmology, University Hospital Essen, University Duisburg-Essen, Germany
| | - Wiebke Hansen
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Anja Eckstein
- Molecular Ophthalmology, Department of Ophthalmology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Utta Berchner-Pfannschmidt
- Molecular Ophthalmology, Department of Ophthalmology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| |
Collapse
|
29
|
Braganza A, Quesnelle K, Bickta J, Reyes C, Wang Y, Jessup M, St Croix C, Arlotti J, Singh SV, Shiva S. Myoglobin induces mitochondrial fusion, thereby inhibiting breast cancer cell proliferation. J Biol Chem 2019; 294:7269-7282. [PMID: 30872402 DOI: 10.1074/jbc.ra118.006673] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 02/21/2019] [Indexed: 01/11/2023] Open
Abstract
Myoglobin is a monomeric heme protein expressed ubiquitously in skeletal and cardiac muscle and is traditionally considered to function as an oxygen reservoir for mitochondria during hypoxia. It is now well established that low concentrations of myoglobin are aberrantly expressed in a significant proportion of breast cancer tumors. Despite being expressed only at low levels in these tumors, myoglobin is associated with attenuated tumor growth and a better prognosis in breast cancer patients, but the mechanism of this myoglobin-mediated protection against further cancer growth remains unclear. Herein, we report a signaling pathway by which myoglobin regulates mitochondrial dynamics and thereby decreases cell proliferation. We demonstrate in vitro that expression of human myoglobin in MDA-MB-231, MDA-MB-468, and MCF7 breast cancer cells induces mitochondrial hyperfusion by up-regulating mitofusins 1 and 2, the predominant catalysts of mitochondrial fusion. This hyperfusion causes cell cycle arrest and subsequent inhibition of cell proliferation. Mechanistically, increased mitofusin expression was due to myoglobin-dependent free-radical production, leading to the oxidation and degradation of the E3 ubiquitin ligase parkin. We recapitulated this pathway in a murine model in which myoglobin-expressing xenografts exhibited decreased tumor volume with increased mitofusin, markers of cell cycle arrest, and decreased parkin expression. Furthermore, in human triple-negative breast tumor tissues, mitofusin and myoglobin levels were positively correlated. Collectively, these results elucidate a new function for myoglobin as a modulator of mitochondrial dynamics and reveal a novel pathway by which myoglobin decreases breast cancer cell proliferation and tumor growth by up-regulating mitofusin levels.
Collapse
Affiliation(s)
| | | | - Janelle Bickta
- the Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, Pennsylvania 15261
| | - Christopher Reyes
- the Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, Pennsylvania 15261
| | - Yinna Wang
- From the Vascular Medicine Institute and
| | | | | | - Julie Arlotti
- Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, and.,University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania 15232
| | - Shivendra V Singh
- Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, and.,University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania 15232
| | - Sruti Shiva
- From the Vascular Medicine Institute and .,Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, and.,Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261
| |
Collapse
|
30
|
Moreira GCM, Boschiero C, Cesar ASM, Reecy JM, Godoy TF, Pértille F, Ledur MC, Moura ASAMT, Garrick DJ, Coutinho LL. Integration of genome wide association studies and whole genome sequencing provides novel insights into fat deposition in chicken. Sci Rep 2018; 8:16222. [PMID: 30385857 PMCID: PMC6212401 DOI: 10.1038/s41598-018-34364-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 10/12/2018] [Indexed: 02/07/2023] Open
Abstract
Excessive fat deposition is a negative factor for poultry production because it reduces feed efficiency, increases the cost of meat production and is a health concern for consumers. We genotyped 497 birds from a Brazilian F2 Chicken Resource Population, using a high-density SNP array (600 K), to estimate the genomic heritability of fat deposition related traits and to identify genomic regions and positional candidate genes (PCGs) associated with these traits. Selection signature regions, haplotype blocks and SNP data from a previous whole genome sequencing study in the founders of this chicken F2 population were used to refine the list of PCGs and to identify potential causative SNPs. We obtained high genomic heritabilities (0.43-0.56) and identified 22 unique QTLs for abdominal fat and carcass fat content traits. These QTLs harbored 26 PCGs involved in biological processes such as fat cell differentiation, insulin and triglyceride levels, and lipid biosynthetic process. Three of these 26 PCGs were located within haplotype blocks there were associated with fat traits, five overlapped with selection signature regions, and 12 contained predicted deleterious variants. The identified QTLs, PCGs and potentially causative SNPs provide new insights into the genetic control of fat deposition and can lead to improved accuracy of selection to reduce excessive fat deposition in chickens.
