1
|
Lee GB, Yang C, Hu F, Hao L. Evaluating sample normalization methods for MS-based multi-omics and the application to a neurodegenerative mouse model. Analyst 2025. [PMID: 39995368 PMCID: PMC11851094 DOI: 10.1039/d4an01573h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Accepted: 02/13/2025] [Indexed: 02/26/2025]
Abstract
Mass spectrometry (MS)-based omics methods have transformed biomedical research with accurate and high-throughput analysis of diverse molecules in biological systems. Recent technological advances also enabled multi-omics to be achieved from the same sample or on a single analytical platform. Sample normalization is a critical step in MS-omics studies but is usually conducted independently for each omics experiment. To bridge this technical gap, we evaluated different sample normalization methods suitable for analyzing proteins, lipids, and metabolites from the same sample for multi-omics analysis. We found that normalizing samples based on tissue weight or protein concentration before or after extraction generated distinct quantitative results. Normalizing samples first by tissue weight before extraction and then by protein concentration after extraction resulted in the lowest sample variation to reveal true biological differences. We then applied this two-step normalization method to investigate multi-omics profiles of mouse brains lacking the GRN gene. Loss-of-function mutations in the GRN gene lead to the deficiency of the progranulin protein and eventually cause neurodegeneration. Comparing the proteomics, lipidomics, and metabolomics profiles of GRN KO and WT mouse brains revealed molecular changes and pathways related to lysosomal dysfunction and neuroinflammation. In summary, we demonstrated the importance of selecting an appropriate normalization method during multi-omics sample preparation. Our normalization method is applicable to all tissue-based multi-omics studies, ensuring reliable and accurate biomolecule quantification for biological comparisons.
Collapse
Affiliation(s)
- Gwang Bin Lee
- Department of Chemistry & Biochemistry, University of Maryland, College Park, MD, 20742, USA.
| | - Cha Yang
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, 14853, USA
| | - Fenghua Hu
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, 14853, USA
| | - Ling Hao
- Department of Chemistry & Biochemistry, University of Maryland, College Park, MD, 20742, USA.
| |
Collapse
|
2
|
Dargham T, Aguilera-Correa JJ, Avellan R, Mallick I, Celik L, Santucci P, Brasseur G, Poncin I, Point V, Audebert S, Camoin L, Daher W, Cavalier JF, Kremer L, Canaan S. A proteomic and functional view of intrabacterial lipid inclusion biogenesis in mycobacteria. mBio 2025:e0147524. [PMID: 39998225 DOI: 10.1128/mbio.01475-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 02/03/2025] [Indexed: 02/26/2025] Open
Abstract
During infection and granuloma formation, pathogenic mycobacteria store triacylglycerol as intrabacterial lipid inclusions (ILIs). This accumulation of nutrients provides a carbon source for bacterial persistence and slows down intracellular metabolism. Mycobacterium abscessus (Mab), a rapidly growing non-tuberculous actinobacterium, produces ILI throughout its infection cycle. Here, Mab was used as a model organism to identify proteins associated with ILI accumulation on a global scale. By using the APEX2 proximity labeling method in an in vitro model for ILI accumulation, we identified 228 proteins possibly implicated in ILI biosynthesis. Fluorescence microscopy of strains overexpressing eight ILI-associated proteins (IAP) candidates fused to superfolder green fluorescent protein showed co-localization with ILI. Genetic inactivation of these potential IAP-encoding genes and subsequent lipid analysis emphasized the importance of MAB_3486 and MAB_4532c as key enzymes influencing triacylglycerol storage. This study underscores the dynamic process of ILI biogenesis and advances our understanding of lipid metabolism in pathogenic mycobacteria. Identifying major IAP in lipid accumulation offers new therapeutic perspectives to control the growth and persistence of pathogenic mycobacteria. IMPORTANCE This study sheds light into the complex process of intracellular lipid accumulation and storage in the survival and persistence of pathogenic mycobacteria, which is of clinical relevance. By identifying the proteins involved in the formation of intrabacterial lipid inclusions and revealing their impact on lipid metabolism, our data may lead to the development of new therapeutic strategies to target and control pathogenic mycobacteria, potentially improving outcomes for patients with mycobacterial infections.
Collapse
Affiliation(s)
- Tonia Dargham
- Aix-Marseille Univ., CNRS, LISM-UMR 7255, IMM FR3479, IM2B, Marseille, France
- IHU Méditerranée Infection, Aix-Marseille Univ., Marseille, France
| | | | - Romain Avellan
- Aix-Marseille Univ., CNRS, LISM-UMR 7255, IMM FR3479, IM2B, Marseille, France
| | - Ivy Mallick
- Aix-Marseille Univ., CNRS, LISM-UMR 7255, IMM FR3479, IM2B, Marseille, France
| | - Léa Celik
- Aix-Marseille Univ., CNRS, LISM-UMR 7255, IMM FR3479, IM2B, Marseille, France
| | - Pierre Santucci
- Aix-Marseille Univ., CNRS, LISM-UMR 7255, IMM FR3479, IM2B, Marseille, France
| | - Gael Brasseur
- Aix-Marseille Univ., CNRS, LCB-UMR 7283, IMM FR3479, IM2B, Marseille, France
| | - Isabelle Poncin
- Aix-Marseille Univ., CNRS, LISM-UMR 7255, IMM FR3479, IM2B, Marseille, France
| | - Vanessa Point
- Aix-Marseille Univ., CNRS, LISM-UMR 7255, IMM FR3479, IM2B, Marseille, France
| | - Stéphane Audebert
- Aix-Marseille Univ., INSERM, CNRS, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Luc Camoin
- Aix-Marseille Univ., INSERM, CNRS, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Wassim Daher
- CNRS UMR 9004, IRIM, Université de Montpellier, Montpellier, France
- INSERM, IRIM, Montpellier, France
| | | | - Laurent Kremer
- CNRS UMR 9004, IRIM, Université de Montpellier, Montpellier, France
- INSERM, IRIM, Montpellier, France
| | - Stéphane Canaan
- Aix-Marseille Univ., CNRS, LISM-UMR 7255, IMM FR3479, IM2B, Marseille, France
| |
Collapse
|
3
|
Kasper M, Karlstetter M, Wildschütz L, Scholz R, Busch M, Bauer D, Meyer Zu Hörste G, Thanos S, Langmann T, Heiligenhaus A. Kinetic changes in microglia-related retinal transcripts in experimental autoimmune uveoretinitis (EAU) of B10.RIII mice. J Neuroinflammation 2025; 22:37. [PMID: 39930455 PMCID: PMC11812248 DOI: 10.1186/s12974-025-03358-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 01/26/2025] [Indexed: 02/13/2025] Open
Abstract
In this study the retinal transcriptome was investigated during the development of experimental autoimmune uveoretinitis (EAU) in mice. EAU was induced by immunizing B10.RIII mice with human interphotoreceptor retinoid binding protein (hIRBP) 161-180 peptide. Genome-wide transcriptional profiles of EAU (day 7, 14 or 21 after immunization) and of control retinas were generated using DNA-microarrays and bioinformatic data mining. Microglia-associated transcripts were identified. Quantitative real-time polymerase chain reaction was performed to validate the expression of differentially expressed genes. Retinal transcript validation revealed that complement and interferon-related pathways, as well as gene clusters specific for antigen-processing and -presentation, and immunosuppression are involved during the course of the disease. Immunofluorescence analysis confirm that upregulated transcripts in EAU are also expressed by retinal microglia. Furthermore, the heterogenous expression patterns observed in retinal microglia, suggests the presence of different subpopulations of retinal microglia in EAU. This study expands our knowledge of the local immune processes involved in EAU pathology.
Collapse
Affiliation(s)
- Maren Kasper
- Department of Ophthalmology at St. Franziskus Hospital, Ophtha-Lab, Hohenzollernring 74, 48145, Münster, Germany.
| | - Marcus Karlstetter
- Chair of Experimental Immunology of the Eye, Department of Ophthalmology, University of Cologne, Cologne, Germany
| | - Lena Wildschütz
- Department of Ophthalmology at St. Franziskus Hospital, Ophtha-Lab, Hohenzollernring 74, 48145, Münster, Germany
| | - Rebecca Scholz
- Chair of Experimental Immunology of the Eye, Department of Ophthalmology, University of Cologne, Cologne, Germany
| | - Martin Busch
- Department of Ophthalmology at St. Franziskus Hospital, Ophtha-Lab, Hohenzollernring 74, 48145, Münster, Germany
| | - Dirk Bauer
- Department of Ophthalmology at St. Franziskus Hospital, Ophtha-Lab, Hohenzollernring 74, 48145, Münster, Germany
| | | | - Solon Thanos
- Institute for Experimental Ophthalmology, Westfalian Wilhelms-University of Münster, Münster, Germany
| | - Thomas Langmann
- Chair of Experimental Immunology of the Eye, Department of Ophthalmology, University of Cologne, Cologne, Germany
| | - Arnd Heiligenhaus
- Department of Ophthalmology at St. Franziskus Hospital, Ophtha-Lab, Hohenzollernring 74, 48145, Münster, Germany
- University of Duisburg-Essen, Duisburg, Germany
| |
Collapse
|
4
|
Singh A, Hu Y, Lopes RF, Lane L, Woldemichael H, Xu C, Udeshi ND, Carr SA, Perrimon N. Cell-death induced immune response and coagulopathy promote cachexia in Drosophila. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.07.631515. [PMID: 39829769 PMCID: PMC11741341 DOI: 10.1101/2025.01.07.631515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Tumors can exert a far-reaching influence on the body, triggering systemic responses that contribute to debilitating conditions like cancer cachexia. To characterize the mechanisms underlying tumor-host interactions, we utilized a BioID-based proximity labeling method to identify proteins secreted by Ykiact adult Drosophila gut tumors into the bloodstream/hemolymph. Among the major proteins identified are coagulation and immune-responsive factors that contribute to the systemic wasting phenotypes associated with Ykiact tumors. The effect of innate immunity factors is mediated by NFκB transcription factors Relish, dorsal, and Dif, which in turn upregulate the expression of the cachectic factors Pvf1, Impl2, and Upd3. In addition, Ykiact tumors secrete Eiger, a TNF-alpha homolog, which activates the JNK signaling pathway in neighboring non-tumor cells, leading to cell death. The release of damage-associated molecular patterns (DAMPs) from these dying cells presumably amplifies the inflammatory response, exacerbating systemic wasting. Targeting the inflammatory response, the JNK pathway, or the production of cachectic factors could potentially alleviate the debilitating effects of cancer cachexia.
Collapse
Affiliation(s)
- Ankita Singh
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 7 02115, USA
| | - Yanhui Hu
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 7 02115, USA
| | - Raphael Fragoso Lopes
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 7 02115, USA
| | - Liz Lane
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 7 02115, USA
| | | | - Charles Xu
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | | | - Steven A. Carr
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Norbert Perrimon
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 7 02115, USA
- HHMI, Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
5
|
Entrenas-García C, Suárez-Cárdenas JM, Fernández-Rodríguez R, Bautista R, Claros MG, Garrido JJ, Zaldívar-López S. miR-215 Modulates Ubiquitination to Impair Inflammasome Activation and Autophagy During Salmonella Typhimurium Infection in Porcine Intestinal Cells. Animals (Basel) 2025; 15:431. [PMID: 39943201 PMCID: PMC11815736 DOI: 10.3390/ani15030431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/25/2025] [Accepted: 01/31/2025] [Indexed: 02/16/2025] Open
Abstract
The host response to S. Typhimurium infection can be post-transcriptionally regulated by miRNAs. In this study, we investigated the role of miR-215 using both in vivo porcine infection models and in vitro intestinal epithelial cell lines. Several miRNAs were found to be dysregulated in the porcine ileum during infection with wild-type and SPI2-defective mutant strains of S. Typhimurium, with some changes being SPI2-dependent. Notably, miR-215 was significantly downregulated during infection. To explore its functional role, gain-of-function experiments were performed by transfecting porcine intestinal epithelial cells (IPEC-J2) with a miR-215-5p mimic, followed by label-free quantitative (LFQ) proteomic analysis. This analysis identified 157 proteins, of which 35 were downregulated in response to miR-215 overexpression, suggesting they are potential targets of this miRNA. Among these, E2 small ubiquitin-like modifier (SUMO)-conjugating enzyme UBC9 and E3 ubiquitin-ligase HUWE1 were identified as key targets, both of which are upregulated during S. Typhimurium infection. The miR-215-mediated downregulation of these proteins resulted in a significant decrease in overall ubiquitination, a process crucial for regulating inflammasome activation and autophagy. Consistently, inflammasome markers caspase 1 (CASP1) and apoptosis-associated speck-like protein containing a CARD (ASC), as well as autophagy markers microtubule-associated protein 1A/1B-light chain 3 (LC3B) and Ras-related protein Rab-11 (RAB11A), showed decreased expression in miR-215 mimic-transfected and infected IPEC-J2 cells. To further validate these findings, human intestinal epithelial cells (HT29) were used as a complementary model, providing additional insights into conserved immune pathways and extending the observations made in the porcine system. Overall, our findings demonstrate that miR-215 plays a significant role in modulating host inflammasome activation and autophagy by targeting proteins involved in ubiquitination during S. Typhimurium infection.
Collapse
Affiliation(s)
- Carmen Entrenas-García
- Immunogenomics and Molecular Pathogenesis Group, UIC Zoonosis and Emergent Diseases ENZOEM, Department of Genetics, University of Cordoba, 14014 Cordoba, Spain; (C.E.-G.); (J.M.S.-C.); (R.F.-R.)
| | - José M. Suárez-Cárdenas
- Immunogenomics and Molecular Pathogenesis Group, UIC Zoonosis and Emergent Diseases ENZOEM, Department of Genetics, University of Cordoba, 14014 Cordoba, Spain; (C.E.-G.); (J.M.S.-C.); (R.F.-R.)
- GA-14 Research Group, Maimónides Biomedical Research Institute of Córdoba (IMIBIC), 14004 Cordoba, Spain
| | - Raúl Fernández-Rodríguez
- Immunogenomics and Molecular Pathogenesis Group, UIC Zoonosis and Emergent Diseases ENZOEM, Department of Genetics, University of Cordoba, 14014 Cordoba, Spain; (C.E.-G.); (J.M.S.-C.); (R.F.-R.)
- GA-14 Research Group, Maimónides Biomedical Research Institute of Córdoba (IMIBIC), 14004 Cordoba, Spain
| | - Rocío Bautista
- Plataforma Andaluza de Bioinformática, Supercomputing and Bioinnovation Center (SCBI), Universidad de Málaga, 29590 Malaga, Spain; (R.B.); (M.G.C.)
- Institute of Biomedical Research in Malaga (IBIMA), IBIMA-RARE, 29590 Malaga, Spain
| | - M. Gonzalo Claros
- Plataforma Andaluza de Bioinformática, Supercomputing and Bioinnovation Center (SCBI), Universidad de Málaga, 29590 Malaga, Spain; (R.B.); (M.G.C.)
- Institute of Biomedical Research in Malaga (IBIMA), IBIMA-RARE, 29590 Malaga, Spain
- Institute for Mediterranean and Subtropical Horticulture “La Mayora” (IHSM-UMA-CSIC), 29590 Malaga, Spain
- Department of Molecular Biology and Biochemistry, Universidad de Málaga, 29010 Malaga, Spain
- CIBER de Enfermedades Raras (CIBERER) U741, 29071 Malaga, Spain
| | - Juan J. Garrido
- Immunogenomics and Molecular Pathogenesis Group, UIC Zoonosis and Emergent Diseases ENZOEM, Department of Genetics, University of Cordoba, 14014 Cordoba, Spain; (C.E.-G.); (J.M.S.-C.); (R.F.-R.)
- GA-14 Research Group, Maimónides Biomedical Research Institute of Córdoba (IMIBIC), 14004 Cordoba, Spain
| | - Sara Zaldívar-López
- Immunogenomics and Molecular Pathogenesis Group, UIC Zoonosis and Emergent Diseases ENZOEM, Department of Genetics, University of Cordoba, 14014 Cordoba, Spain; (C.E.-G.); (J.M.S.-C.); (R.F.-R.)
- GA-14 Research Group, Maimónides Biomedical Research Institute of Córdoba (IMIBIC), 14004 Cordoba, Spain
| |
Collapse
|
6
|
Li Q, Gamallat Y, Rokne JG, Bismar TA, Alhajj R. BioLake: an RNA expression analysis framework for prostate cancer biomarker powered by data lakehouse. BMC Bioinformatics 2025; 26:37. [PMID: 39905288 PMCID: PMC11792420 DOI: 10.1186/s12859-025-06050-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 01/14/2025] [Indexed: 02/06/2025] Open
Abstract
Biomedical researchers must often deal with large amounts of raw data, and analysis of this data might provide significant insights. However, if the raw data size is large, it might be difficult to uncover these insights. In this paper, a data framework named BioLake is presented that provides minimalist interactive methods to help researchers conduct bioinformatics data analysis. Unlike some existing analytical tools on the market, BioLake supports a wide range of web-based bioinformatics data analysis for public datasets, while allowing researchers to analyze their private datasets instantly. The tool also significantly enhances result interpretability by providing the source code and detailed instructions. In terms of data storage design, BioLake adopts the data lakehouse architecture to provide storage scalability and analysis flexibility. To further enhance the analysis efficiency, BioLake supports online analysis for custom data, allowing researchers to upload their own data via a designed procedure without waiting for server-side approval. BioLake allows a one-time upload of custom data of up to 500 MB to ensure that researchers avoid issues with data being too large for upload. In terms of the built-in dataset, BioLake applies reactive continuous data integration, helping the analysis pipeline to get rid of most preprocessing steps. The only pre-built-in dataset of BioLake in the first public version is TCGA-PRAD mRNA expression data for prostate cancer research, which is the primary focus of the development team of BioLake. In summary, BioLake offers a lightweight online tool to facilitate bioinformatic mRNA data analysis with the support of custom online data processing.
Collapse
Affiliation(s)
- Qiaowang Li
- Department of Computer Science, University of Calgary, Calgary, AB, Canada
| | - Yaser Gamallat
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, AB, Canada
- Arnie Charbonneau Cancer Institute and Tom Baker Cancer Center, Calgary, AB, Canada
- Departments of Oncology, Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
| | - Jon George Rokne
- Department of Computer Science, University of Calgary, Calgary, AB, Canada
| | - Tarek A Bismar
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, AB, Canada
- Arnie Charbonneau Cancer Institute and Tom Baker Cancer Center, Calgary, AB, Canada
- Departments of Oncology, Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
- Prostate Cancer Centre, Calgary, AB, Canada
| | - Reda Alhajj
- Department of Computer Science, University of Calgary, Calgary, AB, Canada.
- Department of Computer Engineering, Istanbul Medipol University, Istanbul, Turkey.
- Department of Health Informatics, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
7
|
Robinson JF, Das S, Khan W, Khanam R, Price JT, Rahman A, Ahmed S, Ali SM, Deb S, Deveale B, Dutta A, Gormley M, Hall SC, Hasan ASMT, Hotwani A, Juma MH, Kasaro MP, Khalid J, Kshetrapal P, McMaster MT, Mehmood U, Nisar I, Pervin J, Rahman S, Raqib R, San A, Sarker P, Tuomivaara ST, Zhang G, Zhou Y, Aktar S, Baqui AH, Jehan F, Sazawal S, Stringer JSA, Fisher SJ. High rates of placental inflammation among samples collected by the Multi-Omics for Mothers and Infants consortium. Am J Obstet Gynecol 2025; 232:230.e1-230.e19. [PMID: 38697337 PMCID: PMC11790342 DOI: 10.1016/j.ajog.2024.04.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 04/18/2024] [Accepted: 04/24/2024] [Indexed: 05/04/2024]
Abstract
BACKGROUND The Multi-Omics for Mothers and Infants consortium aims to improve birth outcomes. Preterm birth is a major obstetrical complication globally and causes significant infant and childhood morbidity and mortality. OBJECTIVE We analyzed placental samples (basal plate, placenta or chorionic villi, and the chorionic plate) collected by the 5 Multi-Omics for Mothers and Infants sites, namely The Alliance for Maternal and Newborn Health Improvement Bangladesh, The Alliance for Maternal and Newborn Health Improvement Pakistan, The Alliance for Maternal and Newborn Health Improvement Tanzania, The Global Alliance to Prevent Prematurity and Stillbirth Bangladesh, and The Global Alliance to Prevent Prematurity and Stillbirth Zambia. The goal was to analyze the morphology and gene expression of samples collected from preterm and uncomplicated term births. STUDY DESIGN The teams provided biopsies from 166 singleton preterm (<37 weeks' gestation) and 175 term (≥37 weeks' gestation) deliveries. The samples were fixed in formalin and paraffin embedded. Tissue sections from these samples were stained with hematoxylin and eosin and subjected to morphologic analyses. Other placental biopsies (n=35 preterm, 21 term) were flash frozen, which enabled RNA purification for bulk transcriptomics. RESULTS The morphologic analyses revealed a surprisingly high rate of inflammation that involved the basal plate, placenta or chorionic villi, and the chorionic plate. The rate of inflammation in chorionic villus samples, likely attributable to chronic villitis, ranged from 25% (Pakistan site) to 60% (Zambia site) of cases. Leukocyte infiltration in this location vs in the basal plate or chorionic plate correlated with preterm birth. Our transcriptomic analyses identified 267 genes that were differentially expressed between placentas from preterm vs those from term births (123 upregulated, 144 downregulated). Mapping the differentially expressed genes onto single-cell RNA sequencing data from human placentas suggested that all the component cell types, either singly or in subsets, contributed to the observed dysregulation. Consistent with the histopathologic findings, gene ontology analyses highlighted the presence of leukocyte infiltration or activation and inflammatory responses in both the fetal and maternal compartments. CONCLUSION The relationship between placental inflammation and preterm birth is appreciated in developed countries. In this study, we showed that this link also exists in developing geographies. In addition, among the participating sites, we found geographic- and population-based differences in placental inflammation and preterm birth, suggesting the importance of local factors.
