51
|
Anatolou D, Dovrolis N, Ragia G, Kolios G, Manolopoulos VG. Unpacking COVID-19 Systems Biology in Lung and Whole Blood with Transcriptomics and miRNA Regulators. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2022; 26:608-621. [PMID: 36269619 DOI: 10.1089/omi.2022.0104] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
COVID-19 is a systemic disease affecting tissues and organs, including and beyond the lung. Apart from the current pandemic context, we also have vastly inadequate knowledge of consequences of repeated exposures to SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2), the virus causing COVID-19, in multiple organ systems and the whole organism scales when the disease evolves from a pandemic to an endemic state. This calls for a systems biology and systems medicine approach and unpacking the effects of COVID-19 in lung as well as other tissues. We report here original findings from transcriptomics analyses and differentially expressed genes (DEGs) in lung samples from 60 patients and 27 healthy controls, and in whole blood samples from 255 patients and 103 healthy individuals. A total of 11 datasets with RNA-seq transcriptomic data were obtained from the Gene Expression Omnibus and the European Nucleotide Archive. The identified DEGs were used to construct protein interaction and functional networks and to identify related pathways and miRNAs. We found 35 DEGs common between lung and the whole blood, and importantly, 2 novel genes, namely CYP1B1 and TNFAIP6, which have not been previously implicated with COVID-19. We also identified four novel miRNA potential regulators, hsa-mir-192-5p, hsa-mir-221-3p, hsa-mir-4756-3p, and hsa-mir-10a-5p, implicated in lung or other diseases induced by coronaviruses. In summary, these findings offer new molecular leads and insights to unpack COVID-19 systems biology in a whole organism context and might inform future antiviral drug, diagnostics, and vaccine discovery efforts.
Collapse
Affiliation(s)
- Dimitra Anatolou
- Laboratory of Pharmacology, Department of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
- Individualised Medicine & Pharmacological Research Solutions Center (IMPReS), Alexandroupolis, Greece
| | - Nikolas Dovrolis
- Laboratory of Pharmacology, Department of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
- Individualised Medicine & Pharmacological Research Solutions Center (IMPReS), Alexandroupolis, Greece
| | - Georgia Ragia
- Laboratory of Pharmacology, Department of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
- Individualised Medicine & Pharmacological Research Solutions Center (IMPReS), Alexandroupolis, Greece
| | - George Kolios
- Laboratory of Pharmacology, Department of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
- Individualised Medicine & Pharmacological Research Solutions Center (IMPReS), Alexandroupolis, Greece
| | - Vangelis G Manolopoulos
- Laboratory of Pharmacology, Department of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
- Individualised Medicine & Pharmacological Research Solutions Center (IMPReS), Alexandroupolis, Greece
- Clinical Pharmacology Unit, Academic General Hospital of Alexandroupolis, Alexandroupolis, Greece
| |
Collapse
|
52
|
Analysis of the Plasmid-Based ts Allele of PA0006 Reveals Its Function in Regulation of Cell Morphology and Biosynthesis of Core Lipopolysaccharide in Pseudomonas aeruginosa. Appl Environ Microbiol 2022; 88:e0048022. [PMID: 35762790 PMCID: PMC9317947 DOI: 10.1128/aem.00480-22] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Over 300 essential genes are predicted using transposon sequencing in the genome of Pseudomonas aeruginosa. However, methods for reverse genetic analysis of essential genes are scarce. To address this issue, we developed a three-step protocol consisting of integration of deletion plasmid, introduction of temperature-sensitive rescue plasmid, and excision of integrated-deletion plasmid to construct the plasmid-based temperature-sensitive allele of essential genes. Using PA0006 as an example, we showed that PA0006(Ts) exhibited wild-type cell morphology at permissive temperature but filamentous form at restrictive temperatures. We further showed that the glycerol-mannoheptose-bisphosphate phosphatase GmhB in Escherichia coli shared 32.4% identity with that of PA0006p and functionally complemented the defect of PA0006(Ts) at 42°C. SDS-PAGE and Western blotting indicated the presence and absence of the complete core lipopolysaccharide (LPS) and B-band O-antigen in PA0006(Ts) at 30 and 42°C, respectively. An isolated suppressor sup displayed wild-type-like cell morphology but no complete core LPS or O-antigen. Genome resequencing together with comparative transcriptomic profiling identified a candidate suppressor fructose-bisphosphate phosphatase in which the promoter harbored a SNP and the transcription level was not downregulated at 42°C compared to 30°C in sup. It was further validated that fbp overexpression suppressed the lethality of PA0006(Ts) at 42°C. Taken together, our results demonstrate that PA0006 plays a role in regulation of cell morphology and biosynthesis of core LPS. This three-step protocol for construction of conditional lethal allele in P. aeruginosa should be widely applicable for genetic analysis of other essential genes of interest, including analysis of bypass suppressibility. IMPORTANCE Microbial essential genes encode nondispensable function for cell growth and therefore are ideal targets for the development of new drugs. Essential genes are readily identified using transposon-sequencing technology at the genome scale. However, genetic analysis of essential genes of interest was hampered by limited methodologies. To address this issue, we developed a three-step protocol for construction of conditional allele of essential genes in the opportunistic pathogen Pseudomonas aeruginosa. Using PA0006 as an example, we demonstrated that the plasmid-based PA0006(Ts) mutant exhibited defects in regulation of cell morphology, formation of intact core LPS, and attachment of the O-antigen at restrictive temperatures but not at permissive temperatures. A suppressor of PA0006(Ts) was isolated through spontaneous mutations and showed restored cell morphology but not core oligosaccharide or O-antigen. This method should be widely applicable for phenotype and suppressibility analyses of other essential genes of interest in P. aeruginosa.
Collapse
|
53
|
Wakamatsu T, Ogawa H, Yoshida K, Matsuoka Y, Shizuma K, Imura Y, Tamiya H, Nakai S, Yagi T, Nagata S, Yui Y, Sasagawa S, Takenaka S. Establishment of Organoids From Human Epithelioid Sarcoma With the Air-Liquid Interface Organoid Cultures. Front Oncol 2022; 12:893592. [PMID: 35677170 PMCID: PMC9169059 DOI: 10.3389/fonc.2022.893592] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 04/20/2022] [Indexed: 01/02/2023] Open
Abstract
Background Although biological resources are essential for basic and preclinical research in the oncological field, those of sarcoma are not sufficient for rapid development of the treatment. So far, some sarcoma cell lines have been established, however, the success rate was low and the established sarcoma types were frequently biased. Therefore, an efficient culture method is needed to determine the various types of sarcomas. Organoid culture is a 3-dimentional culture method that enables the recapitulation of the tumor microenvironment and the success rate reported is higher than the 2-dimentional culture. The purpose of this study was to report our newly established organoids from human epithelioid sarcoma using the air-liquid interface organoid culture method. Methods We treated 2 patients with epithelioid sarcoma in our institute. The remaining sarcoma specimens after surgical resection were embedded in collagen type 1 gels according to the air-liquid interface organoid culture method. After serial passages, we xenografted the organoids to NOD-scid IL2Rgnull (NSG) mice. Using the developed tumors, we performed histological and genomic analyses to compare the similarities and differences with the original epithelioid sarcoma from the patient. Results Organoids from the epithelioid sarcoma could be serially cultured and maintained in collagen type 1 gels for more than 3 passages. Developed orthotopic tumor xenografts were detected in the NSG mice. After the process was repeated severally, the patient derived organoid lines from the epithelioid sarcoma were established. The established organoids showed loss of integrase interactor 1 expression with polymerase chain reaction and immunohistochemical analyses. The xenografted organoids of the epithelioid sarcoma had histologically similar phenotypes with the original tumor and genetically resembled it to some degree. Conclusions The present study demonstrated 2 novel established organoid models of epithelioid sarcoma, and our organoid models could be used to investigate the molecular pathogenesis and develop a novel treatment.
