1
|
Davidson JW, Jain R, Kizzar T, Geoghegan G, Nesbitt DJ, Cavanagh A, Abe A, Nyame K, Hunger A, Chao X, James I, Walesewicz H, Baldwin DA, Wade G, Michorowska S, Verma R, Schueler K, Hinkovska-Galcheva V, Shishkova E, Ding WX, Coon JJ, Shayman JA, Abu-Remaileh M, Simcox JA. Hepatic lipid remodeling in cold exposure uncovers direct regulation of bis(monoacylglycero)phosphate lipids by phospholipase A2 group XV. Cell Metab 2025:S1550-4131(25)00253-0. [PMID: 40373767 DOI: 10.1016/j.cmet.2025.04.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/13/2025] [Accepted: 04/21/2025] [Indexed: 05/17/2025]
Abstract
Cold exposure is a selective environmental stress that elicits a rapid metabolic shift to maintain energy homeostasis. In response to cold exposure, the liver rewires the metabolic state, shifting from glucose to lipid catabolism. By probing the liver lipids in cold exposure, we observed that the lysosomal bis(monoacylglycero)phosphate (BMP) lipids were rapidly increased during cold exposure. BMP lipid changes occurred independently of lysosomal abundance but were dependent on the lysosomal transcriptional regulator transcription factor EB (TFEB). Knockdown of Tfeb in hepatocytes decreased BMP lipid levels and led to cold intolerance in mice. We assessed TFEB-binding sites of lysosomal genes and determined that the phospholipase a2 group XV (PLA2G15) regulates BMP lipid catabolism. Decreasing Pla2g15 levels in mice increased BMP lipids, ablated the cold-induced rise in BMP lipids, and improved cold tolerance. Mutation of the catalytic site of PLA2G15 ablated the BMP lipid breakdown. Together, our studies uncover TFEB regulation of BMP lipids through PLA2G15 catabolism.
Collapse
Affiliation(s)
- Jessica W Davidson
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Raghav Jain
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; Howard Hughes Medical Institute, Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Thomas Kizzar
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Gisela Geoghegan
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Daniel J Nesbitt
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Amy Cavanagh
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Akira Abe
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kwamina Nyame
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA; Department of Genetics, Stanford University, Stanford, CA 94305, USA; The Institute for Chemistry, Engineering & Medicine for Human Health (Sarafan ChEM-H), Stanford University, Stanford, CA 94305, USA
| | - Andrea Hunger
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Xiaojuan Chao
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Isabella James
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Helaina Walesewicz
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Dominique A Baldwin
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; Howard Hughes Medical Institute, Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Gina Wade
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Sylwia Michorowska
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; Department of Drug Chemistry, Pharmaceutical and Biomedical Analysis, Faculty of Pharmacy, Medical University of Warsaw, Warsaw, Poland
| | - Rakesh Verma
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kathryn Schueler
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | | | - Evgenia Shishkova
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Joshua J Coon
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; Morgridge Institute for Research, Madison, WI 53715, USA
| | - James A Shayman
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Monther Abu-Remaileh
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA; Department of Genetics, Stanford University, Stanford, CA 94305, USA; The Institute for Chemistry, Engineering & Medicine for Human Health (Sarafan ChEM-H), Stanford University, Stanford, CA 94305, USA
| | - Judith A Simcox
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; Howard Hughes Medical Institute, Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA.
| |
Collapse
|
2
|
Zawadzka M, Gil W, Konieczny A, Krakowska-Jura K, Kijewska M, Stefanowicz P. Affinity on Demand: A One-Pot Method for Synthesis and Sample Enrichment Using TentaGel-Functionalized Resins. ACS OMEGA 2025; 10:18135-18144. [PMID: 40352538 PMCID: PMC12060058 DOI: 10.1021/acsomega.5c02738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Revised: 04/08/2025] [Accepted: 04/11/2025] [Indexed: 05/14/2025]
Abstract
Protein glycation is a nonenzymatic reaction that results in the formation of early glycation products, commonly referred to as Amadori products, which play an important role in diabetes complications. In proteomic research, the analysis of glycated peptides is very challenging due to the low amount of analyte in a biological sample. One of the methods to overcome this is selective enrichment of the sample in the desired analyte. A method for synthesizing functionalized resins with phenylboronic acids has been developed, which allows for the incorporation of different linkers and a variable number of phenylboronic acid moieties, as well as the use of any solid support. Furthermore, the resins are prepared for use in sample enrichment following the completion of the synthesis process and demonstrate a high affinity for glycated peptides. The highest-affinity resin (4PhB-3Lys-TGR) was applied to artificially glycated albumin hydrolyzate and patient serum, and, in addition, it was used in conjunction with a biological sample (i.e., milk) for the selective enrichment of glycated peptides. The bioinformatics analysis provided results that confirmed the high coverage of protein sequences identified in the complex samples based on glycated peptides. This paper presents a novel, fast, simple, and cost-effective one-pot method for the synthesis of functionalized resins, along with a selective method for the enrichment of samples with glycated peptides. We believe that the presented approach is general and, with necessary modifications, could be applied for affinity-based isolation not only of Amadori products but also of carbonyl compounds, thiols, compounds with chelating properties, and others.
Collapse
Affiliation(s)
- Michalina Zawadzka
- Faculty
of Chemistry, University of Wrocław, Joliot-Curie 14, Wrocław 50-383, Poland
| | - Wojciech Gil
- Faculty
of Chemistry, University of Wrocław, Joliot-Curie 14, Wrocław 50-383, Poland
| | - Andrzej Konieczny
- Department
of Nephrology and Transplantation Medicine, Wrocław Medical University, Borowska 213, Wrocław 50-556, Poland
| | - Kornelia Krakowska-Jura
- Department
of Nephrology and Transplantation Medicine, Wrocław Medical University, Borowska 213, Wrocław 50-556, Poland
| | - Monika Kijewska
- Faculty
of Chemistry, University of Wrocław, Joliot-Curie 14, Wrocław 50-383, Poland
| | - Piotr Stefanowicz
- Faculty
of Chemistry, University of Wrocław, Joliot-Curie 14, Wrocław 50-383, Poland
| |
Collapse
|
3
|
Jadhav DB, Roy S. Circadian Proteomics Reassesses the Temporal Regulation of Metabolic Rhythms by Chlamydomonas Clock. PLANT, CELL & ENVIRONMENT 2025; 48:3512-3528. [PMID: 39777639 DOI: 10.1111/pce.15354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 12/17/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025]
Abstract
Circadian clocks execute temporal regulation of metabolism by modulating the timely expression of genes. Clock regulation of mRNA synthesis was envisioned as the primary driver of these daily rhythms. mRNA oscillations often do not concur with the downstream protein oscillations, revealing the importance to study protein oscillations. Chlamydomonas reinhardtii is a well-studied miniature plant model. We quantitatively probed the Chlamydomonas proteome for two subsequent circadian cycles using high throughput SWATH-DIA mass spectrometry. We quantified > 1000 proteins, half of which demonstrate circadian rhythms. Among these rhythmic proteins, > 90% peak around subjective midday or midnight. We uncovered key enzymes involved in Box C/D pathway, amino acid biosynthesis, fatty acid (FA) biosynthesis and peroxisomal β-oxidation of FAs are driven by the clock, which were undocumented from earlier transcriptomic studies. Proteins associated with key biological processes such as photosynthesis, redox, carbon fixation, glycolysis and TCA cycle show extreme temporal regulation. We conclude that circadian proteomics is required to complement transcriptomic studies to understand the complex clock regulation of organismal biology. We believe our study will not only refine and enrich the evaluation of temporal metabolic processes in C. reinhardtii but also provide a novel understanding of clock regulation across species.
Collapse
Affiliation(s)
| | - Sougata Roy
- Department of Biology, Trivedi School of Biosciences, Ashoka University, Sonipat, India
| |
Collapse
|
4
|
Hintze J, Fraumeni R, de Haan N, Miller RL, Saraswat M, Yang Z, Clausen H, Marth JD. Compositional and topological determinants of a physiological Ashwell-Morell receptor ligand. Proc Natl Acad Sci U S A 2025; 122:e2427129122. [PMID: 40208943 PMCID: PMC12012520 DOI: 10.1073/pnas.2427129122] [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: 12/30/2024] [Accepted: 03/06/2025] [Indexed: 04/12/2025] Open
Abstract
The hepatocyte Ashwell-Morell receptor (AMR) is the prototypical mammalian lectin and the first cell receptor isolated. This recycling endocytic receptor of the plasma membrane determines the concentrations of hundreds of circulating glycoproteins in the blood and plays important roles in host responses to and outcomes of infection. The compositional and topological determinants of a physiological AMR ligand have remained unclear with contradictory findings reported. Previous studies established that the AMR binds multivalent galactose on desialylated triantennary or higher-branched N-glycans with little to no binding to galactose on biantennary forms. However, the vast majority of circulating blood glycoproteins are modified by biantennary N-glycans, rendering them unlikely to be ligands bound and eliminated by the AMR. Separately, other studies reported that AMR ligands include sialylated N-glycans, and specifically α2-6, but not α2-3, sialic acid linkages. Herein, we investigated the composition and topology of AMR ligands using a known physiological AMR ligand, intestinal alkaline phosphatase (IAP). Recombinant active IAP was produced in glycoengineered cells with either biantennary or higher valency triantennary and tetra-antennary N-glycan structures, and further with and without either α2-6 or α2-3 sialic acid linkages. These closely homogenous IAP monomer glycoforms assemble as dimers with similar enzymatic activity and were compared in AMR binding and clearance assays. Our results indicate that the AMR does not significantly bind IAP when its N-glycans are predominantly sialylated with either α2-6 or α2-3 sialic acid linkages. Multivalent desialylated AMR ligands may, however, appear when IAP monomers dimerize, resulting in the close proximation of biantennary N-glycans.
Collapse
Affiliation(s)
- John Hintze
- Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA92037
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, CopenhagenN 2200, Denmark
| | - Robert Fraumeni
- Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA92037
| | - Noortje de Haan
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, CopenhagenN 2200, Denmark
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden2300, The Netherlands
| | - Rebecca L. Miller
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, CopenhagenN 2200, Denmark
| | - Mayank Saraswat
- Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA92037
| | - Zhang Yang
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, CopenhagenN 2200, Denmark
| | - Henrik Clausen
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, CopenhagenN 2200, Denmark
| | - Jamey D. Marth
- Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA92037
| |
Collapse
|
5
|
Jirouskova M, Harant K, Cejnar P, Ojha S, Korelova K, Sarnova L, Sticova E, Mayr CH, Schiller HB, Gregor M. Dynamics of compartment-specific proteomic landscapes of hepatotoxic and cholestatic models of liver fibrosis. eLife 2025; 13:RP98023. [PMID: 40197391 PMCID: PMC11978302 DOI: 10.7554/elife.98023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2025] Open
Abstract
Accumulation of extracellular matrix (ECM) in liver fibrosis is associated with changes in protein abundance and composition depending upon etiology of the underlying liver disease. Current efforts to unravel etiology-specific mechanisms and pharmacological targets rely on several models of experimental fibrosis. Here, we characterize and compare dynamics of hepatic proteome remodeling during fibrosis development and spontaneous healing in experimental mouse models of hepatotoxic (carbon tetrachloride [CCl4] intoxication) and cholestatic (3,5-diethoxycarbonyl-1,4-dihydrocollidine [DDC] feeding) injury. Using detergent-based tissue extraction and mass spectrometry, we identified compartment-specific changes in the liver proteome with detailed attention to ECM composition and changes in protein solubility. Our analysis revealed distinct time-resolved CCl4 and DDC signatures, with identified signaling pathways suggesting limited healing and a potential for carcinogenesis associated with cholestasis. Correlation of protein abundance profiles with fibrous deposits revealed extracellular chaperone clusterin with implicated role in fibrosis resolution. Dynamics of clusterin expression was validated in the context of human liver fibrosis. Atomic force microscopy of fibrotic livers complemented proteomics with profiles of disease-associated changes in local liver tissue mechanics. This study determined compartment-specific proteomic landscapes of liver fibrosis and delineated etiology-specific ECM components, providing thus a foundation for future antifibrotic therapies.
Collapse
Affiliation(s)
- Marketa Jirouskova
- Laboratory of Integrative Biology, Institute of Molecular Genetics of the Czech Academy of SciencesPragueCzech Republic
| | - Karel Harant
- Laboratory of Mass Spectrometry, BIOCEV, Faculty of Science, Charles UniversityPragueCzech Republic
| | - Pavel Cejnar
- Department of Mathematics, Informatics and Cybernetics, University of Chemistry and TechnologyPragueCzech Republic
| | - Srikant Ojha
- Laboratory of Integrative Biology, Institute of Molecular Genetics of the Czech Academy of SciencesPragueCzech Republic
- Department of Animal Physiology, Faculty of Science, Charles UniversityPragueCzech Republic
| | - Katerina Korelova
- Laboratory of Integrative Biology, Institute of Molecular Genetics of the Czech Academy of SciencesPragueCzech Republic
| | - Lenka Sarnova
- Laboratory of Integrative Biology, Institute of Molecular Genetics of the Czech Academy of SciencesPragueCzech Republic
| | - Eva Sticova
- Clinical and Transplant Pathology Centre, Institute for Clinical and Experimental MedicinePragueCzech Republic
- Department of Pathology, The Third Faculty of Medicine, Charles University and University Hospital Kralovske VinohradyPragueCzech Republic
| | - Christoph H Mayr
- Helmholtz Munich, Research Unit Precision Regenerative Medicine; Comprehensive Pneumology Center, Member of the German Center for Lung Research (DZL)MunichGermany
| | - Herbert B Schiller
- Helmholtz Munich, Research Unit Precision Regenerative Medicine; Comprehensive Pneumology Center, Member of the German Center for Lung Research (DZL)MunichGermany
- Institute of Experimental Pneumology, LMU University Hospital, Ludwig-Maximilians UniversityMunichGermany
| | - Martin Gregor
- Laboratory of Integrative Biology, Institute of Molecular Genetics of the Czech Academy of SciencesPragueCzech Republic
| |
Collapse
|
6
|
Ott F, Körner C, Krohn K, Fischer J, Damm G, Seehofer D, Berg T, Matz-Soja M. Impact of Hedgehog modulators on signaling pathways in primary murine and human hepatocytes in vitro: insights into liver metabolism. Arch Toxicol 2025; 99:1105-1116. [PMID: 39714734 PMCID: PMC11821798 DOI: 10.1007/s00204-024-03931-y] [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: 08/20/2024] [Accepted: 12/05/2024] [Indexed: 12/24/2024]
Abstract
The Hedgehog (Hh) signaling pathway is essential for maintaining homeostasis during embryogenesis and in adult tissues. In the liver, dysregulation of this pathway often leads to liver cancer development. Recent studies also suggest that disturbances in the Hh pathway can affect liver metabolism in healthy livers through interactions with other signaling pathways, such as the Wnt/β-catenin pathway. As a result, the Hh pathway has emerged as a promising target for therapeutic intervention. However, little is known about the effects of Hh modulators on healthy hepatocytes. In our study, we investigated the effects of the Hh agonists SAG (300 nM) and triamcinolone acetonide (40 µM), as well as the antagonists RU-SKI 43 (100 nM), cyclopamine (5 µM), budesonide (25 µM), GANT61 (0.5 µM), and vismodegib (1 µM) on healthy mouse and human primary hepatocytes in vitro. We employed toxicological, transcriptomic, proteomic, and functional assays, including proliferation and Seahorse assays. Our results show that these compounds significantly impact metabolic pathways such as lipid and glucose metabolism at both transcriptional and protein levels. Mechanistically, our data suggest the involvement of both canonical and non-canonical Hedgehog pathways, a phenomenon not previously described in hepatocytes. These findings highlight the diverse effects of these compounds on signaling and key metabolic functions in the liver, which emphasizes the need to investigate the hepatic Hh cascade and its metabolic control in depth. As the compounds regulate different aspects of metabolism, they need to be carefully studied in appropriate model systems for specific therapeutic use.
Collapse
Affiliation(s)
- Fritzi Ott
- Faculty of Medicine, Rudolf Schönheimer Institute of Biochemistry, Leipzig University, Leipzig, Germany
- Division of Hepatology, Clinic and Polyclinic for Oncology, Gastroenterology, Hepatology, and Pneumology, University Hospital Leipzig, Leipzig, Germany
| | - Christiane Körner
- Faculty of Medicine, Rudolf Schönheimer Institute of Biochemistry, Leipzig University, Leipzig, Germany
- Division of Hepatology, Clinic and Polyclinic for Oncology, Gastroenterology, Hepatology, and Pneumology, University Hospital Leipzig, Leipzig, Germany
| | - Knut Krohn
- Core Unit DNA-Technologies, Leipzig University, Leipzig, Germany
| | - Janett Fischer
- Division of Hepatology, Clinic and Polyclinic for Oncology, Gastroenterology, Hepatology, and Pneumology, University Hospital Leipzig, Leipzig, Germany
| | - Georg Damm
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, Leipzig, Germany
- Saxon Incubator for Clinical Translation (SIKT), Leipzig University, Leipzig, Germany
| | - Daniel Seehofer
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, Leipzig, Germany
| | - Thomas Berg
- Division of Hepatology, Clinic and Polyclinic for Oncology, Gastroenterology, Hepatology, and Pneumology, University Hospital Leipzig, Leipzig, Germany
| | - Madlen Matz-Soja
- Faculty of Medicine, Rudolf Schönheimer Institute of Biochemistry, Leipzig University, Leipzig, Germany.