Collapse
Affiliation(s)
- Joshua F Robinson
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Sciences, University of California, San Francisco, CA
| | - Sayan Das
- Public Health Laboratory Ivo de Carneri, Wawi, Chake, Pemba, Zanzibar, Tanzania
| | - Waqasuddin Khan
- Biorepository and Omics Research Group, Department of Pediatrics and Child Health, Faculty of Health Sciences, Medical College, The Aga Khan University, Stadium Road, Karachi, Pakistan; Department of Pediatrics and Child Health, Faculty of Health Sciences, Medical College, The Aga Khan University, Stadium Road, Karachi, Pakistan
| | - Rasheda Khanam
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Joan T Price
- UNC Global Projects - Zambia, Lusaka, Zambia; Department of Obstetrics and Gynecology, University of North Carolina School of Medicine, Chapel Hill, NC
| | - Anisur Rahman
- Maternal and Child Health Division, International Centre for Diarrhoeal Disease Research Bangladesh, Dhaka, Bangladesh
| | | | - Said Mohammed Ali
- Public Health Laboratory Ivo de Carneri, Wawi, Chake, Pemba, Zanzibar, Tanzania
| | - Saikat Deb
- Public Health Laboratory Ivo de Carneri, Wawi, Chake, Pemba, Zanzibar, Tanzania; Center for Public Health Kinetics, Vinoba Puri, Lajpatnagar II, New Delhi, India
| | - Brian Deveale
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Sciences, University of California, San Francisco, CA; Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA; Department of Urology, University of California, San Francisco, San Francisco, CA
| | - Arup Dutta
- Center for Public Health Kinetics, Vinoba Puri, Lajpatnagar II, New Delhi, India
| | - Matthew Gormley
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Sciences, University of California, San Francisco, CA; Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA; Sandler-Moore Mass Spectrometry Core Facility, University of California, San Francisco, San Francisco, CA
| | - Steven C Hall
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Sciences, University of California, San Francisco, CA; Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA; Sandler-Moore Mass Spectrometry Core Facility, University of California, San Francisco, San Francisco, CA
| | - A S M Tarik Hasan
- Maternal and Child Health Division, International Centre for Diarrhoeal Disease Research Bangladesh, Dhaka, Bangladesh
| | - Aneeta Hotwani
- Department of Pediatrics and Child Health, Faculty of Health Sciences, Medical College, The Aga Khan University, Stadium Road, Karachi, Pakistan
| | - Mohamed Hamid Juma
- Public Health Laboratory Ivo de Carneri, Wawi, Chake, Pemba, Zanzibar, Tanzania
| | - Margaret P Kasaro
- UNC Global Projects - Zambia, Lusaka, Zambia; Department of Obstetrics and Gynecology, University of North Carolina School of Medicine, Chapel Hill, NC; Department of Gynaecology and Obstetrics, University of Zambia School of Medicine, Lusaka, Zambia
| | - Javairia Khalid
- Biorepository and Omics Research Group, Department of Pediatrics and Child Health, Faculty of Health Sciences, Medical College, The Aga Khan University, Stadium Road, Karachi, Pakistan; Department of Pediatrics and Child Health, Faculty of Health Sciences, Medical College, The Aga Khan University, Stadium Road, Karachi, Pakistan
| | - Pallavi Kshetrapal
- Maternal and Child Health, Translational Health Science and Technology Institute, Faridabad, India
| | - Michael T McMaster
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Sciences, University of California, San Francisco, CA; Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA; Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA
| | - Usma Mehmood
- Department of Pediatrics and Child Health, Faculty of Health Sciences, Medical College, The Aga Khan University, Stadium Road, Karachi, Pakistan
| | - Imran Nisar
- Biorepository and Omics Research Group, Department of Pediatrics and Child Health, Faculty of Health Sciences, Medical College, The Aga Khan University, Stadium Road, Karachi, Pakistan; Department of Pediatrics and Child Health, Faculty of Health Sciences, Medical College, The Aga Khan University, Stadium Road, Karachi, Pakistan
| | - Jesmin Pervin
- Maternal and Child Health Division, International Centre for Diarrhoeal Disease Research Bangladesh, Dhaka, Bangladesh
| | - Sayedur Rahman
- Maternal and Child Health Division, International Centre for Diarrhoeal Disease Research Bangladesh, Dhaka, Bangladesh; Projahnmo Research Foundation, Dhaka, Bangladesh
| | - Rubhana Raqib
- International Centre for Diarrhoeal Disease Research Bangladesh, Dhaka, Bangladesh
| | - Ali San
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Sciences, University of California, San Francisco, CA; Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA
| | - Protim Sarker
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research Bangladesh, Dhaka, Bangladesh
| | - Sami T Tuomivaara
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Sciences, University of California, San Francisco, CA; Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA; Sandler-Moore Mass Spectrometry Core Facility, University of California, San Francisco, San Francisco, CA
| | - Ge Zhang
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH; Center for Prevention of Preterm Birth, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; March of Dimes Prematurity Research Center Ohio Collaborative, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Yan Zhou
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Sciences, University of California, San Francisco, CA; Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA
| | - Shaki Aktar
- Maternal and Child Health Division, International Centre for Diarrhoeal Disease Research Bangladesh, Dhaka, Bangladesh
| | - Abdullah H Baqui
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Fyezah Jehan
- Biorepository and Omics Research Group, Department of Pediatrics and Child Health, Faculty of Health Sciences, Medical College, The Aga Khan University, Stadium Road, Karachi, Pakistan; Department of Pediatrics and Child Health, Faculty of Health Sciences, Medical College, The Aga Khan University, Stadium Road, Karachi, Pakistan
| | - Sunil Sazawal
- Center for Public Health Kinetics, Vinoba Puri, Lajpatnagar II, New Delhi, India
| | - Jeffrey S A Stringer
- Department of Obstetrics and Gynecology, University of North Carolina School of Medicine, Chapel Hill, NC
| | - Susan J Fisher
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Sciences, University of California, San Francisco, CA; Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA; Sandler-Moore Mass Spectrometry Core Facility, University of California, San Francisco, San Francisco, CA.
| |
Collapse
|
8
|
Akhlaq R, Ahmed T, Khan T, Yaseen Jeelani SU, Joseph-Chowdhury JSN, Sidoli S, Musharraf SG, Ali A. PX-12 modulates vorinostat-induced acetylation and methylation marks in CAL 27 cells. Epigenomics 2025; 17:79-87. [PMID: 39716806 PMCID: PMC11792842 DOI: 10.1080/17501911.2024.2441652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 12/09/2024] [Indexed: 12/25/2024] Open
Abstract
AIM The hypoxic tumor microenvironment (TME) in oral squamous cell carcinoma (OSCC) is primarily regulated by hypoxia-inducible factor-1 alpha (HIF-1α), impacting histone acetylation and methylation, which contribute to drug resistance. Vorinostat, a histone deacetylase inhibitor (HDACi), de-stabilizes HIF-1α, while PX-12, a thioredoxin-1 (Trx-1) inhibitor, prevents HIF-1α accumulation. Combining HDACi with a Trx-1 inhibitor may enhance efficacy and reduce resistance by increasing reactive oxygen species (ROS) in cancer cells. This study examines how PX-12 influences vorinostat-induced histone modifications under hypoxia in the OSCC cell line CAL 27 using mass spectrometry. MATERIALS AND METHODS The OSCC cell line CAL 27 was used to assess histone post-translational modifications induced by PX-12 and Vorinostat under hypoxic conditions through mass spectrometry. RESULTS The proteomic analysis (ProteomeXchange identifier PXD053244) revealed several crucial histone marks, such as H3K4me1, H3K9ac, H3K9me, H3K14ac, H3K27me, H3K36me, H4K12Ac, and H4K16ac. Along with site-specific histone modifications, exposure of cells to vorinostat and PX-12 alone or in combination affects the global acetylation and methylation levels under hypoxia. CONCLUSION Mass spectrometry-based proteomics highlighted the impact of vorinostat and PX-12 on histone acetylation and methylation, offering valuable insights into the epigenetic mechanisms in OSCC and paving a way for epigenetic-based oral cancer therapeutics.
Collapse
Affiliation(s)
- Rafia Akhlaq
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Tehmina Ahmed
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Tajwali Khan
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Syed Usama Yaseen Jeelani
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | | | - Simone Sidoli
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Syed Ghulam Musharraf
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Arslan Ali
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| |
Collapse
|
9
|
Panahipour L, Kargarpour Z, Mildner M, Kühtreiber H, Gruber R. RNAseq of peripheral blood mononucleated cells exposed to platelet-rich fibrin and enamel matrix derivatives. Sci Rep 2025; 15:3661. [PMID: 39881164 PMCID: PMC11779933 DOI: 10.1038/s41598-025-86791-5] [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: 05/10/2024] [Accepted: 01/14/2025] [Indexed: 01/31/2025] Open
Abstract
Platelet-rich fibrin (PRF) and Enamel Matrix Derivatives (EMD) can support the local regenerative events in periodontal defects. There is reason to suggest that PRF and EMD exert part of their activity by targeting the blood-derived cells accumulating in the early wound healing blastema. However, the impact of PRF and EMD on blood cell response remains to be discovered. To this aim, we have exposed human peripheral blood mononucleated cells (PBMCs) to PRF lysates prepared by a swing-out rotor and EMD, followed by bulk RNA sequencing. A total of 111 and 8 genes are up- and down-regulated by PRF under the premise of an at least log2 two-fold change and a minus log10 significance level of two, respectively. Representative is a characteristic IFN response indicated by various human leukocyte antigens (HLA-DPA1, HLA-DPB1, HLA-DQA1, HLA-DQA2, HLA-DRA, HLA-DRB1, HLA-DRB5), gamma Fc receptors (FCGR1A, FCGR1B, FCGR3B), chemokines (CXCL9-11), and calprotectin (S100A8/9 and S100A12), complement (C1QA/B, C2) and interferon-induced guanylate-binding proteins (GBP1, GBP5). With EMD, 67 and 29 genes are up- and down-regulated, respectively. Characteristic of the upregulated genes are tensins (TNS1 and TNS3). Among the genes downregulated by EMD were epsilon Fc receptors (FCER1A; FCER2), Fc receptor-like proteins (FCRL1, FCRL3) and CX3CR1. Genes commonly upregulated by PRF and EMD were most noticeably NXPH4 and MN1, as well as FN1, MMP14, MERTK, and AXL. Our findings suggest that PRF provokes an inflammatory response, while EMD dampens IgE signaling in peripheral mononucleated blood cells.
Collapse
Affiliation(s)
- Layla Panahipour
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, Sensengasse 2a, 1090, Vienna, Austria
| | - Zahra Kargarpour
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, Sensengasse 2a, 1090, Vienna, Austria
- Department of Pulmonology, Medical University of Vienna, Vienna, Austria
| | - Michael Mildner
- Department of Dermatology, Medical University of Vienna, 1090, Vienna, Austria
| | - Hannes Kühtreiber
- Department of Dermatology, Medical University of Vienna, 1090, Vienna, Austria
| | - Reinhard Gruber
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, Sensengasse 2a, 1090, Vienna, Austria.
- Department of Periodontology, School of Dental Medicine, University of Bern, Bern, 3010, Switzerland.
- Austrian Cluster for Tissue Regeneration, 1200, Vienna, Austria.
| |
Collapse
|
10
|
Santos-García I, Bascuñana P, Brackhan M, Villa M, Eiriz I, Brüning T, Pahnke J. The ABC transporter A7 modulates neuroinflammation via NLRP3 inflammasome in Alzheimer's disease mice. Alzheimers Res Ther 2025; 17:30. [PMID: 39871385 PMCID: PMC11773842 DOI: 10.1186/s13195-025-01673-2] [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: 10/11/2024] [Accepted: 01/06/2025] [Indexed: 01/29/2025]
Abstract
BACKGROUND Specific genetic variants in the ATP-binding cassette transporter A7 locus (ABCA7) are associated with an increased risk of Alzheimer's disease (AD). ABCA7 transports lipids from/across cell membranes, regulates Aβ peptide processing and clearance, and modulates microglial and T-cell functions to maintain immune homeostasis in the brain. During AD pathogenesis, neuroinflammation is one of the key mechanisms involved. Therefore, we wanted to investigate the specific role of ABCA7 in microglial activation via the NLRP3 inflammasome. METHODS We developed the first humanized, Cre-inducible ABCA7flx knock-in mouse model, crossbred it with the APPPS1-21 β-amyloidosis model, and generated constitutive ABCA7ko and microglia Cx3cr1-specific conditional ABCA7ko AD mice. The role of ABCA7 was analyzed using histological, biochemical, molecular and mass spectrometry methods. RESULTS Constitutive knockout of the Abca7 gene in APPPS1 mice increased the levels of Aβ42 and the number of IBA1+ (microglia) and GFAP+ (astrocytes) cells. Changes in the levels of astrocytes and microglia are associated with the activation of the NLRP3 inflammasome and increased levels of proinflammatory cytokines, such as IL1β and TNFα. Interestingly, microglia-specific ABCA7ko restored Aβ42 peptide levels and IBA1+ and GFAP+ and NLRP3-related gene expression to the original APPPS1 mouse levels. In primary glial cell cultures of APPPS1-hA7ko microglia and APPPS1 astrocytes from newborn pups, we observed that conditioned media from LPS-stimulated microglia was able to induce NLRP3 inflammasome expression and proinflammatory cytokine release in astrocytes. CONCLUSIONS Our data suggest that ABCA7 transporters regulate the communication between microglia and astrocytes through the NLRP3 inflammasome and the release of proinflammatory cytokines. This regulation implicates ABCA7 as a key driver ultimately involved in the persistence of the inflammatory response observed in AD.
Collapse
Affiliation(s)
- Irene Santos-García
- Translational Neurodegeneration Research and Neuropathology Lab, Department of Clinical Medicine (KlinMed), Medical Faculty, University of Oslo (UiO) and Section of Neuropathology Research, Department of Pathology (PAT), Clinics for Laboratory Medicine (KLM), Oslo University Hospital (OUS), Sognsvannsveien 20, Oslo, NO-0372, Norway
| | - Pablo Bascuñana
- Translational Neurodegeneration Research and Neuropathology Lab, Department of Clinical Medicine (KlinMed), Medical Faculty, University of Oslo (UiO) and Section of Neuropathology Research, Department of Pathology (PAT), Clinics for Laboratory Medicine (KLM), Oslo University Hospital (OUS), Sognsvannsveien 20, Oslo, NO-0372, Norway
- Brain Mapping Group, Hospital Clínico San Carlos, IdISSC, Madrid, Spain
| | - Mirjam Brackhan
- Translational Neurodegeneration Research and Neuropathology Lab, Department of Clinical Medicine (KlinMed), Medical Faculty, University of Oslo (UiO) and Section of Neuropathology Research, Department of Pathology (PAT), Clinics for Laboratory Medicine (KLM), Oslo University Hospital (OUS), Sognsvannsveien 20, Oslo, NO-0372, Norway
- Brain Mapping Group, Hospital Clínico San Carlos, IdISSC, Madrid, Spain
| | - María Villa
- Translational Neurodegeneration Research and Neuropathology Lab, Department of Clinical Medicine (KlinMed), Medical Faculty, University of Oslo (UiO) and Section of Neuropathology Research, Department of Pathology (PAT), Clinics for Laboratory Medicine (KLM), Oslo University Hospital (OUS), Sognsvannsveien 20, Oslo, NO-0372, Norway
| | - Ivan Eiriz
- Translational Neurodegeneration Research and Neuropathology Lab, Department of Clinical Medicine (KlinMed), Medical Faculty, University of Oslo (UiO) and Section of Neuropathology Research, Department of Pathology (PAT), Clinics for Laboratory Medicine (KLM), Oslo University Hospital (OUS), Sognsvannsveien 20, Oslo, NO-0372, Norway
| | - Thomas Brüning
- Translational Neurodegeneration Research and Neuropathology Lab, Department of Clinical Medicine (KlinMed), Medical Faculty, University of Oslo (UiO) and Section of Neuropathology Research, Department of Pathology (PAT), Clinics for Laboratory Medicine (KLM), Oslo University Hospital (OUS), Sognsvannsveien 20, Oslo, NO-0372, Norway
| | - Jens Pahnke
- Translational Neurodegeneration Research and Neuropathology Lab, Department of Clinical Medicine (KlinMed), Medical Faculty, University of Oslo (UiO) and Section of Neuropathology Research, Department of Pathology (PAT), Clinics for Laboratory Medicine (KLM), Oslo University Hospital (OUS), Sognsvannsveien 20, Oslo, NO-0372, Norway.
- Institute of Nutritional Medicine (INUM), Lübeck Institute of Dermatology (LIED), University of Lübeck (UzL) and University Medical Center Schleswig-Holstein (UKSH), Ratzeburger Allee 160, DE-23538, Lübeck, Germany.
- Department of Neuromedicine and Neuroscience, The Faculty of Medicine and Life Sciences, University of Latvia (LU), Jelgavas iela 3, Rīga, LV-1004, Latvia.
- Department of Neurobiology, School of Neurobiology, Biochemistry and Biophysics, The Georg S. Wise Faculty of Life Sciences, Tel Aviv University (TAU), Ramat Aviv, IL-6997801, Israel.
| |
Collapse
|
11
|
Parikh RY, Nayak D, Lin H, Gangaraju VK. Drosophila Modulo is essential for transposon silencing and developmental robustness. J Biol Chem 2025; 301:108210. [PMID: 39848495 DOI: 10.1016/j.jbc.2025.108210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 12/31/2024] [Accepted: 01/16/2025] [Indexed: 01/25/2025] Open
Abstract
Transposable element (TE) silencing in the germline is crucial for preserving genome integrity; its absence results in sterility and diminished developmental robustness. The Piwi-interacting RNA (piRNA) pathway is the primary small non-coding RNA mechanism by which TEs are silenced in the germline. Three piRNA binding proteins promote the piRNA pathway function in the germline- P-element-induced wimpy testis (Piwi), Aubergine (Aub), and Argonaute 3 (Ago3). Piwi mediates transcriptional silencing of TEs by promoting the deposition of the heterochromatin mark Histone 3 lysine nine trimethylation (H3K9me3) at TE genomic sites. Aub and Ago3 facilitate post-transcriptional silencing of TEs. Proteins and mechanisms that promote piRNA function in TE silencing are still being discovered. This study demonstrates that the Drosophila Modulo protein, a homolog of mammalian Nucleolin and an epigenetic regulator, is crucial for the enrichment of H3K9me3 at TEs. We show that Modulo interacts with Piwi and operates downstream of the Piwi-piRNA complex's entry into the nucleus. Lack of Modulo function impairs Piwi-interacting protein Panoramix's ability to target transposon RNAs. Furthermore, the reduced function of Modulo in the mother undermines developmental robustness and exacerbates neomorphic Kr[If-1]-induced ectopic eye outgrowths in the offspring. Maternal Modulo enhances developmental robustness by inhibiting TE activation and transcriptome variability associated with intrinsic genetic variation. Thus, Modulo is an essential component of the mechanism that operates in the maternal germline to facilitate TE silencing and ensure developmental robustness in the ensuing generation.
Collapse
Affiliation(s)
- Rasesh Y Parikh
- Department of Biochemistry and Molecular Biology and Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Dhananjaya Nayak
- Department of Biochemistry and Molecular Biology and Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Haifan Lin
- Yale Stem Cell Center and Department of Cell Biology, Yale University, New Haven, Connecticut, USA
| | - Vamsi K Gangaraju
- Department of Biochemistry and Molecular Biology and Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina, USA.
| |
Collapse
|
12
|
Jaudas F, Bartenschlager F, Shashikadze B, Santamaria G, Reichart D, Schnell A, Stöckl JB, Degroote RL, Cambra JM, Graeber SY, Bähr A, Kartmann H, Stefanska M, Liu H, Naumann-Bartsch N, Bruns H, Berges J, Hanselmann L, Stirm M, Krebs S, Deeg CA, Blum H, Schulz C, Zawada D, Janda M, Caballero-Posadas I, Kunzelmann K, Moretti A, Laugwitz KL, Kupatt C, Saalmüller A, Fröhlich T, Wolf E, Mall MA, Mundhenk L, Gerner W, Klymiuk N. Perinatal dysfunction of innate immunity in cystic fibrosis. Sci Transl Med 2025; 17:eadk9145. [PMID: 39841805 DOI: 10.1126/scitranslmed.adk9145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 07/13/2024] [Accepted: 12/22/2024] [Indexed: 01/24/2025]
Abstract
In patients with cystic fibrosis (CF), repeated cycles of infection and inflammation eventually lead to fatal lung damage. Although diminished mucus clearance can be restored by highly effective CFTR modulator therapy, inflammation and infection often persist. To elucidate the role of the innate immune system in CF etiology, we investigated a CF pig model and compared these results with those for preschool children with CF. In newborn CF pigs, we observed changes in lung immune cell composition before the onset of infection that were dominated by increased monocyte infiltration, whereas neutrophil numbers remained constant. Flow cytometric and transcriptomic profiling revealed that the infiltrating myeloid cells displayed a more immature status. Cells with comparably immature transcriptomic profiles were enriched in the blood of CF pigs at birth as well as in preschool children with CF. This pattern coincided with decreased CD16 expression in the myeloid cells of both pigs and humans, which translated into lower phagocytic activity and reduced production of reactive oxygen species in both species. These results were indicative of a congenital, translationally conserved, and functionally relevant aberration of the immune system in CF. In newborn wild-type pigs, CFTR transcription in immune cells, including lung-derived and circulating monocytes, isolated from the bone marrow, thymus, spleen, and blood was below the detection limits of highly sensitive assays, suggesting an indirect etiology of the observed effects. Our findings highlight the need for additional immunological treatments to target innate immune deficits in patients with CF.