Collapse
Affiliation(s)
- Toru Wakamatsu
- Department of Musculoskeletal Oncology Service, Osaka International Cancer Institute, Osaka, Japan.,Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hisataka Ogawa
- Nitto joint Research Department for Nucleic Acid Medicine, Research Center, Osaka International Cancer Institute, Osaka, Japan.,Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Keiichi Yoshida
- Next-generation Precision Medicine Research Center, Osaka International Cancer Institute, Osaka, Japan
| | - Yukiko Matsuoka
- Next-generation Precision Medicine Research Center, Osaka International Cancer Institute, Osaka, Japan
| | - Kazuko Shizuma
- Nitto joint Research Department for Nucleic Acid Medicine, Research Center, Osaka International Cancer Institute, Osaka, Japan
| | - Yoshinori Imura
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hironari Tamiya
- Department of Musculoskeletal Oncology Service, Osaka International Cancer Institute, Osaka, Japan.,Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Sho Nakai
- Department of Musculoskeletal Oncology Service, Osaka International Cancer Institute, Osaka, Japan.,Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Toshinari Yagi
- Department of Musculoskeletal Oncology Service, Osaka International Cancer Institute, Osaka, Japan
| | - Shigenori Nagata
- Department of Diagnostic Pathology and Cytology, Osaka International Cancer Institute, Osaka, Japan
| | - Yoshihiro Yui
- Sarcoma Treatment Laboratory, Research Institute, Nozaki Tokushukai Hospital, Osaka, Japan
| | - Satoru Sasagawa
- Molecular Biology Laboratory, Research Institute, Nozaki Tokushukai Hospital, Osaka, Japan
| | - Satoshi Takenaka
- Department of Musculoskeletal Oncology Service, Osaka International Cancer Institute, Osaka, Japan.,Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
54
|
Prieto C, Barrios D, Villaverde A. SingleCAnalyzer: Interactive Analysis of Single Cell RNA-Seq Data on the Cloud. FRONTIERS IN BIOINFORMATICS 2022; 2:793309. [PMID: 36304292 PMCID: PMC9580930 DOI: 10.3389/fbinf.2022.793309] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 05/09/2022] [Indexed: 12/13/2022] Open
Abstract
Single-cell RNA sequencing (scRNA-Seq) enables researchers to quantify the transcriptomes of individual cells. The capacity of researchers to perform this type of analysis has allowed researchers to undertake new scientific goals. The usefulness of scRNA-Seq has depended on the development of new computational biology methods, which have been designed to meeting challenges associated with scRNA-Seq analysis. However, the proper application of these computational methods requires extensive bioinformatics expertise. Otherwise, it is often difficult to obtain reliable and reproducible results. We have developed SingleCAnalyzer, a cloud platform that provides a means to perform full scRNA-Seq analysis from FASTQ within an easy-to-use and self-exploratory web interface. Its analysis pipeline includes the demultiplexing and alignment of FASTQ files, read trimming, sample quality control, feature selection, empty droplets detection, dimensional reduction, cellular type prediction, unsupervised clustering of cells, pseudotime/trajectory analysis, expression comparisons between groups, functional enrichment of differentially expressed genes and gene set expression analysis. Results are presented with interactive graphs, which provide exploratory and analytical features. SingleCAnalyzer is freely available at https://singleCAnalyzer.eu.
Collapse
|
55
|
Katsumura S, Siddiqui N, Goldsmith MR, Cheah JH, Fujikawa T, Minegishi G, Yamagata A, Yabuki Y, Kobayashi K, Shirouzu M, Inagaki T, Huang THM, Musi N, Topisirovic I, Larsson O, Morita M. Deadenylase-dependent mRNA decay of GDF15 and FGF21 orchestrates food intake and energy expenditure. Cell Metab 2022; 34:564-580.e8. [PMID: 35385705 PMCID: PMC9386786 DOI: 10.1016/j.cmet.2022.03.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 10/26/2021] [Accepted: 03/14/2022] [Indexed: 12/11/2022]
Abstract
Hepatokines, secretory proteins from the liver, mediate inter-organ communication to maintain a metabolic balance between food intake and energy expenditure. However, molecular mechanisms by which hepatokine levels are rapidly adjusted following stimuli are largely unknown. Here, we unravel how CNOT6L deadenylase switches off hepatokine expression after responding to stimuli (e.g., exercise and food) to orchestrate energy intake and expenditure. Mechanistically, CNOT6L inhibition stabilizes hepatic Gdf15 and Fgf21 mRNAs, increasing corresponding serum protein levels. The resulting upregulation of GDF15 stimulates the hindbrain to suppress appetite, while increased FGF21 affects the liver and adipose tissues to induce energy expenditure and lipid consumption. Despite the potential of hepatokines to treat metabolic disorders, their administration therapies have been challenging. Using small-molecule screening, we identified a CNOT6L inhibitor enhancing GDF15 and FGF21 hepatokine levels, which dramatically improves diet-induced metabolic syndrome. Our discovery, therefore, lays the foundation for an unprecedented strategy to treat metabolic syndrome.