- Division of Hepatology, Clinic and Polyclinic for Oncology, Gastroenterology, Hepatology, and Pneumology, University Hospital Leipzig, Leipzig, Germany.
| |
Collapse
|
7
|
Lin S, Dong L, De Nardo W, Leeming MG, Cheng Z, Williamson NA, Watt MJ, Montgomery MK. Long-lasting recombinant HEXA treatment improves hepatic steatosis and glycemic control in mild, but not severe, metabolic dysfunction-associated steatohepatitis. Am J Physiol Endocrinol Metab 2025; 328:E377-E394. [PMID: 39925140 DOI: 10.1152/ajpendo.00359.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/24/2024] [Accepted: 01/12/2025] [Indexed: 02/11/2025]
Abstract
The prevalence of metabolic dysfunction-associated steatohepatitis (MASH) is increasing at an alarming rate. To date, only one therapy has been provisionally approved for the treatment of MASH and liver fibrosis, and novel strategies are urgently needed. In addition, the frequent coexistence of MASH and type 2 diabetes has further intensified interest in devising comprehensive therapies to simultaneously tackle both diseases. We have recently shown that increasing hepatic and/or circulating levels of hexosaminidase A (HEXA), a lysosomal enzyme that remodels GM2 to GM3 gangliosides within lipid rafts, offers therapeutic benefits for metabolic dysfunction-associated steatotic liver disease (MASLD) and type 2 diabetes. Taking advantage of the MUP-uPA mouse model of MASH, including both wild-type (WT) mice with mild MASH and MUP-uPA mice with severe MASH and fibrosis, we show that biweekly treatment with a long-lasting HEXA-FC analog improves features of MASLD, including hepatic steatosis and hepatocyte ballooning, in mice with mild MASH, as well as glycemic control across both mouse models. Mechanistically, HEXA-FC enhances hepatic fatty acid oxidation and peripheral glucose disposal while not impacting endogenous glucose production. Together, these outcomes suggest that while HEXA-FC treatment may offer therapeutic benefits in mild MASH and insulin resistance, it is ineffective against severe MASH and liver fibrosis.NEW & NOTEWORTHY The prevalence of metabolic dysfunction-associated steatohepatitis (MASH) and type 2 diabetes is increasing. Here, we show that chronic FC-HEXA recombinant protein treatment reduces hepatic lipid accumulation and improves blood glucose control in mice with mild MASH and insulin resistance.
Collapse
Affiliation(s)
- Sihan Lin
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Li Dong
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Victoria, Australia
| | - William De Nardo
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Michael G Leeming
- Melbourne Mass Spectrometry and Proteomics Facility, Bio21 Molecular Science & Biotechnology Institute, The University of Melbourne, Melbourne, Victoria, Australia
| | - Zhili Cheng
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Nicholas A Williamson
- Melbourne Mass Spectrometry and Proteomics Facility, Bio21 Molecular Science & Biotechnology Institute, The University of Melbourne, Melbourne, Victoria, Australia
| | - Matthew J Watt
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Magdalene K Montgomery
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
8
|
Huschet LA, Kliem FP, Wienand P, Wunderlich CM, Ribeiro A, Bustos-Martínez I, Barco Á, Wunderlich FT, Lech M, Robles MS. FrozONE: quick cell nucleus enrichment for comprehensive proteomics analysis of frozen tissues. Life Sci Alliance 2025; 8:e202403130. [PMID: 39667914 PMCID: PMC11638322 DOI: 10.26508/lsa.202403130] [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: 11/08/2024] [Revised: 12/02/2024] [Accepted: 12/03/2024] [Indexed: 12/14/2024] Open
Abstract
Subcellular fractionation allows for the investigation of compartmentalized processes in individual cellular organelles. Nuclear enrichment methods commonly employ the use of density gradients combined with ultracentrifugation for freshly isolated tissues. Although it is broadly used in combination with proteomics, this approach poses several challenges when it comes to scalability and applicability for frozen material. To overcome these limitations, we developed FrozONE (Frozen Organ Nucleus Enrichment), a nucleus enrichment and proteomics workflow for frozen tissues. By extensively benchmarking our workflow against alternative methods, we showed that FrozONE is a faster, simpler, and more scalable alternative to conventional ultracentrifugation methods. FrozONE allowed for the study, profiling, and classification of nuclear proteomes in different tissues with complex cellular heterogeneity, ensuring optimal nucleus enrichment from different cell types and quantitative resolution for low abundant proteins. In addition to its performance in healthy mouse tissues, FrozONE proved to be very efficient for the characterization of liver nuclear proteome alterations in a pathological condition, diet-induced nonalcoholic steatohepatitis.
Collapse
Affiliation(s)
- Lukas A Huschet
- Institute of Medical Psychology and Biomedical Center (BMC), Faculty of Medicine, LMU, Munich, Germany
| | - Fabian P Kliem
- Institute of Medical Psychology and Biomedical Center (BMC), Faculty of Medicine, LMU, Munich, Germany
| | - Peter Wienand
- Max Planck Institute for Metabolism Research, Center for Molecular Medicine Cologne (CMMC) and Policlinic for Endocrinology, Diabetes, and Preventive Medicine (PEDP), University Hospital Cologne, Cologne, Germany
| | - Claudia M Wunderlich
- Max Planck Institute for Metabolism Research, Center for Molecular Medicine Cologne (CMMC) and Policlinic for Endocrinology, Diabetes, and Preventive Medicine (PEDP), University Hospital Cologne, Cologne, Germany
| | - Andrea Ribeiro
- LMU Klinikum, Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Isabel Bustos-Martínez
- Instituto de Neurociencias (Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas), Alicante, Spain
| | - Ángel Barco
- Instituto de Neurociencias (Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas), Alicante, Spain
| | - F Thomas Wunderlich
- Max Planck Institute for Metabolism Research, Center for Molecular Medicine Cologne (CMMC) and Policlinic for Endocrinology, Diabetes, and Preventive Medicine (PEDP), University Hospital Cologne, Cologne, Germany
| | - Maciej Lech
- LMU Klinikum, Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Maria S Robles
- Institute of Medical Psychology and Biomedical Center (BMC), Faculty of Medicine, LMU, Munich, Germany
| |
Collapse
|
9
|
Davidson JW, Jain R, Kizzar T, Geoghegan G, Nesbitt DJ, Cavanagh A, Abe A, Nyame K, Hunger A, Chao X, James I, Von Bank H, Baldwin DA, Wade G, Michorowska S, Verma R, Scheuler K, Hinkovska-Galcheva V, Shishkova E, Ding WX, Coon JJ, Shayman JA, Abu-Remaileh M, Simcox JA. Modulation of hepatic transcription factor EB activity during cold exposure uncovers direct regulation of bis(monoacylglycero)phosphate lipids by Pla2g15. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.11.03.565498. [PMID: 37986778 PMCID: PMC10659384 DOI: 10.1101/2023.11.03.565498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Cold exposure is a selective environmental stress that elicits a rapid metabolic shift to maintain energy homeostasis. In response to cold exposure, the liver rewires the metabolic state shifting from glucose to lipid catabolism. By probing the liver lipids in cold exposure, we observed that the lysosomal bis(monoacylglycero)phosphate (BMP) lipids were rapidly increased during cold exposure. BMP lipid changes occurred independently of lysosomal abundance but were dependent on the lysosomal transcriptional regulator transcription factor EB (TFEB). Knockdown of TFEB in hepatocytes decreased BMP lipid levels and led to cold intolerance in mice. We assessed TFEB binding sites of lysosomal genes and determined that the phospholipase Pla2g15 regulates BMP lipid catabolism. Knockdown of Pla2g15 in mice increased BMP lipid levels, ablated the cold-induced rise, and improved cold tolerance. Knockout of Pla2g15 in mice and hepatocytes led to increased BMP lipid levels, that were decreased with re-expression of Pla2g15. Mutation of the catalytic site of Pla2g15 ablated the BMP lipid breakdown. Together, our studies uncover TFEB regulation of BMP lipids through Pla2g15 catabolism.
Collapse
|
10
|
Zhang F, Wang Y, Zhu J, Wang J, Li Q, Feng J, Liu M, Li K, Tan J, Luo R, Yang H, Hou Y, He F, Qin J, Ding C, Yang W. Region- and Cell-type-Resolved Multiomic Atlas of the Heart. Mol Cell Proteomics 2025; 24:100922. [PMID: 39921206 DOI: 10.1016/j.mcpro.2025.100922] [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: 08/27/2023] [Revised: 01/02/2025] [Accepted: 02/02/2025] [Indexed: 02/10/2025] Open
Abstract
The heart is a vital muscular organ in vertebrate animals, responsible for maintaining blood circulation through rhythmic contraction. Although previous studies have investigated the heart proteome, the full hierarchical molecular network at cell-type- and region-resolved level, illustrating the specialized roles and crosstalk among different cell-types and regions, remains unclear. Here, we presented an atlas of cell-type-resolved proteome for mouse heart and region-resolved proteome for both mouse and human hearts. In-depth proteomic analysis identified 11,794 proteins across four cell-types and 11,995 proteins across six regions of the mouse heart. To further illustrate protein expression patterns in both physiological and pathological conditions, we conducted proteomic analysis on human heart samples from four regions with dilated cardiomyopathy (DCM). We quantified 8201 proteins in DCM tissue and 8316 proteins in adjacent unaffected myocardium tissue across the four human heart regions. Notably, we found that the retinoic acid synthesis pathway was significantly enriched in the DCM-affected left ventricle, and functional experiments demonstrated that all-trans retinoic acid efficiently rescued Ang II-induced myocardial hypertrophy and transverse aorta constriction-induced heart failure. In conclusion, our datasets uncovered the functional features of different cell-types and their synergistic cooperation centered by cell-type-specific transcription factors (TFs) in different regions, while these TF-TG (target gene) axes were significantly altered in DCM. Additionally, all-trans retinoic acid was demonstrated to be an efficient treatment for heart failure. This work presented a panoramic heart proteome map, offering a valuable resource for future cardiovascular research.
Collapse
Affiliation(s)
- Fan Zhang
- Department of Pediatric Orthopedics, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, China; State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Zhongshan Hospital, Fudan University, Shanghai, China; Department of Pathology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Yunzhi Wang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jiajun Zhu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jinxi Wang
- Laboratory of Molecular Cardiology, CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences (CAS), CAS, Shanghai, China
| | - Qiang Li
- Laboratory of Molecular Cardiology, CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences (CAS), CAS, Shanghai, China
| | - Jinwen Feng
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Mingwei Liu
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Kai Li
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jiliang Tan
- Laboratory of Molecular Cardiology, CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences (CAS), CAS, Shanghai, China
| | - Rongkui Luo
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Huangtian Yang
- Laboratory of Molecular Cardiology, CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences (CAS), CAS, Shanghai, China
| | - Yingyong Hou
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Fuchu He
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Zhongshan Hospital, Fudan University, Shanghai, China; State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Jun Qin
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Zhongshan Hospital, Fudan University, Shanghai, China; State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Chen Ding
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Zhongshan Hospital, Fudan University, Shanghai, China; Departments of Cancer Research Institute, Affiliated Cancer Hospital of Xinjiang Medical University, Xinjiang Key Laboratory of Translational Biomedical Engineering, Urumqi, China.
| | - Wenjun Yang
- Department of Pediatric Orthopedics, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, China.
| |
Collapse
|
11
|
Dong Q, Liu Z, Ma Y, Chen X, Wang X, Tang J, Ma K, Liang C, Wang M, Wu X, Liu Y, Zhou Y, Yang H, Gao M. Adipose tissue deficiency impairs transient lipid accumulation and delays liver regeneration following partial hepatectomy in male Seipin knockout mice. Clin Transl Med 2025; 15:e70238. [PMID: 39980067 PMCID: PMC11842221 DOI: 10.1002/ctm2.70238] [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: 10/08/2024] [Revised: 01/30/2025] [Accepted: 02/11/2025] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND Liver diseases pose significant health challenges, underscoring the importance of understanding liver regeneration mechanisms. Systemic adipose tissue is thought to be a primary source of lipids and energy during this process; however, empirical data on the effects of adipose tissue deficiency are limited. This study investigates the role of adipose tissue in liver regeneration, focusing on transient regeneration-associated steatosis (TRAS) and hepatocyte proliferation using a Seipin knockout mouse model that mimics severe human lipodystrophy. Additionally, the study explores therapeutic strategies through adipose tissue transplantation. METHODS Male Seipin knockout (Seipin-/-) and wild-type (WT) mice underwent 2/3 partial hepatectomy (PHx). Liver and plasma samples were collected at various time points post-surgery. Histological assessments, lipid accumulation analyses and measurements of hepatocyte proliferation markers were conducted. Additionally, normal adipose tissue was transplanted into Seipin-/- mice to evaluate the restoration of liver regeneration. RESULTS Seipin-/- mice exhibited significantly reduced liver regeneration rates and impaired TRAS, as evidenced by histological and lipid measurements. While WT mice demonstrated extensive hepatocyte proliferation at 48 and 72 h post-PHx, characterised by increased mitotic cells, elevated proliferating cell nuclear antigen and Ki67 expression, Seipin-/- mice showed delayed hepatocyte proliferation. Notably, adipose tissue transplantation into Seipin-/- mice restored TRAS and improved liver regeneration and hepatocyte proliferation. Conversely, liver-specific overexpression of Seipin in Seipin-/- mice did not affect TRAS or liver regeneration, indicating that the observed effects are primarily due to adipose tissue deficiency rather than hepatic Seipin itself. CONCLUSIONS Systemic adipose tissue is essential for TRAS and effective liver regeneration following PHx. Its deficiency impairs these processes, while adipose tissue transplantation can restore normal liver function. These findings underscore the critical role of adipose tissue in liver recovery and suggest potential therapeutic strategies for liver diseases associated with lipodystrophies. KEY POINTS Seipin-/- mice, which lack adipose tissue, exhibit significantly impaired TRAS and delayed liver regeneration following partial hepatectomy. Transplantation of normal adipose tissue into Seipin-/- mice restores TRAS and enhances liver regeneration, highlighting the essential role of adipose tissue in these processes. Liver-specific overexpression of Seipin has no effect on TRAS and liver regeneration in Seipin-/- mice.
Collapse
Affiliation(s)
- Qianqian Dong
- Department of Biochemistry and Molecular BiologyThe Key Laboratory of Neural and Vascular BiologyMinistry of Education, The Key Laboratory of Vascular Biology of Hebei Province, Cardiovascular Medical Science Center, Hebei Medical UniversityShijiazhuangHebeiChina
- Department of Clinical LaboratoryThe Second Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
| | - Ziwei Liu
- Department of Biochemistry and Molecular BiologyThe Key Laboratory of Neural and Vascular BiologyMinistry of Education, The Key Laboratory of Vascular Biology of Hebei Province, Cardiovascular Medical Science Center, Hebei Medical UniversityShijiazhuangHebeiChina
- Department of Clinical LaboratoryBethune International Peace HospitalShijiazhuangHebeiChina
| | - Yidan Ma
- Department of Biochemistry and Molecular BiologyThe Key Laboratory of Neural and Vascular BiologyMinistry of Education, The Key Laboratory of Vascular Biology of Hebei Province, Cardiovascular Medical Science Center, Hebei Medical UniversityShijiazhuangHebeiChina
| | - Xin Chen
- Department of Biochemistry and Molecular BiologyThe Key Laboratory of Neural and Vascular BiologyMinistry of Education, The Key Laboratory of Vascular Biology of Hebei Province, Cardiovascular Medical Science Center, Hebei Medical UniversityShijiazhuangHebeiChina
- Department of General SurgeryThe First Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
| | - Xiaowei Wang
- Department of Biochemistry and Molecular BiologyThe Key Laboratory of Neural and Vascular BiologyMinistry of Education, The Key Laboratory of Vascular Biology of Hebei Province, Cardiovascular Medical Science Center, Hebei Medical UniversityShijiazhuangHebeiChina
| | - Jinye Tang
- Department of Biochemistry and Molecular BiologyThe Key Laboratory of Neural and Vascular BiologyMinistry of Education, The Key Laboratory of Vascular Biology of Hebei Province, Cardiovascular Medical Science Center, Hebei Medical UniversityShijiazhuangHebeiChina
| | - Kexin Ma
- Department of Biochemistry and Molecular BiologyThe Key Laboratory of Neural and Vascular BiologyMinistry of Education, The Key Laboratory of Vascular Biology of Hebei Province, Cardiovascular Medical Science Center, Hebei Medical UniversityShijiazhuangHebeiChina
| | - Chenxi Liang
- Department of Biochemistry and Molecular BiologyThe Key Laboratory of Neural and Vascular BiologyMinistry of Education, The Key Laboratory of Vascular Biology of Hebei Province, Cardiovascular Medical Science Center, Hebei Medical UniversityShijiazhuangHebeiChina
| | - Mengyu Wang
- Department of CardiologyFirst Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Xiaoqin Wu
- Department of Integrative Biology and PharmacologyUniversity of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Yang Liu
- Department of Integrative Biology and PharmacologyUniversity of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Yaru Zhou
- Department of EndocrinologyThe Third Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
| | - Hongyuan Yang
- Department of Integrative Biology and PharmacologyUniversity of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Mingming Gao
- Department of Biochemistry and Molecular BiologyThe Key Laboratory of Neural and Vascular BiologyMinistry of Education, The Key Laboratory of Vascular Biology of Hebei Province, Cardiovascular Medical Science Center, Hebei Medical UniversityShijiazhuangHebeiChina
- Department of Integrative Biology and PharmacologyUniversity of Texas Health Science Center at HoustonHoustonTexasUSA
| |
Collapse
|
12
|
Guo T, Steen JA, Mann M. Mass-spectrometry-based proteomics: from single cells to clinical applications. Nature 2025; 638:901-911. [PMID: 40011722 DOI: 10.1038/s41586-025-08584-0] [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: 05/05/2024] [Accepted: 01/02/2025] [Indexed: 02/28/2025]
Abstract
Mass-spectrometry (MS)-based proteomics has evolved into a powerful tool for comprehensively analysing biological systems. Recent technological advances have markedly increased sensitivity, enabling single-cell proteomics and spatial profiling of tissues. Simultaneously, improvements in throughput and robustness are facilitating clinical applications. In this Review, we present the latest developments in proteomics technology, including novel sample-preparation methods, advanced instrumentation and innovative data-acquisition strategies. We explore how these advances drive progress in key areas such as protein-protein interactions, post-translational modifications and structural proteomics. Integrating artificial intelligence into the proteomics workflow accelerates data analysis and biological interpretation. We discuss the application of proteomics to single-cell analysis and spatial profiling, which can provide unprecedented insights into cellular heterogeneity and tissue architecture. Finally, we examine the transition of proteomics from basic research to clinical practice, including biomarker discovery in body fluids and the promise and challenges of implementing proteomics-based diagnostics. This Review provides a broad and high-level overview of the current state of proteomics and its potential to revolutionize our understanding of biology and transform medical practice.