Collapse
Affiliation(s)
- Florian Jaudas
- First Department of Medicine, Cardiology, TUM University Hospital, Technical University of Munich, School of Medicine and Health, Munich 81675, Germany
- Chair of Molecular Animal Breeding and Biotechnology, Gene Center, LMU Munich, Munich 81377, Germany
- Center of Innovative Medical Models (CiMM), LMU Munich, Oberschleissheim 85764, Germany
- Medizinische Klinik und Poliklinik I, LMU Munich, Munich 81377, Germany
| | | | - Bachuki Shashikadze
- Laboratory for Functional Genome Analysis, Gene Center, LMU Munich, Munich 81377, Germany
| | - Gianluca Santamaria
- First Department of Medicine, Cardiology, TUM University Hospital, Technical University of Munich, School of Medicine and Health, Munich 81675, Germany
- Department of Experimental and Clinical Medicine, University "Magna Graecia" of Catanzaro, Catanzaro 88100, Italy
| | - Daniel Reichart
- Medizinische Klinik und Poliklinik I, LMU Munich, Munich 81377, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich 80802, Germany
- Gene Center Munich, LMU Munich, Munich 81377, Germany
| | - Alexander Schnell
- Department for Pediatric and Adolescent Medicine, University Hospital Erlangen, Erlangen 91054, Germany
| | - Jan Bernd Stöckl
- Laboratory for Functional Genome Analysis, Gene Center, LMU Munich, Munich 81377, Germany
| | - Roxane L Degroote
- Chair of Physiology, Department of Veterinary Sciences, LMU Munich, Planegg 82152, Germany
| | - Josep M Cambra
- First Department of Medicine, Cardiology, TUM University Hospital, Technical University of Munich, School of Medicine and Health, Munich 81675, Germany
- Center of Innovative Medical Models (CiMM), LMU Munich, Oberschleissheim 85764, Germany
| | - Simon Y Graeber
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine and Cystic Fibrosis Center, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 13353, Germany
- German Center for Lung Research (DZL), associated partner site, Berlin 13353, Germany
- German Center for Child and Adolescent Health (DZKJ), partner site, Berlin 13353, Germany
| | - Andrea Bähr
- First Department of Medicine, Cardiology, TUM University Hospital, Technical University of Munich, School of Medicine and Health, Munich 81675, Germany
- Center of Innovative Medical Models (CiMM), LMU Munich, Oberschleissheim 85764, Germany
| | - Heike Kartmann
- Medizinische Klinik und Poliklinik I, LMU Munich, Munich 81377, Germany
- Gene Center Munich, LMU Munich, Munich 81377, Germany
| | - Monika Stefanska
- Medizinische Klinik und Poliklinik I, LMU Munich, Munich 81377, Germany
| | - Huan Liu
- Medizinische Klinik und Poliklinik I, LMU Munich, Munich 81377, Germany
| | - Nora Naumann-Bartsch
- Department for Pediatric and Adolescent Medicine, University Hospital Erlangen, Erlangen 91054, Germany
| | - Heiko Bruns
- Department of Pediatrics and Adolescent Medicine, FAU Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen 91054, Germany
| | - Johannes Berges
- Department of Pediatrics and Adolescent Medicine, FAU Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen 91054, Germany
| | - Lea Hanselmann
- Institute of Veterinary Pathology, Freie Universität Berlin, Berlin 14163, Germany
| | - Michael Stirm
- Chair of Molecular Animal Breeding and Biotechnology, Gene Center, LMU Munich, Munich 81377, Germany
- Center of Innovative Medical Models (CiMM), LMU Munich, Oberschleissheim 85764, Germany
| | - Stefan Krebs
- Laboratory for Functional Genome Analysis, Gene Center, LMU Munich, Munich 81377, Germany
| | - Cornelia A Deeg
- Chair of Physiology, Department of Veterinary Sciences, LMU Munich, Planegg 82152, Germany
| | - Helmut Blum
- Laboratory for Functional Genome Analysis, Gene Center, LMU Munich, Munich 81377, Germany
| | - Christian Schulz
- Medizinische Klinik und Poliklinik I, LMU Munich, Munich 81377, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich 80802, Germany
- Department of Immunopharmacology, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, Heidelberg University, Mannheim 68167, Germany
| | - Dorota Zawada
- First Department of Medicine, Cardiology, TUM University Hospital, Technical University of Munich, School of Medicine and Health, Munich 81675, Germany
| | - Melanie Janda
- Chair of Molecular Animal Breeding and Biotechnology, Gene Center, LMU Munich, Munich 81377, Germany
| | | | - Karl Kunzelmann
- Institut für Physiologie, Universität Regensburg, Regensburg 93053, Germany
| | - Alessandra Moretti
- First Department of Medicine, Cardiology, TUM University Hospital, Technical University of Munich, School of Medicine and Health, Munich 81675, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich 80802, Germany
- Chair of Regenerative Medicine in Cardiovascular Disease, TUM University Hospital, Technical University of Munich, School of Medicine and Health, Munich 81675, Germany
| | - Karl-Ludwig Laugwitz
- First Department of Medicine, Cardiology, TUM University Hospital, Technical University of Munich, School of Medicine and Health, Munich 81675, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich 80802, Germany
| | - Christian Kupatt
- First Department of Medicine, Cardiology, TUM University Hospital, Technical University of Munich, School of Medicine and Health, Munich 81675, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich 80802, Germany
| | - Armin Saalmüller
- Institute of Immunology, University of Veterinary Medicine, Vienna 1210, Austria
| | - Thomas Fröhlich
- Laboratory for Functional Genome Analysis, Gene Center, LMU Munich, Munich 81377, Germany
| | - Eckhard Wolf
- Chair of Molecular Animal Breeding and Biotechnology, Gene Center, LMU Munich, Munich 81377, Germany
- Center of Innovative Medical Models (CiMM), LMU Munich, Oberschleissheim 85764, Germany
- Gene Center Munich, LMU Munich, Munich 81377, Germany
| | - Marcus A Mall
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine and Cystic Fibrosis Center, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 13353, Germany
- German Center for Lung Research (DZL), associated partner site, Berlin 13353, Germany
- German Center for Child and Adolescent Health (DZKJ), partner site, Berlin 13353, Germany
| | - Lars Mundhenk
- Institute of Veterinary Pathology, Freie Universität Berlin, Berlin 14163, Germany
| | - Wilhelm Gerner
- Institute of Immunology, University of Veterinary Medicine, Vienna 1210, Austria
| | - Nikolai Klymiuk
- First Department of Medicine, Cardiology, TUM University Hospital, Technical University of Munich, School of Medicine and Health, Munich 81675, Germany
- Center of Innovative Medical Models (CiMM), LMU Munich, Oberschleissheim 85764, Germany
| |
Collapse
|
13
|
Piñeiro-Ramil M, Gómez-Seoane I, Rodríguez-Cendal AI, Sanjurjo-Rodríguez C, Riva-Mendoza S, Fuentes-Boquete I, De Toro-Santos J, Señarís-Rodríguez J, Díaz-Prado S. Disease-Associated Signatures Persist in Extracellular Vesicles from Reprogrammed Cells of Osteoarthritis Patients. Int J Mol Sci 2025; 26:870. [PMID: 39940641 PMCID: PMC11816895 DOI: 10.3390/ijms26030870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/07/2025] [Accepted: 01/15/2025] [Indexed: 02/16/2025] Open
Abstract
Osteoarthritis (OA) is a prevalent joint disorder that lacks effective therapies to halt cartilage degeneration. Mesenchymal stromal cell (MSC)-derived small extracellular vesicles (sEVs) are being investigated as promising chondroprotective agents. Compared to primary MSCs, induced pluripotent stem cell (iPSC)-derived MSCs (MLCs) offer superior scalability and enhanced paracrine activity. The aim of this study was to explore the feasibility of using autologous MLC-derived sEVs as a potential therapeutic strategy for OA through the analysis of their protein cargo. iPSCs from an OA patient and a healthy donor were differentiated into MLCs. sEVs were isolated from these MLCs and characterized, with a particular focus on their protein cargo. Both iPSC lines were successfully differentiated into MLCs, which secreted sEVs with comparable size distributions and yields. The analysis of differentially expressed proteins revealed a high abundance of proteins associated with OA pathology and cartilage degradation in sEVs from OA MLCs compared to those from healthy MLCs. The persistence of OA-associated protein signatures in autologous MLC-derived sEVs may limit their therapeutic efficacy. These findings underscore the importance of carefully evaluating disease-specific protein profiles in sEVs for regenerative applications.
Collapse
Affiliation(s)
- María Piñeiro-Ramil
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Instituto de Investigación Biomédica de A Coruña (INIBIC), Fundación Pública Gallega de Investigación Biomédica INIBIC, Complexo Hospitalario Universitario de A Coruña (CHUAC), Servizo Galego de Saúde (SERGAS), 15006 A Coruña, Spain; (M.P.-R.); (I.G.-S.); (A.I.R.-C.); (I.F.-B.); (J.D.T.-S.); (J.S.-R.)
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidade da Coruña (UDC), 15006 A Coruña, Spain
- Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña (UDC), 15008 A Coruña, Spain
| | - Iván Gómez-Seoane
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Instituto de Investigación Biomédica de A Coruña (INIBIC), Fundación Pública Gallega de Investigación Biomédica INIBIC, Complexo Hospitalario Universitario de A Coruña (CHUAC), Servizo Galego de Saúde (SERGAS), 15006 A Coruña, Spain; (M.P.-R.); (I.G.-S.); (A.I.R.-C.); (I.F.-B.); (J.D.T.-S.); (J.S.-R.)
| | - Ana Isabel Rodríguez-Cendal
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Instituto de Investigación Biomédica de A Coruña (INIBIC), Fundación Pública Gallega de Investigación Biomédica INIBIC, Complexo Hospitalario Universitario de A Coruña (CHUAC), Servizo Galego de Saúde (SERGAS), 15006 A Coruña, Spain; (M.P.-R.); (I.G.-S.); (A.I.R.-C.); (I.F.-B.); (J.D.T.-S.); (J.S.-R.)
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidade da Coruña (UDC), 15006 A Coruña, Spain
- Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña (UDC), 15008 A Coruña, Spain
| | - Clara Sanjurjo-Rodríguez
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Instituto de Investigación Biomédica de A Coruña (INIBIC), Fundación Pública Gallega de Investigación Biomédica INIBIC, Complexo Hospitalario Universitario de A Coruña (CHUAC), Servizo Galego de Saúde (SERGAS), 15006 A Coruña, Spain; (M.P.-R.); (I.G.-S.); (A.I.R.-C.); (I.F.-B.); (J.D.T.-S.); (J.S.-R.)
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidade da Coruña (UDC), 15006 A Coruña, Spain
- Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña (UDC), 15008 A Coruña, Spain
| | - Selva Riva-Mendoza
- Grupo de Investigación en Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Fundación Pública Gallega de Investigación Biomédica INIBIC, Complexo Hospitalario Universitario de A Coruña (CHUAC), Servizo Galego de Saúde (SERGAS), 15006 A Coruña, Spain;
| | - Isaac Fuentes-Boquete
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Instituto de Investigación Biomédica de A Coruña (INIBIC), Fundación Pública Gallega de Investigación Biomédica INIBIC, Complexo Hospitalario Universitario de A Coruña (CHUAC), Servizo Galego de Saúde (SERGAS), 15006 A Coruña, Spain; (M.P.-R.); (I.G.-S.); (A.I.R.-C.); (I.F.-B.); (J.D.T.-S.); (J.S.-R.)
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidade da Coruña (UDC), 15006 A Coruña, Spain
- Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña (UDC), 15008 A Coruña, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
| | - Javier De Toro-Santos
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Instituto de Investigación Biomédica de A Coruña (INIBIC), Fundación Pública Gallega de Investigación Biomédica INIBIC, Complexo Hospitalario Universitario de A Coruña (CHUAC), Servizo Galego de Saúde (SERGAS), 15006 A Coruña, Spain; (M.P.-R.); (I.G.-S.); (A.I.R.-C.); (I.F.-B.); (J.D.T.-S.); (J.S.-R.)
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidade da Coruña (UDC), 15006 A Coruña, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
- Servicio de Reumatología, Complexo Hospitalario Universitario de A Coruña (CHUAC), Servizo Galego de Saúde (SERGAS), 15006 A Coruña, Spain
| | - José Señarís-Rodríguez
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Instituto de Investigación Biomédica de A Coruña (INIBIC), Fundación Pública Gallega de Investigación Biomédica INIBIC, Complexo Hospitalario Universitario de A Coruña (CHUAC), Servizo Galego de Saúde (SERGAS), 15006 A Coruña, Spain; (M.P.-R.); (I.G.-S.); (A.I.R.-C.); (I.F.-B.); (J.D.T.-S.); (J.S.-R.)
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidade da Coruña (UDC), 15006 A Coruña, Spain
- Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña (UDC), 15008 A Coruña, Spain
- Servicio de Cirugía Ortopédica y Traumatología, Complexo Hospitalario Universitario de A Coruña (CHUAC), Servizo Galego de Saúde (SERGAS), 15006 A Coruña, Spain
| | - Silvia Díaz-Prado
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Instituto de Investigación Biomédica de A Coruña (INIBIC), Fundación Pública Gallega de Investigación Biomédica INIBIC, Complexo Hospitalario Universitario de A Coruña (CHUAC), Servizo Galego de Saúde (SERGAS), 15006 A Coruña, Spain; (M.P.-R.); (I.G.-S.); (A.I.R.-C.); (I.F.-B.); (J.D.T.-S.); (J.S.-R.)
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidade da Coruña (UDC), 15006 A Coruña, Spain
- Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña (UDC), 15008 A Coruña, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
| |
Collapse
|
14
|
Kar S, Deis R, Ahmad A, Bogoch Y, Dominitz A, Shvaizer G, Sasson E, Mytlis A, Ben-Zvi A, Elkouby YM. The Balbiani body is formed by microtubule-controlled molecular condensation of Buc in early oogenesis. Curr Biol 2025; 35:315-332.e7. [PMID: 39793567 DOI: 10.1016/j.cub.2024.11.056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 10/01/2024] [Accepted: 11/22/2024] [Indexed: 01/13/2025]
Abstract
Vertebrate oocyte polarity has been observed for two centuries and is essential for embryonic axis formation and germline specification, yet its underlying mechanisms remain unknown. In oocyte polarization, critical RNA-protein (RNP) granules delivered to the oocyte's vegetal pole are stored by the Balbiani body (Bb), a membraneless organelle conserved across species from insects to humans. However, the mechanisms of Bb formation are still unclear. Here, we elucidate mechanisms of Bb formation in zebrafish through developmental biomolecular condensation. Using super-resolution microscopy, live imaging, biochemical, and genetic analyses in vivo, we demonstrate that Bb formation is driven by molecular condensation through phase separation of the essential intrinsically disordered protein Bucky ball (Buc). Live imaging, molecular analyses, and fluorescence recovery after photobleaching (FRAP) experiments in vivo reveal Buc-dependent changes in the Bb condensate's dynamics and apparent material properties, transitioning from liquid-like condensates to a solid-like stable compartment. Furthermore, we identify a multistep regulation by microtubules that controls Bb condensation: first through dynein-mediated trafficking of early condensing Buc granules, then by scaffolding condensed granules, likely through molecular crowding, and finally by caging the mature Bb to prevent overgrowth and maintain shape. These regulatory steps ensure the formation of a single intact Bb, which is considered essential for oocyte polarization and embryonic development. Our work offers insight into the long-standing question of the origins of embryonic polarity in non-mammalian vertebrates, supports a paradigm of cellular control over molecular condensation by microtubules, and highlights biomolecular condensation as a key process in female reproduction.
Collapse
Affiliation(s)
- Swastik Kar
- Department of Developmental Biology and Cancer Research, The Hebrew University of Jerusalem Faculty of Medicine, Ein-Kerem Campus, Jerusalem 9112102, Israel; Institute for Medical Research, Israel-Canada (IMRIC), Ein-Kerem Campus, Jerusalem 9112102, Israel
| | - Rachael Deis
- Department of Developmental Biology and Cancer Research, The Hebrew University of Jerusalem Faculty of Medicine, Ein-Kerem Campus, Jerusalem 9112102, Israel; Institute for Medical Research, Israel-Canada (IMRIC), Ein-Kerem Campus, Jerusalem 9112102, Israel
| | - Adam Ahmad
- Department of Developmental Biology and Cancer Research, The Hebrew University of Jerusalem Faculty of Medicine, Ein-Kerem Campus, Jerusalem 9112102, Israel; Institute for Medical Research, Israel-Canada (IMRIC), Ein-Kerem Campus, Jerusalem 9112102, Israel
| | - Yoel Bogoch
- Department of Developmental Biology and Cancer Research, The Hebrew University of Jerusalem Faculty of Medicine, Ein-Kerem Campus, Jerusalem 9112102, Israel; Institute for Medical Research, Israel-Canada (IMRIC), Ein-Kerem Campus, Jerusalem 9112102, Israel
| | - Avichai Dominitz
- Department of Developmental Biology and Cancer Research, The Hebrew University of Jerusalem Faculty of Medicine, Ein-Kerem Campus, Jerusalem 9112102, Israel; Institute for Medical Research, Israel-Canada (IMRIC), Ein-Kerem Campus, Jerusalem 9112102, Israel
| | - Gal Shvaizer
- Department of Developmental Biology and Cancer Research, The Hebrew University of Jerusalem Faculty of Medicine, Ein-Kerem Campus, Jerusalem 9112102, Israel; Institute for Medical Research, Israel-Canada (IMRIC), Ein-Kerem Campus, Jerusalem 9112102, Israel
| | - Esther Sasson
- Department of Developmental Biology and Cancer Research, The Hebrew University of Jerusalem Faculty of Medicine, Ein-Kerem Campus, Jerusalem 9112102, Israel; Institute for Medical Research, Israel-Canada (IMRIC), Ein-Kerem Campus, Jerusalem 9112102, Israel
| | - Avishag Mytlis
- Department of Developmental Biology and Cancer Research, The Hebrew University of Jerusalem Faculty of Medicine, Ein-Kerem Campus, Jerusalem 9112102, Israel; Institute for Medical Research, Israel-Canada (IMRIC), Ein-Kerem Campus, Jerusalem 9112102, Israel
| | - Ayal Ben-Zvi
- Department of Developmental Biology and Cancer Research, The Hebrew University of Jerusalem Faculty of Medicine, Ein-Kerem Campus, Jerusalem 9112102, Israel; Institute for Medical Research, Israel-Canada (IMRIC), Ein-Kerem Campus, Jerusalem 9112102, Israel
| | - Yaniv M Elkouby
- Department of Developmental Biology and Cancer Research, The Hebrew University of Jerusalem Faculty of Medicine, Ein-Kerem Campus, Jerusalem 9112102, Israel; Institute for Medical Research, Israel-Canada (IMRIC), Ein-Kerem Campus, Jerusalem 9112102, Israel.
| |
Collapse
|
15
|
Dhillon-Richardson RM, Haugan AK, Lyons LW, McKenna JK, Bronner ME, Martik ML. Reactivation of an Embryonic Cardiac Neural Crest Transcriptional Subcircuit During Zebrafish Heart Regeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.16.633462. [PMID: 39868190 PMCID: PMC11760256 DOI: 10.1101/2025.01.16.633462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
During vertebrate development, the heart primarily arises from mesoderm, with crucial contributions from cardiac neural crest cells that migrate to the heart and form a variety of cardiovascular derivatives. Here, by integrating bulk and single cell RNA-seq with ATAC-seq, we identify a gene regulatory subcircuit specific to migratory cardiac crest cells composed of key transcription factors egr1, sox9a, tfap2a and ets1. Notably, we show that cells expressing the canonical neural crest gene sox10 are essential for proper cardiac regeneration in adult zebrafish. Furthermore, expression of all transcription factors from the migratory cardiac crest gene subcircuit are reactivated after injury at the wound edge. Together, our results uncover a developmental gene regulatory network that is important for cardiac neural crest fate determination, with key factors reactivated during regeneration.
Collapse
Affiliation(s)
| | - Alexandra K. Haugan
- Department of Molecular and Cell Biology, University of California, Berkeley; Berkeley, CA, USA
| | - Luke W. Lyons
- Department of Molecular and Cell Biology, University of California, Berkeley; Berkeley, CA, USA
| | - Joseph K. McKenna
- Department of Molecular and Cell Biology, University of California, Berkeley; Berkeley, CA, USA
| | | | - Megan L. Martik
- Department of Molecular and Cell Biology, University of California, Berkeley; Berkeley, CA, USA
- Division of Biology, California Institute of Technology; Pasadena, CA, USA
| |
Collapse
|
16
|
Venkataraman M, Infante V, Sabat G, Sanos-Giles K, Ané JM, Pfleger BF. A Novel Membrane-Associated Protein Aids Bacterial Colonization of Maize. ACS Synth Biol 2025; 14:206-215. [PMID: 39707987 PMCID: PMC11747777 DOI: 10.1021/acssynbio.4c00489] [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] [Indexed: 12/23/2024]
Abstract
The soil environment affected by plant roots and their exudates, termed the rhizosphere, significantly impacts crop health and is an attractive target for engineering desirable agricultural traits. Engineering microbes in the rhizosphere is one approach to improving crop yields that directly minimizes the number of genetic modifications made to plants. Soil microbes have the potential to assist with nutrient acquisition, heat tolerance, and drought response if they can persist in the rhizosphere in the correct numbers. Unfortunately, the mechanisms by which microbes adhere and persist on plant roots are poorly understood, limiting their application. This study examined the membrane proteome shift upon adherence to roots in two bacteria of interest, Klebsiella variicola and Pseudomonas putida. From this surface proteome data, we identified a novel membrane protein from a nonlaboratory isolate of P. putida that increases binding to maize roots using unlabeled proteomics. When this protein was moved from the environmental isolate to a common lab strain (P. putida KT2440), we observed increased binding capabilities of P. putida KT2440 to both abiotic mimic surfaces and maize roots. We observed a similar increased binding capability to maize roots when the protein was heterologously expressed in K. variicola and Stutzerimonas stutzeri. With the discovery of this novel binding protein, we outline a strategy for harnessing natural selection and wild isolates to build more persistent strains of bacteria for field applications and plant growth promotion.