Collapse
Affiliation(s)
- Sakie Katsumura
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Nadeem Siddiqui
- Department of Biochemistry and Goodman Cancer Research Centre, McGill University, Montreal, QC H3A 1A3, Canada
| | | | - Jaime H Cheah
- High Throughput Sciences Facility, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Teppei Fujikawa
- Center for Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Genki Minegishi
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan
| | - Atsushi Yamagata
- RIKEN Center for Biosystems Dynamics Research, Yokohama, Kanagawa 230-0045, Japan
| | - Yukako Yabuki
- RIKEN Center for Biosystems Dynamics Research, Yokohama, Kanagawa 230-0045, Japan
| | - Kaoru Kobayashi
- Department of Biopharmaceutics, Graduate School of Clinical Pharmacy, Meiji Pharmaceutical University, Kiyose-shi, Tokyo 204-8588, Japan
| | - Mikako Shirouzu
- RIKEN Center for Biosystems Dynamics Research, Yokohama, Kanagawa 230-0045, Japan
| | - Takeshi Inagaki
- Laboratory of Epigenetics and Metabolism, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi-shi, Gunma 371-8512, Japan
| | - Tim H-M Huang
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Nicolas Musi
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; San Antonio Geriatric Research, Education, and Clinical Center, South Texas Veterans Health Care System, San Antonio, TX 78229, USA
| | - Ivan Topisirovic
- Lady Davis Institute, Sir Mortimer B. Davis Jewish General Hospital, Montreal, QC H3A 1A3, Canada; Gerald Bronfman Department of Oncology, Division of Experimental Medicine and Department of Biochemistry, McGill University, Montreal, QC H3A 1A3, Canada
| | - Ola Larsson
- Department of Oncology-Pathology, Science for Life Laboratory, Karolinska Institute, 171 65 Stockholm, Sweden
| | - Masahiro Morita
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA.
| |
Collapse
|
56
|
Yamada A, Ohtsuki K, Shiga N, Green JA, Matsuno Y, Imakawa K. Epithelial-mesenchymal transition and bi- and multi-nucleated trophoblast cell formation in ovine conceptuses during the peri-implantation period. J Reprod Dev 2022; 68:110-117. [PMID: 34980711 PMCID: PMC8979805 DOI: 10.1262/jrd.2021-088] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT), which is common in cancer metastasis, is also observed during developmental processes such as embryo implantation into the maternal endometrium in
humans and rodents. However, this process has not been well characterized in the non-invasive type of implantation that occurs in ruminants. To understand whether EMT occurs in ruminant
ungulates, ovine conceptuses (embryo plus extraembryonic membranes) from days 15 (P15: pre-attachment), 17 (P17: during attachment), and 21 (P21: post-attachment, day 0 = day of estrus) were
evaluated. RNA-seq analysis revealed that the expression of EMT-related transcripts increased on P21. Real-time PCR and western blotting analyses indicated that levels of transcripts and
proteins indicative of mesenchyme-related molecules increased on P21, but a minor expression of epithelium-related molecules remained. Immunohistochemical analysis revealed that E-cadherin
(CDH1) was localized in the elongated trophectoderm on P15 and P17. On P21, CDH1 was localized to the trophectoderm and on the conceptus cells undergoing differentiation. Vimentin (VIM) was
localized in the uterine stroma on P15 and P17, and its expression was observed at the edge of elongating trophoblast on P21. Further, it was found that some bi-nucleated trophoblast cells
were present on P17; however, numerous bi- and multi-nucleated trophoblast cells on the uterine epithelium or next to the uterine stroma were found on P21. A minor expression of
pregnancy-associated glycoprotein (PAG) transcripts was found on P15 and P17, but a definitive expression of PAGs, transcripts, and proteins was found on P21. Although
further investigation is required, these observations indicate that bi-nucleated trophoblast cell formation begins on the day conceptus implantation to the maternal endometrium is initiated,
followed by EMT in trophoblast cells. These results suggest that these sequential events are required if pregnancy is to be established in ruminants.
Collapse
Affiliation(s)
- Ayami Yamada
- Research Institute of Agriculture, Tokai University, Kumamoto 862-8652, Japan
| | - Kaito Ohtsuki
- Research Institute of Agriculture, Tokai University, Kumamoto 862-8652, Japan
| | - Natsumi Shiga
- Research Institute of Agriculture, Tokai University, Kumamoto 862-8652, Japan
| | - Jonathan A Green
- Animal Science Research Center, University of Missouri-Columbia, Columbia, MO 65211, USA
| | - Yuta Matsuno
- Research Institute of Agriculture, Tokai University, Kumamoto 862-8652, Japan
| | - Kazuhiko Imakawa
- Research Institute of Agriculture, Tokai University, Kumamoto 862-8652, Japan
| |
Collapse
|
57
|
Yeh CC, Liu HM, Lee MC, Leu YL, Chiang WH, Chang HH, Lee TY. Phytochemical‑rich herbal formula ATG‑125 protects against sucrose‑induced gastrocnemius muscle atrophy by rescuing Akt signaling and improving mitochondrial dysfunction in young adult mice. Mol Med Rep 2021; 25:57. [PMID: 34913071 PMCID: PMC8711025 DOI: 10.3892/mmr.2021.12572] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 11/18/2021] [Indexed: 11/06/2022] Open
Abstract
The antioxidant capability of herbal remedies has attracted widespread attention, but their molecular mechanisms in a muscle atrophy model have not been explored. The aim of the present study was to compare the bioactivity of sucrose challenged mice following treatment with ATG‑125. Here, through a combination of transcriptomic and biomedical analysis, herbal formula ATG‑125, a phytochemical‑rich formula, was identified as a protective factor against muscle atrophy in sucrose challenged mice. Gene ontology (GO) identified differentially expressed genes that were primarily enriched in the 'negative regulation of proteolysis', 'cellular amino acid metabolic process', 'lipoprotein particle' and 'cell cycle', all of which were associated with the ATG‑125‑mediated prevention of muscle atrophy, particularly with regard to mitochondrial biogenesis. In skeletal muscle, a set of mitochondrial‑related genes, including angiopoietin‑like 4, nicotinamide riboside kinase 2 (Nmrk2), pyruvate dehydrogenase lipoamide kinase isozyme 4, Asc‑type amino acid transporter 1 and mitochondrial uncoupling protein 3 (Ucp3) were markedly upregulated following ATG‑125 intervention. An increase in Nmrk2 and Ucp3 expression were noted after ATG‑125 treatment, in parallel with upregulation of the 'nicotinate and nicotinamide metabolism' pathway, as determined using the Kyoto Encyclopedia of Genes and Genomes (KEGG). Furthermore, KEGG pathway analysis revealed the downregulation of 'complement and coagulation cascades', 'cholesterol metabolism', 'biosynthesis of amino acids' and 'PPAR signaling pathway', which were associated with the downregulation of serine (or cysteine) peptidase inhibitor clade A member (Serpina)3, Serpina1b, Serpina1d, Serpina1e, apolipoprotein (Apo)a1 and Apoa2, all of which were cardiovascular and diabetes‑associated risk factors and were regulated by ATG‑125. In addition, ATG‑125 treatment resulted in downregulated mRNA expression levels of ATPase sarcoplasmic/endoplasmic reticulum Ca2+ transporting 2, troponin‑I1, troponin‑C1 and troponin‑T1 in young adult gastrocnemius muscle compared with the sucrose group. Nuclear factor‑κB‑hypoxia inducible factor‑1α‑TGFβ receptor type‑II‑vascular endothelial growth factor staining indicated that ATG‑125 decreased sucrose‑induced chronic inflammation. ATG‑125 was sufficient to prevent muscle atrophy, and this protective effect may be mediated through upregulation of AKT phosphorylation, upregulating the insulin growth factor‑1R‑insulin receptor substrate‑PI3K‑AKT pathway, which in turn resulted in a forkhead box O‑dependent decrease in protein degradation pathways, including regulation of atrogin1 and E3 ubiquitin‑protein ligase TRIM63. Peroxisome‑proliferator activated receptor γ coactivator 1α (PGC1α) was decreased in young adult mice challenged with sucrose. ATG‑125 treatment significantly increased PGC1α and significantly increased UCP‑1,2,3 expression levels, which suggested ATG‑125 poised the mitochondria for uncoupling of respiration. This effect is consistent with the increased SIRT1 levels and may explain an increase in mitochondria biogenesis. Taken together, the present study showed that ATG‑125, as an integrator of protein synthesis and degradative pathways, prevented muscle wasting.