Collapse
Affiliation(s)
- Tiannan Guo
- State Key Laboratory of Medical Proteomics, School of Medicine, Westlake University, Hangzhou, China.
- Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China.
- Research Center for Industries of the Future, School of Life Sciences, Westlake University, Hangzhou, China.
| | - Judith A Steen
- Department of Neurology, Harvard Medical School, Boston, MA, USA.
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA.
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany.
- NNF Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
13
|
Li G, Ma Y, Zhang S, Lin W, Yao X, Zhou Y, Zhao Y, Rao Q, Qu Y, Gao Y, Chen L, Zhang Y, Han F, Sun M, Zhao C. A mechanistic systems biology model of brain microvascular endothelial cell signaling reveals dynamic pathway-based therapeutic targets for brain ischemia. Redox Biol 2024; 78:103415. [PMID: 39520909 PMCID: PMC11584692 DOI: 10.1016/j.redox.2024.103415] [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: 10/21/2024] [Revised: 10/31/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
Ischemic stroke is a significant threat to human health. Currently, there is a lack of effective treatments for stroke, and progress in new neuron-centered drug target development is relatively slow. On the other hand, studies have demonstrated that brain microvascular endothelial cells (BMECs) are crucial components of the neurovascular unit and play pivotal roles in ischemic stroke progression. To better understand the complex multifaceted roles of BMECs in the regulation of ischemic stroke pathophysiology and facilitate BMEC-based drug target discovery, we utilized a transcriptomics-informed systems biology modeling approach and constructed a mechanism-based computational multipathway model to systematically investigate BMEC function and its modulatory potential. Extensive multilevel data regarding complex BMEC pathway signal transduction and biomarker expression under various pathophysiological conditions were used for quantitative model calibration and validation, and we generated dynamic BMEC phenotype maps in response to various stroke-related stimuli to identify potential determinants of BMEC fate under stress conditions. Through high-throughput model sensitivity analyses and virtual target perturbations in model-based single cells, our model predicted that targeting succinate could effectively reverse the detrimental cell phenotype of BMECs under oxygen and glucose deprivation/reoxygenation, a condition that mimics stroke pathogenesis, and we experimentally validated the utility of this new target in terms of regulating inflammatory factor production, free radical generation and tight junction protection in vitro and in vivo. Our work is the first that complementarily couples transcriptomic analysis with mechanistic systems-level pathway modeling in the study of BMEC function and endothelium-based therapeutic targets in ischemic stroke.
Collapse
Affiliation(s)
- Geli Li
- School of Pharmacy, Nanjing Medical University, 210000, Nanjing, China; Gusu School, Nanjing Medical University, 215000, Suzhou, China
| | - Yuchen Ma
- School of Pharmacy, Nanjing Medical University, 210000, Nanjing, China
| | - Sujie Zhang
- School of Pharmacy, Nanjing Medical University, 210000, Nanjing, China
| | - Wen Lin
- School of Pharmacy, Nanjing Medical University, 210000, Nanjing, China
| | - Xinyi Yao
- School of Pharmacy, Nanjing Medical University, 210000, Nanjing, China
| | - Yating Zhou
- The First Affiliated Hospital of Nanjing Medical University, 210000, Nanjing, China
| | - Yanyong Zhao
- School of Pharmacy, Nanjing Medical University, 210000, Nanjing, China
| | - Qi Rao
- School of Pharmacy, Nanjing Medical University, 210000, Nanjing, China
| | - Yuchen Qu
- School of Pharmacy, Nanjing Medical University, 210000, Nanjing, China
| | - Yuan Gao
- QSPMed Technologies, 210000, Nanjing, China
| | - Lianmin Chen
- The First Affiliated Hospital of Nanjing Medical University, 210000, Nanjing, China
| | - Yu Zhang
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, 21205, Baltimore, USA
| | - Feng Han
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine, Drug Target and Drug Discovery Center, School of Pharmacy, Nanjing Medical University, 210000, Nanjing, China.
| | - Meiling Sun
- School of Basic Medical Sciences, Nanjing Medical University, 210000, Nanjing, China.
| | - Chen Zhao
- School of Pharmacy, Nanjing Medical University, 210000, Nanjing, China; The First Affiliated Hospital of Nanjing Medical University, 210000, Nanjing, China.
| |
Collapse
|
14
|
Miao X, Alidadipour A, Saed V, Sayyadi F, Jadidi Y, Davoudi M, Amraee F, Jadidi N, Afrisham R. Hepatokines: unveiling the molecular and cellular mechanisms connecting hepatic tissue to insulin resistance and inflammation. Acta Diabetol 2024; 61:1339-1361. [PMID: 39031190 DOI: 10.1007/s00592-024-02335-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 07/06/2024] [Indexed: 07/22/2024]
Abstract
Insulin resistance arising from Non-Alcoholic Fatty Liver Disease (NAFLD) stands as a prevalent global ailment, a manifestation within societies stemming from individuals' suboptimal dietary habits and lifestyles. This form of insulin resistance emerges as a pivotal factor in the development of type 2 diabetes mellitus (T2DM). Emerging evidence underscores the significant role of hepatokines, as hepatic-secreted hormone-like entities, in the genesis of insulin resistance and eventual onset of type 2 diabetes. Hepatokines exert influence over extrahepatic metabolism regulation. Their principal functions encompass impacting adipocytes, pancreatic cells, muscles, and the brain, thereby playing a crucial role in shaping body metabolism through signaling to target tissues. This review explores the most important hepatokines, each with distinct influences. Our review shows that Fetuin-A promotes lipid-induced insulin resistance by acting as an endogenous ligand for Toll-like receptor 4 (TLR-4). FGF21 reduces inflammation in diabetes by blocking the nuclear translocation of nuclear factor-κB (NF-κB) in adipocytes and adipose tissue, while also improving glucose metabolism. ANGPTL6 enhances AMPK and insulin signaling in muscle, and suppresses gluconeogenesis. Follistatin can influence insulin resistance and inflammation by interacting with members of the TGF-β family. Adropin show a positive correlation with phosphoenolpyruvate carboxykinase 1 (PCK1), a key regulator of gluconeogenesis. This article delves into hepatokines' impact on NAFLD, inflammation, and T2DM, with a specific focus on insulin resistance. The aim is to comprehend the influence of these recently identified hormones on disease development and their underlying physiological and pathological mechanisms.
Collapse
Affiliation(s)
- Xiaolei Miao
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, 437100, China
| | - Arian Alidadipour
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Vian Saed
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Firooze Sayyadi
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Yasaman Jadidi
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Davoudi
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Amraee
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Nastaran Jadidi
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Afrisham
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
15
|
Zhao D, Huang Z, Li X, Wang H, Hou Q, Wang Y, Yan F, Yang W, Liu D, Yi S, Han C, Hao Y, Tang L. GDF2 and BMP10 coordinate liver cellular crosstalk to maintain liver health. eLife 2024; 13:RP95811. [PMID: 39453386 PMCID: PMC11509670 DOI: 10.7554/elife.95811] [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: 10/26/2024] Open
Abstract
The liver is the largest solid organ in the body and is primarily composed of hepatocytes (HCs), endothelial cells (ECs), Kupffer cells (KCs), and hepatic stellate cells (HSCs), which spatially interact and cooperate with each other to maintain liver homeostasis. However, the complexity and molecular mechanisms underlying the crosstalk between these different cell types remain to be revealed. Here, we generated mice with conditional deletion of Gdf2 (also known as Bmp9) and Bmp10 in different liver cell types and demonstrated that HSCs were the major source of GDF2 and BMP10 in the liver. Using transgenic ALK1 (receptor for GDF2 and BMP10) reporter mice, we found that ALK1 is expressed on KCs and ECs other than HCs and HSCs, and GDF2 and BMP10 secreted by HSCs promote the differentiation of KCs and ECs and maintain their identity. Pdgfb expression was significantly upregulated in KCs and ECs after Gdf2 and Bmp10 deletion, ultimately leading to HSCs activation and liver fibrosis. ECs express several angiocrine factors, such as BMP2, BMP6, Wnt2, and Rspo3, to regulate HC iron metabolism and metabolic zonation. We found that these angiocrine factors were significantly decreased in ECs from Gdf2/Bmp10HSC-KO mice, which further resulted in liver iron overload and disruption of HC zonation. In summary, we demonstrated that HSCs play a central role in mediating liver cell-cell crosstalk via the production of GDF2 and BMP10, highlighting the important role of intercellular interaction in organ development and homeostasis.
Collapse
Affiliation(s)
- Dianyuan Zhao
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of LifeomicsBeijingChina
| | - Ziwei Huang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of LifeomicsBeijingChina
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical UniversityHefeiChina
| | - Xiaoyu Li
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of LifeomicsBeijingChina
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical UniversityHefeiChina
| | - Huan Wang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of LifeomicsBeijingChina
| | - Qingwei Hou
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of LifeomicsBeijingChina
- School of Basic Medicine, Qingdao UniversityQingdaoChina
| | - Yuyao Wang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of LifeomicsBeijingChina
- School of Basic Medicine, Qingdao UniversityQingdaoChina
| | - Fang Yan
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of LifeomicsBeijingChina
| | - Wenting Yang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of LifeomicsBeijingChina
| | - Di Liu
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of LifeomicsBeijingChina
| | - Shaoqiong Yi
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of LifeomicsBeijingChina
| | - Chunguang Han
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of LifeomicsBeijingChina
| | - Yanan Hao
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of LifeomicsBeijingChina
| | - Li Tang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of LifeomicsBeijingChina
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical UniversityHefeiChina
- School of Basic Medicine, Qingdao UniversityQingdaoChina
| |
Collapse
|
16
|
Lin C, Jans A, Wolters JC, Mohamed MR, Van der Vorst EPC, Trautwein C, Bartneck M. Targeting Ligand Independent Tropism of siRNA-LNP by Small Molecules for Directed Therapy of Liver or Myeloid Immune Cells. Adv Healthc Mater 2024; 13:e2202670. [PMID: 36617516 DOI: 10.1002/adhm.202202670] [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: 10/16/2022] [Revised: 12/15/2022] [Indexed: 01/10/2023]
Abstract
Hepatic clearance of lipid nanoparticles (LNP) with encapsulated nucleic acids restricts their therapeutic applicability. Therefore, tools for regulating hepatic clearance are of high interest for nucleic acid delivery. To this end, this work employs wild-type (WT) and low-density lipoprotein receptor (Ldlr)-/- mice pretreated with either a leukotriene B4 receptor inhibitor (BLT1i) or a high-density lipoprotein receptor inhibitor (HDLRi) prior to the injection of siRNA-LNP. This work is able to demonstrate significantly increased hepatic uptake of siRNA-LNP by the BLT1i in Ldlr-/- mice by in vivo imaging and discover an induction of specific uptake-related proteins. Irrespective of the inhibitors and Ldlr deficiency, the siRNA-LNP induced RNA-binding and transport-related proteins in liver, including haptoglobin (HP) that is also identified as most upregulated serum protein. This work observes a downregulation of proteins functioning in hepatic detoxification and of serum opsonins. Most strikingly, the HDLRi reduces hepatic uptake and increases siRNA accumulation in spleen and myeloid immune cells of blood and liver. RNA sequencing demonstrates leukocyte recruitment by the siRNA-LNP and the HDLRi through induction of chemokine ligands in liver tissue. The data provide insights into key mechanisms of siRNA-LNP biodistribution and indicate that the HDLRi has potential for extrahepatic and leukocyte targeting.
Collapse
Affiliation(s)
- Cheng Lin
- Department of Internal Medicine III, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074, Aachen, Germany
- Department of Rheumatology and Shanghai Institute of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Alexander Jans
- Department of Internal Medicine III, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Justina Clarinda Wolters
- Department of Pediatrics, Section Systems Medicine of Metabolism and Signaling, University of Groningen, University Medical Center Groningen, Groningen, 9713 AV, The Netherlands
| | - Mohamed Ramadan Mohamed
- Department of Internal Medicine III, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Emiel P C Van der Vorst
- Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, 52074, Aachen, Germany
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074, Aachen, Germany
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich, 80336, Munich, Germany
| | - Christian Trautwein
- Department of Internal Medicine III, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Matthias Bartneck
- Department of Internal Medicine III, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074, Aachen, Germany
| |
Collapse
|
17
|
Tohme C, Haykal T, Yang R, Austin TJ, Loughran P, Geller DA, Simmons RL, Tohme S, Yazdani HO. ZLN005, a PGC-1α Activator, Protects the Liver against Ischemia-Reperfusion Injury and the Progression of Hepatic Metastases. Cells 2024; 13:1448. [PMID: 39273020 PMCID: PMC11393917 DOI: 10.3390/cells13171448] [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: 07/30/2024] [Revised: 08/22/2024] [Accepted: 08/25/2024] [Indexed: 09/15/2024] Open
Abstract
BACKGROUND Exercise can promote sustainable protection against cold and warm liver ischemia-reperfusion injury (IRI) and tumor metastases. We have shown that this protection is by the induction of hepatic mitochondrial biogenesis pathway. In this study, we hypothesize that ZLN005, a PGC-1α activator, can be utilized as an alternative therapeutic strategy. METHODS Eight-week-old mice were pretreated with ZLN005 and subjected to liver warm IRI. To establish a liver metastatic model, MC38 cancer cells (1 × 106) were injected into the spleen, followed by splenectomy and liver IRI. RESULTS ZLN005-pretreated mice showed a significant decrease in IRI-induced tissue injury as measured by serum ALT/AST/LDH levels and tissue necrosis. ZLN005 pretreatment decreased ROS generation and cell apoptosis at the site of injury, with a significant decrease in serum pro-inflammatory cytokines, innate immune cells infiltration, and intrahepatic neutrophil extracellular trap (NET) formation. Moreover, mitochondrial mass was significantly upregulated in hepatocytes and maintained after IRI. This was confirmed in murine and human hepatocytes treated with ZLN005 in vitro under normoxic and hypoxic conditions. Additionally, ZLN005 preconditioning significantly attenuated tumor burden and increased the percentage of intratumoral cytotoxic T cells. CONCLUSIONS Our study highlights the effective protection of ZLN005 pretreatment as a therapeutic alternative in terms of acute liver injury and tumor metastases.
Collapse
Affiliation(s)
- Celine Tohme
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (C.T.); (T.H.); (R.Y.); (T.J.A.); (P.L.); (D.A.G.); (R.L.S.)
| | - Tony Haykal
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (C.T.); (T.H.); (R.Y.); (T.J.A.); (P.L.); (D.A.G.); (R.L.S.)
| | - Ruiqi Yang
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (C.T.); (T.H.); (R.Y.); (T.J.A.); (P.L.); (D.A.G.); (R.L.S.)
- School of Medicine, Tsinghua University, Beijing 100084, China
| | - Taylor J. Austin
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (C.T.); (T.H.); (R.Y.); (T.J.A.); (P.L.); (D.A.G.); (R.L.S.)
| | - Patricia Loughran
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (C.T.); (T.H.); (R.Y.); (T.J.A.); (P.L.); (D.A.G.); (R.L.S.)
- Center for Biologic Imaging, Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - David A. Geller
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (C.T.); (T.H.); (R.Y.); (T.J.A.); (P.L.); (D.A.G.); (R.L.S.)
| | - Richard L. Simmons
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (C.T.); (T.H.); (R.Y.); (T.J.A.); (P.L.); (D.A.G.); (R.L.S.)
| | - Samer Tohme
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (C.T.); (T.H.); (R.Y.); (T.J.A.); (P.L.); (D.A.G.); (R.L.S.)
| | - Hamza O. Yazdani
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (C.T.); (T.H.); (R.Y.); (T.J.A.); (P.L.); (D.A.G.); (R.L.S.)
| |
Collapse
|
18
|
Venetos NM, Stomberski CT, Qian Z, Premont RT, Stamler JS. Activation of hepatic acetyl-CoA carboxylase by S-nitrosylation in response to diet. J Lipid Res 2024; 65:100542. [PMID: 38641009 PMCID: PMC11126798 DOI: 10.1016/j.jlr.2024.100542] [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: 01/12/2024] [Revised: 03/25/2024] [Accepted: 04/08/2024] [Indexed: 04/21/2024] Open
Abstract
Nitric oxide (NO), produced primarily by nitric oxide synthase enzymes, is known to influence energy metabolism by stimulating fat uptake and oxidation. The effects of NO on de novo lipogenesis (DNL), however, are less clear. Here we demonstrate that hepatic expression of endothelial nitric oxide synthase is reduced following prolonged administration of a hypercaloric high-fat diet. This results in marked reduction in the amount of S-nitrosylation of liver proteins including notably acetyl-CoA carboxylase (ACC), the rate-limiting enzyme in DNL. We further show that ACC S-nitrosylation markedly increases enzymatic activity. Diminished endothelial nitric oxide synthase expression and ACC S-nitrosylation may thus represent a physiological adaptation to caloric excess by constraining lipogenesis. Our findings demonstrate that S-nitrosylation of liver proteins is subject to dietary control and suggest that DNL is coupled to dietary and metabolic conditions through ACC S-nitrosylation.
Collapse
Affiliation(s)
- Nicholas M Venetos
- Department of Medicine, Institute for Transformative Molecular Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Colin T Stomberski
- Department of Medicine, Institute for Transformative Molecular Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Zhaoxia Qian
- Department of Medicine, Institute for Transformative Molecular Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Richard T Premont
- Department of Medicine, Institute for Transformative Molecular Medicine, Case Western Reserve University, Cleveland, OH, USA; Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Jonathan S Stamler
- Department of Medicine, Institute for Transformative Molecular Medicine, Case Western Reserve University, Cleveland, OH, USA; Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA.
| |
Collapse
|
19
|
Kim G, Chen Z, Li J, Luo J, Castro-Martinez F, Wisniewski J, Cui K, Wang Y, Sun J, Ren X, Crawford SE, Becerra SP, Zhu J, Liu T, Wang S, Zhao K, Wu C. Gut-liver axis calibrates intestinal stem cell fitness. Cell 2024; 187:914-930.e20. [PMID: 38280375 PMCID: PMC10923069 DOI: 10.1016/j.cell.2024.01.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 10/25/2023] [Accepted: 01/02/2024] [Indexed: 01/29/2024]
Abstract
The gut and liver are recognized to mutually communicate through the biliary tract, portal vein, and systemic circulation. However, it remains unclear how this gut-liver axis regulates intestinal physiology. Through hepatectomy and transcriptomic and proteomic profiling, we identified pigment epithelium-derived factor (PEDF), a liver-derived soluble Wnt inhibitor, which restrains intestinal stem cell (ISC) hyperproliferation to maintain gut homeostasis by suppressing the Wnt/β-catenin signaling pathway. Furthermore, we found that microbial danger signals resulting from intestinal inflammation can be sensed by the liver, leading to the repression of PEDF production through peroxisome proliferator-activated receptor-α (PPARα). This repression liberates ISC proliferation to accelerate tissue repair in the gut. Additionally, treating mice with fenofibrate, a clinical PPARα agonist used for hypolipidemia, enhances colitis susceptibility due to PEDF activity. Therefore, we have identified a distinct role for PEDF in calibrating ISC expansion for intestinal homeostasis through reciprocal interactions between the gut and liver.