Collapse
Affiliation(s)
- Maya Venkataraman
- Department of Chemical and Biological Engineering, University of Wisconsin – Madison, Madison, WI, USA 53706
| | - Valentina Infante
- Department of Bacteriology, University of Wisconsin – Madison, Madison, WI, USA 53706
| | - Grzegorz Sabat
- Biotechnology Center, University of Wisconsin – Madison, Madison, WI, USA 53706
| | - Kai Sanos-Giles
- Department of Chemical and Biological Engineering, University of Wisconsin – Madison, Madison, WI, USA 53706
| | - Jean-Michel Ané
- Department of Bacteriology, University of Wisconsin – Madison, Madison, WI, USA 53706
- Department of Plant and Agroecosystem Sciences, University of Wisconsin – Madison, Madison, WI, USA 53705
| | - Brian F. Pfleger
- Department of Chemical and Biological Engineering, University of Wisconsin – Madison, Madison, WI, USA 53706
| |
Collapse
|
17
|
Subramanian V, Juhr D, Giansanti P, Grumbach IM. Long-Term Effects of Radiation Therapy on Cerebral Microvessel Proteome: A Six-Month Post-Exposure Analysis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.13.632491. [PMID: 39868171 PMCID: PMC11760261 DOI: 10.1101/2025.01.13.632491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Background Radiation therapy (RT) treats primary and metastatic brain tumors, with about one million Americans surviving beyond six months post-treatment. However, up to 90% of survivors experience RT-induced cognitive impairment. Emerging evidence links cognitive decline to RT-induced endothelial dysfunction in brain microvessels, yet in vivo studies of endothelial injury remain limited. Investigating the molecular and cellular pathways connecting RT, endothelial dysfunction, and cognitive impairment is vital for developing targeted interventions. This study examines proteomic changes in cerebral microvessels following RT. Methods We conducted a comprehensive quantitative analysis comparing the proteome in cerebral microvessels from five control mice and five irradiated mice (12 Gy) 6 months after RT. Bioinformatics analyses included gene ontology (GO) enrichment, Mitocarta analysis, Ingenuity Pathway Analysis (IPA), and iPathwayGuide. Predictions from the analyses were validated by western blotting. Results Our data identified significant dysregulation of 414 proteins following RT, with 157 upregulated and 257 downregulated. Gene ontology analysis indicated that the majority of the dysregulated proteins were part of various metabolic pathways. Cross referencing with Mitocarta revealed a significant presence of mitochondrial proteins among the dysregulated proteins, indicating potential mitochondrial metabolic dysfunction. Further investigation with IPA analysis uncovered 76 enriched canonical pathways, 34 transcription regulators, 6 nuclear receptors, and 5 growth factors involved in RT-induced damage responses in cerebral microvessels. IPA canonical pathway analysis predicted mitochondrial dysfunction due to inhibition of various metabolic pathways in the irradiated group. Validation with western blotting confirmed the bioinformatics predictions from the proteomic dataset. Conclusions Our data show significant proteomic changes in cerebral microvessels 6 months post-radiation, including oxidative phosphorylation, the TCA cycle, and glycolysis, suggesting metabolic mechanisms of RT-induced microvascular dysfunction.
Collapse
Affiliation(s)
- Vikram Subramanian
- Abboud Cardiovascular Research Center, Department of Internal Medicine, Carver College of Medicine, University of Iowa
| | - Denise Juhr
- Abboud Cardiovascular Research Center, Department of Internal Medicine, Carver College of Medicine, University of Iowa
| | - Piero Giansanti
- Bavarian Center for Biomolecular Mass Spectrometry at Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Isabella M. Grumbach
- Abboud Cardiovascular Research Center, Department of Internal Medicine, Carver College of Medicine, University of Iowa
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Carver College of Medicine, University of Iowa
- Iowa City VA Healthcare System, Iowa City, IA
| |
Collapse
|
18
|
Richter A, Mörl H, Thielemann M, Kleemann M, Geißen R, Schwarz R, Albertz C, Koch P, Petzold A, Kroll T, Groth M, Hartmann N, Herpin A, Englert C. The master male sex determinant Gdf6Y of the turquoise killifish arose through allelic neofunctionalization. Nat Commun 2025; 16:540. [PMID: 39788971 PMCID: PMC11718055 DOI: 10.1038/s41467-025-55899-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 01/03/2025] [Indexed: 01/30/2025] Open
Abstract
Although sex determination is a fundamental process in vertebrate development, it is very plastic. Diverse genes became major sex determinants in teleost fishes. Deciphering how individual sex-determining genes orchestrate sex determination can reveal new actors in sexual development. Here, we demonstrate that the Y-chromosomal copy of the TGF-β family member gdf6 (gdf6Y) in Nothobranchius furzeri, an emerging model organism in aging research, gained the function of the male sex determinant through allelic diversification while retaining the skeletal developmental function shared with the X-chromosomal gdf6 allele (gdf6X). Concerning sex determination, gdf6Y is expressed by somatic supporting cells of the developing testes. There it induces the male sex in a germ cell-independent manner in contrast to sex determination in zebrafish and the medaka. Looking for downstream effectors of Gdf6Y, we identified besides TGF-β signaling modulators, especially the inhibitor of DNA binding genes id1/2/3, the mRNA decay activator zfp36l2 as a new GDF6 signaling target.
Collapse
Affiliation(s)
- Annekatrin Richter
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany.
| | - Hanna Mörl
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Maria Thielemann
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
- BianoGMP GmbH, Gera, Germany
| | - Markus Kleemann
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
- Abbott Rapid Diagnostics Jena GmbH, Jena, Germany
| | - Raphael Geißen
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Robert Schwarz
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Carolin Albertz
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Philipp Koch
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Andreas Petzold
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
- DRESDEN-concept e. V., Technical University (TU) Dresden, Dresden, Germany
| | - Torsten Kroll
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Marco Groth
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Nils Hartmann
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
- Institute of Pathology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Amaury Herpin
- INRAE, UR1037 Laboratory of Fish Physiology and Genomics, Campus de Beaulieu, Rennes, France
| | - Christoph Englert
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany.
- Institute of Biochemistry and Biophysics, Friedrich Schiller University Jena, Jena, Germany.
| |
Collapse
|
19
|
Yoneda K, Sendo S, Okano T, Shimizu H, Yamada H, Nishimura K, Ueda Y, Saegusa J. Impact of dysregulated microbiota-derived C18 polyunsaturated fatty acid metabolites on arthritis severity in mice with collagen-induced arthritis. Front Immunol 2025; 15:1444892. [PMID: 39850876 PMCID: PMC11754244 DOI: 10.3389/fimmu.2024.1444892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 12/09/2024] [Indexed: 01/25/2025] Open
Abstract
Objective We aimed to evaluate microbiome and microbiota-derived C18 dietary polyunsaturated fatty acids (PUFAs), such as conjugated linoleic acid (CLA), and to investigate their differences that correlate with arthritis severity in collagen-induced arthritis (CIA) mice. Methods On day 84 after induction, during the chronic phase of arthritis, cecal samples were analyzed using 16S rRNA sequencing, and plasma and cecal digesta were evaluated using liquid chromatography-tandem mass spectrometry. Differences in microbial composition between 10 control (Ctrl) and 29 CIA mice or between the mild and severe subgroups based on arthritis scores were identified. The cecal metabolite profile and its correlation with the microbiome were evaluated with respect to arthritis severity. Results The hydroxy and oxo metabolite levels were higher in CIA mice than in Ctrl mice, some of which, including 10-hydroxy-cis-6-18:1, were positively correlated with arthritis scores. The 9-trans,11-trans CLA levels in CIA mice had a negative linear correlation with arthritis scores. Microbial diversity was lower in severe CIA mice than in mild CIA or Ctrl mice. The abundance of Lactobacillus relatively increased in the severe subgroup of CIA mice compared with that in the mild subgroup and was positively correlated with arthritis severity. Conclusion Alterations in gut microbiota and microbiota-derived C18 PUFA metabolites are associated in CIA mice and correlated with arthritis scores, indicating that plasma or fecal C18 PUFA metabolites can be potential biomarkers for arthritis severity and dysbiosis.
Collapse
Affiliation(s)
- Katsuhiko Yoneda
- Department of Rheumatology and Clinical Immunology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Sho Sendo
- Department of Rheumatology and Clinical Immunology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Takaichi Okano
- Department of Rheumatology and Clinical Immunology, Kobe University Graduate School of Medicine, Kobe, Japan
| | | | - Hirotaka Yamada
- Department of Rheumatology and Clinical Immunology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Keisuke Nishimura
- Department of Rheumatology and Clinical Immunology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yo Ueda
- Department of Rheumatology and Clinical Immunology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Jun Saegusa
- Department of Rheumatology and Clinical Immunology, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
20
|
Sridharan K, Sivaramakrishnan G. Interaction between dipeptidyl-peptidase-4 inhibitors and drugs acting on renin angiotensin aldosterone system for the risk of angioedema: a pharmacovigilance assessment using disproportionality and interaction analyses. Diabetol Metab Syndr 2025; 17:7. [PMID: 39773243 PMCID: PMC11706060 DOI: 10.1186/s13098-024-01570-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 12/28/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Dipeptidyl peptidase-4 inhibitors (DPP-4is) and drugs interfering with the renin-angiotensin-aldosterone system (RAAS) are frequently co-prescribed in type 2 diabetes management. Both drug classes have been independently associated with angioedema, raising concerns about potential interaction risks. This study aimed to evaluate the safety signals and interaction patterns for angioedema associated with DPP-4is alone and in combination with RAAS-interfering drugs. METHODS We conducted a comprehensive pharmacovigilance analysis using the United States Food and Drug Administration Adverse Event Reporting System (USFDA AERS) database. Disproportionality analyses employing both frequentist (Reporting Odds Ratio, Proportional Reporting Ratio) and Bayesian approaches were performed. Drug-drug interactions were assessed using multiplicative drug-drug interaction model. Additionally, we reviewed published case reports of DPP-4i-associated angioedema. RESULTS Analysis of 29,163,222 reports identified 588 cases of DPP-4i-associated angioedema. Significant safety signals were detected for DPP-4i monotherapies, while combinations with RAAS-interfering drugs demonstrated stronger signals through both frequentist and Bayesian analyses. Significant interaction signals were observed for sitagliptin/irbesartan, sitagliptin/valsartan, linagliptin/valsartan and alogliptin/lisinopril combinations. Alogliptin/lisinopril and sitagliptin/irbesartan combinations showed the highest risk profiles. Angioedema occurred predominantly in elderly patients (> 65 years) and females. Sixteen case reports corroborated the findings from the database assessment. Clinical outcomes were comparable between monotherapy and combination therapy groups. CONCLUSION This pharmacovigilance analysis reveals significant safety signals for angioedema with specific DPP-4i combinations with RAAS-interfering drugs, suggesting potential drug-drug interactions. These findings emphasize the need for careful patient monitoring, particularly in vulnerable populations, when prescribing these combinations. Further prospective studies are warranted to validate these findings and establish definitive causal relationships.
Collapse
Affiliation(s)
- Kannan Sridharan
- Department of Pharmacology & Therapeutics, College of Medicine & Health Sciences, Arabian Gulf University, Manama, Kingdom of Bahrain.
| | | |
Collapse
|
21
|
Taylor J, Lal S, Braet F, McLachlan CS, Li A. The molecular and cellular landscape of hypertrophic cardiomyopathy phenotypes: transition from obstructive to end-stage heart failure. J Mol Med (Berl) 2025; 103:113-123. [PMID: 39774683 DOI: 10.1007/s00109-024-02508-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 12/10/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025]
Abstract
Hypertrophic cardiomyopathy (HCM) is a myocardial disorder which commonly presents as an obstructive or end-stage disease. This study aims to investigate the transcriptomic changes related to cardiac cell-specific expression profiles that underpin the molecular transition between the HCM phenotypes. This study utilizes bioinformatics meta-analysis to integrate independent datasets to generate a comprehensive gene expression profile of obstructive HCM and end-stage HCM phenotypes compared to donor hearts. Gene set enrichment and cellular deconvolution were applied to identify ontologies and pathways related to each phenotype and to enumerate cell abundances. The intersection between cell lineage genes and meta-genes was identified to explore the cellular contribution to the phenotypic molecular signatures. Meta-analysis revealed, enhanced muscle function and myocardial remodeling, alongside impaired immune and inflammatory processes in obstructive HCM. In contrast, enriched tissue matrix remodeling pathways and altered metabolic and signaling cascades were identified in end-stage HCM, indicating a shift towards cellular dysfunction and loss of homeostasis. These molecular profiles were associated with an altered cellular landscape, with increased cardiomyocytes and lower immune cell populations in obstructive samples but increased fibroblasts and smooth muscle cells in end-stage HCM, implicating extensive tissue remodeling. This study provides novel insights into the cellular contributions of contractile, immune, homeostatic, and vascular alterations underpinning each of the HCM phenotypes. KEY MESSAGES: HCM phenotypes are characterized by distinct molecular and cellular profiles. Obstructive HCM has an enriched contractile profile underpinned by an expanded cardiomyocyte population. End-stage HCM shifts the cellular profile towards extracellular and vascular remodeling.
Collapse
Affiliation(s)
- Jude Taylor
- Centre for Healthy Futures, Torrens University Australia, Surry Hills, NSW, 2010, Australia
| | - Sean Lal
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, 2006, Australia
| | - Filip Braet
- School of Medical Sciences (Molecular and Cellular Biomedicine), University of Sydney, Sydney, NSW, 2006, Australia
- Australian Centre for Microscopy and Microanalysis, University of Sydney, Sydney, NSW, 2006, Australia
| | - Craig S McLachlan
- Centre for Healthy Futures, Torrens University Australia, Surry Hills, NSW, 2010, Australia
| | - Amy Li
- Centre for Healthy Futures, Torrens University Australia, Surry Hills, NSW, 2010, Australia.
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, 2006, Australia.
- Department of Rural Clinical Sciences, La Trobe Rural Health School, La Trobe University, Bendigo, VIC, 3550, Australia.
| |
Collapse
|
22
|
Chaudhary A, Patil P, Raina P, Kaul-Ghanekar R. Matairesinol repolarizes M2 macrophages to M1 phenotype to induce apoptosis in triple-negative breast cancer cells. Immunopharmacol Immunotoxicol 2024:1-15. [PMID: 39722605 DOI: 10.1080/08923973.2024.2425028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 10/27/2024] [Indexed: 12/28/2024]
Abstract
OBJECTIVE Triple-Negative Breast Cancer (TNBC), the most challenging subtype of Breast Cancer (BC), currently lacks targeted therapy, presenting a significant therapeutic gap in its management. Tumor Associated Macrophages (TAMs) play a significant role in TNBC progression and could be targeted by repolarizing them from M2 to M1 phenotype. Matairesinol (MAT), a plant lignan, has been shown to exhibit anticancer, anti-inflammatory and immunomodulatory activities. In this study, we explored how MAT-induced repolarization of THP-1-derived M2 macrophages towards the M1 phenotype, which could effectively target the TNBC cell line, MDA-MB-231. METHODS The differential expression of genes in THP-1-derived macrophages at mRNA levels was evaluated by RNAseq assay. An inverted microscope equipped with a CMOS camera was utilized to capture the morphological variations in THP-1 cells and THP-1-derived macrophages. Relative mRNA expression of M1 and M2 specific marker genes was quantified by qRT-PCR. Cell viability and induction of apoptosis were evaluated by 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT) and 5,5',6,6'-tetrachloro-1,1',3,3'-tetraethylbenzimidazolylcarbocyanine iodide (JC-1 dye) assays, respectively. RESULTS MAT reduced the viability of M2a and M2d macrophages and repolarized them to M1 phenotype. Conditioned medium (CM) from MAT-treated M2a and M2d macrophages significantly reduced the viability of TNBC cells by apoptosis. CONCLUSION Targeting M2 macrophages is an important strategy to regulate cancer progression. Our study provides evidence that MAT may be a promising drug candidate for developing novel anti-TNBC therapy. However, further studies are warranted to thoroughly elucidate the molecular mechanism of action of MAT and evaluate its therapeutic potential in TNBC in vitro and in vivo models.
Collapse
Affiliation(s)
- Amol Chaudhary
- Cancer Research Lab, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, India
| | - Prajakta Patil
- Cancer Research Lab, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, India
| | - Prerna Raina
- Cancer Research Lab, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, India
- Analytical Department (ADT), Lupin Limited, Pune, India
| | - Ruchika Kaul-Ghanekar
- Cancer Research Lab, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, India
- Symbiosis Centre for Research and Innovation (SCRI); Symbiosis International Deemed University (SIU), Pune, India
- Cancer Research Lab, Symbiosis School of Biological Sciences (SSBS), Symbiosis International Deemed University (SIU), Pune, India
| |
Collapse
|
23
|
Panahipour L, Imani A, dos Santos Sanches N, Kühtreiber H, Mildner M, Gruber R. RNA Sequencing Revealed a Weak Response of Gingival Fibroblasts Exposed to Hyaluronic Acid. Bioengineering (Basel) 2024; 11:1307. [PMID: 39768125 PMCID: PMC11726844 DOI: 10.3390/bioengineering11121307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/11/2024] [Accepted: 12/16/2024] [Indexed: 01/16/2025] Open
Abstract
Hyaluronic acid was proposed to support soft tissue recession surgery and guided tissue regeneration. The molecular mechanisms through which hyaluronic acid modulates the response of connective tissue cells remain elusive. To elucidate the impact of hyaluronic acid on the connective tissue cells, we used bulk RNA sequencing to determine the changes in the genetic signature of gingival fibroblasts exposed to 1.6% cross-linked hyaluronic acid and 0.2% natural hyaluronic acid. Transcriptome-wide changes were modest. Even when implementing a minimum of 1.5 log2 fold-change and a significance threshold of 1.0 -log10, only a dozenth of genes were differentially expressed. Upregulated genes were PLK3, SLC16A6, IL6, HBEGF, DGKE, DUSP4, PTGS2, FOXC2, ATAD2B, NFATC2, and downregulated genes were MMP24 and PLXNA2. RT-PCR analysis supported the impact of hyaluronic acid on increasing the expression of a selected gene panel. The findings from bulk RNA sequencing suggest that gingival fibroblasts experience weak changes in their transcriptome when exposed to hyaluronic acid.
Collapse
Affiliation(s)
- Layla Panahipour
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, Sensengasse 2a, 1090 Vienna, Austria; (L.P.); (A.I.); (N.d.S.S.)
| | - Atefe Imani
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, Sensengasse 2a, 1090 Vienna, Austria; (L.P.); (A.I.); (N.d.S.S.)
| | - Natália dos Santos Sanches
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, Sensengasse 2a, 1090 Vienna, Austria; (L.P.); (A.I.); (N.d.S.S.)
- Department of Diagnosis and Surgery, Araçatuba Dental School of Sao Paulo, Sao Paulo 16015-050, Brazil
| | - Hannes Kühtreiber
- Department of Dermatology, Medical University of Vienna, 1090 Vienna, Austria; (H.K.); (M.M.)
| | - Michael Mildner
- Department of Dermatology, Medical University of Vienna, 1090 Vienna, Austria; (H.K.); (M.M.)
| | - Reinhard Gruber
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, Sensengasse 2a, 1090 Vienna, Austria; (L.P.); (A.I.); (N.d.S.S.)
- Department of Periodontology, School of Dental Medicine, University of Bern, 3010 Bern, Switzerland
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| |
Collapse
|
24
|
Brezovská B, Narasimhan S, Šiková M, Šanderová H, Kovaľ T, Borah N, Shoman M, Pospíšilová D, Vaňková Hausnerová V, Tužinčin D, Černý M, Komárek J, Janoušková M, Kambová M, Halada P, Křenková A, Hubálek M, Trundová M, Dohnálek J, Hnilicová J, Žídek L, Krásný L. MoaB2, a newly identified transcription factor, binds to σ A in Mycobacterium smegmatis. J Bacteriol 2024; 206:e0006624. [PMID: 39499088 PMCID: PMC11656743 DOI: 10.1128/jb.00066-24] [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: 02/22/2024] [Accepted: 09/18/2024] [Indexed: 11/07/2024] Open
Abstract
In mycobacteria, σA is the primary sigma factor. This essential protein binds to RNA polymerase (RNAP) and mediates transcription initiation of housekeeping genes. Our knowledge about this factor in mycobacteria is limited. Here, we performed an unbiased search for interacting partners of Mycobacterium smegmatis σA. The search revealed a number of proteins; prominent among them was MoaB2. The σA-MoaB2 interaction was validated and characterized by several approaches, revealing that it likely does not require RNAP and is specific, as alternative σ factors (e.g., closely related σB) do not interact with MoaB2. The structure of MoaB2 was solved by X-ray crystallography. By immunoprecipitation and nuclear magnetic resonance, the unique, unstructured N-terminal domain of σA was identified to play a role in the σA-MoaB2 interaction. Functional experiments then showed that MoaB2 inhibits σA-dependent (but not σB-dependent) transcription and may increase the stability of σA in the cell. We propose that MoaB2, by sequestering σA, has a potential to modulate gene expression. In summary, this study has uncovered a new binding partner of mycobacterial σA, paving the way for future investigation of this phenomenon.IMPORTANCEMycobacteria cause serious human diseases such as tuberculosis and leprosy. The mycobacterial transcription machinery is unique, containing transcription factors such as RbpA, CarD, and the RNA polymerase (RNAP) core-interacting small RNA Ms1. Here, we extend our knowledge of the mycobacterial transcription apparatus by identifying MoaB2 as an interacting partner of σA, the primary sigma factor, and characterize its effects on transcription and σA stability. This information expands our knowledge of interacting partners of subunits of mycobacterial RNAP, providing opportunities for future development of antimycobacterial compounds.