Collapse
Affiliation(s)
- Ching-Chuan Yeh
- Graduate Institute of Traditional Chinese Medicine, School of Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan, R.O.C
| | - Hsuan-Miao Liu
- Graduate Institute of Traditional Chinese Medicine, School of Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan, R.O.C
| | - Ming-Chung Lee
- Brion Research Institute of Taiwan, New Taipei City 23143, Taiwan, R.O.C
| | - Yann-Lii Leu
- Graduate Institute of Nature Products, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan, R.O.C
| | - Wei-Han Chiang
- Department of Rehabilitation, Cheng‑Hsin General Hospital, Taipei 11283, Taiwan, R.O.C
| | - Hen-Hong Chang
- Graduate Institute of Integrated Medicine, China Medical University, Taichung 40402, Taiwan, R.O.C
| | - Tzung-Yan Lee
- Graduate Institute of Traditional Chinese Medicine, School of Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan, R.O.C
| |
Collapse
|
58
|
Impact of spaceflight and artificial gravity on sulfur metabolism in mouse liver: sulfur metabolomic and transcriptomic analysis. Sci Rep 2021; 11:21786. [PMID: 34750416 PMCID: PMC8575787 DOI: 10.1038/s41598-021-01129-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 10/11/2021] [Indexed: 12/30/2022] Open
Abstract
Spaceflight induces hepatic damage, partially owing to oxidative stress caused by the space environment such as microgravity and space radiation. We examined the roles of anti-oxidative sulfur-containing compounds on hepatic damage after spaceflight. We analyzed the livers of mice on board the International Space Station for 30 days. During spaceflight, half of the mice were exposed to artificial earth gravity (1 g) using centrifugation cages. Sulfur-metabolomics of the livers of mice after spaceflight revealed a decrease in sulfur antioxidants (ergothioneine, glutathione, cysteine, taurine, thiamine, etc.) and their intermediates (cysteine sulfonic acid, hercynine, N-acethylserine, serine, etc.) compared to the controls on the ground. Furthermore, RNA-sequencing showed upregulation of gene sets related to oxidative stress and sulfur metabolism, and downregulation of gene sets related to glutathione reducibility in the livers of mice after spaceflight, compared to controls on the ground. These changes were partially mitigated by exposure to 1 g centrifugation. For the first time, we observed a decrease in sulfur antioxidants based on a comprehensive analysis of the livers of mice after spaceflight. Our data suggest that a decrease in sulfur-containing compounds owing to both microgravity and other spaceflight environments (radiation and stressors) contributes to liver damage after spaceflight.
Collapse
|
59
|
Vuelta E, Ordoñez JL, Alonso-Pérez V, Méndez L, Hernández-Carabias P, Saldaña R, Sevilla J, Sebastián E, Muntión S, Sánchez-Guijo F, Hernández-Rivas JM, García-Tuñón I, Sánchez-Martín M. CRISPR-Cas9 Technology as a Tool to Target Gene Drivers in Cancer: Proof of Concept and New Opportunities to Treat Chronic Myeloid Leukemia. CRISPR J 2021; 4:519-535. [PMID: 34406033 DOI: 10.1089/crispr.2021.0009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Chronic myeloid leukemia (CML) is a hematopoietic malignancy produced by a unique oncogenic event involving the constitutively active tyrosine-kinase (TK) BCR/ABL1. TK inhibitors (TKI) changed its prognosis and natural history. Unfortunately, ABL1 remains unaffected by TKIs. Leukemic stem cells (LSCs) remain, and resistant mutations arise during treatment. To address this problem, we have designed a therapeutic CRISPR-Cas9 deletion system targeting BCR/ABL1. The system was efficiently electroporated to cell lines, LSCs from a CML murine model, and LSCs from CML patients at diagnosis, generating a specific ABL1 null mutation at high efficiency and allowing the edited leukemic cells to be detected and tracked. The CRISPR-Cas9 deletion system triggered cell proliferation arrest and apoptosis in murine and human CML cell lines. Patient and murine-derived xenografts with CRISPR-edited LSCs in NOD SCID gamma niches revealed that normal multipotency and repopulation ability of CRISPR edited LSCs were fully restored. Normal hematopoiesis was restored, avoiding myeloid bias. To the best of our knowledge, we show for the first time how a CRISPR-Cas9 deletion system efficiently interrupts BCR/ABL1 oncogene in primary LSCs to bestow a therapeutic benefit. This study is a proof of concept for genome editing in all those diseases, like CML, sustained by a single oncogenic event, opening up new therapeutic opportunities.