Collapse
Affiliation(s)
- Girak Kim
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Zuojia Chen
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jian Li
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jialie Luo
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Felipe Castro-Martinez
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jan Wisniewski
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kairong Cui
- Laboratory of Epigenome Biology, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yan Wang
- Mass Spectrometry Facility, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jialei Sun
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xiaobai Ren
- Department of Ophthalmology, Mary M. and Sash A. Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Stanford, CA 94304, USA
| | - Susan E Crawford
- Department of Surgery, North Shore University Research Institute, University of Chicago Pritzker School of Medicine, Chicago, IL 60637, USA
| | - S Patricia Becerra
- Section of Protein Structure and Function, Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jimin Zhu
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Taotao Liu
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Sui Wang
- Department of Ophthalmology, Mary M. and Sash A. Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Stanford, CA 94304, USA
| | - Keji Zhao
- Laboratory of Epigenome Biology, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Chuan Wu
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
20
|
Zhang Y, Qiang Y, Li H, Li G, Lu L, Dao M, Karniadakis GE, Popel AS, Zhao C. Signaling-biophysical modeling unravels mechanistic control of red blood cell phagocytosis by macrophages in sickle cell disease. PNAS NEXUS 2024; 3:pgae031. [PMID: 38312226 PMCID: PMC10833451 DOI: 10.1093/pnasnexus/pgae031] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 01/09/2024] [Indexed: 02/06/2024]
Abstract
Red blood cell (RBC) aging manifests through progressive changes in cell morphology, rigidity, and expression of membrane proteins. To maintain the quality of circulating blood, splenic macrophages detect the biochemical signals and biophysical changes of RBCs and selectively clear them through erythrophagocytosis. In sickle cell disease (SCD), RBCs display alterations affecting their interaction with macrophages, leading to aberrant phagocytosis that may cause life-threatening spleen sequestration crises. To illuminate the mechanistic control of RBC engulfment by macrophages in SCD, we integrate a system biology model of RBC-macrophage signaling interactions with a biophysical model of macrophage engulfment, as well as in vitro phagocytosis experiments using the spleen-on-a-chip technology. Our modeling framework accurately predicts the phagocytosis dynamics of RBCs under different disease conditions, reveals patterns distinguishing normal and sickle RBCs, and identifies molecular targets including Src homology 2 domain-containing protein tyrosine phosphatase-1 (SHP1) and cluster of differentiation 47 (CD47)/signal regulatory protein α (SIRPα) as therapeutic targets to facilitate the controlled clearance of sickle RBCs in the spleen.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Yuhao Qiang
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - He Li
- School of Chemical, Materials and Biomedical Engineering, University of Georgia, Athens, GA 30602, USA
| | - Guansheng Li
- School of Engineering, Brown University, Providence, RI 02912, USA
| | - Lu Lu
- Department of Statistics and Data Science, Yale University, New Haven, CT 06520, USA
| | - Ming Dao
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | - Aleksander S Popel
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Chen Zhao
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| |
Collapse
|
21
|
Zhou C, Li W, Liang Z, Wu X, Cheng S, Peng J, Zeng K, Li W, Lan P, Yang X, Xiong L, Zeng Z, Zheng X, Huang L, Fan W, Liu Z, Xing Y, Kang L, Liu H. Mutant KRAS-activated circATXN7 fosters tumor immunoescape by sensitizing tumor-specific T cells to activation-induced cell death. Nat Commun 2024; 15:499. [PMID: 38216551 PMCID: PMC10786880 DOI: 10.1038/s41467-024-44779-1] [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: 04/18/2023] [Accepted: 01/05/2024] [Indexed: 01/14/2024] Open
Abstract
Mutant KRAS (KRASMUT) is often exploited by cancers to shape tumor immunity, but the underlying mechanisms are not fully understood. Here we report that tumor-specific cytotoxic T lymphocytes (CTLs) from KRASMUT cancers are sensitive to activation-induced cell death (AICD). circATXN7, an NF-κB-interacting circular RNA, governs T cell sensitivity to AICD by inactivating NF-κB. Mechanistically, histone lactylation derived from KRASMUT tumor cell-produced lactic acid directly activates transcription of circATXN7, which binds to NF-κB p65 subunit and masks the p65 nuclear localization signal motif, thereby sequestering it in the cytoplasm. Clinically, circATXN7 upregulation in tumor-specific CTLs correlates with adverse clinical outcomes and immunotherapeutic resistance. Genetic ablation of circAtxn7 in CD8+ T cells leads to mutant-selective tumor inhibition, while also increases anti-PD1 efficacy in multiple tumor models in female mice. Furthermore, targeting circATXN7 in adoptively transferred tumor-reactive CTLs improves their antitumor activities. These findings provide insight into how lymphocyte-expressed circRNAs contribute to T-cell fate decisions and anticancer immunotherapies.
Collapse
Affiliation(s)
- Chi Zhou
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Wenxin Li
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhenxing Liang
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xianrui Wu
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Sijing Cheng
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jianhong Peng
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Kaixuan Zeng
- Precision Medical Research Institute, the Second Affiliated Hospital of Xi' an Jiaotong University, Xi'an, China
| | - Weihao Li
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Ping Lan
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xin Yang
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Li Xiong
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Ziwei Zeng
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiaobin Zheng
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Liang Huang
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Wenhua Fan
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Zhanzhen Liu
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yue Xing
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
| | - Liang Kang
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Huashan Liu
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, the Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China.
| |
Collapse
|
22
|
Wang J, An H, Ding M, Liu Y, Wang S, Jin Q, Wu Q, Dong H, Guo Q, Tian X, Liu J, Zhang J, Zhu T, Li J, Shao Z, Briles DE, Veening JW, Zheng H, Zhang L, Zhang JR. Liver macrophages and sinusoidal endothelial cells execute vaccine-elicited capture of invasive bacteria. Sci Transl Med 2023; 15:eade0054. [PMID: 38117903 DOI: 10.1126/scitranslmed.ade0054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 11/29/2023] [Indexed: 12/22/2023]
Abstract
Vaccination has substantially reduced the morbidity and mortality of bacterial diseases, but mechanisms of vaccine-elicited pathogen clearance remain largely undefined. We report that vaccine-elicited immunity against invasive bacteria mainly operates in the liver. In contrast to the current paradigm that migrating phagocytes execute vaccine-elicited immunity against blood-borne pathogens, we found that invasive bacteria are captured and killed in the liver of vaccinated host via various immune mechanisms that depend on the protective potency of the vaccine. Vaccines with relatively lower degrees of protection only activated liver-resident macrophage Kupffer cells (KCs) by inducing pathogen-binding immunoglobulin M (IgM) or low amounts of IgG. IgG-coated pathogens were directly captured by KCs via multiple IgG receptors FcγRs, whereas IgM-opsonized bacteria were indirectly bound to KCs via complement receptors of immunoglobulin superfamily (CRIg) and complement receptor 3 (CR3) after complement C3 activation at the bacterial surface. Conversely, the more potent vaccines engaged both KCs and liver sinusoidal endothelial cells by inducing higher titers of functional IgG antibodies. Endothelial cells (ECs) captured densely IgG-opsonized pathogens by the low-affinity IgG receptor FcγRIIB in a "zipper-like" manner and achieved bacterial killing predominantly in the extracellular milieu via an undefined mechanism. KC- and endothelial cell-based capture of antibody-opsonized bacteria also occurred in FcγR-humanized mice. These vaccine protection mechanisms in the liver not only provide a comprehensive explanation for vaccine-/antibody-boosted immunity against invasive bacteria but also may serve as in vivo functional readouts of vaccine efficacy.
Collapse
Affiliation(s)
- Juanjuan Wang
- Center for Infectious Disease Research, Department of Basic Medical Science, School of Medicine, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Haoran An
- Center for Infectious Disease Research, Department of Basic Medical Science, School of Medicine, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Ming Ding
- Center for Infectious Disease Research, Department of Basic Medical Science, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Yanhong Liu
- Center for Infectious Disease Research, Department of Basic Medical Science, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Shaomeng Wang
- Center for Infectious Disease Research, Department of Basic Medical Science, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Qian Jin
- Center for Infectious Disease Research, Department of Basic Medical Science, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Qi Wu
- Center for Infectious Disease Research, Department of Basic Medical Science, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Haodi Dong
- Center for Infectious Disease Research, Department of Basic Medical Science, School of Medicine, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Qile Guo
- Center for Infectious Disease Research, Department of Basic Medical Science, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Xianbin Tian
- Center for Infectious Disease Research, Department of Basic Medical Science, School of Medicine, Tsinghua University, Beijing 100084, China
| | | | | | - Tao Zhu
- Cansino Biologics, Tianjin 300301, China
| | | | - Zhujun Shao
- National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102299, China
| | - David E Briles
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Jan-Willem Veening
- Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, Lausanne 1015, Switzerland
| | | | - Linqi Zhang
- Center for Infectious Disease Research, Department of Basic Medical Science, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Jing-Ren Zhang
- Center for Infectious Disease Research, Department of Basic Medical Science, School of Medicine, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| |
Collapse
|
23
|
Makhmut A, Qin D, Fritzsche S, Nimo J, König J, Coscia F. A framework for ultra-low-input spatial tissue proteomics. Cell Syst 2023; 14:1002-1014.e5. [PMID: 37909047 DOI: 10.1016/j.cels.2023.10.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 08/03/2023] [Accepted: 10/06/2023] [Indexed: 11/02/2023]
Abstract
Spatial proteomics combining microscopy-based cell phenotyping with ultrasensitive mass-spectrometry-based proteomics is an emerging and powerful concept to study cell function and heterogeneity in (patho)physiology. However, optimized workflows that preserve morphological information for phenotype discovery and maximize proteome coverage of few or even single cells from laser microdissected tissue are currently lacking. Here, we report a robust and scalable workflow for the proteomic analysis of ultra-low-input archival material. Benchmarking in murine liver resulted in up to 2,000 quantified proteins from single hepatocyte contours and nearly 5,000 proteins from 50-cell regions. Applied to human tonsil, we profiled 146 microregions including T and B lymphocyte niches and quantified cell-type-specific markers, cytokines, and transcription factors. These data also highlighted proteome dynamics within activated germinal centers, illuminating sites undergoing B cell proliferation and somatic hypermutation. This approach has broad implications in biomedicine, including early disease profiling and drug target and biomarker discovery. A record of this paper's transparent peer review process is included in the supplemental information.
Collapse
Affiliation(s)
- Anuar Makhmut
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Spatial Proteomics Group, Berlin, Germany
| | - Di Qin
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Spatial Proteomics Group, Berlin, Germany
| | - Sonja Fritzsche
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Spatial Proteomics Group, Berlin, Germany
| | - Jose Nimo
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Spatial Proteomics Group, Berlin, Germany
| | - Janett König
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Spatial Proteomics Group, Berlin, Germany
| | - Fabian Coscia
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Spatial Proteomics Group, Berlin, Germany.
| |
Collapse
|
24
|
Kojima Y, Mishiro-Sato E, Fujishita T, Satoh K, Kajino-Sakamoto R, Oze I, Nozawa K, Narita Y, Ogata T, Matsuo K, Muro K, Taketo MM, Soga T, Aoki M. Decreased liver B vitamin-related enzymes as a metabolic hallmark of cancer cachexia. Nat Commun 2023; 14:6246. [PMID: 37803016 PMCID: PMC10558488 DOI: 10.1038/s41467-023-41952-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 09/20/2023] [Indexed: 10/08/2023] Open
Abstract
Cancer cachexia is a complex metabolic disorder accounting for ~20% of cancer-related deaths, yet its metabolic landscape remains unexplored. Here, we report a decrease in B vitamin-related liver enzymes as a hallmark of systemic metabolic changes occurring in cancer cachexia. Metabolomics of multiple mouse models highlights cachexia-associated reductions of niacin, vitamin B6, and a glycine-related subset of one-carbon (C1) metabolites in the liver. Integration of proteomics and metabolomics reveals that liver enzymes related to niacin, vitamin B6, and glycine-related C1 enzymes dependent on B vitamins decrease linearly with their associated metabolites, likely reflecting stoichiometric cofactor-enzyme interactions. The decrease of B vitamin-related enzymes is also found to depend on protein abundance and cofactor subtype. These metabolic/proteomic changes and decreased protein malonylation, another cachexia feature identified by protein post-translational modification analysis, are reflected in blood samples from mouse models and gastric cancer patients with cachexia, underscoring the clinical relevance of our findings.
Collapse
Affiliation(s)
- Yasushi Kojima
- Division of Pathophysiology, Aichi Cancer Center Research Institute, 1-1 Kanokoden, Chikusa-ku, Nagoya, Aichi, 464-8681, Japan.
| | - Emi Mishiro-Sato
- Division of Pathophysiology, Aichi Cancer Center Research Institute, 1-1 Kanokoden, Chikusa-ku, Nagoya, Aichi, 464-8681, Japan
| | - Teruaki Fujishita
- Division of Pathophysiology, Aichi Cancer Center Research Institute, 1-1 Kanokoden, Chikusa-ku, Nagoya, Aichi, 464-8681, Japan
| | - Kiyotoshi Satoh
- Institute for Advanced Biosciences, Keio University, 246-2 Mizukami, Kakuganji, Tsuruoka, Yamagata, 997-0052, Japan
| | - Rie Kajino-Sakamoto
- Division of Pathophysiology, Aichi Cancer Center Research Institute, 1-1 Kanokoden, Chikusa-ku, Nagoya, Aichi, 464-8681, Japan
| | - Isao Oze
- Division of Cancer Epidemiology and Prevention, Aichi Cancer Center Research Institute, 1-1 Kanokoden, Chikusa-ku, Nagoya, Aichi, 464-8681, Japan
| | - Kazuki Nozawa
- Department of Clinical Oncology, Aichi Cancer Center Hospital, 1-1 Kanokoden, Chikusa-ku, Nagoya, Aichi, 464-8681, Japan
| | - Yukiya Narita
- Department of Clinical Oncology, Aichi Cancer Center Hospital, 1-1 Kanokoden, Chikusa-ku, Nagoya, Aichi, 464-8681, Japan
| | - Takatsugu Ogata
- Department of Clinical Oncology, Aichi Cancer Center Hospital, 1-1 Kanokoden, Chikusa-ku, Nagoya, Aichi, 464-8681, Japan
| | - Keitaro Matsuo
- Division of Cancer Epidemiology and Prevention, Aichi Cancer Center Research Institute, 1-1 Kanokoden, Chikusa-ku, Nagoya, Aichi, 464-8681, Japan
| | - Kei Muro
- Department of Clinical Oncology, Aichi Cancer Center Hospital, 1-1 Kanokoden, Chikusa-ku, Nagoya, Aichi, 464-8681, Japan
| | - Makoto Mark Taketo
- Colon Cancer Project, Kyoto University Hospital-iACT, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Tomoyoshi Soga
- Institute for Advanced Biosciences, Keio University, 246-2 Mizukami, Kakuganji, Tsuruoka, Yamagata, 997-0052, Japan
| | - Masahiro Aoki
- Division of Pathophysiology, Aichi Cancer Center Research Institute, 1-1 Kanokoden, Chikusa-ku, Nagoya, Aichi, 464-8681, Japan.
- Department of Cancer Physiology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan.
| |
Collapse
|
25
|
Hui ST, Gong L, Swichkow C, Blencowe M, Kaminska D, Diamante G, Pan C, Dalsania M, French SW, Magyar CE, Pajukanta P, Pihlajamäki J, Boström KI, Yang X, Lusis AJ. Role of Matrix Gla Protein in Transforming Growth Factor-β Signaling and Nonalcoholic Steatohepatitis in Mice. Cell Mol Gastroenterol Hepatol 2023; 16:943-960. [PMID: 37611662 PMCID: PMC10632746 DOI: 10.1016/j.jcmgh.2023.08.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 08/15/2023] [Accepted: 08/16/2023] [Indexed: 08/25/2023]
Abstract
BACKGROUND & AIMS Nonalcoholic steatohepatitis (NASH) is a complex disease involving both genetic and environmental factors in its onset and progression. We analyzed NASH phenotypes in a genetically diverse cohort of mice, the Hybrid Mouse Diversity Panel, to identify genes contributing to disease susceptibility. METHODS A "systems genetics" approach, involving integration of genetic, transcriptomic, and phenotypic data, was used to identify candidate genes and pathways in a mouse model of NASH. The causal role of Matrix Gla Protein (MGP) was validated using heterozygous MGP knockout (Mgp+/-) mice. The mechanistic role of MGP in transforming growth factor-beta (TGF-β) signaling was examined in the LX-2 stellate cell line by using a loss of function approach. RESULTS Local cis-acting regulation of MGP was correlated with fibrosis, suggesting a causal role in NASH, and this was validated using loss of function experiments in 2 models of diet-induced NASH. Using single-cell RNA sequencing, Mgp was found to be primarily expressed in hepatic stellate cells and dendritic cells in mice. Knockdown of MGP expression in stellate LX-2 cells led to a blunted response to TGF-β stimulation. This was associated with reduced regulatory SMAD phosphorylation and TGF-β receptor ALK1 expression as well as increased expression of inhibitory SMAD6. Hepatic MGP expression was found to be significantly correlated with the severity of fibrosis in livers of patients with NASH, suggesting relevance to human disease. CONCLUSIONS MGP regulates liver fibrosis and TGF-β signaling in hepatic stellate cells and contributes to NASH pathogenesis.