Collapse
Affiliation(s)
- Barbora Brezovská
- Laboratory of Microbial Genetics and Gene Expression, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Subhash Narasimhan
- Central European Institute of Technology (CEITEC), Masaryk University, Brno, Czechia
- Faculty of Science, National Centre for Biomolecular Research, Masaryk University, Brno, Czechia
| | - Michaela Šiková
- Laboratory of Microbial Genetics and Gene Expression, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Hana Šanderová
- Laboratory of Microbial Genetics and Gene Expression, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Tomáš Kovaľ
- Institute of Biotechnology of the Czech Academy of Sciences, Centre BIOCEV, Vestec, Czechia
| | - Nabajyoti Borah
- Laboratory of Microbial Genetics and Gene Expression, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Mahmoud Shoman
- Laboratory of Microbial Genetics and Gene Expression, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Debora Pospíšilová
- Laboratory of Microbial Genetics and Gene Expression, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Viola Vaňková Hausnerová
- Laboratory of Microbial Genetics and Gene Expression, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
- Laboratory of Regulatory RNAs, Faculty of Science, Charles University, Prague, Czechia
| | - Dávid Tužinčin
- Central European Institute of Technology (CEITEC), Masaryk University, Brno, Czechia
- Faculty of Science, National Centre for Biomolecular Research, Masaryk University, Brno, Czechia
| | - Martin Černý
- Central European Institute of Technology (CEITEC), Masaryk University, Brno, Czechia
- Faculty of Science, National Centre for Biomolecular Research, Masaryk University, Brno, Czechia
| | - Jan Komárek
- Central European Institute of Technology (CEITEC), Masaryk University, Brno, Czechia
- Faculty of Science, National Centre for Biomolecular Research, Masaryk University, Brno, Czechia
| | - Martina Janoušková
- Laboratory of Microbial Genetics and Gene Expression, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Milada Kambová
- Laboratory of Microbial Genetics and Gene Expression, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Petr Halada
- Institute of Microbiology of the Czech Academy of Sciences, Centre BIOCEV, Vestec, Czechia
| | - Alena Křenková
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czechia
| | - Martin Hubálek
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czechia
| | - Mária Trundová
- Institute of Biotechnology of the Czech Academy of Sciences, Centre BIOCEV, Vestec, Czechia
| | - Jan Dohnálek
- Institute of Biotechnology of the Czech Academy of Sciences, Centre BIOCEV, Vestec, Czechia
| | - Jarmila Hnilicová
- Laboratory of Microbial Genetics and Gene Expression, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
- Laboratory of Regulatory RNAs, Faculty of Science, Charles University, Prague, Czechia
| | - Lukáš Žídek
- Faculty of Science, National Centre for Biomolecular Research, Masaryk University, Brno, Czechia
| | - Libor Krásný
- Laboratory of Microbial Genetics and Gene Expression, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|
25
|
Randhawa A, Sinha T, Das M, Yazdani SS. AMPK Activates Cellulase Secretion in Penicillium funiculosum by Downregulating P-HOG1 MAPK Levels. J Basic Microbiol 2024:e2400658. [PMID: 39702928 DOI: 10.1002/jobm.202400658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 11/26/2024] [Indexed: 12/21/2024]
Abstract
Cellulase production for hydrolyzing plant cell walls is energy-intensive in filamentous fungi during nutrient scarcity. AMP-activated protein kinase (AMPK), encoded by snf1, is known to be the nutrient and energy sensor in eukaryotes. Previous studies on AMPK identified its role in alternate carbon utilization in pathogenic fungi. However, the precise role of AMPK in cellulase production remains elusive. In the present study, we employed gene-deletion analysis, quantitative proteomics and chemical-genetic approaches to investigate the role of AMPK in cellulase synthesis in Penicillium funiculosum. Gene-deletion analysis revealed that AMPK does not promote transcription and translation but is essential for cellulase secretion in a calcium-dependent manner. Proteomic analysis of the snf1-deleted (Δsnf1) strain confirmed trapped cellulase inside the mycelia and identified HOG1 MAPK activation as the most significant Ca2+-induced signaling event during carbon stress in Δsnf1. Western blot analysis analysis revealed that the phosphorylated HOG1 (P-HOG1)/HOG1 MAPK ratio maintained by Ca2+-signaling/Ca2+-activated AMPK, respectively, forms a secretion checkpoint for cellulases, and disturbing this equilibrium blocks cellulase secretion. The proteomic analysis also indicated a massive increase in mTORC1-activated anabolic pathways during carbon stress in Δsnf1. Our study suggests that AMPK maintains homeostasis by acting as a global repressor during carbon stress.
Collapse
Affiliation(s)
- Anmoldeep Randhawa
- Microbial Engineering Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
- Department of Microbiology, Amity University Punjab, Mohali, India
| | - Tulika Sinha
- Microbial Engineering Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Maitreyee Das
- Microbial Engineering Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Syed Shams Yazdani
- Microbial Engineering Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| |
Collapse
|
26
|
Thompson SL, Crowder SM, Hekmatara M, Sechrest ER, Deng WT, Robichaux MA. P23H rhodopsin aggregation in the ER causes synaptic protein imbalance in rod photoreceptors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.18.619115. [PMID: 39484588 PMCID: PMC11526887 DOI: 10.1101/2024.10.18.619115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Rod photoreceptor neurons in the retina detect scotopic light through the visual pigment rhodopsin (Rho) in their outer segments (OS). Efficient Rho trafficking to the OS through the inner rod compartments is critical for long-term rod health. Given the importance of protein trafficking to the OS, less is known about the trafficking of rod synaptic proteins. Furthermore, the subcellular impact of Rho mislocalization on rod synapses (i.e., "spherules") has not been investigated. In this study we used super-resolution and electron microscopies, along with proteomics, to perform a subcellular analysis of Rho synaptic mislocalization in P23H-Rho-RFP mutant mice. We discovered that mutant P23H-Rho-RFP protein mislocalized in distinct ER aggregations within the spherule cytoplasm, which we confirmed with AAV overexpression. Additionally, we found synaptic protein abundance differences in P23H-Rho-RFP mice. By comparison, Rho mislocalized along the spherule plasma membrane in WT and rd10 mutant rods, in which there was no synaptic protein disruption. Throughout the study, we also identified a network of ER membranes within WT rod presynaptic spherules. Together, our findings indicate that photoreceptor synaptic proteins are sensitive to ER dysregulation.
Collapse
Affiliation(s)
- Samantha L Thompson
- Department of Ophthalmology & Visual Sciences and Department of Biochemistry & Molecular Medicine, West Virginia University, Morgantown, WV 26506, United States
| | - Sophie M Crowder
- Department of Ophthalmology & Visual Sciences and Department of Biochemistry & Molecular Medicine, West Virginia University, Morgantown, WV 26506, United States
| | - Maryam Hekmatara
- Department of Ophthalmology & Visual Sciences and Department of Biochemistry & Molecular Medicine, West Virginia University, Morgantown, WV 26506, United States
| | - Emily R Sechrest
- Department of Ophthalmology & Visual Sciences and Department of Biochemistry & Molecular Medicine, West Virginia University, Morgantown, WV 26506, United States
| | - Wen-Tao Deng
- Department of Ophthalmology & Visual Sciences and Department of Biochemistry & Molecular Medicine, West Virginia University, Morgantown, WV 26506, United States
| | - Michael A Robichaux
- Department of Ophthalmology & Visual Sciences and Department of Biochemistry & Molecular Medicine, West Virginia University, Morgantown, WV 26506, United States
| |
Collapse
|
27
|
Luh D, Ghezellou P, Heiles S, Gramberg S, Haeberlein S, Spengler B. Glycolipidomics of Liver Flukes and Host Tissues during Fascioliasis: Insights from Mass Spectrometry Imaging. ACS Infect Dis 2024; 10:4233-4245. [PMID: 39510517 DOI: 10.1021/acsinfecdis.4c00551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
Fascioliasis, a zoonotic disease caused by liver flukes of the genus Fasciola, poses significant health threats to both humans and livestock. While some infections remain asymptomatic, others can lead to fatal outcomes, particularly during the acute phase characterized by the migration of immature parasites causing severe liver damage. Through the combination of data acquired via high-spatial-resolution atmospheric-pressure scanning microprobe matrix-assisted laser desorption/ionization mass spectrometry imaging (AP-SMALDI MSI) and nanohydrophilic interaction chromatography tandem mass spectrometry, we investigated glycosphingolipids (GSLs) in both adult and immature parasite stages as well as the host liver and bile duct to unravel the intricacies of the host-pathogen interplay and associated pathology. Several GSLs showed characteristic distribution patterns within the parasite depending on the fatty acid composition of their ceramides, notably including GSLs carrying very long-chain fatty acids. Additionally, GSL compositions within the tegument of immature versus adult parasites varied, suggestive of tissue remodeling upon maturation. AP-SMALDI MSI further enabled the identification of GSLs potentially involved in in vivo interactions between the host and immature parasites. Moreover, our experiments unveiled alterations in other lipid classes during Fasciola infection, providing a broader understanding of lipidomic changes associated with the disease. Collectively, our findings contribute to a deeper comprehension of the molecular intricacies underlying fascioliasis, with a specific focus on GSLs.
Collapse
Affiliation(s)
- David Luh
- Institute of Inorganic and Analytical Chemistry, Justus Liebig University Giessen, Giessen 35392, Germany
| | - Parviz Ghezellou
- Institute of Inorganic and Analytical Chemistry, Justus Liebig University Giessen, Giessen 35392, Germany
| | - Sven Heiles
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., Dortmund 44139, Germany
- Lipidomics, Faculty of Chemistry, University of Duisburg-Essen, Essen 45141, Germany
| | - Svenja Gramberg
- Institute of Parasitology, Biomedical Research Center Seltersberg (BFS), Justus Liebig University Giessen, Giessen 35392, Germany
| | - Simone Haeberlein
- Institute of Parasitology, Biomedical Research Center Seltersberg (BFS), Justus Liebig University Giessen, Giessen 35392, Germany
| | - Bernhard Spengler
- Institute of Inorganic and Analytical Chemistry, Justus Liebig University Giessen, Giessen 35392, Germany
| |
Collapse
|
28
|
Panahipour L, Micucci C, Gruber R. Inflammatory Response of THP1 and U937 Cells: The RNAseq Approach. Cells 2024; 13:2062. [PMID: 39768153 PMCID: PMC11674919 DOI: 10.3390/cells13242062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/09/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
THP1 and U937 are monocytic cell lines that are common bioassays to reflect monocyte and macrophage activities in inflammation research. However, THP-1 is a human monocytic leukemia cell line, and U937 originates from pleural effusion of histiocytic lymphoma; thus, even though they serve as bioassay in inflammation research, their response to agonists is not identical. Consequently, there has yet to be a consensus about the panel of strongly regulated genes in THP1 and U937 cells representing the inflammatory response to LPS and IFNG. Therefore, we have performed an RNAseq of THP1 and U937 exposed to LPS and IFNG to identify the most sensitive genes and the unique properties of each individual cell line. When applying a highly stringent threshold, we could identify 43, 8 up and 94, 103 down-regulated genes in THP1 and U937 cells, respectively. In THP1 cells, among the most strongly up-regulated genes are CCL1, CXCL2, CXCL3, IL1A, IL1B, IL6, and PTGES. In U937 cells, the strongest up-regulated genes include CSF2, CSF3, CXCL2, CXCL5, CXCL6, IL1A, IL19, IL36G, IL6, ITGA1, ITGA2, and PTGS2. Even though THP1 is considerably less responsive than U937, there are genes commonly upregulated by LPS and IFNG, including the CCL1, CCL3, CCL20, CXCL2, CXCL3, CXCL8, as well as IL1A, IL1B, IL23A, IL6, and genes of prostaglandin synthesis PTGES and PTGS2. Downregulated genes are limited to NRGN and CD36. This head-to-head comparison revealed that THP1 is less responsive than U937 cells to LPS and IFNG and identified a panel of highly regulated genes that can be applied in bioassays in inflammation research. Our data further propose bulk RNAseq as a standard method in bioassay research.
Collapse
Affiliation(s)
- Layla Panahipour
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria; (L.P.); (C.M.)
| | - Chiara Micucci
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria; (L.P.); (C.M.)
| | - Reinhard Gruber
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria; (L.P.); (C.M.)
- Department of Periodontology, School of Dental Medicine, University of Bern, 3010 Bern, Switzerland
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| |
Collapse
|
29
|
Teles RHG, Villarinho NJ, Yamagata AS, Hiroki CT, de Oliveira MC, Terçarioli GR, Jaeger RG, Meybohm P, Burek M, Freitas VM. Valosin-containing protein (VCP), a component of tumor-derived extracellular vesicles, impairs the barrier integrity of brain microvascular endothelial cells. BBA ADVANCES 2024; 7:100130. [PMID: 39802400 PMCID: PMC11722580 DOI: 10.1016/j.bbadva.2024.100130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 12/06/2024] [Accepted: 12/10/2024] [Indexed: 01/16/2025] Open
Abstract
Metastases are the leading cause of cancer-related deaths, and their origin is not fully elucidated. Recently, studies have shown that extracellular vesicles (EVs), particularly small extracellular vesicles (sEV), can disrupt the homeostasis of organs, promoting the development of a secondary tumor. However, the role of sEV in brain endothelium and their association with metastasis related to breast cancer is unknown. Thus, this study aimed to investigate sEV-triggered changes in the phosphorylation state of proteins on the surface of brain endothelial cells, as they form the first barrier in contact with circulating tumor cells and EVs, and once identified, to modulate its interactors and effects from this through different functional assays. We used the most aggressive breast cancer cell line, MDA-MB-231, and its brain-seeking variant, MDA-MB-231-br. From these cells, small and large extracellular vesicles were harvested to treat hCMEC/D3 cells, an immortalized cell line from the human brain microvasculature. Higher levels of phosphorylation of VEGFR1 and VEGFR2 were found in hCMEC/D3 cells treated with MDA-MB-231-br sEV. By computational analysis, the Valosin-Containing Protein (VCP) was predicted to be an important sEV cargo affecting the VEGFR2 intracellular trafficking, validated by western blotting analysis. Then, VCP was modulated by cell transfection or chemical inhibition in hCMEC/D3 cells and assessed in different functional in vitro assays evidencing a significant effect on the functionality of these cells. Thus, this study demonstrates that the VCP-containing sEVs induce modifications at different phosphor sites of VEGFR2 and effectively modulate the state of brain microvascular endothelial cells.
Collapse
Affiliation(s)
- Ramon Handerson Gomes Teles
- University of São Paulo, Department of Cell and Developmental Biology, Institute of Biomedical Sciences (ICB), São Paulo, 05508-000, Brazil
- University Hospital Würzburg, Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, 97080 Würzburg, Germany
- University Würzburg, Graduate School of Life Sciences, Campus Hubland Nord, 97074 Würzburg, Germany
| | - Nicolas Jones Villarinho
- University of São Paulo, Department of Cell and Developmental Biology, Institute of Biomedical Sciences (ICB), São Paulo, 05508-000, Brazil
| | - Ana Sayuri Yamagata
- University of São Paulo, Department of Cell and Developmental Biology, Institute of Biomedical Sciences (ICB), São Paulo, 05508-000, Brazil
| | - Camila Tamy Hiroki
- University of São Paulo, Department of Cell and Developmental Biology, Institute of Biomedical Sciences (ICB), São Paulo, 05508-000, Brazil
| | - Murilo Camargo de Oliveira
- University of São Paulo, Department of Cell and Developmental Biology, Institute of Biomedical Sciences (ICB), São Paulo, 05508-000, Brazil
| | - Gisela Ramos Terçarioli
- University of São Paulo, Department of Cell and Developmental Biology, Institute of Biomedical Sciences (ICB), São Paulo, 05508-000, Brazil
| | - Ruy Gastaldoni Jaeger
- University of São Paulo, Department of Cell and Developmental Biology, Institute of Biomedical Sciences (ICB), São Paulo, 05508-000, Brazil
| | - Patrick Meybohm
- University Hospital Würzburg, Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, 97080 Würzburg, Germany
| | - Malgorzata Burek
- University Hospital Würzburg, Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, 97080 Würzburg, Germany
| | - Vanessa Morais Freitas
- University of São Paulo, Department of Cell and Developmental Biology, Institute of Biomedical Sciences (ICB), São Paulo, 05508-000, Brazil
| |
Collapse
|
30
|
Ortiz-Melo MT, Campos JE, Sánchez-Guzmán E, Herrera-Aguirre ME, Castro-Muñozledo F. Regulation of corneal epithelial differentiation: miR-141-3p promotes the arrest of cell proliferation and enhances the expression of terminal phenotype. PLoS One 2024; 19:e0315296. [PMID: 39642122 PMCID: PMC11623785 DOI: 10.1371/journal.pone.0315296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 11/22/2024] [Indexed: 12/08/2024] Open
Abstract
In recent years, different laboratories have provided evidence on the role of miRNAs in regulation of corneal epithelial metabolism, permeability and wound healing, as well as their alteration after surgery and in some ocular pathologies. We searched the available databases reporting miRNA expression in the human eye, looking for miRNAs highly expressed in central cornea, which could be crucial for maintenance of the epithelial phenotype. Using the rabbit RCE1(5T5) cell line as a model of corneal epithelial differentiation, we describe the participation of miR-141-3p as a possible negative regulator of the proliferative/migratory phenotype in corneal epithelial cells. The expression of miR-141-3p followed a time course similar to the differentiation-linked KRT3 cytokeratin, being delayed 24-48 hours relative to PAX6 expression; such result suggested that miR-141-3p only regulates the expression of terminal phenotype. Inhibition of miR-141-3p led to increased cell proliferation and motility, and induced the expression of molecular makers characteristic of an Epithelial Mesenchymal Transition (EMT). Comparison between the transcriptional profile of cells in which miR-141-3p was knocked down, and the transcriptomes from proliferative non-differentiated and differentiated stratified epithelia suggest that miR-141-3p is involved in the expression of terminal differentiation mediating the arrest of cell proliferation and inhibiting the EMT in highly motile early differentiating cells.
Collapse
Affiliation(s)
- María Teresa Ortiz-Melo
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, México City, México
- Unidad de Investigación en Biomedicina (UBIMED), Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla, Estado de México, México
| | - Jorge E. Campos
- Unidad de Biotecnología y Prototipos (UBIPRO), Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla, Estado de México, México
| | - Erika Sánchez-Guzmán
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, México City, México
| | - María Esther Herrera-Aguirre
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, México City, México
| | - Federico Castro-Muñozledo
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, México City, México
| |
Collapse
|
31
|
Abou-Kandil A, Tröger-Görler S, Pschibul A, Krüger T, Rosin M, Schmidt F, Akbarimoghaddam P, Sarkar A, Cseresnyés Z, Shadkchan Y, Heinekamp T, Gräler MH, Barber AE, Walther G, Figge MT, Brakhage AA, Osherov N, Kniemeyer O. The proteomic response of Aspergillus fumigatus to amphotericin B (AmB) reveals the involvement of the RTA-like protein RtaA in AmB resistance. MICROLIFE 2024; 6:uqae024. [PMID: 39790482 PMCID: PMC11707875 DOI: 10.1093/femsml/uqae024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 11/19/2024] [Accepted: 12/04/2024] [Indexed: 01/12/2025]
Abstract
The polyene antimycotic amphotericin B (AmB) and its liposomal formulation AmBisome belong to the treatment options of invasive aspergillosis caused by Aspergillus fumigatus. Increasing resistance to AmB in clinical isolates of Aspergillus species is a growing concern, but mechanisms of AmB resistance remain unclear. In this study, we conducted a proteomic analysis of A. fumigatus exposed to sublethal concentrations of AmB and AmBisome. Both antifungals induced significantly increased levels of proteins involved in aromatic acid metabolism, transmembrane transport, and secondary metabolite biosynthesis. One of the most upregulated proteins was RtaA, a member of the RTA-like protein family, which includes conserved fungal membrane proteins with putative functions as transporters or translocases. Accordingly, we found that RtaA is mainly located in the cytoplasmic membrane and to a minor extent in vacuolar-like structures. Deletion of rtaA led to increased polyene sensitivity and its overexpression resulted in modest resistance. Interestingly, rtaA expression was only induced by exposure to the polyenes AmB and nystatin, but not by itraconazole and caspofungin. Orthologues of rtaA were also induced by AmB exposure in A. lentulus and A. terreus. Deletion of rtaA did not significantly change the ergosterol content of A. fumigatus, but decreased fluorescence intensity of the sterol-binding stain filipin. This suggests that RtaA is involved in sterol and lipid trafficking, possibly by transporting the target ergosterol to or from lipid droplets. These findings reveal the contribution of RtaA to polyene resistance in A. fumigatus, and thus provide a new putative target for antifungal drug development.