Collapse
Affiliation(s)
- Elena Vuelta
- Unidad de Diagnóstico Molecular y Celular del Cáncer, Instituto Biología Molecular y Celular del Cáncer (USAL/CSIC), Salamanca, Spain; Hospital Universitario de Salamanca, Salamanca, Spain
- Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain; Hospital Universitario de Salamanca, Salamanca, Spain
- Servicio de Transgénesis, NUCLEUS, Universidad de Salamanca, Salamanca, Spain; Hospital Universitario de Salamanca, Salamanca, Spain
| | - José Luis Ordoñez
- Unidad de Diagnóstico Molecular y Celular del Cáncer, Instituto Biología Molecular y Celular del Cáncer (USAL/CSIC), Salamanca, Spain; Hospital Universitario de Salamanca, Salamanca, Spain
- IBSAL, Instituto de Investigación Biomédica de Salamanca, Salamanca, Spain; Hospital Universitario de Salamanca, Salamanca, Spain
| | - Verónica Alonso-Pérez
- Unidad de Diagnóstico Molecular y Celular del Cáncer, Instituto Biología Molecular y Celular del Cáncer (USAL/CSIC), Salamanca, Spain; Hospital Universitario de Salamanca, Salamanca, Spain
- IBSAL, Instituto de Investigación Biomédica de Salamanca, Salamanca, Spain; Hospital Universitario de Salamanca, Salamanca, Spain
| | - Lucía Méndez
- Servicio de Transgénesis, NUCLEUS, Universidad de Salamanca, Salamanca, Spain; Hospital Universitario de Salamanca, Salamanca, Spain
| | - Patricia Hernández-Carabias
- Servicio de Transgénesis, NUCLEUS, Universidad de Salamanca, Salamanca, Spain; Hospital Universitario de Salamanca, Salamanca, Spain
| | - Raquel Saldaña
- Servicio de Hematología, Hospital de Jerez, Cádiz, Spain; Hospital Universitario de Salamanca, Salamanca, Spain
| | - Julián Sevilla
- Hospital Infantil Universitario Niño Jesús, Madrid, Spain; Hospital Universitario de Salamanca, Salamanca, Spain
| | - Elena Sebastián
- Hospital Infantil Universitario Niño Jesús, Madrid, Spain; Hospital Universitario de Salamanca, Salamanca, Spain
| | - Sandra Muntión
- Unidad de Diagnóstico Molecular y Celular del Cáncer, Instituto Biología Molecular y Celular del Cáncer (USAL/CSIC), Salamanca, Spain; Hospital Universitario de Salamanca, Salamanca, Spain
- Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain; Hospital Universitario de Salamanca, Salamanca, Spain
- Hospital Infantil Universitario Niño Jesús, Madrid, Spain; Hospital Universitario de Salamanca, Salamanca, Spain
- RETIC TerCel y CIBERONC, ISCIII, Madrid, Spain; and Hospital Universitario de Salamanca, Salamanca, Spain
- Servicio de Hematología, Hospital Universitario de Salamanca, Salamanca, Spain
| | - Fermín Sánchez-Guijo
- Unidad de Diagnóstico Molecular y Celular del Cáncer, Instituto Biología Molecular y Celular del Cáncer (USAL/CSIC), Salamanca, Spain; Hospital Universitario de Salamanca, Salamanca, Spain
- Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain; Hospital Universitario de Salamanca, Salamanca, Spain
- Hospital Infantil Universitario Niño Jesús, Madrid, Spain; Hospital Universitario de Salamanca, Salamanca, Spain
- RETIC TerCel y CIBERONC, ISCIII, Madrid, Spain; and Hospital Universitario de Salamanca, Salamanca, Spain
- Servicio de Hematología, Hospital Universitario de Salamanca, Salamanca, Spain
| | - Jesús María Hernández-Rivas
- Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain; Hospital Universitario de Salamanca, Salamanca, Spain
- IBSAL, Instituto de Investigación Biomédica de Salamanca, Salamanca, Spain; Hospital Universitario de Salamanca, Salamanca, Spain
- Servicio de Hematología, Hospital Universitario de Salamanca, Salamanca, Spain
| | - Ignacio García-Tuñón
- Unidad de Diagnóstico Molecular y Celular del Cáncer, Instituto Biología Molecular y Celular del Cáncer (USAL/CSIC), Salamanca, Spain; Hospital Universitario de Salamanca, Salamanca, Spain
- IBSAL, Instituto de Investigación Biomédica de Salamanca, Salamanca, Spain; Hospital Universitario de Salamanca, Salamanca, Spain
| | - Manuel Sánchez-Martín
- Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain; Hospital Universitario de Salamanca, Salamanca, Spain
- Servicio de Transgénesis, NUCLEUS, Universidad de Salamanca, Salamanca, Spain; Hospital Universitario de Salamanca, Salamanca, Spain
- IBSAL, Instituto de Investigación Biomédica de Salamanca, Salamanca, Spain; Hospital Universitario de Salamanca, Salamanca, Spain
| |
Collapse
|
60
|
Crespo-Facorro B, Ruiz-Veguilla M, Vázquez-Bourgon J, Sánchez-Hidalgo AC, Garrido-Torres N, Cisneros JM, Prieto C, Sainz J. Aripiprazole as a Candidate Treatment of COVID-19 Identified Through Genomic Analysis. Front Pharmacol 2021; 12:646701. [PMID: 33762960 PMCID: PMC7982825 DOI: 10.3389/fphar.2021.646701] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Accepted: 02/08/2021] [Indexed: 01/08/2023] Open
Abstract
Background: Antipsychotics modulate expression of inflammatory cytokines and inducible inflammatory enzymes. Elopiprazole (a phenylpiperazine antipsychotic drug in phase 1) has been characterized as a therapeutic drug to treat SARS-CoV-2 infection in a repurposing study. We aim to investigate the potential effects of aripiprazole (an FDA approved phenylpiperazine) on COVID-19-related immunological parameters. Methods: Differential gene expression profiles of non-COVID-19 vs. COVID-19 RNA-Seq samples (CRA002390 project in GSA database) and drug-naïve patients with non-affective psychosis at baseline and after three months of aripiprazole treatment were identified. An integrative transcriptomic analyses of aripiprazole effects on differentially expressed genes in COVID-19 patients was performed. Findings: 82 out the 377 genes (21.7%) with expression significantly altered by aripiprazole have also their expression altered in COVID-19 patients and in 93.9% of these genes their expression is reverted by aripiprazole. The number of common genes with expression altered in both analyses is significantly higher than expected (Fisher's Exact Test, two tail; p value = 3.2e-11). 11 KEGG pathways were significantly enriched with genes with altered expression both in COVID-19 patients and aripiprazole medicated non-affective psychosis patients (p adj<0.05). The most significant pathways were associated to immune responses and mechanisms of hyperinflammation-driven pathology (i.e.,"inflammatory bowel disease (IBD)" (the most significant pathway with a p adj of 0.00021), "Th1 and Th2 cell differentiation" and "B cell receptor signaling pathway") that have been also associated with COVID19 clinical outcome. Interpretation: This exploratory investigation may provide further support to the notion that a protective effect is exerted by aripiprazole (phenylpiperazine) by modulating the expression of genes that have shown to be altered in COVID-19 patients. Along with many ongoing studies and clinical trials, repurposing available medications could be of use in countering SARS-CoV-2 infection, but require further studies and trials.