Collapse
Affiliation(s)
- Simon T Hui
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California.
| | - Lili Gong
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California; Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Chantle Swichkow
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Montgomery Blencowe
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, California
| | - Dorota Kaminska
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Graciel Diamante
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, California
| | - Calvin Pan
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Meet Dalsania
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Samuel W French
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Clara E Magyar
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Päivi Pajukanta
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Jussi Pihlajamäki
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland; Department of Medicine, Endocrinology, and Clinical Nutrition, Kuopio University Hospital, Kuopio, Finland
| | - Kristina I Boström
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Xia Yang
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, California
| | - Aldons J Lusis
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California.
| |
Collapse
|
26
|
Barber TM, Kabisch S, Pfeiffer AFH, Weickert MO. Metabolic-Associated Fatty Liver Disease and Insulin Resistance: A Review of Complex Interlinks. Metabolites 2023; 13:757. [PMID: 37367914 PMCID: PMC10304744 DOI: 10.3390/metabo13060757] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/12/2023] [Accepted: 06/14/2023] [Indexed: 06/28/2023] Open
Abstract
Metabolic-associated fatty liver disease (MAFLD) has now surpassed alcohol excess as the most common cause of chronic liver disease globally, affecting one in four people. Given its prevalence, MAFLD is an important cause of cirrhosis, even though only a small proportion of patients with MAFLD ultimately progress to cirrhosis. MAFLD suffers as a clinical entity due to its insidious and often asymptomatic onset, lack of an accurate and reliable non-invasive diagnostic test, and lack of a bespoke therapy that has been designed and approved for use specifically in MAFLD. MAFLD sits at a crossroads between the gut and the periphery. The development of MAFLD (including activation of the inflammatory cascade) is influenced by gut-related factors that include the gut microbiota and intactness of the gut mucosal wall. The gut microbiota may interact directly with the liver parenchyma (through translocation via the portal vein), or indirectly through the release of metabolic metabolites that include secondary bile acids, trimethylamine, and short-chain fatty acids (such as propionate and acetate). In turn, the liver mediates the metabolic status of peripheral tissues (including insulin sensitivity) through a complex interplay of hepatokines, liver-secreted metabolites, and liver-derived micro RNAs. As such, the liver plays a key central role in influencing overall metabolic status. In this concise review, we provide an overview of the complex mechanisms whereby MAFLD influences the development of insulin resistance within the periphery, and gut-related factors impact on the development of MAFLD. We also discuss lifestyle strategies for optimising metabolic liver health.
Collapse
Affiliation(s)
- Thomas M. Barber
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism, University Hospitals Coventry and Warwickshire, Clifford Bridge Road, Coventry CV2 2DX, UK
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
- NIHR CRF Human Metabolism Research Unit, University Hospitals Coventry and Warwickshire, Clifford Bridge Road, Coventry CV2 2DX, UK
| | - Stefan Kabisch
- Department of Endocrinology and Metabolic Medicine, Campus Benjamin Franklin, Charité University Medicine, Hindenburgdamm 30, 12203 Berlin, Germany
- Deutsches Zentrum für Diabetesforschung e.V., Geschäftsstelle am Helmholtz-Zentrum München, Ingolstädter Landstraße, 85764 Neuherberg, Germany
| | - Andreas F. H. Pfeiffer
- Department of Endocrinology and Metabolic Medicine, Campus Benjamin Franklin, Charité University Medicine, Hindenburgdamm 30, 12203 Berlin, Germany
- Deutsches Zentrum für Diabetesforschung e.V., Geschäftsstelle am Helmholtz-Zentrum München, Ingolstädter Landstraße, 85764 Neuherberg, Germany
| | - Martin O. Weickert
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism, University Hospitals Coventry and Warwickshire, Clifford Bridge Road, Coventry CV2 2DX, UK
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
- NIHR CRF Human Metabolism Research Unit, University Hospitals Coventry and Warwickshire, Clifford Bridge Road, Coventry CV2 2DX, UK
- Centre for Sport, Exercise and Life Sciences, Faculty of Health & Life Sciences, Coventry University, Coventry CV1 5FB, UK
| |
Collapse
|
27
|
Stokes T, Cen HH, Kapranov P, Gallagher IJ, Pitsillides AA, Volmar C, Kraus WE, Johnson JD, Phillips SM, Wahlestedt C, Timmons JA. Transcriptomics for Clinical and Experimental Biology Research: Hang on a Seq. ADVANCED GENETICS (HOBOKEN, N.J.) 2023; 4:2200024. [PMID: 37288167 PMCID: PMC10242409 DOI: 10.1002/ggn2.202200024] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Indexed: 06/09/2023]
Abstract
Sequencing the human genome empowers translational medicine, facilitating transcriptome-wide molecular diagnosis, pathway biology, and drug repositioning. Initially, microarrays are used to study the bulk transcriptome; but now short-read RNA sequencing (RNA-seq) predominates. Positioned as a superior technology, that makes the discovery of novel transcripts routine, most RNA-seq analyses are in fact modeled on the known transcriptome. Limitations of the RNA-seq methodology have emerged, while the design of, and the analysis strategies applied to, arrays have matured. An equitable comparison between these technologies is provided, highlighting advantages that modern arrays hold over RNA-seq. Array protocols more accurately quantify constitutively expressed protein coding genes across tissue replicates, and are more reliable for studying lower expressed genes. Arrays reveal long noncoding RNAs (lncRNA) are neither sparsely nor lower expressed than protein coding genes. Heterogeneous coverage of constitutively expressed genes observed with RNA-seq, undermines the validity and reproducibility of pathway analyses. The factors driving these observations, many of which are relevant to long-read or single-cell sequencing are discussed. As proposed herein, a reappreciation of bulk transcriptomic methods is required, including wider use of the modern high-density array data-to urgently revise existing anatomical RNA reference atlases and assist with more accurate study of lncRNAs.
Collapse
Affiliation(s)
- Tanner Stokes
- Faculty of ScienceMcMaster UniversityHamiltonL8S 4L8Canada
| | - Haoning Howard Cen
- Life Sciences InstituteUniversity of British ColumbiaVancouverV6T 1Z3Canada
| | | | - Iain J Gallagher
- School of Applied SciencesEdinburgh Napier UniversityEdinburghEH11 4BNUK
| | | | | | | | - James D. Johnson
- Life Sciences InstituteUniversity of British ColumbiaVancouverV6T 1Z3Canada
| | | | | | - James A. Timmons
- Miller School of MedicineUniversity of MiamiMiamiFL33136USA
- William Harvey Research InstituteQueen Mary University LondonLondonEC1M 6BQUK
- Augur Precision Medicine LTDStirlingFK9 5NFUK
| |
Collapse
|
28
|
Dolly A, Pötgens SA, Thibaut MM, Neyrinck AM, de Castro GS, Galbert C, Lefevre C, Wyart E, Gomes SP, Gonçalves DC, Lanthier N, Baldin P, Huot JR, Bonetto A, Seelaender M, Delzenne NM, Sokol H, Bindels LB. Impairment of aryl hydrocarbon receptor signalling promotes hepatic disorders in cancer cachexia. J Cachexia Sarcopenia Muscle 2023; 14:1569-1582. [PMID: 37127348 PMCID: PMC10235873 DOI: 10.1002/jcsm.13246] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 03/02/2023] [Accepted: 04/04/2023] [Indexed: 05/03/2023] Open
Abstract
BACKGROUND The aryl hydrocarbon receptor (AHR) is expressed in the intestine and liver, where it has pleiotropic functions and target genes. This study aims to explore the potential implication of AHR in cancer cachexia, an inflammatory and metabolic syndrome contributing to cancer death. Specifically, we tested the hypothesis that targeting AHR can alleviate cachectic features, particularly through the gut-liver axis. METHODS AHR pathways were explored in multiple tissues from four experimental mouse models of cancer cachexia (C26, BaF3, MC38 and APCMin/+ ) and from non-cachectic mice (sham-injected mice and non-cachexia-inducing [NC26] tumour-bearing mice), as well as in liver biopsies from cancer patients. Cachectic mice were treated with an AHR agonist (6-formylindolo(3,2-b)carbazole [FICZ]) or an antibody neutralizing interleukin-6 (IL-6). Key mechanisms were validated in vitro on HepG2 cells. RESULTS AHR activation, reflected by the expression of Cyp1a1 and Cyp1a2, two major AHR target genes, was deeply reduced in all models (C26 and BaF3, P < 0.001; MC38 and APCMin/+ , P < 0.05) independently of anorexia. This reduction occurred early in the liver (P < 0.001; before the onset of cachexia), compared to the ileum and skeletal muscle (P < 0.01; pre-cachexia stage), and was intrinsically related to cachexia (C26 vs. NC26, P < 0.001). We demonstrate a differential modulation of AHR activation in the liver (through the IL-6/hypoxia-inducing factor 1α pathway) compared to the ileum (attributed to the decreased levels of indolic AHR ligands, P < 0.001), and the muscle. In cachectic mice, FICZ treatment reduced hepatic inflammation: expression of cytokines (Ccl2, P = 0.005; Cxcl2, P = 0.018; Il1b, P = 0.088) with similar trends at the protein levels, expression of genes involved in the acute-phase response (Apcs, P = 0.040; Saa1, P = 0.002; Saa2, P = 0.039; Alb, P = 0.003), macrophage activation (Cd68, P = 0.038) and extracellular matrix remodelling (Fga, P = 0.008; Pcolce, P = 0.025; Timp1, P = 0.003). We observed a decrease in blood glucose in cachectic mice (P < 0.0001), which was also improved by FICZ treatment (P = 0.026) through hepatic transcriptional promotion of a key marker of gluconeogenesis, namely, G6pc (C26 vs. C26 + FICZ, P = 0.029). Strikingly, these benefits on glycaemic disorders occurred independently of an amelioration of the gut barrier dysfunction. In cancer patients, the hepatic expression of G6pc was correlated to Cyp1a1 (Spearman's ρ = 0.52, P = 0.089) and Cyp1a2 (Spearman's ρ = 0.67, P = 0.020). CONCLUSIONS With this set of studies, we demonstrate that impairment of AHR signalling contributes to hepatic inflammatory and metabolic disorders characterizing cancer cachexia, paving the way for innovative therapeutic strategies in this context.
Collapse
Affiliation(s)
- Adeline Dolly
- Metabolism and Nutrition Research Group, Louvain Drug Research InstituteUCLouvain, Université catholique de LouvainBrusselsBelgium
| | - Sarah A. Pötgens
- Metabolism and Nutrition Research Group, Louvain Drug Research InstituteUCLouvain, Université catholique de LouvainBrusselsBelgium
| | - Morgane M. Thibaut
- Metabolism and Nutrition Research Group, Louvain Drug Research InstituteUCLouvain, Université catholique de LouvainBrusselsBelgium
| | - Audrey M. Neyrinck
- Metabolism and Nutrition Research Group, Louvain Drug Research InstituteUCLouvain, Université catholique de LouvainBrusselsBelgium
| | - Gabriela S. de Castro
- Cancer Metabolism Research Group, Department of Surgery, LIM26 HC‐USPUniversity of São PauloSão PauloBrazil
- Departamento de Biologia Celular e do Desenvolvimento, Instituto de Ciências BiomédicasUniversidade de São PauloSão PauloBrazil
| | - Chloé Galbert
- Sorbonne Université, INSERM, Centre de Recherche Saint‐Antoine, CRSA, AP‐HP, Saint Antoine Hospital, Gastroenterology DepartmentParisFrance
- Paris Center for Microbiome Medicine (PaCeMM) FHUParisFrance
| | - Camille Lefevre
- Metabolism and Nutrition Research Group, Louvain Drug Research InstituteUCLouvain, Université catholique de LouvainBrusselsBelgium
| | - Elisabeth Wyart
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology CenterUniversity of TorinoTurinItaly
| | - Silvio P. Gomes
- Departamento de Cirurgia, Faculdade de Medicina VeterinariaUniversidade de São PauloSão PauloBrazil
| | | | - Nicolas Lanthier
- Service d'Hépato‐Gastroentérologie, Cliniques universitaires Saint‐LucUCLouvainBrusselsBelgium
- Laboratory of Hepatogastroenterology, Institut de Recherche Expérimentale et CliniqueUCLouvainBrusselsBelgium
| | - Pamela Baldin
- Service d'Anatomie Pathologique, Cliniques Universitaires Saint‐LucUCLouvainBrusselsBelgium
| | - Joshua R. Huot
- Department of Anatomy, Cell Biology & PhysiologyIndiana University School of MedicineIndianapolisINUSA
| | - Andrea Bonetto
- Department of PathologyUniversity of Colorado Anschutz Medical CampusAuroraCOUSA
| | - Marília Seelaender
- Cancer Metabolism Research Group, Department of Surgery, LIM26 HC‐USPUniversity of São PauloSão PauloBrazil
| | - Nathalie M. Delzenne
- Metabolism and Nutrition Research Group, Louvain Drug Research InstituteUCLouvain, Université catholique de LouvainBrusselsBelgium
| | - Harry Sokol
- Sorbonne Université, INSERM, Centre de Recherche Saint‐Antoine, CRSA, AP‐HP, Saint Antoine Hospital, Gastroenterology DepartmentParisFrance
- Paris Center for Microbiome Medicine (PaCeMM) FHUParisFrance
- INRAE, UMR1319 Micalis and AgroParisTechJouy‐en‐JosasFrance
| | - Laure B. Bindels
- Metabolism and Nutrition Research Group, Louvain Drug Research InstituteUCLouvain, Université catholique de LouvainBrusselsBelgium
- WELBIO DepartmentWEL Research InstituteWavreBelgium
| |
Collapse
|
29
|
González-Moreno L, Santamaría-Cano A, Paradela A, Martínez-Chantar ML, Martín MÁ, Pérez-Carreras M, García-Picazo A, Vázquez J, Calvo E, González-Aseguinolaza G, Saheki T, del Arco A, Satrústegui J, Contreras L. Exogenous aralar/slc25a12 can replace citrin/slc25a13 as malate aspartate shuttle component in liver. Mol Genet Metab Rep 2023; 35:100967. [PMID: 36967723 PMCID: PMC10031141 DOI: 10.1016/j.ymgmr.2023.100967] [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: 03/09/2023] [Accepted: 03/10/2023] [Indexed: 03/18/2023] Open
Abstract
The deficiency of CITRIN, the liver mitochondrial aspartate-glutamate carrier (AGC), is the cause of four human clinical phenotypes, neonatal intrahepatic cholestasis caused by CITRIN deficiency (NICCD), silent period, failure to thrive and dyslipidemia caused by CITRIN deficiency (FTTDCD), and citrullinemia type II (CTLN2). Clinical symptoms can be traced back to disruption of the malate-aspartate shuttle due to the lack of citrin. A potential therapy for this condition is the expression of aralar, the AGC present in brain, to replace citrin. To explore this possibility we have first verified that the NADH/NAD+ ratio increases in hepatocytes from citrin(-/-) mice, and then found that exogenous aralar expression reversed the increase in NADH/NAD+ observed in these cells. Liver mitochondria from citrin (-/-) mice expressing liver specific transgenic aralar had a small (~ 4-6 nmoles x mg prot-1 x min-1) but consistent increase in malate aspartate shuttle (MAS) activity over that of citrin(-/-) mice. These results support the functional replacement between AGCs in the liver. To explore the significance of AGC replacement in human therapy we studied the relative levels of citrin and aralar in mouse and human liver through absolute quantification proteomics. We report that mouse liver has relatively high aralar levels (citrin/aralar molar ratio of 7.8), whereas human liver is virtually devoid of aralar (CITRIN/ARALAR ratio of 397). This large difference in endogenous aralar levels partly explains the high residual MAS activity in liver of citrin(-/-) mice and why they fail to recapitulate the human disease, but supports the benefit of increasing aralar expression to improve the redox balance capacity of human liver, as an effective therapy for CITRIN deficiency.
Collapse
Key Words
- (BNGE), Blue native gel electrophoresis
- AGC, aspartate-glutamate carrier
- AQUA, Absolute Quantification methods
- Aspartate-glutamate carrier
- CD, CITRIN Deficiency
- CTNL2, citrullinemia type II
- Citrin deficiency
- DAB, 3,3-diaminobenzidine
- FBS, Fetal Bovine serum
- FTTDCD, failure to thrive and dyslipidemia caused by CITRIN Deficiency
- GOT, aspartate transaminase
- GPD2, mitochondrial glycerol phosphate dehydrogenase
- GPS, glycerol phosphate shuttle
- Hepatocyte
- IM, imaging medium
- LC-MS, liquid chromatography mass spectrometry
- LNP, lipid nanoparticles
- MAS, malate aspartate shuttle
- Malate-aspartate shuttle
- Mitochondria
- NAA, N-Acetyl-aspartate
- NICCD, neonatal intrahepatic cholestasis caused by CITRIN Deficiency
- OXPHOS, oxidative phosphorylation
- PFA, paraformaldehyde
- PRM, parallel reaction monitoring
- SDS, sodium dodecyl sulfate
- TBS, Tris-Buffered saline.