Collapse
Affiliation(s)
- Ammar Abou-Kandil
- Tel-Aviv University, Department of Clinical Microbiology and Immunology, Faculty of Medical and Health Sciences, Ramat-Aviv, 69987 Tel-Aviv, Israel
| | - Sophie Tröger-Görler
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute (HKI), Adolf-Reichwein-Str. 23, 07745 Jena, Germany
| | - Annica Pschibul
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute (HKI), Adolf-Reichwein-Str. 23, 07745 Jena, Germany
| | - Thomas Krüger
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute (HKI), Adolf-Reichwein-Str. 23, 07745 Jena, Germany
| | - Maira Rosin
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute (HKI), Adolf-Reichwein-Str. 23, 07745 Jena, Germany
- Institute of Microbiology, Friedrich Schiller University (FSU), 07743 Jena, Germany
| | - Franziska Schmidt
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute (HKI), Adolf-Reichwein-Str. 23, 07745 Jena, Germany
- Institute of Microbiology, Friedrich Schiller University (FSU), 07743 Jena, Germany
| | - Parastoo Akbarimoghaddam
- Research Group Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology (HKI), 07745 Jena, Germany
| | - Arjun Sarkar
- Research Group Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology (HKI), 07745 Jena, Germany
| | - Zoltán Cseresnyés
- Research Group Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology (HKI), 07745 Jena, Germany
| | - Yana Shadkchan
- Tel-Aviv University, Department of Clinical Microbiology and Immunology, Faculty of Medical and Health Sciences, Ramat-Aviv, 69987 Tel-Aviv, Israel
| | - Thorsten Heinekamp
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute (HKI), Adolf-Reichwein-Str. 23, 07745 Jena, Germany
| | - Markus H Gräler
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, 07747 Jena, Germany
- Center for Molecular Biomedicine (CMB), Jena University Hospital, 07745 Jena, Germany
- Center for Sepsis Control and Care (CSCC), Jena University Hospital, 07747 Jena, Germany
| | - Amelia E Barber
- Institute of Microbiology, Friedrich Schiller University (FSU), 07743 Jena, Germany
- Junior Research Group Fungal Informatics, Friedrich Schiller University, 07745 Jena, Germany
| | - Grit Walther
- National Reference Centre for Invasive Fungal Infections (NRZMyk), 07745 Jena, Germany
| | - Marc Thilo Figge
- Institute of Microbiology, Friedrich Schiller University (FSU), 07743 Jena, Germany
- Research Group Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology (HKI), 07745 Jena, Germany
| | - Axel A Brakhage
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute (HKI), Adolf-Reichwein-Str. 23, 07745 Jena, Germany
- Institute of Microbiology, Friedrich Schiller University (FSU), 07743 Jena, Germany
| | - Nir Osherov
- Tel-Aviv University, Department of Clinical Microbiology and Immunology, Faculty of Medical and Health Sciences, Ramat-Aviv, 69987 Tel-Aviv, Israel
| | - Olaf Kniemeyer
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute (HKI), Adolf-Reichwein-Str. 23, 07745 Jena, Germany
| |
Collapse
|
32
|
Czechowicz P, Gebert M, Bartoszewska S, Kalinowski L, Collawn JF, Bartoszewski R. The Yin and Yang of hsa-miR-1244 expression levels during activation of the UPR control cell fate. Cell Commun Signal 2024; 22:577. [PMID: 39623432 PMCID: PMC11610070 DOI: 10.1186/s12964-024-01967-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 11/28/2024] [Indexed: 12/06/2024] Open
Abstract
Regulation of endoplasmic reticulum (ER) homeostasis plays a critical role in maintaining cell survival. When ER stress occurs, a network of three pathways called the unfolded protein response (UPR) is activated to reestablish homeostasis. While it is known that there is cross-talk between these pathways, how this complex network is regulated is not entirely clear. Using human cancer and non-cancer cell lines, two different genome-wide approaches, and two different ER stress models, we searched for miRNAs that were decreased during the UPR and surprisingly found only one, miR-1244, that was found under all these conditions. We also verified that ER-stress related downregulation of miR-1244 expression occurred with 5 different ER stressors and was confirmed in another human cell line (HeLa S3). These analyses demonstrated that the outcome of this reduction during ER stress supported both IRE1 signaling and elevated BIP expression. Further analysis using inhibitors specific for IRE1, ATF6, and PERK also revealed that this miRNA is impacted by all three pathways of the UPR. This is the first example of a complex mechanism by which this miRNA serves as a regulatory check point for all 3 pathways that is switched off during UPR activation. In summary, the results indicate that ER stress reduction of miR-1244 expression contributes to the pro-survival arm of UPR.
Collapse
Affiliation(s)
- Paulina Czechowicz
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, F. Joliot-Curie 14a Street, Wroclaw, 50- 383, Poland
| | - Magdalena Gebert
- Department of Medical Laboratory Diagnostics-Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, Gdansk, Poland
| | - Sylwia Bartoszewska
- Department of Inorganic Chemistry, Medical University of Gdansk, Gdansk, Poland
| | - Leszek Kalinowski
- Department of Medical Laboratory Diagnostics-Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, Gdansk, Poland
- BioTechMed Center, Department of Mechanics of Materials and Structures, Gdansk University of Technology, Gdansk, Poland
| | - James F Collawn
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, USA
| | - Rafal Bartoszewski
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, F. Joliot-Curie 14a Street, Wroclaw, 50- 383, Poland.
| |
Collapse
|
33
|
Zhao N, Michelucci A, Pietrangelo L, Malik S, Groom L, Leigh J, O'Connor TN, Takano T, Kingsley PD, Palis J, Boncompagni S, Protasi F, Dirksen RT. An Orai1 gain-of-function tubular aggregate myopathy mouse model phenocopies key features of the human disease. EMBO J 2024; 43:5941-5971. [PMID: 39420094 PMCID: PMC11612304 DOI: 10.1038/s44318-024-00273-4] [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: 01/24/2024] [Revised: 09/19/2024] [Accepted: 09/27/2024] [Indexed: 10/19/2024] Open
Abstract
Tubular aggregate myopathy (TAM) is a heritable myopathy primarily characterized by progressive muscle weakness, elevated levels of creatine kinase (CK), hypocalcemia, exercise intolerance, and the presence of tubular aggregates (TAs). Here, we generated a knock-in mouse model based on a human gain-of-function mutation which results in a severe, early-onset form of TAM, by inducing a glycine-to-serine point mutation in the ORAI1 pore (Orai1G100S/+ or GS mice). By 8 months of age, GS mice exhibited significant muscle weakness, exercise intolerance, elevated CK levels, hypocalcemia, and robust TA presence. Unexpectedly, constitutive Ca2+ entry in mutant mice was observed in muscle only during early development and was abolished in adult skeletal muscle, partly due to reduced ORAI1 expression. Consistent with proteomic results, significant mitochondrial damage and dysfunction was observed in skeletal muscle of GS mice. Thus, GS mice represent a powerful model for investigation of the pathophysiological mechanisms that underlie key TAM symptoms, as well as those compensatory responses that limit the damaging effects of uncontrolled ORAI1-mediated Ca2+ influx.
Collapse
Affiliation(s)
- Nan Zhao
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Antonio Michelucci
- Department of Chemistry, Biology, and Biotechnology, University of Perugia, Perugia, Italy
| | - Laura Pietrangelo
- CAST, Center for Advanced Studies and Technology & DMSI, Department of Medicine and Aging Sciences, University Gabriele d'Annunzio of Chieti-Pescara, Chieti, Italy
| | - Sundeep Malik
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Linda Groom
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Jennifer Leigh
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Thomas N O'Connor
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Takahiro Takano
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Paul D Kingsley
- Department of Pediatrics, University of Rochester Medical Center, Rochester, NY, USA
| | - James Palis
- Department of Pediatrics, University of Rochester Medical Center, Rochester, NY, USA
| | - Simona Boncompagni
- CAST, Center for Advanced Studies and Technology & DNICS, Department of Neuroscience and Clinical Sciences, University Gabriele d'Annunzio of Chieti-Pescara, Chieti, Italy
| | - Feliciano Protasi
- CAST, Center for Advanced Studies and Technology & DMSI, Department of Medicine and Aging Sciences, University Gabriele d'Annunzio of Chieti-Pescara, Chieti, Italy
| | - Robert T Dirksen
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
34
|
Cobo‐Vuilleumier N, Rodríguez‐Fernandez S, López‐Noriega L, Lorenzo PI, Franco JM, Lachaud CC, Vazquez EM, Legido RA, Dorronsoro A, López‐Férnandez‐Sobrino R, Fernández‐Santos B, Serrano CE, Salas‐Lloret D, van Overbeek N, Ramos‐Rodriguez M, Mateo‐Rodríguez C, Hidalgo L, Marin‐Canas S, Nano R, Arroba AI, Caro AC, Vertegaal ACO, Martín‐Montalvo A, Martín F, Aguilar‐Diosdado M, Piemonti L, Pasquali L, Prieto RG, Sánchez MIG, Eizirik DL, Martínez‐Brocca MA, Vives‐Pi M, Gauthier BR. LRH-1/NR5A2 targets mitochondrial dynamics to reprogram type 1 diabetes macrophages and dendritic cells into an immune tolerance phenotype. Clin Transl Med 2024; 14:e70134. [PMID: 39702941 PMCID: PMC11659195 DOI: 10.1002/ctm2.70134] [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: 07/12/2024] [Revised: 11/01/2024] [Accepted: 12/05/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND The complex aetiology of type 1 diabetes (T1D), characterised by a detrimental cross-talk between the immune system and insulin-producing beta cells, has hindered the development of effective disease-modifying therapies. The discovery that the pharmacological activation of LRH-1/NR5A2 can reverse hyperglycaemia in mouse models of T1D by attenuating the autoimmune attack coupled to beta cell survival/regeneration prompted us to investigate whether immune tolerisation could be translated to individuals with T1D by LRH-1/NR5A2 activation and improve islet survival. METHODS Peripheral blood mononuclear cells (PBMCs) were isolated from individuals with and without T1D and derived into various immune cells, including macrophages and dendritic cells. Cell subpopulations were then treated or not with BL001, a pharmacological agonist of LRH-1/NR5A2, and processed for: (1) Cell surface marker profiling, (2) cytokine secretome profiling, (3) autologous T-cell proliferation, (4) RNAseq and (5) proteomic analysis. BL001-target gene expression levels were confirmed by quantitative PCR. Mitochondrial function was evaluated through the measurement of oxygen consumption rate using a Seahorse XF analyser. Co-cultures of PBMCs and iPSCs-derived islet organoids were performed to assess the impact of BL001 on beta cell viability. RESULTS LRH-1/NR5A2 activation induced a genetic and immunometabolic reprogramming of T1D immune cells, marked by reduced pro-inflammatory markers and cytokine secretion, along with enhanced mitohormesis in pro-inflammatory M1 macrophages and mitochondrial turnover in mature dendritic cells. These changes induced a shift from a pro-inflammatory to an anti-inflammatory/tolerogenic state, resulting in the inhibition of CD4+ and CD8+ T-cell proliferation. BL001 treatment also increased CD4+/CD25+/FoxP3+ regulatory T-cells and Th2 cells within PBMCs while decreasing CD8+ T-cell proliferation. Additionally, BL001 alleviated PBMC-induced apoptosis and maintained insulin expression in human iPSC-derived islet organoids. CONCLUSION These findings demonstrate the potential of LRH-1/NR5A2 activation to modulate immune responses and support beta cell viability in T1D, suggesting a new therapeutic approach. KEY POINTS LRH-1/NR5A2 activation in inflammatory cells of individuals with type 1 diabetes (T1D) reduces pro-inflammatory cell surface markers and cytokine release. LRH-1/NR5A2 promotes a mitohormesis-induced immuno-resistant phenotype to pro-inflammatory macrophages. Mature dendritic cells acquire a tolerogenic phenotype via LRH-1/NR5A2-stimulated mitochondria turnover. LRH-1/NR5A2 agonistic activation expands a CD4+/CD25+/FoxP3+ T-cell subpopulation. Pharmacological activation of LRH-1/NR5A2 improves the survival iPSC-islets-like organoids co-cultured with PBMCs from individuals with T1D.
Collapse
|
35
|
Breithofer J, Bulfon D, Fawzy N, Tischitz M, Zitta C, Hartig L, Grabner GF, Pirchheim A, Hackl H, Taschler U, Lass A, Tam-Amersdorfer C, Strobl H, Kratky D, Zimmermann R. Phospholipase A2 group IVD mediates the transacylation of glycerophospholipids and acylglycerols. J Lipid Res 2024; 65:100685. [PMID: 39490928 PMCID: PMC11621493 DOI: 10.1016/j.jlr.2024.100685] [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: 07/25/2024] [Revised: 10/11/2024] [Accepted: 10/16/2024] [Indexed: 11/05/2024] Open
Abstract
In mammalian cells, glycerolipids are mainly synthesized using acyl-CoA-dependent mechanisms. The acyl-CoA-independent transfer of fatty acids between lipids, designated as transacylation reaction, represents an additional mechanism for lipid remodeling and synthesis pathways. Here, we demonstrate that human and mouse phospholipase A2 group IVD (PLA2G4D) catalyzes transacylase reactions using both phospholipids and acylglycerols as substrates. In the presence of monoglycerol and diacylglycerol (MAG and DAG), purified PLA2G4D generates DAG and triacylglycerol, respectively. The enzyme also transfers fatty acids between phospholipids and from phospholipids to acylglycerols. Overexpression of PLA2G4D in COS7 cells enhances the incorporation of polyunsaturated fatty acids into triacylglycerol stores and induces the accumulation of lysophospholipids. In the presence of exogenously added MAG, the enzyme strongly increases cellular DAG formation, while MAG levels are decreased. PLA2G4D is not or poorly detectable in commonly used cell lines. It is expressed in keratinocytes, where it is strongly upregulated by proinflammatory cytokines. Pla2g4d-deficient mouse keratinocytes exhibit complex lipidomic changes in response to cytokine treatment, indicating that PLA2G4D is involved in the remodeling of the lipidome under inflammatory conditions. Transcriptomic analysis revealed that PLA2G4D modulates fundamental biological processes including cell proliferation, differentiation, and signaling. Together, our observations demonstrate that PLA2G4D has broad substrate specificity for fatty acid donor and acceptor lipids, allowing the acyl-CoA-independent synthesis of both phospholipids and acylglycerols. Loss-of-function studies indicate that PLA2G4D affects metabolic and signaling pathways in keratinocytes, which is associated with complex lipidomic and transcriptomic alterations.
Collapse
Affiliation(s)
| | - Dominik Bulfon
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Nermeen Fawzy
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Martin Tischitz
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Clara Zitta
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Lennart Hartig
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Gernot F Grabner
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Anita Pirchheim
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Hubert Hackl
- Institute of Bioinformatics, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Ulrike Taschler
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Achim Lass
- Institute of Molecular Biosciences, University of Graz, Graz, Austria; BioTechMed-Graz, Graz, Austria; Field of Excellence BioHealth, University of Graz, Graz, Austria
| | - Carmen Tam-Amersdorfer
- Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Immunology, Medical University of Graz, Graz, Austria
| | - Herbert Strobl
- Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Immunology, Medical University of Graz, Graz, Austria
| | - Dagmar Kratky
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria; BioTechMed-Graz, Graz, Austria
| | - Robert Zimmermann
- Institute of Molecular Biosciences, University of Graz, Graz, Austria; BioTechMed-Graz, Graz, Austria; Field of Excellence BioHealth, University of Graz, Graz, Austria.
| |
Collapse
|
36
|
Ahlström FH, Viisanen H, Karhinen L, Velagapudi V, Blomqvist KJ, Lilius TO, Rauhala PV, Kalso EA. Gene expression in the dorsal root ganglion and the cerebrospinal fluid metabolome in polyneuropathy and opioid tolerance in rats. IBRO Neurosci Rep 2024; 17:38-51. [PMID: 38933596 PMCID: PMC11201153 DOI: 10.1016/j.ibneur.2024.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 05/13/2024] [Accepted: 05/20/2024] [Indexed: 06/28/2024] Open
Abstract
First-line pharmacotherapy for peripheral neuropathic pain (NP) of diverse pathophysiology consists of antidepressants and gabapentinoids, but only a minority achieve sufficient analgesia with these drugs. Opioids are considered third-line analgesics in NP due to potential severe and unpredictable adverse effects in long-term use. Also, opioid tolerance and NP may have shared mechanisms, raising further concerns about opioid use in NP. We set out to further elucidate possible shared and separate mechanisms after chronic morphine treatment and oxaliplatin-induced and diabetic polyneuropathies, and to identify potential diagnostic markers and therapeutic targets. We analysed thermal nociceptive behaviour, the transcriptome of dorsal root ganglia (DRG) and the metabolome of cerebrospinal fluid (CSF) in these three conditions, in rats. Several genes were differentially expressed, most following oxaliplatin and least after chronic morphine treatment, compared with saline-treated rats. A few genes were differentially expressed in the DRGs in all three models (e.g. Csf3r and Fkbp5). Some, e.g. Alox15 and Slc12a5, were differentially expressed in both diabetic and oxaliplatin models. Other differentially expressed genes were associated with nociception, inflammation, and glial cells. The CSF metabolome was most significantly affected in the diabetic rats. Interestingly, we saw changes in nicotinamide metabolism, which has been associated with opioid addiction and withdrawal, in the CSF of morphine-tolerant rats. Our results offer new hypotheses for the pathophysiology and treatment of NP and opioid tolerance. In particular, the role of nicotinamide metabolism in opioid addiction deserves further study.
Collapse
Affiliation(s)
- Fredrik H.G. Ahlström
- Department of Pharmacology, Faculty of Medicine, Biomedicum 1, University of Helsinki, Haartmaninkatu 8, 00014, Finland
- Individualized Drug Therapy Research Programme, Faculty of Medicine, Biomedicum 1, University of Helsinki, Haartmaninkatu 8, 00014, Finland
| | - Hanna Viisanen
- Department of Pharmacology, Faculty of Medicine, Biomedicum 1, University of Helsinki, Haartmaninkatu 8, 00014, Finland
- Individualized Drug Therapy Research Programme, Faculty of Medicine, Biomedicum 1, University of Helsinki, Haartmaninkatu 8, 00014, Finland
| | - Leena Karhinen
- Department of Pharmacology, Faculty of Medicine, Biomedicum 1, University of Helsinki, Haartmaninkatu 8, 00014, Finland
| | - Vidya Velagapudi
- Metabolomics Unit, Institute for Molecular Medicine Finland FIMM, University of Helsinki, P.O. Box 20, FI-00014, Finland
| | - Kim J. Blomqvist
- Department of Pharmacology, Faculty of Medicine, Biomedicum 1, University of Helsinki, Haartmaninkatu 8, 00014, Finland
- Individualized Drug Therapy Research Programme, Faculty of Medicine, Biomedicum 1, University of Helsinki, Haartmaninkatu 8, 00014, Finland
| | - Tuomas O. Lilius
- Department of Pharmacology, Faculty of Medicine, Biomedicum 1, University of Helsinki, Haartmaninkatu 8, 00014, Finland
- Individualized Drug Therapy Research Programme, Faculty of Medicine, Biomedicum 1, University of Helsinki, Haartmaninkatu 8, 00014, Finland
- Department of Clinical Pharmacology, University of Helsinki and Helsinki University Hospital, Tukholmankatu 8C, 00014, Finland
- Department of Emergency Medicine and Services, University of Helsinki and HUS Helsinki University Hospital, Haartmaninkatu 4, Helsinki 00290, Finland
| | - Pekka V. Rauhala
- Department of Pharmacology, Faculty of Medicine, Biomedicum 1, University of Helsinki, Haartmaninkatu 8, 00014, Finland
- Individualized Drug Therapy Research Programme, Faculty of Medicine, Biomedicum 1, University of Helsinki, Haartmaninkatu 8, 00014, Finland
| | - Eija A. Kalso
- Department of Pharmacology, Faculty of Medicine, Biomedicum 1, University of Helsinki, Haartmaninkatu 8, 00014, Finland
- SleepWell Research Programme, Faculty of Medicine, , University of Helsinki, Haartmaninkatu 3, 00014, Finland
- Department of Anaesthesiology and Intensive Care Medicine, Helsinki University Hospital and University of Helsinki, HUS, Stenbäckinkatu 9, P.O. Box 440, 00029, Finland
| |
Collapse
|
37
|
Leonard MR, Jones DM, Read KA, Pokhrel S, Tuazon JA, Warren RT, Yount JS, Oestreich KJ. Aiolos promotes CXCR3 expression on Th1 cells via positive regulation of IFN-γ/STAT1 signaling. JCI Insight 2024; 10:e180287. [PMID: 39560988 PMCID: PMC11721307 DOI: 10.1172/jci.insight.180287] [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: 02/12/2024] [Accepted: 11/13/2024] [Indexed: 11/20/2024] Open
Abstract
CD4+ T helper 1 (Th1) cells coordinate adaptive immune responses to intracellular pathogens, including viruses. Key to this function is the ability of Th1 cells to migrate within secondary lymphoid tissues, as well as to sites of inflammation, which relies on signals received through the chemokine receptor CXCR3. CXCR3 expression is driven by the Th1 lineage-defining transcription factor T-bet and the cytokine-responsive STAT family members STAT1 and STAT4. Here, we identify the Ikaros zinc finger (IkZF) transcription factor Aiolos (Ikzf3) as an additional positive regulator of CXCR3 both in vitro and in vivo using a murine model of influenza virus infection. Mechanistically, we found that Aiolos-deficient CD4+ T cells exhibited decreased expression of key components of the IFN-γ/STAT1 signaling pathway, including JAK2 and STAT1. Consequently, Aiolos deficiency resulted in decreased levels of STAT1 tyrosine phosphorylation and reduced STAT1 enrichment at the Cxcr3 promoter. We further found that Aiolos and STAT1 formed a positive feedback loop via reciprocal regulation of each other downstream of IFN-γ signaling. Collectively, our study demonstrates that Aiolos promotes CXCR3 expression on Th1 cells by propagating the IFN-γ/STAT1 cytokine signaling pathway.