Collapse
Affiliation(s)
| | - Clarissa Boschiero
- Department of Animal Science, University of São Paulo, Piracicaba, SP, Brazil
| | | | - James M Reecy
- Department of Animal Science, Iowa State University, Ames, IA, USA
| | | | - Fábio Pértille
- Department of Animal Science, University of São Paulo, Piracicaba, SP, Brazil
| | | | | | - Dorian J Garrick
- School of Agriculture, Massey University, Ruakura, Hamilton, New Zealand
| | | |
Collapse
|
31
|
Chintapalli SV, Anishkin A, Adams SH. Exploring the entry route of palmitic acid and palmitoylcarnitine into myoglobin. Arch Biochem Biophys 2018; 655:56-66. [PMID: 30092229 DOI: 10.1016/j.abb.2018.07.024] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 07/23/2018] [Accepted: 07/31/2018] [Indexed: 11/26/2022]
Abstract
Myoglobin, besides its role in oxygen turnover, has gained recognition as a potential regulator of lipid metabolism. Previously, we confirmed the interaction of fatty acids and acylcarnitines with Oxy-Myoglobin, using both molecular dynamic simulations and Isothermal Titration Calorimetry studies. However, those studies were limited to testing only the binding sites derived from homology to fatty acid binding proteins and predictions using automated docking. To explore the entry mechanisms of the lipid ligands into myoglobin, we conducted molecular dynamic simulations of murine Oxy- and Deoxy-Mb structures with palmitate or palmitoylcarnitine starting at different positions near the protein surface. The simulations indicated that both ligands readily (under ∼10-20 ns) enter the Oxy-Mb structure through a dynamic area ("portal region") near heme, known to be the entry point for small molecule gaseous ligands like O2, CO and NO. The entry is not observed with Deoxy-Mb where lipid ligands move away from protein surface, due to a compaction of the entry portal and the heme-containing crevice in the Mb protein upon O2 removal. The results suggest quick spontaneous binding of lipids to Mb driven by hydrophobic interactions, strongly enhanced by oxygenation, and consistent with the emergent role of Mb in lipid metabolism.
Collapse
Affiliation(s)
- Sree V Chintapalli
- Arkansas Children's Nutrition Center -and- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, USA.
| | - Andriy Anishkin
- Department of Biology, University of Maryland, College Park, USA
| | - Sean H Adams
- Arkansas Children's Nutrition Center -and- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, USA
| |
Collapse
|
32
|
Zhou T, Prather ER, Garrison DE, Zuo L. Interplay between ROS and Antioxidants during Ischemia-Reperfusion Injuries in Cardiac and Skeletal Muscle. Int J Mol Sci 2018; 19:ijms19020417. [PMID: 29385043 PMCID: PMC5855639 DOI: 10.3390/ijms19020417] [Citation(s) in RCA: 132] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 01/14/2018] [Accepted: 01/21/2018] [Indexed: 12/16/2022] Open
Abstract
Ischemia reperfusion (IR), present in myocardial infarction or extremity injuries, is a major clinical issue and leads to substantial tissue damage. Molecular mechanisms underlying IR injury in striated muscles involve the production of reactive oxygen species (ROS). Excessive ROS accumulation results in cellular oxidative stress, mitochondrial dysfunction, and initiation of cell death by activation of the mitochondrial permeability transition pore. Elevated ROS levels can also decrease myofibrillar Ca2+ sensitivity, thereby compromising muscle contractile function. Low levels of ROS can act as signaling molecules involved in the protective pathways of ischemic preconditioning (IPC). By scavenging ROS, antioxidant therapies aim to prevent IR injuries with positive treatment outcomes. Novel therapies such as postconditioning and pharmacological interventions that target IPC pathways hold great potential in attenuating IR injuries. Factors such as aging and diabetes could have a significant impact on the severity of IR injuries. The current paper aims to provide a comprehensive review on the multifaceted roles of ROS in IR injuries, with a focus on cardiac and skeletal muscle, as well as recent advancement in ROS-related therapies.
Collapse
Affiliation(s)
- Tingyang Zhou
- Radiologic Sciences and Respiratory Therapy Division, School of Health and Rehabilitation Sciences, The Ohio State University College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA.
- Interdisciplinary Biophysics Graduate Program, The Ohio State University, Columbus, OH 43210, USA.
| | - Evan R Prather
- Radiologic Sciences and Respiratory Therapy Division, School of Health and Rehabilitation Sciences, The Ohio State University College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA.
| | - Davis E Garrison
- Radiologic Sciences and Respiratory Therapy Division, School of Health and Rehabilitation Sciences, The Ohio State University College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA.
| | - Li Zuo
- Radiologic Sciences and Respiratory Therapy Division, School of Health and Rehabilitation Sciences, The Ohio State University College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA.
- Interdisciplinary Biophysics Graduate Program, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
33
|
Angelini A, Pi X, Xie L. Dioxygen and Metabolism; Dangerous Liaisons in Cardiac Function and Disease. Front Physiol 2017; 8:1044. [PMID: 29311974 PMCID: PMC5732914 DOI: 10.3389/fphys.2017.01044] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 11/29/2017] [Indexed: 12/19/2022] Open
Abstract
The heart must consume a significant amount of energy to sustain its contractile activity. Although the fuel demands are huge, the stock remains very low. Thus, in order to supply its daily needs, the heart must have amazing adaptive abilities, which are dependent on dioxygen availability. However, in myriad cardiovascular diseases, “fuel” depletion and hypoxia are common features, leading cardiomyocytes to favor low-dioxygen-consuming glycolysis rather than oxidation of fatty acids. This metabolic switch makes it challenging to distinguish causes from consequences in cardiac pathologies. Finally, despite the progress achieved in the past few decades, medical treatments have not improved substantially, either. In such a situation, it seems clear that much remains to be learned about cardiac diseases. Therefore, in this review, we will discuss how reconciling dioxygen availability and cardiac metabolic adaptations may contribute to develop full and innovative strategies from bench to bedside.
Collapse
Affiliation(s)
- Aude Angelini
- Department of Medicine-Athero and Lipo, Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, United States
| | - Xinchun Pi
- Department of Medicine-Athero and Lipo, Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, United States
| | - Liang Xie
- Department of Medicine-Athero and Lipo, Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|