- hCitrin, human citrin
Collapse
Affiliation(s)
- Luis González-Moreno
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, 28049 Madrid, Spain
- Instituto Universitario de Biología Molecular, (IUBM), and Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid, Madrid, Spain
- Instituto de Investigaciones Sanitarias Fundación Jiménez Díaz (IIS-FJD), Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Andrea Santamaría-Cano
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, 28049 Madrid, Spain
- Instituto Universitario de Biología Molecular, (IUBM), and Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid, Madrid, Spain
- Instituto de Investigaciones Sanitarias Fundación Jiménez Díaz (IIS-FJD), Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Alberto Paradela
- Centro Nacional de Biotecnología (CNB), CSIC. C/Darwin 3, 28049 Madrid, Spain
| | - María Luz Martínez-Chantar
- Liver Disease Lab, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), 48160 Derio, Bizkaia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Miguel Á. Martín
- Grupo Enfermedades Mitocondriales y Neuromusculares, Instituto de Investigación Hospital 12 de Octubre (imas12), Madrid, Spain
- Servicio de Genética, Hospital Universitario 12 de Octubre, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain
| | | | - Alberto García-Picazo
- Departamento de Cirugía General Aparato Digestivo, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Jesús Vázquez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Enrique Calvo
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Gloria González-Aseguinolaza
- Gene Therapy and Regulation of Gene Expression Program, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Spain
- IdiSNA Navarra Institute for Health Research, 31008 Pamplona, Spain
| | | | - Araceli del Arco
- Instituto Universitario de Biología Molecular, (IUBM), and Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid, Madrid, Spain
- Instituto de Investigaciones Sanitarias Fundación Jiménez Díaz (IIS-FJD), Universidad Autónoma de Madrid, 28049 Madrid, Spain
- Facultad de Ciencias Ambientales y Bioquímica, Universidad de Castilla la Mancha, Toledo 45071, Spain
- Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina, Toledo 45071, Spain
| | - Jorgina Satrústegui
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, 28049 Madrid, Spain
- Instituto Universitario de Biología Molecular, (IUBM), and Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid, Madrid, Spain
- Instituto de Investigaciones Sanitarias Fundación Jiménez Díaz (IIS-FJD), Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Laura Contreras
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, 28049 Madrid, Spain
- Instituto Universitario de Biología Molecular, (IUBM), and Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid, Madrid, Spain
- Instituto de Investigaciones Sanitarias Fundación Jiménez Díaz (IIS-FJD), Universidad Autónoma de Madrid, 28049 Madrid, Spain
| |
Collapse
|
30
|
Hasper J, Welle K, Hryhorenko J, Ghaemmaghami S, Buchwalter A. Turnover and replication analysis by isotope labeling (TRAIL) reveals the influence of tissue context on protein and organelle lifetimes. Mol Syst Biol 2023; 19:e11393. [PMID: 36929723 PMCID: PMC10090950 DOI: 10.15252/msb.202211393] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 02/13/2023] [Accepted: 02/16/2023] [Indexed: 03/18/2023] Open
Abstract
The lifespans of proteins range from minutes to years within mammalian tissues. Protein lifespan is relevant to organismal aging, as long-lived proteins accrue damage over time. It is unclear how protein lifetime is shaped by tissue context, where both cell turnover and proteolytic degradation contribute to protein turnover. We develop turnover and replication analysis by 15 N isotope labeling (TRAIL) to quantify protein and cell lifetimes with high precision and demonstrate that cell turnover, sequence-encoded features, and environmental factors modulate protein lifespan across tissues. Cell and protein turnover flux are comparable in proliferative tissues, while protein turnover outpaces cell turnover in slowly proliferative tissues. Physicochemical features such as hydrophobicity, charge, and disorder influence protein turnover in slowly proliferative tissues, but protein turnover is much less sequence-selective in highly proliferative tissues. Protein lifetimes vary nonrandomly across tissues after correcting for cell turnover. Multiprotein complexes such as the ribosome have consistent lifetimes across tissues, while mitochondria, peroxisomes, and lipid droplets have variable lifetimes. TRAIL can be used to explore how environment, aging, and disease affect tissue homeostasis.
Collapse
Affiliation(s)
- John Hasper
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Kevin Welle
- University of Rochester Mass Spectrometry Resource Laboratory, Rochester, NY, USA
| | - Jennifer Hryhorenko
- University of Rochester Mass Spectrometry Resource Laboratory, Rochester, NY, USA
| | - Sina Ghaemmaghami
- University of Rochester Mass Spectrometry Resource Laboratory, Rochester, NY, USA.,Department of Biology, University of Rochester, Rochester, NY, USA
| | - Abigail Buchwalter
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA.,Department of Physiology, University of California, San Francisco, San Francisco, CA, USA.,Chan Zuckerberg Biohub, San Francisco, CA, USA
| |
Collapse
|
31
|
Madsen S, Nelson ME, Deshpande V, Humphrey SJ, Cooke KC, Howell A, Diaz-Vegas A, Burchfield JG, Stöckli J, James DE. Deep Proteome Profiling of White Adipose Tissue Reveals Marked Conservation and Distinct Features Between Different Anatomical Depots. Mol Cell Proteomics 2023; 22:100508. [PMID: 36787876 PMCID: PMC10014311 DOI: 10.1016/j.mcpro.2023.100508] [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: 08/26/2022] [Revised: 01/26/2023] [Accepted: 02/08/2023] [Indexed: 02/16/2023] Open
Abstract
White adipose tissue is deposited mainly as subcutaneous adipose tissue (SAT), often associated with metabolic protection, and abdominal/visceral adipose tissue, which contributes to metabolic disease. To investigate the molecular underpinnings of these differences, we conducted comprehensive proteomics profiling of whole tissue and isolated adipocytes from these two depots across two diets from C57Bl/6J mice. The adipocyte proteomes from lean mice were highly conserved between depots, with the major depot-specific differences encoded by just 3% of the proteome. Adipocytes from SAT (SAdi) were enriched in pathways related to mitochondrial complex I and beiging, whereas visceral adipocytes (VAdi) were enriched in structural proteins and positive regulators of mTOR presumably to promote nutrient storage and cellular expansion. This indicates that SAdi are geared toward higher catabolic activity, while VAdi are more suited for lipid storage. By comparing adipocytes from mice fed chow or Western diet (WD), we define a core adaptive proteomics signature consisting of increased extracellular matrix proteins and decreased fatty acid metabolism and mitochondrial Coenzyme Q biosynthesis. Relative to SAdi, VAdi displayed greater changes with WD including a pronounced decrease in mitochondrial proteins concomitant with upregulation of apoptotic signaling and decreased mitophagy, indicating pervasive mitochondrial stress. Furthermore, WD caused a reduction in lipid handling and glucose uptake pathways particularly in VAdi, consistent with adipocyte de-differentiation. By overlaying the proteomics changes with diet in whole adipose tissue and isolated adipocytes, we uncovered concordance between adipocytes and tissue only in the visceral adipose tissue, indicating a unique tissue-specific adaptation to sustained WD in SAT. Finally, an in-depth comparison of isolated adipocytes and 3T3-L1 proteomes revealed a high degree of overlap, supporting the utility of the 3T3-L1 adipocyte model. These deep proteomes provide an invaluable resource highlighting differences between white adipose depots that may fine-tune their unique functions and adaptation to an obesogenic environment.
Collapse
Affiliation(s)
- Søren Madsen
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia; Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Marin E Nelson
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia; Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Vinita Deshpande
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia; Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Sean J Humphrey
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia; Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Kristen C Cooke
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia; Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Anna Howell
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia; Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Alexis Diaz-Vegas
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia; Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - James G Burchfield
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia; Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Jacqueline Stöckli
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia; Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - David E James
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia; Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia; Faculty of Medicine and Health, University of Sydney, Camperdown, New South Wales, Australia.
| |
Collapse
|
32
|
Liu Y, Trnka MJ, He L, Burlingame AL, Correia MA. In-Cell Chemical Crosslinking Identifies Hotspots for SQSTM-1/p62-IκBα Interaction That Underscore a Critical Role of p62 in Limiting NF-κB Activation Through IκBα Stabilization. Mol Cell Proteomics 2023; 22:100495. [PMID: 36634736 PMCID: PMC9947424 DOI: 10.1016/j.mcpro.2023.100495] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 01/02/2023] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
We have previously documented that in liver cells, the multifunctional protein scaffold p62/SQSTM1 is closely associated with IκBα, an inhibitor of the transcriptional activator NF-κB. Such an intimate p62-IκBα association we now document leads to a marked 18-fold proteolytic IκBα-stabilization, enabling its nuclear entry and termination of the NF-κB-activation cycle. In p62-/--cells, such termination is abrogated resulting in the nuclear persistence and prolonged activation of NF-κB following inflammatory stimuli. Utilizing various approaches both classic (structural deletion, site-directed mutagenesis) as well as novel (in-cell chemical crosslinking), coupled with proteomic analyses, we have defined the precise structural hotspots of p62-IκBα association. Accordingly, we have identified such IκBα hotspots to reside around N-terminal (K38, K47, and K67) and C-terminal (K238/C239) residues in its fifth ankyrin repeat domain. These sites interact with two hotspots in p62: One in its PB-1 subdomain around K13, and the other comprised of a positively charged patch (R183/R186/K187/K189) between its ZZ- and TB-subdomains. APEX proximity analyses upon IκBα-cotransfection of cells with and without p62 have enabled the characterization of the p62 influence on IκBα-protein-protein interactions. Interestingly, consistent with p62's capacity to proteolytically stabilize IκBα, its presence greatly impaired IκBα's interactions with various 20S/26S proteasomal subunits. Furthermore, consistent with p62 interaction with IκBα on an interface opposite to that of its NF-κB-interacting interface, p62 failed to significantly affect IκBα-NF-κB interactions. These collective findings together with the known dynamic p62 nucleocytoplasmic shuttling leads us to speculate that it may be involved in "piggy-back" nuclear transport of IκBα following its NF-κB-elicited transcriptional activation and de novo synthesis, required for termination of the NF-κB-activation cycle. Consequently, mice carrying a liver-specific deletion of p62-residues 68 to 252 reveal age-dependent-enhanced liver inflammation. Our findings reveal yet another mode of p62-mediated pathophysiologically relevant regulation of NF-κB.
Collapse
Affiliation(s)
- Yi Liu
- Department of Cellular & Molecular Pharmacology, University of California San Francisco, San Francisco, California, USA
| | - Michael J Trnka
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California, USA
| | - Liang He
- Department of Cellular & Molecular Pharmacology, University of California San Francisco, San Francisco, California, USA
| | - A L Burlingame
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California, USA
| | - Maria Almira Correia
- Department of Cellular & Molecular Pharmacology, University of California San Francisco, San Francisco, California, USA; Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California, USA; Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA; The Liver Center, University of California San Francisco, San Francisco, California, USA.
| |
Collapse
|
33
|
McGuire MR, Espenshade PJ. PGRMC1: An enigmatic heme-binding protein. Pharmacol Ther 2023; 241:108326. [PMID: 36463977 PMCID: PMC9839567 DOI: 10.1016/j.pharmthera.2022.108326] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/22/2022] [Accepted: 11/28/2022] [Indexed: 12/03/2022]
Abstract
Progesterone Receptor Membrane Component 1 (PGRMC1) is a heme-binding protein that has been implicated in a wide range of cell and tissue functions, including cytochromes P450 activity, heme homeostasis, cancer, female reproduction, and protein quality control. Despite an extensive body of literature, a relative lack of mechanistic insight means that how PGRMC1 functions in these different aspects of biology is largely unknown. This review provides an overview of the PGRMC1 literature, highlighting what information is rigorously supported by experimental evidence and where additional investigation is warranted. The central role of PGRMC1 in supporting cytochrome P450 activity is discussed at length. Building on existing models of PGRMC1 function, a speculative model is proposed using the reviewed literature in which PGRMC1 functions as a heme chaperone to shuttle heme from its site of synthesis in the mitochondrion to other subcellular compartments. By spotlighting knowledge gaps, this review will motivate investigators to better understand this enigmatic protein.
Collapse
Affiliation(s)
- Meredith R McGuire
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Peter J Espenshade
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Oncology, Johns Hopkins University School of Medicine, 725 N. Wolfe St., Physiology 107B, Baltimore, MD 21205, USA.
| |
Collapse
|
34
|
Integrated proteomic and transcriptomic landscape of macrophages in mouse tissues. Nat Commun 2022; 13:7389. [PMID: 36450731 PMCID: PMC9712610 DOI: 10.1038/s41467-022-35095-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 11/18/2022] [Indexed: 12/03/2022] Open
Abstract
Macrophages are involved in tissue homeostasis and are critical for innate immune responses, yet distinct macrophage populations in different tissues exhibit diverse gene expression patterns and biological processes. While tissue-specific macrophage epigenomic and transcriptomic profiles have been reported, proteomes of different macrophage populations remain poorly characterized. Here we use mass spectrometry and bulk RNA sequencing to assess the proteomic and transcriptomic patterns, respectively, of 10 primary macrophage populations from seven mouse tissues, bone marrow-derived macrophages and the cell line RAW264.7. The results show distinct proteomic landscape and protein copy numbers between tissue-resident and recruited macrophages. Construction of a hierarchical regulatory network finds cell-type-specific transcription factors of macrophages serving as hubs for denoting tissue and functional identity of individual macrophage subsets. Finally, Il18 is validated to be essential in distinguishing molecular signatures and cellular function features between tissue-resident and recruited macrophages in the lung and liver. In summary, these deposited datasets and our open proteome server ( http://macrophage.mouseprotein.cn ) integrating all information will provide a valuable resource for future functional and mechanistic studies of mouse macrophages.
Collapse
|
35
|
Li R, Bhandari S, Martinez-Zubiaurre I, Bruun JA, Urbarova I, Smedsrød B, Simón-Santamaría J, Sørensen KK. Changes in the proteome and secretome of rat liver sinusoidal endothelial cells during early primary culture and effects of dexamethasone. PLoS One 2022; 17:e0273843. [PMID: 36054185 PMCID: PMC9439253 DOI: 10.1371/journal.pone.0273843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 08/16/2022] [Indexed: 11/28/2022] Open
Abstract
Introduction Liver sinusoidal endothelial cells (LSECs) are specialized fenestrated scavenger endothelial cells involved in the elimination of modified plasma proteins and tissue turnover waste macromolecules from blood. LSECs also participate in liver immune responses. A challenge when studying LSEC biology is the rapid loss of the in vivo phenotype in culture. In this study, we have examined biological processes and pathways affected during early-stage primary culture of rat LSECs and checked for cell responses to the pro-inflammatory cytokine interleukin (IL)-1β and the anti-inflammatory drug dexamethasone. Methods LSECs from male Sprague Dawley rats were cultured on type I collagen in 5% oxygen atmosphere in DMEM with serum-free supplements for 2 and 24 h. Quantitative proteomics using tandem mass tag technology was used to examine proteins in cells and supernatants. Validation was done with qPCR, ELISA, multiplex immunoassay, and caspase 3/7 assay. Cell ultrastructure was examined by scanning electron microscopy, and scavenger function by quantitative endocytosis assays. Results LSECs cultured for 24 h showed a characteristic pro-inflammatory phenotype both in the presence and absence of IL-1β, with upregulation of cellular responses to cytokines and interferon-γ, cell-cell adhesion, and glycolysis, increased expression of fatty acid binding proteins (FABP4, FABP5), and downregulation of several membrane receptors (STAB1, STAB2, LYVE1, CLEC4G) and proteins in pyruvate metabolism, citric acid cycle, fatty acid elongation, amino acid metabolism, and oxidation-reduction processes. Dexamethasone inhibited apoptosis and improved LSEC viability in culture, repressed inflammatory and immune regulatory pathways and secretion of IL-1β and IL-6, and further upregulated FABP4 and FABP5 compared to time-matched controls. The LSEC porosity and endocytic activity were reduced at 24 h both with and without dexamethasone but the dexamethasone-treated cells showed a less stressed phenotype. Conclusion Rat LSECs become activated towards a pro-inflammatory phenotype during early culture. Dexamethasone represses LSEC activation, inhibits apoptosis, and improves cell viability.
Collapse
Affiliation(s)
- Ruomei Li
- Department of Medical Biology, UiT–The Arctic University of Norway, Tromsø, Norway
| | - Sabin Bhandari
- Department of Medical Biology, UiT–The Arctic University of Norway, Tromsø, Norway
| | | | - Jack-Ansgar Bruun
- Department of Medical Biology, UiT–The Arctic University of Norway, Tromsø, Norway
| | - Ilona Urbarova
- Department of Community Medicine, UiT–The Arctic University of Norway, Tromsø, Norway
| | - Bård Smedsrød
- Department of Medical Biology, UiT–The Arctic University of Norway, Tromsø, Norway
| | | | | |
Collapse
|
36
|
Zhang Z, Xun Y, Rong S, Yan L, SoRelle JA, Li X, Tang M, Keller K, Ludwig S, Moresco EMY, Beutler B. Loss of immunity-related GTPase GM4951 leads to nonalcoholic fatty liver disease without obesity. Nat Commun 2022; 13:4136. [PMID: 35842425 PMCID: PMC9288484 DOI: 10.1038/s41467-022-31812-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 07/05/2022] [Indexed: 11/30/2022] Open
Abstract
Obesity and diabetes are well known risk factors for nonalcoholic fatty liver disease (NAFLD), but the genetic factors contributing to the development of NAFLD remain poorly understood. Here we describe two semi-dominant allelic missense mutations (Oily and Carboniferous) of Predicted gene 4951 (Gm4951) identified from a forward genetic screen in mice. GM4951 deficient mice developed NAFLD on high fat diet (HFD) with no changes in body weight or glucose metabolism. Moreover, HFD caused a reduction in the level of Gm4951, which in turn promoted the development of NAFLD. Predominantly expressed in hepatocytes, GM4951 was verified as an interferon inducible GTPase. The NAFLD in Gm4951 knockout mice was associated with decreased lipid oxidation in the liver and no defect in hepatic lipid secretion. After lipid loading, hepatocyte GM4951 translocated to lipid droplets (LDs), bringing with it hydroxysteroid 17β-dehydrogenase 13 (HSD17B13), which in the absence of GM4951 did not undergo this translocation. We identified a rare non-obese mouse model of NAFLD caused by GM4951 deficiency and define a critical role for GTPase-mediated translocation in hepatic lipid metabolism.