Collapse
Affiliation(s)
- Melissa R. Leonard
- Department of Microbial Infection and Immunity, The Ohio State University College of Medicine and Wexner Medical Center, Columbus, Ohio, USA
- Combined Anatomic Pathology Residency/PhD Program, The Ohio State University College of Veterinary Medicine, Columbus, Ohio, USA
| | - Devin M. Jones
- Department of Microbial Infection and Immunity, The Ohio State University College of Medicine and Wexner Medical Center, Columbus, Ohio, USA
- Biomedical Sciences Graduate Program and
| | - Kaitlin A. Read
- Department of Microbial Infection and Immunity, The Ohio State University College of Medicine and Wexner Medical Center, Columbus, Ohio, USA
- Biomedical Sciences Graduate Program and
| | - Srijana Pokhrel
- Department of Microbial Infection and Immunity, The Ohio State University College of Medicine and Wexner Medical Center, Columbus, Ohio, USA
| | - Jasmine A. Tuazon
- Department of Microbial Infection and Immunity, The Ohio State University College of Medicine and Wexner Medical Center, Columbus, Ohio, USA
- Biomedical Sciences Graduate Program and
- Medical Scientist Training Program, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Robert T. Warren
- Department of Microbial Infection and Immunity, The Ohio State University College of Medicine and Wexner Medical Center, Columbus, Ohio, USA
| | - Jacob S. Yount
- Department of Microbial Infection and Immunity, The Ohio State University College of Medicine and Wexner Medical Center, Columbus, Ohio, USA
- Infectious Diseases Institute, The Ohio State University College of Medicine and Wexner Medical Center, Columbus, Ohio, USA
| | - Kenneth J. Oestreich
- Department of Microbial Infection and Immunity, The Ohio State University College of Medicine and Wexner Medical Center, Columbus, Ohio, USA
- Infectious Diseases Institute, The Ohio State University College of Medicine and Wexner Medical Center, Columbus, Ohio, USA
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA
| |
Collapse
|
38
|
Annalora AJ, Coburn JL, Jozic A, Iversen PL, Marcus CB. Global transcriptome modulation by xenobiotics: the role of alternative splicing in adaptive responses to chemical exposures. Hum Genomics 2024; 18:127. [PMID: 39558396 PMCID: PMC11572221 DOI: 10.1186/s40246-024-00694-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 11/08/2024] [Indexed: 11/20/2024] Open
Abstract
BACKGROUND Xenobiotic exposures can extensively influence the expression and alternative splicing of drug-metabolizing enzymes, including cytochromes P450 (CYPs), though their transcriptome-wide impact on splicing remains underexplored. This study used a well-characterized splicing event in the Cyp2b2 gene to validate a sandwich-cultured primary rat hepatocyte model for studying global splicing in vitro. Using endpoint PCR, RNA sequencing, and bioinformatics tools (rSeqDiff, rMATs, IGV), we analyzed differential gene expression and splicing in CYP and nuclear receptor genes, as well as the entire transcriptome, to understand how xenobiotic exposures shape alternative splicing and activate xenosensors. METHODS Primary rat hepatocytes in sandwich culture were exposed to two methylenedioxybenzene (MDB) congeners and carbamazepine, with gene expression and splicing assessed. A 3D-clustergram integrating KEGG pathway analysis with differential gene expression provided distinct splicing landscapes for each xenobiotic, showing that splicing diversity does not always align with gene expression changes. RESULTS Endpoint PCR revealed a Cyp2b2v to wild-type Cyp2b2 splicing ratio near 1:1 (100%) under most conditions, while RNA-seq showed a stable baseline closer to 40%. C6-MDB reduced this ratio to ~ 50% by PCR and ~ 39% by RNA-seq, showing slight method-dependent variations yet consistent trends. In contrast, exon 6 skipping in Cyp1a1 occurred only with MDB exposure, implicating AHR activation. Xenobiotic treatments also induced alternative splicing in defensome and stress-responsive genes, including the phase II enzyme Gstm3, Albumin, Orm1, and Fxyd1, highlighting their roles in xenobiotic response modulation. Significant splicing changes in factors such as SRSF1, SRSF7, and METTL3 suggest a coordinated feedback loop involving epitranscriptomic modulation and cross-talk within SR protein networks, refining splice site selection, transcript stability, and cellular fate. CONCLUSIONS This study demonstrates how xenobiotic structural features influence gene expression and splicing, revealing splicing patterns that expand our understanding of transcriptome diversity and function. By identifying regulatory mechanisms, including AHR activation, epitranscriptomic modulation, and crosstalk within SR protein networks, that shape adaptive responses to xenobiotic stress, this work offers insights into the splicing and transcriptional networks that maintain cellular homeostasis. These findings provide predictive biomarkers for toxic exposures and highlight the potential of splicing profiles as diagnostic tools for assessing the health impacts of chemical exposure.
Collapse
Affiliation(s)
- Andrew J Annalora
- Department of Environmental and Molecular Toxicology, Oregon State University, 2750 SW Campus Way, Corvallis, OR, 97331, USA.
| | - Jacki L Coburn
- Department of Environmental and Molecular Toxicology, Oregon State University, 2750 SW Campus Way, Corvallis, OR, 97331, USA
| | - Antony Jozic
- Department of Environmental and Molecular Toxicology, Oregon State University, 2750 SW Campus Way, Corvallis, OR, 97331, USA
| | - Patrick L Iversen
- Department of Environmental and Molecular Toxicology, Oregon State University, 2750 SW Campus Way, Corvallis, OR, 97331, USA
| | - Craig B Marcus
- Department of Environmental and Molecular Toxicology, Oregon State University, 2750 SW Campus Way, Corvallis, OR, 97331, USA
| |
Collapse
|
39
|
Macadangdang BR, Wang Y, Woodward C, Revilla JI, Shaw BM, Sasaninia K, Makanani SK, Berruto C, Ahuja U, Miller JF. Targeted protein evolution in the gut microbiome by diversity-generating retroelements. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.15.621889. [PMID: 39605476 PMCID: PMC11601372 DOI: 10.1101/2024.11.15.621889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Diversity-generating retroelements (DGRs) accelerate evolution by rapidly diversifying variable proteins. The human gastrointestinal microbiota harbors the greatest density of DGRs known in nature, suggesting they play adaptive roles in this environment. We identified >1,100 unique DGRs among human-associated Bacteroides species and discovered a subset that diversify adhesive components of Type V pili and related proteins. We show that Bacteroides DGRs are horizontally transferred across species, that some are highly active while others are tightly controlled, and that they preferentially alter the functional characteristics of ligand-binding residues on adhesive organelles. Specific variable protein sequences are enriched when Bacteroides strains compete with other commensal bacteria in gnotobiotic mice. Analysis of >2,700 DGRs from diverse phyla in mother-infant pairs shows that Bacteroides DGRs are preferentially transferred to vaginally delivered infants where they actively diversify. Our observations provide a foundation for understanding the roles of stochastic, targeted genome plasticity in shaping host-associated microbial communities.
Collapse
Affiliation(s)
- Benjamin R. Macadangdang
- Division of Neonatology and Developmental Biology, Department of Pediatrics, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA, United States
- California NanoSystems Institute, Los Angeles, CA, United States
| | - Yanling Wang
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, California, United States
| | - Cora Woodward
- California NanoSystems Institute, Los Angeles, CA, United States
| | - Jessica I. Revilla
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Bennett M. Shaw
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Kayvan Sasaninia
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, California, United States
| | - Sara K. Makanani
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, California, United States
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, United States
| | - Chiara Berruto
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, California, United States
| | - Umesh Ahuja
- California NanoSystems Institute, Los Angeles, CA, United States
| | - Jeff F. Miller
- California NanoSystems Institute, Los Angeles, CA, United States
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, California, United States
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, United States
- Lead contact
| |
Collapse
|
40
|
Erinjeri AP, Wang X, Williams R, Chiozzi RZ, Thalassinos K, Labbadia J. HSF-1 promotes longevity through ubiquilin-1-dependent mitochondrial network remodelling. Nat Commun 2024; 15:9797. [PMID: 39532882 PMCID: PMC11557981 DOI: 10.1038/s41467-024-54136-x] [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: 10/23/2023] [Accepted: 11/04/2024] [Indexed: 11/16/2024] Open
Abstract
Increased activity of the heat shock factor, HSF-1, suppresses proteotoxicity and enhances longevity. However, the precise mechanisms by which HSF-1 promotes lifespan are unclear. Using an RNAi screen, we identify ubiquilin-1 (ubql-1) as an essential mediator of lifespan extension in worms overexpressing hsf-1. We find that hsf-1 overexpression leads to transcriptional downregulation of all components of the CDC-48-UFD-1-NPL-4 complex, which is central to both endoplasmic reticulum and mitochondria associated protein degradation, and that this is complemented by UBQL-1-dependent turnover of NPL-4.1. As a consequence, mitochondrial network dynamics are altered, leading to increased lifespan. Together, our data establish that HSF-1 mediates lifespan extension through mitochondrial network adaptations that occur in response to down-tuning of components associated with organellar protein degradation pathways.
Collapse
Affiliation(s)
- Annmary Paul Erinjeri
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, Division of Biosciences, University College London, London, UK
| | - Xunyan Wang
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, Division of Biosciences, University College London, London, UK
| | - Rhianna Williams
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, Division of Biosciences, University College London, London, UK
| | - Riccardo Zenezini Chiozzi
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, London, UK
- UCL Mass Spectrometry Science Technology Platform, Division of Biosciences, University College London, London, UK
| | - Konstantinos Thalassinos
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, London, UK
- UCL Mass Spectrometry Science Technology Platform, Division of Biosciences, University College London, London, UK
- Institute of Structural and Molecular Biology, Birkbeck College, University of London, London, UK
| | - Johnathan Labbadia
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, Division of Biosciences, University College London, London, UK.
| |
Collapse
|
41
|
Gélinas Bélanger J, Copley TR, Hoyos-Villegas V, O’Donoughue L. Integrated eQTL mapping approach reveals genomic regions regulating candidate genes of the E8-r3 locus in soybean. FRONTIERS IN PLANT SCIENCE 2024; 15:1463300. [PMID: 39600900 PMCID: PMC11589821 DOI: 10.3389/fpls.2024.1463300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 10/21/2024] [Indexed: 11/29/2024]
Abstract
Deciphering the gene regulatory networks of critical quantitative trait loci associated with early maturity provides information for breeders to unlock soybean's (Glycine max (L.) Merr.) northern potential and expand its cultivation range. The E8-r3 locus is a genomic region regulating the number of days to maturity under constant short-day photoperiodic conditions in two early-maturing soybean populations (QS15524F2:F3 and QS15544RIL) belonging to maturity groups MG00 and MG000. In this study, we developed a combinatorial expression quantitative trait loci mapping approach using three algorithms (ICIM, IM, and GCIM) to identify the regions that regulate three candidate genes of the E8-r3 locus (Glyma.04G167900/GmLHCA4a, Glyma.04G166300/GmPRR1a, and Glyma.04G159300/GmMDE04). Using this approach, a total of 2,218 trans (2,061 genes)/7 cis (7 genes) and 4,073 trans (2,842 genes)/3,083 cis (2,418 genes) interactions were mapped in the QS15524F2:F3 and QS15544RIL populations, respectively. From these interactions, we successfully identified two hotspots (F2_GM15:49,385,092-49,442,237 and F2_GM18:1,434,182-1,935,386) and three minor regions (RIL_GM04:17,227,512-20,251,662, RIL_GM04:31,408,946-31,525,671 and RIL_GM13:37,289,785-38,620,690) regulating the candidate genes of E8-r3 and several of their homologs. Based on co-expression network and single nucleotide variant analyses, we identified ALTERED PHLOEM DEVELOPMENT (Glyma.15G263700) and DOMAIN-CONTAINING PROTEIN 21 (Glyma.18G025600) as the best candidates for the F2_GM15:49,385,092-49,442,237 and F2_GM18:1,434,182-1,935,386 hotspots. These findings demonstrate that a few key regions are involved in the regulation of the E8-r3 candidates GmLHCA4a, GmPRR1a, and GmMDE04.
Collapse
Affiliation(s)
- Jérôme Gélinas Bélanger
- Soybean Breeding and Genetics Lab, Centre de recherche sur les grains (CÉROM) Inc., St-Mathieu-de-Beloeil, QC, Canada
- Department of Plant Science, McGill University, Montréal, QC, Canada
| | - Tanya Rose Copley
- Soybean Breeding and Genetics Lab, Centre de recherche sur les grains (CÉROM) Inc., St-Mathieu-de-Beloeil, QC, Canada
| | | | - Louise O’Donoughue
- Soybean Breeding and Genetics Lab, Centre de recherche sur les grains (CÉROM) Inc., St-Mathieu-de-Beloeil, QC, Canada
| |
Collapse
|
42
|
Calina TG, Perez E, Grafenhorst E, Benhamida J, Schallenberg S, Popescu A, Koch I, Janik T, Chen B, Ihlow J, Roessler S, Goeppert B, Sinn B, Bahra M, Calin GA, Taube ET, Pelzer U, Neumann CCM, Horst D, Knutsen E, Capper D, Dragomir MP. DNA methylation classifier to diagnose pancreatic ductal adenocarcinoma metastases from different anatomical sites. Clin Epigenetics 2024; 16:156. [PMID: 39523345 PMCID: PMC11550539 DOI: 10.1186/s13148-024-01768-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND We have recently constructed a DNA methylation classifier that can discriminate between pancreatic ductal adenocarcinoma (PAAD) liver metastasis and intrahepatic cholangiocarcinoma (iCCA) with high accuracy (PAAD-iCCA-Classifier). PAAD is one of the leading causes of cancer of unknown primary and diagnosis is based on exclusion of other malignancies. Therefore, our focus was to investigate whether the PAAD-iCCA-Classifier can be used to diagnose PAAD metastases from other sites. METHODS For this scope, the anomaly detection filter of the initial classifier was expanded by 8 additional mimicker carcinomas, amounting to a total of 10 carcinomas in the negative class. We validated the updated version of the classifier on a validation set, which consisted of a biological cohort (n = 3579) and a technical one (n = 15). We then assessed the performance of the classifier on a test set, which included a positive control cohort of 16 PAAD metastases from various sites and a cohort of 124 negative control samples consisting of 96 breast cancer metastases from 18 anatomical sites and 28 carcinoma metastases to the brain. RESULTS The updated PAAD-iCCA-Classifier achieved 98.21% accuracy on the biological validation samples, and on the technical validation ones it reached 100%. The classifier also correctly identified 15/16 (93.75%) metastases of the positive control as PAAD, and on the negative control, it correctly classified 122/124 samples (98.39%) for a 97.85% overall accuracy on the test set. We used this DNA methylation dataset to explore the organotropism of PAAD metastases and observed that PAAD liver metastases are distinct from PAAD peritoneal carcinomatosis and primary PAAD, and are characterized by specific copy number alterations and hypomethylation of enhancers involved in epithelial-mesenchymal-transition. CONCLUSIONS The updated PAAD-iCCA-Classifier (available at https://classifier.tgc-research.de/ ) can accurately classify PAAD samples from various metastatic sites and it can serve as a diagnostic aid.
Collapse
Affiliation(s)
- Teodor G Calina
- TGC Ventures UG, Berlin, Germany
- Faculty of Physics, Babeș-Bolyai University, Cluj-Napoca, Romania
| | - Eilís Perez
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin School of Integrative Oncology (BSIO), Charite - Universitätsmedizin Berlin (CVK), Berlin, Germany
| | - Elena Grafenhorst
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Jamal Benhamida
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Simon Schallenberg
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | | | - Ines Koch
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Tobias Janik
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - BaoQing Chen
- State Key Laboratory of Oncology in South China, Department of Radiation Oncology, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, People's Republic of China
- Guangdong Esophageal Cancer Research Institute, Guangzhou, Guangdong, People's Republic of China
| | - Jana Ihlow
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
| | - Stephanie Roessler
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Benjamin Goeppert
- Institute of Pathology and Neuropathology, Hospital RKH Kliniken Ludwigsburg, Ludwigsburg, Germany
- Institute of Tissue Medicine and Pathology (ITMP), University Bern, Bern, Switzerland
| | - Bruno Sinn
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Marcus Bahra
- Department of Surgical Oncology and Robotics, Krankenhaus Waldfriede, Lehrkrankenhaus der Charité, Berlin, Germany
| | - George A Calin
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The RNA Interference and Non-Coding RNA Center, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Eliane T Taube
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Uwe Pelzer
- Department of Hematology, Oncology and Tumor Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Christopher C M Neumann
- Department of Hematology, Oncology and Tumor Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - David Horst
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Erik Knutsen
- Department of Medical Biology, Faculty of Health Sciences, UiT The Artic University of Norway, Tromsø, Norway
| | - David Capper
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Mihnea P Dragomir
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany.
- German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Berlin Institute of Health, Berlin, Germany.
| |
Collapse
|
43
|
Dansu DK, Selcen I, Sauma S, Prentice E, Huang D, Li M, Moyon S, Casaccia P. Histone H4 acetylation differentially modulates proliferation in adult oligodendrocyte progenitors. J Cell Biol 2024; 223:e202308064. [PMID: 39133301 PMCID: PMC11318668 DOI: 10.1083/jcb.202308064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 06/18/2024] [Accepted: 07/29/2024] [Indexed: 08/13/2024] Open
Abstract
Adult oligodendrocyte progenitors (aOPCs) generate myelinating oligodendrocytes like neonatal progenitors (nOPCs), and they also display unique functional features. Here, using unbiased histone proteomics analysis and ChIP sequencing analysis of PDGFRα+ OPCs sorted from neonatal and adult Pdgfra-H2B-EGFP reporter mice, we identify the activating H4K8ac histone mark as enriched in the aOPCs. We detect increased occupancy of the H4K8ac activating mark at chromatin locations corresponding to genes related to the progenitor state (e.g., Hes5, Gpr17), metabolic processes (e.g., Txnip, Ptdgs), and myelin components (e.g., Cnp, Mog). aOPCs showed higher levels of transcripts related to lipid metabolism and myelin, and lower levels of transcripts related to cell cycle and proliferation compared with nOPCs. In addition, pharmacological inhibition of histone acetylation decreased the expression of the H4K8ac target genes in aOPCs and decreased their proliferation. Overall, this study identifies acetylation of the histone H4K8 as a regulator of the proliferative capacity of aOPCs.
Collapse
Affiliation(s)
- David K. Dansu
- Neuroscience Initiative, Advanced Science Research Center at the City University of New York, New York, NY, USA
- Graduate Program in Biochemistry, The Graduate Center of The City University of New York, New York, NY, USA
| | - Ipek Selcen
- Neuroscience Initiative, Advanced Science Research Center at the City University of New York, New York, NY, USA
- Graduate Program in Biochemistry, The Graduate Center of The City University of New York, New York, NY, USA
| | - Sami Sauma
- Neuroscience Initiative, Advanced Science Research Center at the City University of New York, New York, NY, USA
- Graduate Program in Biology, The Graduate Center of The City University of New York, New York, NY, USA
| | - Emily Prentice
- Neuroscience Initiative, Advanced Science Research Center at the City University of New York, New York, NY, USA
- Graduate Program in Biology, The Graduate Center of The City University of New York, New York, NY, USA
| | - Dennis Huang
- Neuroscience Initiative, Advanced Science Research Center at the City University of New York, New York, NY, USA
- Graduate Program in Biology, The Graduate Center of The City University of New York, New York, NY, USA
| | - Meng Li
- Norris Medical Library, University of Southern California, Los Angeles, CA, USA
| | - Sarah Moyon
- Neuroscience Initiative, Advanced Science Research Center at the City University of New York, New York, NY, USA
- Institute of NeuroPhysiopathology (INP) UMR7051, Aix-Marseille University, CNRS, Marseille, France
| | - Patrizia Casaccia
- Neuroscience Initiative, Advanced Science Research Center at the City University of New York, New York, NY, USA
- Graduate Program in Biochemistry, The Graduate Center of The City University of New York, New York, NY, USA
- Graduate Program in Biology, The Graduate Center of The City University of New York, New York, NY, USA
| |
Collapse
|
44
|
Trejo Vazquez JA, Towle R, Farnsworth DA, Sarafan M, Lockwood WW, Garnis C. Extracellular Vesicles from Lung Adenocarcinoma Cells Induce Activation of Different Cancer-Associated Fibroblast Subtypes. Biomedicines 2024; 12:2523. [PMID: 39595089 PMCID: PMC11591910 DOI: 10.3390/biomedicines12112523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 10/19/2024] [Accepted: 10/29/2024] [Indexed: 11/28/2024] Open
Abstract
Background: Lung cancer, including the major subtype lung adenocarcinoma (LUAD), is the leading cause of cancer deaths worldwide, largely due to metastasis. Improving survival rates requires new treatment strategies and a deeper understanding of the mechanisms that drive tumor progression within the tumor microenvironment (TME). This study investigated the impact of extracellular vesicles (EVs) derived from LUAD cells on lung fibroblasts. Methods: EVs were isolated from LUAD cell lines via ultracentrifugation and characterized using nanoparticle tracking analysis and Western blotting. Lung fibroblasts were treated with PBS, TGFβ, or EVs, and their activation was assessed through protein (Western blotting) and RNA analyses (RNA seq and RT-qPCR). Results: The results confirmed the TGFβ induced activation and showed that LUAD EVs could also activate fibroblasts, increasing cancer-associated fibroblast (CAF) markers. While EV-induced CAF activation displayed unique features, like an increase in proliferation-related genes, the EV and TGFβ treatments also shared some differentially expressed genes. The EV groups induced a higher expression of ECM remodeling and EMT-associated genes, but some of those genes were also upregulated in the TGFβ group. Mesenchymal genes POSTN and SPOCK1 were significantly upregulated in TGFβ- and EV-treated fibroblasts. Their secretion as proteins from the TGFβ- and EV-induced CAFs was not significant, confirmed through ELISA. Conclusions: These findings suggest that LUAD EVs play a role in CAF activation through both shared and distinct pathways compared to canonical TGFβ activation, potentially identifying novel gene expressions involved in CAF activation. Additionally, optimal protein secretion conditions of confirmed CAF-upregulated genes need to be established to determine their contribution to the TME.