Collapse
Affiliation(s)
- Benedicto Crespo-Facorro
- Department of Psychiatry, School of Medicine, University Hospital Virgen del Rocio-IBIS, Sevilla, Spain
- Spanish Network for Research in Mental Health (CIBERSAM), Sevilla, Spain
| | - Miguel Ruiz-Veguilla
- Department of Psychiatry, School of Medicine, University Hospital Virgen del Rocio-IBIS, Sevilla, Spain
- Spanish Network for Research in Mental Health (CIBERSAM), Sevilla, Spain
| | - Javier Vázquez-Bourgon
- Spanish Network for Research in Mental Health (CIBERSAM), Sevilla, Spain
- Department of Psychiatry, University Hospital Marques de Valdecilla - Instituto de Investigacion Marques de Valdecilla (IDIVAL), Santander, Spain
- Department of Medicine and Psychiatry, School of Medicine, University of Cantabria, Santander, Spain
| | - Ana C. Sánchez-Hidalgo
- Spanish Network for Research in Mental Health (CIBERSAM), Sevilla, Spain
- Seville Biomedical Research Centre (IBiS), Sevilla, Spain
| | - Nathalia Garrido-Torres
- Department of Psychiatry, School of Medicine, University Hospital Virgen del Rocio-IBIS, Sevilla, Spain
| | - Jose M. Cisneros
- Department of Infectious Diseases, Microbiology and Preventive Medicine, Institute of Biomedicine of Seville, University Hospital Virgen del Rocio, University of Seville, Salamanca, Spain
- Spanish Network for Research in Infectious Diseases (REIPI), Madrid, Spain
| | - Carlos Prieto
- Bioinformatics Service, Nucleus, University of Salamanca, Salamanca, Spain
| | - Jesus Sainz
- Spanish National Research Council (CSIC), Institute of Biomedicine and Biotechnology of Cantabria, Santander, Spain
| |
Collapse
|
61
|
Abdellatif M, Trummer-Herbst V, Koser F, Durand S, Adão R, Vasques-Nóvoa F, Freundt JK, Voglhuber J, Pricolo MR, Kasa M, Türk C, Aprahamian F, Herrero-Galán E, Hofer SJ, Pendl T, Rech L, Kargl J, Anto-Michel N, Ljubojevic-Holzer S, Schipke J, Brandenberger C, Auer M, Schreiber R, Koyani CN, Heinemann A, Zirlik A, Schmidt A, von Lewinski D, Scherr D, Rainer PP, von Maltzahn J, Mühlfeld C, Krüger M, Frank S, Madeo F, Eisenberg T, Prokesch A, Leite-Moreira AF, Lourenço AP, Alegre-Cebollada J, Kiechl S, Linke WA, Kroemer G, Sedej S. Nicotinamide for the treatment of heart failure with preserved ejection fraction. Sci Transl Med 2021; 13:eabd7064. [PMID: 33568522 PMCID: PMC7611499 DOI: 10.1126/scitranslmed.abd7064] [Citation(s) in RCA: 129] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 11/05/2020] [Accepted: 01/12/2021] [Indexed: 12/17/2022]
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a highly prevalent and intractable form of cardiac decompensation commonly associated with diastolic dysfunction. Here, we show that diastolic dysfunction in patients with HFpEF is associated with a cardiac deficit in nicotinamide adenine dinucleotide (NAD+). Elevating NAD+ by oral supplementation of its precursor, nicotinamide, improved diastolic dysfunction induced by aging (in 2-year-old C57BL/6J mice), hypertension (in Dahl salt-sensitive rats), or cardiometabolic syndrome (in ZSF1 obese rats). This effect was mediated partly through alleviated systemic comorbidities and enhanced myocardial bioenergetics. Simultaneously, nicotinamide directly improved cardiomyocyte passive stiffness and calcium-dependent active relaxation through increased deacetylation of titin and the sarcoplasmic reticulum calcium adenosine triphosphatase 2a, respectively. In a long-term human cohort study, high dietary intake of naturally occurring NAD+ precursors was associated with lower blood pressure and reduced risk of cardiac mortality. Collectively, these results suggest NAD+ precursors, and especially nicotinamide, as potential therapeutic agents to treat diastolic dysfunction and HFpEF in humans.
Collapse
Affiliation(s)
- Mahmoud Abdellatif
- Department of Cardiology, Medical University of Graz, Graz 8036, Austria
| | | | - Franziska Koser
- Institute of Physiology II, University of Münster, Münster 48149, Germany
| | - Sylvère Durand
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif 94805, France
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, INSERM U1138, Institut Universitaire de France, Paris 75006, France
| | - Rui Adão
- Department of Cardiology, Medical University of Graz, Graz 8036, Austria
- Department of Surgery and Physiology, Cardiovascular Research and Development Centre (UnIC), Faculty of Medicine, University of Porto, Porto 4200-319, Portugal
| | - Francisco Vasques-Nóvoa
- Department of Surgery and Physiology, Cardiovascular Research and Development Centre (UnIC), Faculty of Medicine, University of Porto, Porto 4200-319, Portugal
| | - Johanna K Freundt
- Institute of Physiology II, University of Münster, Münster 48149, Germany
| | - Julia Voglhuber
- Department of Cardiology, Medical University of Graz, Graz 8036, Austria
- BioTechMed Graz, Graz 8010, Austria
| | | | - Michael Kasa
- Department of Cardiology, Medical University of Graz, Graz 8036, Austria
| | - Clara Türk
- Institute for Genetics, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, Cologne 50931, Germany
- Center for Molecular Medicine (CMMC), University of Cologne, Cologne 50931, Germany
| | - Fanny Aprahamian
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif 94805, France
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, INSERM U1138, Institut Universitaire de France, Paris 75006, France
| | - Elías Herrero-Galán
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid 28029, Spain
| | - Sebastian J Hofer
- Institute of Molecular Biosciences, University of Graz, NAWI Graz, Graz 8010, Austria
| | - Tobias Pendl
- Institute of Molecular Biosciences, University of Graz, NAWI Graz, Graz 8010, Austria
| | - Lavinia Rech
- Department of Cardiology, Medical University of Graz, Graz 8036, Austria
| | - Julia Kargl
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, Graz 8010, Austria
| | | | - Senka Ljubojevic-Holzer
- Department of Cardiology, Medical University of Graz, Graz 8036, Austria
- BioTechMed Graz, Graz 8010, Austria
| | - Julia Schipke
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover 30625, Germany
| | - Christina Brandenberger
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover 30625, Germany
| | - Martina Auer
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Medical University of Graz, Graz 8010, Austria
- Division of Cell Biology, Histology and Embryology, Medical University of Graz, Graz 8010, Austria
| | - Renate Schreiber
- Institute of Molecular Biosciences, University of Graz, NAWI Graz, Graz 8010, Austria
| | - Chintan N Koyani
- Department of Cardiology, Medical University of Graz, Graz 8036, Austria
| | - Akos Heinemann
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, Graz 8010, Austria
| | - Andreas Zirlik
- Department of Cardiology, Medical University of Graz, Graz 8036, Austria
| | - Albrecht Schmidt
- Department of Cardiology, Medical University of Graz, Graz 8036, Austria
| | - Dirk von Lewinski
- Department of Cardiology, Medical University of Graz, Graz 8036, Austria
| | - Daniel Scherr
- Department of Cardiology, Medical University of Graz, Graz 8036, Austria
| | - Peter P Rainer
- Department of Cardiology, Medical University of Graz, Graz 8036, Austria
- BioTechMed Graz, Graz 8010, Austria
| | | | - Christian Mühlfeld
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover 30625, Germany
| | - Marcus Krüger
- Institute for Genetics, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, Cologne 50931, Germany
- Center for Molecular Medicine (CMMC), University of Cologne, Cologne 50931, Germany
| | - Saša Frank
- BioTechMed Graz, Graz 8010, Austria
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Medical University of Graz, Graz 8010, Austria
| | - Frank Madeo
- BioTechMed Graz, Graz 8010, Austria
- Institute of Molecular Biosciences, University of Graz, NAWI Graz, Graz 8010, Austria
| | - Tobias Eisenberg
- BioTechMed Graz, Graz 8010, Austria
- Institute of Molecular Biosciences, University of Graz, NAWI Graz, Graz 8010, Austria
| | - Andreas Prokesch
- BioTechMed Graz, Graz 8010, Austria
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Medical University of Graz, Graz 8010, Austria
- Division of Cell Biology, Histology and Embryology, Medical University of Graz, Graz 8010, Austria
| | - Adelino F Leite-Moreira
- Department of Surgery and Physiology, Cardiovascular Research and Development Centre (UnIC), Faculty of Medicine, University of Porto, Porto 4200-319, Portugal
| | - André P Lourenço
- Department of Surgery and Physiology, Cardiovascular Research and Development Centre (UnIC), Faculty of Medicine, University of Porto, Porto 4200-319, Portugal
| | | | - Stefan Kiechl
- Department of Neurology, Medical University of Innsbruck, Innsbruck 6020, Austria
- VASCage, Research Centre for Promoting Vascular Health in the Ageing Community, Innsbruck 6020, Austria
| | - Wolfgang A Linke
- Institute of Physiology II, University of Münster, Münster 48149, Germany
| | - Guido Kroemer
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif 94805, France.