Collapse
Affiliation(s)
- Zhao Zhang
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA. .,Division of Endocrinology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| | - Yu Xun
- grid.267313.20000 0000 9482 7121Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA ,grid.267313.20000 0000 9482 7121Division of Endocrinology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
| | - Shunxing Rong
- grid.267313.20000 0000 9482 7121Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA ,grid.267313.20000 0000 9482 7121Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
| | - Lijuan Yan
- grid.267313.20000 0000 9482 7121Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
| | - Jeffrey A. SoRelle
- grid.267313.20000 0000 9482 7121Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
| | - Xiaohong Li
- grid.267313.20000 0000 9482 7121Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
| | - Miao Tang
- grid.267313.20000 0000 9482 7121Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
| | - Katie Keller
- grid.267313.20000 0000 9482 7121Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
| | - Sara Ludwig
- grid.267313.20000 0000 9482 7121Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
| | - Eva Marie Y. Moresco
- grid.267313.20000 0000 9482 7121Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
| | - Bruce Beutler
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
37
|
Lao Y, Li Y, Wang W, Ren L, Qian X, He F, Chen X, Jiang Y. A Cytological Atlas of the Human Liver Proteome from PROTEOME SKY-LIVER Hu 2.0, a Publicly Available Database. J Proteome Res 2022; 21:1916-1929. [PMID: 35820117 DOI: 10.1021/acs.jproteome.2c00190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The liver plays a unique role as a metabolic center of the body, and also performs other important functions such as detoxification and immune response. Here, we establish a cell type-resolved healthy human liver proteome including hepatocytes (HCs), hepatic stellate cells (HSCs), Kupffer cells (KCs), and liver sinusoidal endothelial cells (LSECs) by high-resolution mass spectrometry. Overall, we quantify total 8354 proteins for four cell types and over 6000 proteins for each cell type. Analysis of this data set and regulatory pathway reveals the cellular labor division in the human liver follows the pattern that parenchymal cells make the main components of pathways, but nonparenchymal cells trigger these pathways. Human liver cells show some novel molecular features: HCs maintain KCs and LSECs homeostasis by producing cholesterol and ketone bodies; HSCs participate in xenobiotics metabolism as an agent deliverer; KCs and LSECs mediate immune response through MHC class II-TLRs and MHC class I-TGFβ cascade, respectively; and KCs play a central role in diurnal rhythms regulation through sensing diurnal IGF and temperature flux. Together, this work expands our understandings of liver physiology and provides a useful resource for future analyses of normal and diseased livers.
Collapse
Affiliation(s)
- Yuanxiang Lao
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center of Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China.,Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Yanyan Li
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center of Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China.,Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Wei Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center of Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Liangliang Ren
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center of Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Xiaohong Qian
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center of Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Fuchu He
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center of Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Xinguo Chen
- Institute of Liver Transplantation, The Third Medical Center, Chinese PLA General Hospital, Beijing 100039, China
| | - Ying Jiang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center of Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China.,Anhui Medical University, Hefei 230031, China
| |
Collapse
|
38
|
Deciphering signal transduction networks in the liver by mechanistic mathematical modelling. Biochem J 2022; 479:1361-1374. [PMID: 35748700 PMCID: PMC9246346 DOI: 10.1042/bcj20210548] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 06/10/2022] [Accepted: 06/10/2022] [Indexed: 11/17/2022]
Abstract
In health and disease, liver cells are continuously exposed to cytokines and growth factors. While individual signal transduction pathways induced by these factors were studied in great detail, the cellular responses induced by repeated or combined stimulations are complex and less understood. Growth factor receptors on the cell surface of hepatocytes were shown to be regulated by receptor interactions, receptor trafficking and feedback regulation. Here, we exemplify how mechanistic mathematical modelling based on quantitative data can be employed to disentangle these interactions at the molecular level. Crucial is the analysis at a mechanistic level based on quantitative longitudinal data within a mathematical framework. In such multi-layered information, step-wise mathematical modelling using submodules is of advantage, which is fostered by sharing of standardized experimental data and mathematical models. Integration of signal transduction with metabolic regulation in the liver and mechanistic links to translational approaches promise to provide predictive tools for biology and personalized medicine.
Collapse
|
39
|
Mund A, Brunner AD, Mann M. Unbiased spatial proteomics with single-cell resolution in tissues. Mol Cell 2022; 82:2335-2349. [PMID: 35714588 DOI: 10.1016/j.molcel.2022.05.022] [Citation(s) in RCA: 132] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 05/05/2022] [Accepted: 05/18/2022] [Indexed: 12/19/2022]
Abstract
Mass spectrometry (MS)-based proteomics has become a powerful technology to quantify the entire complement of proteins in cells or tissues. Here, we review challenges and recent advances in the LC-MS-based analysis of minute protein amounts, down to the level of single cells. Application of this technology revealed that single-cell transcriptomes are dominated by stochastic noise due to the very low number of transcripts per cell, whereas the single-cell proteome appears to be complete. The spatial organization of cells in tissues can be studied by emerging technologies, including multiplexed imaging and spatial transcriptomics, which can now be combined with ultra-sensitive proteomics. Combined with high-content imaging, artificial intelligence and single-cell laser microdissection, MS-based proteomics provides an unbiased molecular readout close to the functional level. Potential applications range from basic biological questions to precision medicine.
Collapse
Affiliation(s)
- Andreas Mund
- Proteomics Program, The Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Faculty of Health and Medical Sciences, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Andreas-David Brunner
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany; Boehringer Ingelheim Pharma GmbH & Co. KG, Drug Discovery Sciences, Birkendorfer Str. 65, D-88397, Biberach Riss, Germany
| | - Matthias Mann
- Proteomics Program, The Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Faculty of Health and Medical Sciences, Blegdamsvej 3B, 2200 Copenhagen, Denmark; Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany.
| |
Collapse
|
40
|
Wang S, García-Seisdedos D, Prakash A, Kundu DJ, Collins A, George N, Fexova S, Moreno P, Papatheodorou I, Jones AR, Vizcaíno JA. Integrated view and comparative analysis of baseline protein expression in mouse and rat tissues. PLoS Comput Biol 2022; 18:e1010174. [PMID: 35714157 PMCID: PMC9246241 DOI: 10.1371/journal.pcbi.1010174] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 06/30/2022] [Accepted: 05/05/2022] [Indexed: 11/19/2022] Open
Abstract
The increasingly large amount of proteomics data in the public domain enables, among other applications, the combined analyses of datasets to create comparative protein expression maps covering different organisms and different biological conditions. Here we have reanalysed public proteomics datasets from mouse and rat tissues (14 and 9 datasets, respectively), to assess baseline protein abundance. Overall, the aggregated dataset contained 23 individual datasets, including a total of 211 samples coming from 34 different tissues across 14 organs, comprising 9 mouse and 3 rat strains, respectively. In all cases, we studied the distribution of canonical proteins between the different organs. The number of canonical proteins per dataset ranged from 273 (tendon) and 9,715 (liver) in mouse, and from 101 (tendon) and 6,130 (kidney) in rat. Then, we studied how protein abundances compared across different datasets and organs for both species. As a key point we carried out a comparative analysis of protein expression between mouse, rat and human tissues. We observed a high level of correlation of protein expression among orthologs between all three species in brain, kidney, heart and liver samples, whereas the correlation of protein expression was generally slightly lower between organs within the same species. Protein expression results have been integrated into the resource Expression Atlas for widespread dissemination.
Collapse
Affiliation(s)
- Shengbo Wang
- European Molecular Biology Laboratory - European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | - David García-Seisdedos
- European Molecular Biology Laboratory - European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
- Open Targets, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Ananth Prakash
- European Molecular Biology Laboratory - European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
- Open Targets, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Deepti Jaiswal Kundu
- European Molecular Biology Laboratory - European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Andrew Collins
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Nancy George
- European Molecular Biology Laboratory - European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Silvie Fexova
- European Molecular Biology Laboratory - European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Pablo Moreno
- European Molecular Biology Laboratory - European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Irene Papatheodorou
- European Molecular Biology Laboratory - European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
- Open Targets, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Andrew R. Jones
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Juan Antonio Vizcaíno
- European Molecular Biology Laboratory - European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
- Open Targets, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| |
Collapse
|
41
|
Niu L, Geyer PE, Gupta R, Santos A, Meier F, Doll S, Wewer Albrechtsen NJ, Klein S, Ortiz C, Uschner FE, Schierwagen R, Trebicka J, Mann M. Dynamic human liver proteome atlas reveals functional insights into disease pathways. Mol Syst Biol 2022; 18:e10947. [PMID: 35579278 PMCID: PMC9112488 DOI: 10.15252/msb.202210947] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 12/12/2022] Open
Abstract
Deeper understanding of liver pathophysiology would benefit from a comprehensive quantitative proteome resource at cell type resolution to predict outcome and design therapy. Here, we quantify more than 150,000 sequence-unique peptides aggregated into 10,000 proteins across total liver, the major liver cell types, time course of primary cell cultures, and liver disease states. Bioinformatic analysis reveals that half of hepatocyte protein mass is comprised of enzymes and 23% of mitochondrial proteins, twice the proportion of other liver cell types. Using primary cell cultures, we capture dynamic proteome remodeling from tissue states to cell line states, providing useful information for biological or pharmaceutical research. Our extensive data serve as spectral library to characterize a human cohort of non-alcoholic steatohepatitis and cirrhosis. Dramatic proteome changes in liver tissue include signatures of hepatic stellate cell activation resembling liver cirrhosis and providing functional insights. We built a web-based dashboard application for the interactive exploration of our resource (www.liverproteome.org).
Collapse
Affiliation(s)
- Lili Niu
- Novo Nordisk Foundation Center for Protein ResearchFaculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
- Department of Proteomics and Signal TransductionMax Planck Institute of BiochemistryMartinsriedGermany
| | - Philipp E Geyer
- Department of Proteomics and Signal TransductionMax Planck Institute of BiochemistryMartinsriedGermany
- Present address:
OmicEra Diagnostics GmbHPlaneggGermany
| | - Rajat Gupta
- Novo Nordisk Foundation Center for Protein ResearchFaculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
- Present address:
Pfizer Worldwide Research and DevelopmentSan DiegoCAUSA
| | - Alberto Santos
- Novo Nordisk Foundation Center for Protein ResearchFaculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
- Center for Health Data ScienceFaculty of Health SciencesUniversity of CopenhagenCopenhagenDenmark
- Big Data InstituteNuffield Department of MedicineUniversity of OxfordOxfordUK
| | - Florian Meier
- Department of Proteomics and Signal TransductionMax Planck Institute of BiochemistryMartinsriedGermany
- Present address:
Functional ProteomicsJena University HospitalJenaGermany
| | - Sophia Doll
- Department of Proteomics and Signal TransductionMax Planck Institute of BiochemistryMartinsriedGermany
- Present address:
OmicEra Diagnostics GmbHPlaneggGermany
| | - Nicolai J Wewer Albrechtsen
- Novo Nordisk Foundation Center for Protein ResearchFaculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
- Department of Clinical BiochemistryRigshospitaletUniversity of CopenhagenCopenhagenDenmark
| | - Sabine Klein
- Department of Internal Medicine IGoethe University Clinic FrankfurtFrankfurtGermany
- Department of Internal Medicine BWW University MünsterMünsterGermany
| | - Cristina Ortiz
- Department of Internal Medicine IGoethe University Clinic FrankfurtFrankfurtGermany
| | - Frank E Uschner
- Department of Internal Medicine IGoethe University Clinic FrankfurtFrankfurtGermany
- Department of Internal Medicine BWW University MünsterMünsterGermany
| | - Robert Schierwagen
- Department of Internal Medicine IGoethe University Clinic FrankfurtFrankfurtGermany
- Department of Internal Medicine BWW University MünsterMünsterGermany
| | - Jonel Trebicka
- Department of Internal Medicine IGoethe University Clinic FrankfurtFrankfurtGermany
- Department of Internal Medicine BWW University MünsterMünsterGermany
- European Foundation for the Study of Chronic Failure, EFCLIFBarcelonaSpain
| | - Matthias Mann
- Novo Nordisk Foundation Center for Protein ResearchFaculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
- Department of Proteomics and Signal TransductionMax Planck Institute of BiochemistryMartinsriedGermany
| |
Collapse
|
42
|
Niu Y, Jiang H, Yin H, Wang F, Hu R, Hu X, Peng B, Shu Y, Li Z, Chen S, Guo F. Hepatokine ERAP1 Disturbs Skeletal Muscle Insulin Sensitivity Via Inhibiting USP33-Mediated ADRB2 Deubiquitination. Diabetes 2022; 71:921-933. [PMID: 35192681 DOI: 10.2337/db21-0857] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 02/15/2022] [Indexed: 11/13/2022]
Abstract
Chronic inflammation in liver induces insulin resistance systemically and in other tissues, including the skeletal muscle (SM); however, the underlying mechanisms remain largely unknown. RNA sequencing of primary hepatocytes from wild-type mice fed long-term high-fat diet (HFD), which have severe chronic inflammation and insulin resistance revealed that the expression of hepatokine endoplasmic reticulum aminopeptidase 1 (ERAP1) was upregulated by a HFD. Increased ERAP1 levels were also observed in interferon-γ-treated primary hepatocytes. Furthermore, hepatic ERAP1 overexpression attenuated systemic and SM insulin sensitivity, whereas hepatic ERAP1 knockdown had the opposite effects, with corresponding changes in serum ERAP1 levels. Mechanistically, ERAP1 functions as an antagonist-like factor, which interacts with β2 adrenergic receptor (ADRB2) and reduces its expression by decreasing ubiquitin-specific peptidase 33-mediated deubiquitination and thereby interrupts ADRB2-stimulated insulin signaling in the SM. The findings of this study indicate ERAP1 is an inflammation-induced hepatokine that impairs SM insulin sensitivity. Its inhibition may provide a therapeutic strategy for insulin resistance-related diseases, such as type 2 diabetes.
Collapse
Affiliation(s)
- Yuguo Niu
- Zhongshan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai, China
- Chinese Academy of Sciences (CAS) Key Laboratory of Nutrition, Metabolism and Food Safety, Innovation Center for Intervention of Chronic Disease and Promotion of Health, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Haizhou Jiang
- Chinese Academy of Sciences (CAS) Key Laboratory of Nutrition, Metabolism and Food Safety, Innovation Center for Intervention of Chronic Disease and Promotion of Health, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Hanrui Yin
- Chinese Academy of Sciences (CAS) Key Laboratory of Nutrition, Metabolism and Food Safety, Innovation Center for Intervention of Chronic Disease and Promotion of Health, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Fenfen Wang
- Chinese Academy of Sciences (CAS) Key Laboratory of Nutrition, Metabolism and Food Safety, Innovation Center for Intervention of Chronic Disease and Promotion of Health, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Ronggui Hu
- Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Xiaoming Hu
- Zhongshan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Bo Peng
- Zhongshan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Yousheng Shu
- Zhongshan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Zhigang Li
- Chinese Academy of Sciences (CAS) Key Laboratory of Nutrition, Metabolism and Food Safety, Innovation Center for Intervention of Chronic Disease and Promotion of Health, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Shanghai Chen
- Zhongshan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Feifan Guo
- Zhongshan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai, China
- Chinese Academy of Sciences (CAS) Key Laboratory of Nutrition, Metabolism and Food Safety, Innovation Center for Intervention of Chronic Disease and Promotion of Health, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
43
|
An H, Qian C, Huang Y, Li J, Tian X, Feng J, Hu J, Fang Y, Jiao F, Zeng Y, Huang X, Meng X, Liu X, Lin X, Zeng Z, Guilliams M, Beschin A, Chen Y, Wu Y, Wang J, Oggioni MR, Leong J, Veening JW, Deng H, Zhang R, Wang H, Wu J, Cui Y, Zhang JR. Functional vulnerability of liver macrophages to capsules defines virulence of blood-borne bacteria. J Exp Med 2022; 219:e20212032. [PMID: 35258552 PMCID: PMC8908791 DOI: 10.1084/jem.20212032] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 12/22/2021] [Accepted: 01/24/2022] [Indexed: 12/12/2022] Open
Abstract
Many encapsulated bacteria use capsules to cause invasive diseases. However, it remains largely unknown how the capsules enhance bacterial virulence under in vivo infection conditions. Here we show that the capsules primarily target the liver to enhance bacterial survival at the onset of blood-borne infections. In a mouse sepsis model, the capsules enabled human pathogens Streptococcus pneumoniae and Escherichia coli to circumvent the recognition of liver-resident macrophage Kupffer cells (KCs) in a capsular serotype-dependent manner. In contrast to effective capture of acapsular bacteria by KCs, the encapsulated bacteria are partially (low-virulence types) or completely (high-virulence types) "untouchable" for KCs. We finally identified the asialoglycoprotein receptor (ASGR) as the first known capsule receptor on KCs to recognize the low-virulence serotype-7F and -14 pneumococcal capsules. Our data identify the molecular interplay between the capsules and KCs as a master controller of the fate and virulence of encapsulated bacteria, and suggest that the interplay is targetable for therapeutic control of septic infections.
Collapse
Affiliation(s)
- Haoran An
- Center for Infectious Disease Research, Department of Basic Medical Science, School of Medicine, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Chenyun Qian
- Center for Infectious Disease Research, Department of Basic Medical Science, School of Medicine, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Yijia Huang
- Center for Infectious Disease Research, Department of Basic Medical Science, School of Medicine, Tsinghua University, Beijing, China
| | - Jing Li
- Center for Infectious Disease Research, Department of Basic Medical Science, School of Medicine, Tsinghua University, Beijing, China
| | - Xianbin Tian
- Center for Infectious Disease Research, Department of Basic Medical Science, School of Medicine, Tsinghua University, Beijing, China
| | - Jiaying Feng
- Center for Infectious Disease Research, Department of Basic Medical Science, School of Medicine, Tsinghua University, Beijing, China
| | - Jiao Hu
- Center for Infectious Disease Research, Department of Basic Medical Science, School of Medicine, Tsinghua University, Beijing, China
| | - Yujie Fang
- Center for Infectious Disease Research, Department of Basic Medical Science, School of Medicine, Tsinghua University, Beijing, China
| | - Fangfang Jiao
- Center for Infectious Disease Research, Department of Basic Medical Science, School of Medicine, Tsinghua University, Beijing, China
| | - Yuna Zeng
- Center for Infectious Disease Research, Department of Basic Medical Science, School of Medicine, Tsinghua University, Beijing, China
| | - Xueting Huang
- Center for Infectious Disease Research, Department of Basic Medical Science, School of Medicine, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Xianbin Meng
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Xue Liu
- Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Xin Lin
- Center for Infectious Disease Research, Department of Basic Medical Science, School of Medicine, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Zhutian Zeng
- School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Martin Guilliams
- Laboratory of Myeloid Cell Biology in Tissue Homeostasis and Regeneration, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Faculty of Science, Ghent University, Ghent, Belgium
| | - Alain Beschin
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
- Laboratory of Cellular and Molecular Immunology, Vrije University Brussel, Brussels, Belgium
| | - Yongwen Chen
- Institute of Immunology, Third Military Medical University, Chongqing, China
| | - Yuzhang Wu
- Institute of Immunology, Third Military Medical University, Chongqing, China
| | - Jing Wang
- Shanghai Institute of Immunology, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | | | - John Leong
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA
| | - Jan-Willem Veening
- Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Haiteng Deng
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Rong Zhang
- The Second Affiliated Hospital of Zhejiang University, Zhejiang University, Hangzhou, China
| | - Hui Wang
- Department of Clinical Laboratory, Peking University People’s Hospital, Beijing, China
| | - Jiang Wu
- Beijing Center for Disease Control and Prevention, Beijing, China
| | - Yan Cui
- Department of General Surgery, Strategic Support Force Medical Center, Beijing, China
| | - Jing-Ren Zhang
- Center for Infectious Disease Research, Department of Basic Medical Science, School of Medicine, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| |
Collapse
|
44
|
Hemojuvelin deficiency promotes liver mitochondrial dysfunction and predisposes mice to hepatocellular carcinoma. Commun Biol 2022; 5:153. [PMID: 35194137 PMCID: PMC8863832 DOI: 10.1038/s42003-022-03108-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 02/01/2022] [Indexed: 11/11/2022] Open
Abstract
Hemojuvelin (HJV) enhances signaling to the iron hormone hepcidin and its deficiency causes iron overload, a risk factor for hepatocellular carcinoma (HCC). We utilized Hjv−/− mice to dissect mechanisms for hepatocarcinogenesis. We show that suboptimal treatment with diethylnitrosamine (DEN) triggers HCC only in Hjv−/− but not wt mice. Liver proteomics data were obtained by mass spectrometry. Hierarchical clustering analysis revealed that Hjv deficiency and DEN elicit similar liver proteomic responses, including induction of mitochondrial proteins. Dietary iron overload of wt mice does not recapitulate the liver proteomic phenotype of Hjv−/− animals, which is only partially corrected by iron depletion. Consistent with these data, primary Hjv−/− hepatocytes exhibit mitochondrial hyperactivity, while aged Hjv−/− mice develop spontaneous HCC. Moreover, low expression of HJV or hepcidin (HAMP) mRNAs predicts poor prognosis in HCC patients. We conclude that Hjv has a hepatoprotective function and its deficiency in mice promotes mitochondrial dysfunction and hepatocarcinogenesis. Hemojuvelin (HJV), a BMP co-receptor promoting hepcidin expression in the liver, has a hepatoprotective function and its deficiency in mice triggers mitochondrial dysfunction and hepatocarcinogenesis.