Collapse
Affiliation(s)
- Jessica Angelina Trejo Vazquez
- Department of Integrative Oncology, British Columbia Cancer Research Center, Vancouver, BC V5Z1L3, Canada; (J.A.T.V.); (R.T.)
- Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC V5Z1L3, Canada
| | - Rebecca Towle
- Department of Integrative Oncology, British Columbia Cancer Research Center, Vancouver, BC V5Z1L3, Canada; (J.A.T.V.); (R.T.)
| | - Dylan Andrew Farnsworth
- Department of Integrative Oncology, British Columbia Cancer Research Center, Vancouver, BC V5Z1L3, Canada; (J.A.T.V.); (R.T.)
- Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC V5Z1L3, Canada
| | - Masih Sarafan
- Department of Integrative Oncology, British Columbia Cancer Research Center, Vancouver, BC V5Z1L3, Canada; (J.A.T.V.); (R.T.)
| | - William Wallace Lockwood
- Department of Integrative Oncology, British Columbia Cancer Research Center, Vancouver, BC V5Z1L3, Canada; (J.A.T.V.); (R.T.)
- Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC V5Z1L3, Canada
| | - Cathie Garnis
- Department of Integrative Oncology, British Columbia Cancer Research Center, Vancouver, BC V5Z1L3, Canada; (J.A.T.V.); (R.T.)
- Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC V5Z1L3, Canada
- Division of Otolaryngology, Department of Surgery, University of British Columbia, Vancouver, BC V5Z1M9, Canada
| |
Collapse
|
45
|
Feierman ER, Paranjapye A, Su S, Qiu Q, Wu H, Korb E. Histone variant H2BE controls activity-dependent gene expression and homeostatic scaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.01.620920. [PMID: 39553997 PMCID: PMC11565944 DOI: 10.1101/2024.11.01.620920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
A cell's ability to respond and adapt to environmental stimuli relies in part on transcriptional programs controlled by histone proteins. Histones affect transcription through numerous mechanisms including through replacement with variant forms that carry out specific functions. We recently identified the first widely expressed H2B histone variant, H2BE and found that it promotes transcription and is critical for neuronal function and long-term memory. However, how H2BE is regulated by extracellular stimuli and whether it controls activity-dependent transcription and cellular plasticity remain unknown. We used CUT&Tag and RNA-sequencing of primary neurons, single-nucleus sequencing of cortical tissue, and multielectrode array recordings to interrogate the expression of H2BE in response to stimuli and the role of H2BE in activity-dependent gene expression and plasticity. We find that unlike Further, we show that neurons lacking H2BE are unable to mount proper long-term activity-dependent transcriptional responses both in cultured neurons and in animal models. Lastly, we demonstrate that H2BE knockout neurons fail to undergo the electrophysiological changes associated with homeostatic plasticity in neurons after long-term stimulation. In summary, these data demonstrate that H2BE expression is inversely correlated to activity and necessary for long-term activity-dependent responses, revealing the first instance of a histone variant involved in the homeostatic plasticity response in neurons.
Collapse
|
46
|
Górska AM, Santos-García I, Eiriz I, Brüning T, Nyman T, Pahnke J. Evaluation of cerebrospinal fluid (CSF) and interstitial fluid (ISF) mouse proteomes for the validation and description of Alzheimer's disease biomarkers. J Neurosci Methods 2024; 411:110239. [PMID: 39102902 DOI: 10.1016/j.jneumeth.2024.110239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 07/22/2024] [Accepted: 07/26/2024] [Indexed: 08/07/2024]
Abstract
BACKGROUND Mass spectrometry (MS)-based cerebrospinal fluid (CSF) proteomics is an important method for discovering biomarkers of neurodegenerative diseases. CSF serves as a reservoir for interstitial fluid (ISF), and extensive communication between the two fluid compartments helps to remove waste products from the brain. NEW METHOD We performed proteomic analyses of both CSF and ISF fluid compartments using intracerebral microdialysis to validate and detect novel biomarkers of Alzheimer's disease (AD) in APPtg and C57Bl/6J control mice. RESULTS We identified up to 625 proteins in ISF and 4483 proteins in CSF samples. By comparing the biofluid profiles of APPtg and C57Bl/6J mice, we detected 37 and 108 significantly up- and downregulated candidates, respectively. In ISF, 7 highly regulated proteins, such as Gfap, Aldh1l1, Gstm1, and Txn, have already been implicated in AD progression, whereas in CSF, 9 out of 14 highly regulated proteins, such as Apba2, Syt12, Pgs1 and Vsnl1, have also been validated to be involved in AD pathogenesis. In addition, we also detected new interesting regulated proteins related to the control of synapses and neurotransmission (Kcna2, Cacng3, and Clcn6) whose roles as AD biomarkers should be further investigated. COMPARISON WITH EXISTING METHODS This newly established combined protocol provides better insight into the mutual communication between ISF and CSF as an analysis of tissue or CSF compartments alone. CONCLUSIONS The use of multiple fluid compartments, ISF and CSF, for the detection of their biological communication enables better detection of new promising AD biomarkers.
Collapse
Affiliation(s)
- Anna Maria Górska
- Translational Neurodegeneration Research and Neuropathology Lab, Department of Clinical Medicine (KlinMed), Medical Faculty, University of Oslo (UiO) and Section of Neuropathology Research, Department of Pathology, Clinics for Laboratory Medicine (KLM), Oslo University Hospital (OUS), Sognsvannsveien 20, Oslo NO-0372, Norway.
| | - Irene Santos-García
- Translational Neurodegeneration Research and Neuropathology Lab, Department of Clinical Medicine (KlinMed), Medical Faculty, University of Oslo (UiO) and Section of Neuropathology Research, Department of Pathology, Clinics for Laboratory Medicine (KLM), Oslo University Hospital (OUS), Sognsvannsveien 20, Oslo NO-0372, Norway.
| | - Ivan Eiriz
- Translational Neurodegeneration Research and Neuropathology Lab, Department of Clinical Medicine (KlinMed), Medical Faculty, University of Oslo (UiO) and Section of Neuropathology Research, Department of Pathology, Clinics for Laboratory Medicine (KLM), Oslo University Hospital (OUS), Sognsvannsveien 20, Oslo NO-0372, Norway.
| | - Thomas Brüning
- Translational Neurodegeneration Research and Neuropathology Lab, Department of Clinical Medicine (KlinMed), Medical Faculty, University of Oslo (UiO) and Section of Neuropathology Research, Department of Pathology, Clinics for Laboratory Medicine (KLM), Oslo University Hospital (OUS), Sognsvannsveien 20, Oslo NO-0372, Norway.
| | - Tuula Nyman
- Proteomics Core Facility, Department of Immunology, Oslo University Hospital (OUS) and University of Oslo (UiO), Faculty of Medicine, Sognsvannsveien 20, Oslo NO-0372, Norway.
| | - Jens Pahnke
- Translational Neurodegeneration Research and Neuropathology Lab, Department of Clinical Medicine (KlinMed), Medical Faculty, University of Oslo (UiO) and Section of Neuropathology Research, Department of Pathology, Clinics for Laboratory Medicine (KLM), Oslo University Hospital (OUS), Sognsvannsveien 20, Oslo NO-0372, Norway; Institute of Nutritional Medicine (INUM) and Lübeck Institute of Dermatology (LIED), University of Lübeck (UzL) and University Medical Center Schleswig-Holstein (UKSH), Ratzeburger Allee 160, Lübeck D-23538, Germany; Department of Pharmacology, Faculty of Medicine and Life Sciences, University of Latvia, Jelgavas iela 3, Rīga LV-1004, Latvia; School of Neurobiology, Biochemistry and Biophysics, The Georg S. Wise Faculty of Life Sciences, Tel Aviv University, Ramat Aviv IL-6997801, Israel.
| |
Collapse
|
47
|
Nowak TA, Fernandes C, Malfetano J, Lasek-Nesselquist E, Combs M, Strle K, Burke RL, Lin YP. Microbial genetic variation impacts host eco-immunological strategies and microparasite fitness in Lyme borreliae-reptile system. Ticks Tick Borne Dis 2024; 15:102410. [PMID: 39541748 PMCID: PMC11815320 DOI: 10.1016/j.ttbdis.2024.102410] [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: 07/08/2024] [Revised: 09/23/2024] [Accepted: 10/10/2024] [Indexed: 11/16/2024]
Abstract
Tolerance and resistance are two host eco-immunological strategies in response to microparasite invasion. In the strategy of "resistance", host responses are induced to decrease microparasite replication while the "tolerance" strategy allows hosts coexistence with microparasites by minimizing responses to avoid immune-mediated damage. The causative agent of Lyme disease is a group of genotypically diverse bacterial species, Borrelia burgdorferi sensu lato (Bb), which is transmitted by Ixodes ticks and persists in different reservoir animals. In North America, eastern fence lizards (Sceloporus undulatus) can be fed on by Ixodes ticks but are incompetent to one genotype of Bb (i.e., ospC type A). However, field-collected lizards showed evidence of previous infection by Bb strains with undefined genotypes. Supporting this evidence, we introduced three genotypically different Bb strains individually to eastern fence lizards and found a Bb genotype-dependent manner of infectivity. We compared liver transcriptomics and observed elevated immune responses triggered by a lizard-incompetent Bb strain (strain B31). We showed two lizard-competent strains with one having no immunomodulation (strain B379) but the other developing upregulated immune responses (strain 297). These results suggest that genetic variation in microparasites both induces different host strategies for dealing with infection and determines microparasite fitness in the hosts. These findings demonstrate that Bb and eastern fence lizards can serve as a model to investigate the mechanisms underlying eco-immunological strategies of tolerance vs. resistance during host-microparasite interaction.
Collapse
Affiliation(s)
- Tristan A Nowak
- Division of Infectious Diseases, Wadsworth Center, NYSDOH, Albany, NY, USA; Department of Biomedical Sciences, SUNY Albany, Albany, NY, USA
| | - Carly Fernandes
- Division of Infectious Diseases, Wadsworth Center, NYSDOH, Albany, NY, USA; Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA, USA
| | - Jill Malfetano
- Division of Infectious Diseases, Wadsworth Center, NYSDOH, Albany, NY, USA
| | - Erica Lasek-Nesselquist
- Department of Biomedical Sciences, SUNY Albany, Albany, NY, USA; Bioinformatics Core, Wadsworth Center, NYSDOH, Albany, NY, USA
| | - Matthew Combs
- National Wildlife Research Center, United State Department of Agriculture, Fort Collins, CO, USA
| | - Klemen Strle
- Department of Molecular Biology and Microbiology, School of Medicine, Tufts University, Boston, MA, USA
| | - Russell L Burke
- Department of Biology, Hofstra University, Hempstead, NY, USA
| | - Yi-Pin Lin
- Division of Infectious Diseases, Wadsworth Center, NYSDOH, Albany, NY, USA; Department of Biomedical Sciences, SUNY Albany, Albany, NY, USA; Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA, USA.
| |
Collapse
|
48
|
McGrail JP, Mondolfi AP, Ramírez JD, Vidal S, García‐Sastre A, Palacios G, Sanchez‐Seco MP, Guerra S. Comparative Analysis of 2022 Outbreak MPXV and Previous Clade II MPXV. J Med Virol 2024; 96:e70023. [PMID: 39466906 PMCID: PMC11600476 DOI: 10.1002/jmv.70023] [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: 07/24/2024] [Revised: 09/20/2024] [Accepted: 10/10/2024] [Indexed: 10/30/2024]
Abstract
The 2022-2024 outbreak of MPOX is an important worldwide public health issue that has triggered significant concerns in the scientific community. MPOX is caused by monkeypox virus (MPXV) belonging to the Poxviridae family. The study of MPXV presents a multifaceted challenge due to the diverse viral formThis study was supported by ISIDORe consortium and Agencia Estatal de Investigación.s produced by this pathogen. Notably the intracellular mature viruses (MVs) primarily contribute to localized lesions and host-to-host transmission, while the extracellular enveloped viruses (EVs) are associated with systemic infection. Clinically, MPOX manifests as a vesiculopustular rash that initially emerges on the face and trunk, subsequently spreading throughout the body, with heightened severity observed in immunocompromised individuals. Results obtained in this manuscript indicate that the 2022 outbreak MPXV has a significantly slower viral cycle compared with previous Clade II strains, with WRAIR 7-61 being more intermediate and USA 2003 producing highest viral titers. Additionally, proteomic and phospho-proteomic analysis displays differences in protein expression between these three strains. These findings highlight key differences between the current Lineage B.1 MPXV and previous strains. Further studies will be undertaken to demonstrate if these differences are important for the apparent increased human-to-human transmission mechanisms observed in the Clade IIb MPXV outbreak.
Collapse
Affiliation(s)
- Joseph Patrick McGrail
- Department of Preventive Medicine, Public Health and MicrobiologyUniversidad Autónoma de MadridMadridSpain
| | - Alberto Paniz Mondolfi
- Department of Pathology, Molecular, and Cell‐Based MedicineIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Juan David Ramírez
- Department of Pathology, Molecular, and Cell‐Based MedicineIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Centro de Investigaciones en Microbiología y Biotecnología‐UR (CIMBIUR), Facultad de Ciencias NaturalesUniversidad del RosarioBogotáColombia
| | - Santiago Vidal
- Department of MicrobiologyIcahn School of Medicine at Mount SinaiNew YorkUSA
- Global Health and Emerging Pathogens InstituteIcahn School of Medicine at Mount SinaiNew YorkUSA
| | - Adolfo García‐Sastre
- Department of MicrobiologyIcahn School of Medicine at Mount SinaiNew YorkUSA
- Global Health and Emerging Pathogens InstituteIcahn School of Medicine at Mount SinaiNew YorkUSA
- Department of Medicine, Division of Infectious DiseasesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- The Tisch Cancer InstituteIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of Pathology, Molecular and Cell‐Based MedicineIcahn School of Medicine at Mount SinaiNew YorkUSA
- The Icahn Genomics InstituteIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Gustavo Palacios
- Department of MicrobiologyIcahn School of Medicine at Mount SinaiNew YorkUSA
- Global Health and Emerging Pathogens InstituteIcahn School of Medicine at Mount SinaiNew YorkUSA
- Department of Medicine, Division of Infectious DiseasesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- The Tisch Cancer InstituteIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Mari Paz Sanchez‐Seco
- Centro Nacional de MicrobiologíaInstituto de Salud Carlos IIIMadridSpain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC)Instituto de Salud Carlos IIIMadridSpain
| | - Susana Guerra
- Department of Preventive Medicine, Public Health and MicrobiologyUniversidad Autónoma de MadridMadridSpain
- Department of MicrobiologyIcahn School of Medicine at Mount SinaiNew YorkUSA
- Global Health and Emerging Pathogens InstituteIcahn School of Medicine at Mount SinaiNew YorkUSA
| |
Collapse
|
49
|
Peterson PP, Choi JT, Fu C, Cowen LE, Sun S, Bahn YS, Heitman J. The Cryptococcus neoformans STRIPAK complex controls genome stability, sexual development, and virulence. PLoS Pathog 2024; 20:e1012735. [PMID: 39561188 PMCID: PMC11614259 DOI: 10.1371/journal.ppat.1012735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 12/03/2024] [Accepted: 11/07/2024] [Indexed: 11/21/2024] Open
Abstract
The eukaryotic serine/threonine protein phosphatase PP2A is a heterotrimeric enzyme composed of a scaffold A subunit, a regulatory B subunit, and a catalytic C subunit. Of the four known B subunits, the B"' subunit (known as striatin) interacts with the multi-protein striatin-interacting phosphatase and kinase (STRIPAK) complex. Orthologs of STRIPAK components were identified in Cryptococcus neoformans, namely PP2AA/Tpd3, PP2AC/Pph22, PP2AB/Far8, STRIP/Far11, SLMAP/Far9, and Mob3. Structural modeling, protein domain analysis, and detected protein-protein interactions suggest C. neoformans STRIPAK is assembled similarly to the human and fungal orthologs. Here, STRIPAK components Pph22, Far8, and Mob3 were functionally characterized. Whole-genome sequencing revealed that mutations in STRIPAK complex subunits lead to increased segmental and chromosomal aneuploidy, suggesting STRIPAK functions in maintaining genome stability. We demonstrate that PPH22 is a haploinsufficient gene: heterozygous PPH22/pph22Δ mutant diploid strains exhibit defects in hyphal growth and sporulation and have a significant fitness disadvantage when grown in competition against a wild-type diploid. Deletion mutants pph22Δ, far8Δ, and mob3Δ exhibit defects in mating and sexual differentiation, including impaired hyphae, basidia, and basidiospore production. Loss of either PPH22 or FAR8 in a haploid background leads to growth defects at 30°C, severely reduced growth at elevated temperature, abnormal cell morphology, and impaired virulence. Additionally, pph22Δ strains frequently accumulate suppressor mutations that result in overexpression of another putative PP2A catalytic subunit, PPG1. The pph22Δ and far8Δ mutants are also unable to grow in the presence of the calcineurin inhibitors cyclosporine A or FK506, and thus these mutations are synthetically lethal with loss of calcineurin activity. Conversely, mob3Δ mutants display increased thermotolerance, capsule production, and melanization, and are hypervirulent in a murine infection model. Taken together, these findings reveal that the C. neoformans STRIPAK complex plays an important role in genome stability, vegetative growth, sexual development, and virulence in this prominent human fungal pathogen.
Collapse
Affiliation(s)
- Patricia P. Peterson
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Jin-Tae Choi
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Ci Fu
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Leah E. Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Sheng Sun
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Yong-Sun Bahn
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Joseph Heitman
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, United States of America
| |
Collapse
|
50
|
Zhang T, Li Y, Pan L, Sha J, Bailey M, Faure-Kumar E, Williams CK, Wohlschlegel J, Magaki S, Niu C, Lee Y, Su YC, Li X, Vinters HV, Geschwind DH. Brain-wide alterations revealed by spatial transcriptomics and proteomics in COVID-19 infection. NATURE AGING 2024; 4:1598-1618. [PMID: 39543407 DOI: 10.1038/s43587-024-00730-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 09/25/2024] [Indexed: 11/17/2024]
Abstract
Understanding the pathophysiology of neurological symptoms observed after severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) infection is essential to optimizing outcomes and therapeutics. To date, small sample sizes and narrow molecular profiling have limited the generalizability of findings. In this study, we profiled multiple cortical and subcortical regions in postmortem brains of patients with coronavirus disease 2019 (COVID-19) and controls with matched pulmonary pathology (total n = 42) using spatial transcriptomics, bulk gene expression and proteomics. We observed a multi-regional antiviral response without direct active SARS-CoV2 infection. We identified dysregulation of mitochondrial and synaptic pathways in deep-layer excitatory neurons and upregulation of neuroinflammation in glia, consistent across both mRNA and protein. Remarkably, these alterations overlapped substantially with changes in age-related neurodegenerative diseases, including Parkinson's disease and Alzheimer's disease. Our work, combining multiple experimental and analytical methods, demonstrates the brain-wide impact of severe acute/subacute COVID-19, involving both cortical and subcortical regions, shedding light on potential therapeutic targets within pathways typically associated with pathological aging and neurodegeneration.
Collapse
Affiliation(s)
- Ting Zhang
- Section of Neuropathology, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yunfeng Li
- Translational Pathology Core Laboratory, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Liuliu Pan
- Technology Access Program, Bruker Spatial Technology, Seattle, WA, USA
- Duality Biologics, Shanghai, China
| | - Jihui Sha
- Proteome Research Center, University of California, Los Angeles, Los Angeles, CA, USA
| | - Michael Bailey
- Proof of Principle Team, Translational Science, Bruker Spatial Technology, Seattle, WA, USA
| | - Emmanuelle Faure-Kumar
- Center for Systems Biomedicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Christopher Kazu Williams
- Section of Neuropathology, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - James Wohlschlegel
- Proteome Research Center, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Shino Magaki
- Section of Neuropathology, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Chao Niu
- Technology Center for Genomics & Bioinformatics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yoojin Lee
- Technology Center for Genomics & Bioinformatics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yu-Chyuan Su
- Technology Center for Genomics & Bioinformatics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Xinmin Li
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Technology Center for Genomics & Bioinformatics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Harry V Vinters
- Section of Neuropathology, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
| | - Daniel H Geschwind
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
- Institute for Precision Health, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|