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, INSERM U1138, Institut Universitaire de France, Paris 75006, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris 75015, France
- Suzhou Institute for Systems Medicine, Chinese Academy of Sciences, Suzhou 215000, China
- Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, Solna 17164, Sweden
| | - Simon Sedej
- Department of Cardiology, Medical University of Graz, Graz 8036, Austria.
- BioTechMed Graz, Graz 8010, Austria
- Faculty of Medicine, University of Maribor, Maribor 2000, Slovenia
| |
Collapse
|
62
|
Garrido-Rodriguez M, Lopez-Lopez D, Ortuno FM, Peña-Chilet M, Muñoz E, Calzado MA, Dopazo J. A versatile workflow to integrate RNA-seq genomic and transcriptomic data into mechanistic models of signaling pathways. PLoS Comput Biol 2021; 17:e1008748. [PMID: 33571195 PMCID: PMC7904194 DOI: 10.1371/journal.pcbi.1008748] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 02/24/2021] [Accepted: 01/30/2021] [Indexed: 12/13/2022] Open
Abstract
MIGNON is a workflow for the analysis of RNA-Seq experiments, which not only efficiently manages the estimation of gene expression levels from raw sequencing reads, but also calls genomic variants present in the transcripts analyzed. Moreover, this is the first workflow that provides a framework for the integration of transcriptomic and genomic data based on a mechanistic model of signaling pathway activities that allows a detailed biological interpretation of the results, including a comprehensive functional profiling of cell activity. MIGNON covers the whole process, from reads to signaling circuit activity estimations, using state-of-the-art tools, it is easy to use and it is deployable in different computational environments, allowing an optimized use of the resources available.
Collapse
Affiliation(s)
- Martín Garrido-Rodriguez
- Clinical Bioinformatics Area, Fundación Progreso y Salud (FPS), Hospital Virgen del Rocío, Sevilla, Spain
- Departamento de Biología Celular, Fisiología e Inmunología, Universidad de Córdoba, Córdoba, Spain
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Daniel Lopez-Lopez
- Clinical Bioinformatics Area, Fundación Progreso y Salud (FPS), Hospital Virgen del Rocío, Sevilla, Spain
- Computational Systems Medicine, Institute of Biomedicine of Seville (IBIS), Sevilla, Spain
| | - Francisco M. Ortuno
- Clinical Bioinformatics Area, Fundación Progreso y Salud (FPS), Hospital Virgen del Rocío, Sevilla, Spain
- Computational Systems Medicine, Institute of Biomedicine of Seville (IBIS), Sevilla, Spain
| | - María Peña-Chilet
- Clinical Bioinformatics Area, Fundación Progreso y Salud (FPS), Hospital Virgen del Rocío, Sevilla, Spain
- Computational Systems Medicine, Institute of Biomedicine of Seville (IBIS), Sevilla, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), FPS, Hospital Virgen del Rocío, Sevilla, Spain
| | - Eduardo Muñoz
- Departamento de Biología Celular, Fisiología e Inmunología, Universidad de Córdoba, Córdoba, Spain
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Marco A. Calzado
- Departamento de Biología Celular, Fisiología e Inmunología, Universidad de Córdoba, Córdoba, Spain
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Joaquin Dopazo
- Clinical Bioinformatics Area, Fundación Progreso y Salud (FPS), Hospital Virgen del Rocío, Sevilla, Spain
- Computational Systems Medicine, Institute of Biomedicine of Seville (IBIS), Sevilla, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), FPS, Hospital Virgen del Rocío, Sevilla, Spain
- FPS/ELIXIR-es, Hospital Virgen del Rocío, Sevilla, Spain
| |
Collapse
|
63
|
Firoj Mahmud AKM, Delhomme N, Nandi S, Fällman M. ProkSeq for complete analysis of RNA-Seq data from prokaryotes. Bioinformatics 2020; 37:126-128. [PMID: 33367516 PMCID: PMC8034529 DOI: 10.1093/bioinformatics/btaa1063] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 11/19/2020] [Accepted: 12/11/2020] [Indexed: 12/02/2022] Open
Abstract
Summary Since its introduction, RNA-Seq technology has been used extensively in studies of pathogenic bacteria to identify and quantify differences in gene expression across multiple samples from bacteria exposed to different conditions. With some exceptions, tools for studying gene expression, determination of differential gene expression, downstream pathway analysis and normalization of data collected in extreme biological conditions is still lacking. Here, we describe ProkSeq, a user-friendly, fully automated RNA-Seq data analysis pipeline designed for prokaryotes. ProkSeq provides a wide variety of options for analysing differential expression, normalizing expression data and visualizing data and results. Availability and implementation ProkSeq is implemented in Python and is published under the MIT source license. The pipeline is available as a Docker container https://hub.docker.com/repository/docker/snandids/prokseq-v2.0, or can be used through Anaconda: https://anaconda.org/snandiDS/prokseq. The code is available on Github: https://github.com/snandiDS/prokseq and a detailed user documentation, including a manual and tutorial can be found at https://prokseqV20.readthedocs.io. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- A K M Firoj Mahmud
- Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå Centre for Microbial Research (UCMR), Department of Molecular Biology, Umeå University, SE-901 87, Umeå, Sweden
| | - Nicolas Delhomme
- Department of Forest Genetics and Plant Physiology, Umeå Plant Science Centre (UPSC), Swedish University of Agricultural Sciences
| | - Soumyadeep Nandi
- Amity Institute of Integrative Sciences and Health, Amity University Haryana, Gurgaon, 122413, Haryana, India
| | - Maria Fällman
- Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå Centre for Microbial Research (UCMR), Department of Molecular Biology, Umeå University, SE-901 87, Umeå, Sweden
| |
Collapse
|
64
|
Sánchez de la Nieta R, Antoraz S, Alzate JF, Santamaría RI, Díaz M. Antibiotic Production and Antibiotic Resistance: The Two Sides of AbrB1/B2, a Two-Component System of Streptomyces coelicolor. Front Microbiol 2020; 11:587750. [PMID: 33162964 PMCID: PMC7581861 DOI: 10.3389/fmicb.2020.587750] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 09/22/2020] [Indexed: 11/13/2022] Open
Abstract
Antibiotic resistance currently presents one of the biggest threats to humans. The development and implementation of strategies against the spread of superbugs is a priority for public health. In addition to raising social awareness, approaches such as the discovery of new antibiotic molecules and the elucidation of resistance mechanisms are common measures. Accordingly, the two-component system (TCS) of Streptomyces coelicolor AbrB1/B2, offer amenable ways to study both antibiotic production and resistance. Global transcriptomic comparisons between the wild-type strain S. coelicolor M145 and the mutant ΔabrB, using RNA-Seq, showed that the AbrB1/B2 TCS is implicated in the regulation of different biological processes associated with stress responses, primary and secondary metabolism, and development and differentiation. The ΔabrB mutant showed the up-regulation of antibiotic biosynthetic gene clusters and the down-regulation of the vancomycin resistance gene cluster, according to the phenotypic observations of increased antibiotic production of actinorhodin and undecylprodigiosin, and greater susceptibility to vancomycin. The role of AbrB1/B2 in vancomycin resistance has also been shown by an in silico analysis, which strongly indicates that AbrB1/B2 is a homolog of VraR/S from Staphylococcus aureus and LiaR/S from Enterococcus faecium/Enterococcus faecalis, both of which are implied in vancomycin resistance in these pathogenic organisms that present a serious threat to public health. The results obtained are interesting from a biotechnological perspective since, on one hand, this TCS is a negative regulator of antibiotic production and its high degree of conservation throughout Streptomyces spp. makes it a valuable tool for improving antibiotic production and the discovery of cryptic metabolites with antibiotic action. On the other hand, AbrB1/B2 contributes to vancomycin resistance and is a homolog of VraR/S and LiaR/S, important regulators in clinically relevant antibiotic-resistant bacteria. Therefore, the study of AbrB1/B2 could provide new insight into the mechanism of this type of resistance.
Collapse
Affiliation(s)
- Ricardo Sánchez de la Nieta
- Instituto de Biología Funcional y Genómica/Departamento de Microbiología y Genética, Consejo Superior de Investigaciones Científicas/Universidad de Salamanca, Salamanca, Spain
| | - Sergio Antoraz
- Instituto de Biología Funcional y Genómica/Departamento de Microbiología y Genética, Consejo Superior de Investigaciones Científicas/Universidad de Salamanca, Salamanca, Spain
| | - Juan F Alzate
- Departamento de Microbiología y Parasitología, Facultad de Medicina, Centro Nacional de Secuenciación Genómica, Sede de Investigación Universitaria, Universidad de Antioquia, Medellín, Colombia
| | - Ramón I Santamaría
- Instituto de Biología Funcional y Genómica/Departamento de Microbiología y Genética, Consejo Superior de Investigaciones Científicas/Universidad de Salamanca, Salamanca, Spain
| | - Margarita Díaz
- Instituto de Biología Funcional y Genómica/Departamento de Microbiología y Genética, Consejo Superior de Investigaciones Científicas/Universidad de Salamanca, Salamanca, Spain
| |
Collapse
|
65
|
Vikhorev PG, Vikhoreva NN, Yeung W, Li A, Lal S, dos Remedios CG, Blair CA, Guglin M, Campbell KS, Yacoub MH, de Tombe P, Marston SB. Titin-truncating mutations associated with dilated cardiomyopathy alter length-dependent activation and its modulation via phosphorylation. Cardiovasc Res 2020; 118:241-253. [PMID: 33135063 PMCID: PMC8752363 DOI: 10.1093/cvr/cvaa316] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 10/20/2020] [Indexed: 12/18/2022] Open
Abstract
Abstract
Aims
Dilated cardiomyopathy (DCM) is associated with mutations in many genes encoding sarcomere proteins. Truncating mutations in the titin gene TTN are the most frequent. Proteomic and functional characterizations are required to elucidate the origin of the disease and the pathogenic mechanisms of TTN-truncating variants.
Methods and results
We isolated myofibrils from DCM hearts carrying truncating TTN mutations and measured the Ca2+ sensitivity of force and its length dependence. Simultaneous measurement of force and adenosine triphosphate (ATP) consumption in skinned cardiomyocytes was also performed. Phosphorylation levels of troponin I (TnI) and myosin binding protein-C (MyBP-C) were manipulated using protein kinase A and λ phosphatase. mRNA sequencing was employed to overview gene expression profiles. We found that Ca2+ sensitivity of myofibrils carrying TTN mutations was significantly higher than in myofibrils from donor hearts. The length dependence of the Ca2+ sensitivity was absent in DCM myofibrils with TTN-truncating variants. No significant difference was found in the expression level of TTN mRNA between the DCM and donor groups. TTN exon usage and splicing were also similar. However, we identified down-regulation of genes encoding Z-disk proteins, while the atrial-specific regulatory myosin light chain gene, MYL7, was up-regulated in DCM patients with TTN-truncating variants.
Conclusion
Titin-truncating mutations lead to decreased length-dependent activation and increased elasticity of myofibrils. Phosphorylation levels of TnI and MyBP-C seen in the left ventricles are essential for the length-dependent changes in Ca2+ sensitivity in healthy donors, but they are reduced in DCM patients with TTN-truncating variants. A decrease in expression of Z-disk proteins may explain the observed decrease in myofibril passive stiffness and length-dependent activation.
Collapse
Affiliation(s)
- Petr G Vikhorev
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Natalia N Vikhoreva
- Heart Science Centre, Magdi Yacoub Institute, Harefield Hospital, London UB9 6JH, UK
| | - WaiChun Yeung
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Amy Li
- Department of Pharmacy and Biomedical Sciences, La Trobe University, Bendigo, VIC 3550, Australia
| | - Sean Lal
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, NSW 2006, Australia
| | - Cristobal G dos Remedios
- Division of Molecular Cardiology and Biophysics, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
| | - Cheavar A Blair
- Division of Cardiovascular Medicine, Department of Physiology, University of Kentucky, Lexington, KY, USA
| | - Maya Guglin
- Division of Cardiovascular Medicine, Department of Physiology, University of Kentucky, Lexington, KY, USA
| | - Kenneth S Campbell
- Division of Cardiovascular Medicine, Department of Physiology, University of Kentucky, Lexington, KY, USA
| | - Magdi H Yacoub
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Pieter de Tombe
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London W12 0NN, UK
- Heart Science Centre, Magdi Yacoub Institute, Harefield Hospital, London UB9 6JH, UK
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, USA
| | - Steven B Marston
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London W12 0NN, UK
| |
Collapse
|