Collapse
|
45
|
cRGD enables rapid phagocytosis of liposomal vancomycin for intracellular bacterial clearance. J Control Release 2022; 344:202-213. [DOI: 10.1016/j.jconrel.2022.02.030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 02/18/2022] [Accepted: 02/23/2022] [Indexed: 12/24/2022]
|
46
|
Xue WY, Zhang L, Liu CM, Gao Y, Li SJ, Huai ZY, Dai J, Wang YY. Research progress on the relationship between TM6SF2 rs58542926 polymorphism and non-alcoholic fatty liver disease. Expert Rev Gastroenterol Hepatol 2022; 16:97-107. [PMID: 35057689 DOI: 10.1080/17474124.2022.2032661] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION nonalcoholic fatty liver disease is a common liver disease with a global average prevalence of about 25%. In addition to the incidence of NAFLD being related to obesity, diabetes, hyperlipidemia, etc., genetic factors also have an important impact on the incidence of NAFLD. AREAS COVERED Current experimental results and clinical studies show that the transmembrane 6 superfamily member 2 (TM6SF2) gene plays an important role in the pathogenesis of NAFLD. The research on genetic polymorphism of TM6SF2 gene mainly focuses on rs58542926 locus (rs58542926 c.449 C > T, p. Glu167Lys, E167K). The Mutations of this site might increase the risk of NAFLD in carriers. EXPERT OPINION The mutation of this site causes the disorder of triglyceride metabolism in the liver, which leads to the deposition of a large amount of lipids in the liver, and further induces the incidence of NAFLD. With the study of the mechanism of TM6SF2 gene polymorphism in the pathogenesis of NAFLD, it is helpful to understand the molecular mechanism of the pathogenesis of NAFLD, which has a great value for the treatment of NAFLD.
Collapse
Affiliation(s)
- Wan-Ying Xue
- School of Life Sciences, Bengbu Medical College, Bengbu, Anhui, China
| | - Li Zhang
- Department of Infectious Diseases, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Chuan-Miao Liu
- Department of Infectious Diseases, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Yu Gao
- School of Life Sciences, Bengbu Medical College, Bengbu, Anhui, China
| | - Shu-Jing Li
- School of Life Sciences, Bengbu Medical College, Bengbu, Anhui, China
| | - Zi-You Huai
- School of Life Sciences, Bengbu Medical College, Bengbu, Anhui, China
| | - Jing Dai
- School of Life Sciences, Bengbu Medical College, Bengbu, Anhui, China
| | - Yuan-Yuan Wang
- School of Life Sciences, Bengbu Medical College, Bengbu, Anhui, China
| |
Collapse
|
47
|
Xie B, Shi X, Li Y, Xia B, Zhou J, Du M, Xing X, Bai L, Liu E, Alvarez F, Jin L, Deng S, Mitchell GA, Pan D, Li M, Wu J. Deficiency of ASGR1 in pigs recapitulates reduced risk factor for cardiovascular disease in humans. PLoS Genet 2021; 17:e1009891. [PMID: 34762653 PMCID: PMC8584755 DOI: 10.1371/journal.pgen.1009891] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 10/18/2021] [Indexed: 11/18/2022] Open
Abstract
Genetic variants in the asialoglycoprotein receptor 1 (ASGR1) are associated with a reduced risk of cardiovascular disease (CVD) in humans. However, the underlying molecular mechanism remains elusive. Given the cardiovascular similarities between pigs and humans, we generated ASGR1-deficient pigs using the CRISPR/Cas9 system. These pigs show age-dependent low levels of non-HDL-C under standard diet. When received an atherogenic diet for 6 months, ASGR1-deficient pigs show lower levels of non-HDL-C and less atherosclerotic lesions than that of controls. Furthermore, by analysis of hepatic transcriptome and in vivo cholesterol metabolism, we show that ASGR1 deficiency reduces hepatic de novo cholesterol synthesis by downregulating 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR), and increases cholesterol clearance by upregulating the hepatic low-density lipoprotein receptor (LDLR), which together contribute to the low levels of non-HDL-C. Despite the cardioprotective effect, we unexpectedly observed mild to moderate hepatic injury in ASGR1-deficient pigs, which has not been documented in humans with ASGR1 variants. Thus, targeting ASGR1 might be an effective strategy to reduce hypercholesterolemia and atherosclerosis, whereas further clinical evidence is required to assess its hepatic impact. Previous studies have reported an association between ASGR1 variants and CVD in humans. However, the underlying mechanism is unknown. We used ASGR1-deficient pig to recapitulate the reduced risk features of CVD in humans with ASGR1 variants, indicating that ASGR1 inhibition could be an effective strategy to treat atherosclerotic CVD. Our results highlight the demand for taking advantage of genetically modified large animal models to investigate the pathogenesis and therapeutic development of CVD in humans. Unexpectedly, we demonstrate the first link between ASGR1 deficiency and liver injury, a feature that has not been documented in humans with ASGR1 variants. These results suggest that ASGR1 might be an effective target for reducing CVD, whereas revealing a genetic predisposition to liver disease in humans with ASGR1 variants.
Collapse
Affiliation(s)
- Baocai Xie
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Xiaochen Shi
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Yan Li
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Bo Xia
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Jia Zhou
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, Chengdu, Sichuan, China
| | - Minjie Du
- Chengdu Clonorgan Biotechnology Co. LTD, Chengdu, Sichuan, China
| | - Xiangyang Xing
- Chengdu Clonorgan Biotechnology Co. LTD, Chengdu, Sichuan, China
| | - Liang Bai
- Institute of Cardiovascular Sciences, Health Science Center, Xi’an Jiao Tong University, Xi’an, Shaanxi, China
| | - Enqi Liu
- Institute of Cardiovascular Sciences, Health Science Center, Xi’an Jiao Tong University, Xi’an, Shaanxi, China
| | - Fernando Alvarez
- Divisions of Gastroenterology, Hepatology and Nurition, University of Montreal and Centre Hospitalier Universitaire Sainte-Justine, Montreal, Quebec, Canada
| | - Long Jin
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Shaoping Deng
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, Chengdu, Sichuan, China
| | - Grant A. Mitchell
- Divisions of Medical Genetics, Department of Pediatrics, University of Montreal and Centre Hospitalier Universitaire Sainte-Justine, Montreal, Quebec, Canada
| | - Dengke Pan
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, Chengdu, Sichuan, China
- * E-mail: (DP); (ML); (JW)
| | - Mingzhou Li
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
- * E-mail: (DP); (ML); (JW)
| | - Jiangwei Wu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
- * E-mail: (DP); (ML); (JW)
| |
Collapse
|
48
|
Cunningham RP, Moore MP, Dashek RJ, Meers GM, Takahashi T, Sheldon RD, Wheeler AA, Diaz-Arias A, Ibdah JA, Parks EJ, Thyfault JP, Rector RS. Critical Role for Hepatocyte-Specific eNOS in NAFLD and NASH. Diabetes 2021; 70:2476-2491. [PMID: 34380696 PMCID: PMC8564406 DOI: 10.2337/db20-1228] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 07/30/2021] [Indexed: 11/13/2022]
Abstract
Regulation of endothelial nitric oxide synthase (eNOS) in hepatocytes may be an important target in nonalcoholic fatty liver disease (NAFLD) development and progression to nonalcoholic steatohepatitis (NASH). In this study, we show genetic deletion and viral knockdown of hepatocyte-specific eNOS exacerbated hepatic steatosis and inflammation, decreased hepatic mitochondrial fatty acid oxidation and respiration, increased mitochondrial H2O2 emission, and impaired the hepatic mitophagic (BNIP3 and LC3II) response. Conversely, overexpressing eNOS in hepatocytes in vitro and in vivo increased hepatocyte mitochondrial respiration and attenuated Western diet-induced NASH. Moreover, patients with elevated NAFLD activity score (histology score of worsening steatosis, hepatocyte ballooning, and inflammation) exhibited reduced hepatic eNOS expression, which correlated with reduced hepatic mitochondrial fatty acid oxidation and lower hepatic protein expression of mitophagy protein BNIP3. The current study reveals an important molecular role for hepatocyte-specific eNOS as a key regulator of NAFLD/NASH susceptibility and mitochondrial quality control with direct clinical correlation to patients with NASH.
Collapse
Affiliation(s)
- Rory P Cunningham
- Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO
| | - Mary P Moore
- Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO
| | - Ryan J Dashek
- Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO
- Comparative Medicine Program, University of Missouri, Columbia, MO
| | - Grace M Meers
- Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO
| | - Takamune Takahashi
- Division of Nephrology and Hypertension, Vanderbilt University School of Medicine, Nashville, TN
| | - Ryan D Sheldon
- Metabolic and Nutritional Programming, Center for Cancer and Cell Biology, Van Andel Institute, Grand Rapids, MI
| | | | | | - Jamal A Ibdah
- Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, MO
| | - Elizabeth J Parks
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, MO
| | - John P Thyfault
- Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS
- Kansas City VA Medical Center, Kansas City, MO
| | - R Scott Rector
- Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, MO
| |
Collapse
|
49
|
McGuire MR, Mukhopadhyay D, Myers SL, Mosher EP, Brookheart RT, Kammers K, Sehgal A, Selen ES, Wolfgang MJ, Bumpus NN, Espenshade PJ. Progesterone receptor membrane component 1 (PGRMC1) binds and stabilizes cytochromes P450 through a heme-independent mechanism. J Biol Chem 2021; 297:101316. [PMID: 34678314 PMCID: PMC8591507 DOI: 10.1016/j.jbc.2021.101316] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 10/12/2021] [Accepted: 10/18/2021] [Indexed: 12/03/2022] Open
Abstract
Progesterone receptor membrane component 1 (PGRMC1) is a heme-binding protein implicated in a wide range of cellular functions. We previously showed that PGRMC1 binds to cytochromes P450 in yeast and mammalian cells and supports their activity. Recently, the paralog PGRMC2 was shown to function as a heme chaperone. The extent of PGRMC1 function in cytochrome P450 biology and whether PGRMC1 is also a heme chaperone are unknown. Here, we examined the function of Pgrmc1 in mouse liver using a knockout model and found that Pgrmc1 binds and stabilizes a broad range of cytochromes P450 in a heme-independent manner. Proteomic and transcriptomic studies demonstrated that Pgrmc1 binds more than 13 cytochromes P450 and supports maintenance of cytochrome P450 protein levels posttranscriptionally. In vitro assays confirmed that Pgrmc1 KO livers exhibit reduced cytochrome P450 activity consistent with reduced enzyme levels. Mechanistic studies in cultured cells demonstrated that PGRMC1 stabilizes cytochromes P450 and that binding and stabilization do not require PGRMC1 binding to heme. Importantly, Pgrmc1-dependent stabilization of cytochromes P450 is physiologically relevant, as Pgrmc1 deletion protected mice from acetaminophen-induced liver injury. Finally, evaluation of Y113F mutant Pgrmc1, which lacks the axial heme iron-coordinating hydroxyl group, revealed that proper iron coordination is not required for heme binding, but is required for binding to ferrochelatase, the final enzyme in heme biosynthesis. PGRMC1 was recently identified as the causative mutation in X-linked isolated pediatric cataract formation. Together, these results demonstrate a heme-independent function for PGRMC1 in cytochrome P450 stability that may underlie clinical phenotypes.
Collapse
Affiliation(s)
- Meredith R McGuire
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Debaditya Mukhopadhyay
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Stephanie L Myers
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Eric P Mosher
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Rita T Brookheart
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kai Kammers
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Alfica Sehgal
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ebru S Selen
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Michael J Wolfgang
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Namandjé N Bumpus
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Peter J Espenshade
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
50
|
Wang VE, Blaser BW, Patel RK, Behbehani GK, Rao AA, Durbin-Johnson B, Jiang T, Logan AC, Settles M, Mannis GN, Olin R, Damon LE, Martin TG, Sayre PH, Gaensler KM, McMahon E, Flanders M, Weinberg V, Ye CJ, Carbone DP, Munster PN, Fragiadakis GK, McCormick F, Andreadis C. Inhibition of MET Signaling with Ficlatuzumab in Combination with Chemotherapy in Refractory AML: Clinical Outcomes and High-Dimensional Analysis. Blood Cancer Discov 2021; 2:434-449. [PMID: 34514432 DOI: 10.1158/2643-3230.bcd-21-0055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Acute myeloid leukemia patients refractory to induction therapy or relapsed within one year have poor outcomes. Autocrine production of hepatocyte growth factor by myeloid blasts drives leukemogenesis in pre-clinical models. A phase Ib trial evaluated ficlatuzumab, a first-in-class anti-HGF antibody, in combination with cytarabine in this high-risk population. Dose-limiting toxicities were not observed, and 20 mg/kg was established as the recommended phase II dose. The most frequent treatment-related adverse event was febrile neutropenia. Among 17 evaluable patients, the overall response rate was 53%, all complete remissions. Phospho-proteomic mass cytometry showed potent on-target suppression of p-MET after ficlatuzumab treatment and that attenuation of p-S6 was associated with clinical response. Multiplexed single cell RNA sequencing using prospectively acquired patient specimens identified interferon response genes as adverse predictive factors. The ficlatuzumab and cytarabine combination is well-tolerated with favorable efficacy. High-dimensional analyses at single-cell resolution represent promising approaches for identifying biomarkers of response and mechanisms of resistance in prospective clinical studies.
Collapse
Affiliation(s)
- Victoria E Wang
- Department of Medicine, University of California, San Francisco, CA 94158, USA.,Helen Diller Comprehensive Cancer Center, University of California, San Francisco, CA 94158, USA
| | - Bradley W Blaser
- The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Ravi K Patel
- CoLabs, University of California, San Francisco, CA 94143, USA
| | - Gregory K Behbehani
- The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Arjun A Rao
- CoLabs, University of California, San Francisco, CA 94143, USA
| | | | - Tommy Jiang
- Department of Medicine, University of California, San Francisco, CA 94158, USA.,Helen Diller Comprehensive Cancer Center, University of California, San Francisco, CA 94158, USA
| | - Aaron C Logan
- Department of Medicine, University of California, San Francisco, CA 94158, USA.,Helen Diller Comprehensive Cancer Center, University of California, San Francisco, CA 94158, USA
| | - Matthew Settles
- Bioinformatics Core, Genome Center, University of California, Davis, CA 95616, USA
| | - Gabriel N Mannis
- Department of Medicine, University of California, San Francisco, CA 94158, USA
| | - Rebecca Olin
- Department of Medicine, University of California, San Francisco, CA 94158, USA.,Helen Diller Comprehensive Cancer Center, University of California, San Francisco, CA 94158, USA
| | - Lloyd E Damon
- Department of Medicine, University of California, San Francisco, CA 94158, USA.,Helen Diller Comprehensive Cancer Center, University of California, San Francisco, CA 94158, USA
| | - Thomas G Martin
- Department of Medicine, University of California, San Francisco, CA 94158, USA.,Helen Diller Comprehensive Cancer Center, University of California, San Francisco, CA 94158, USA
| | - Peter H Sayre
- Department of Medicine, University of California, San Francisco, CA 94158, USA.,Helen Diller Comprehensive Cancer Center, University of California, San Francisco, CA 94158, USA
| | - Karin M Gaensler
- Department of Medicine, University of California, San Francisco, CA 94158, USA.,Helen Diller Comprehensive Cancer Center, University of California, San Francisco, CA 94158, USA
| | - Emma McMahon
- Department of Medicine, University of California, San Francisco, CA 94158, USA.,Helen Diller Comprehensive Cancer Center, University of California, San Francisco, CA 94158, USA
| | - Michael Flanders
- Department of Medicine, University of California, San Francisco, CA 94158, USA.,Helen Diller Comprehensive Cancer Center, University of California, San Francisco, CA 94158, USA
| | - Vivian Weinberg
- Department of Medicine, University of California, San Francisco, CA 94158, USA
| | - Chun J Ye
- Department of Medicine, University of California, San Francisco, CA 94158, USA
| | - David P Carbone
- The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Pamela N Munster
- Department of Medicine, University of California, San Francisco, CA 94158, USA.,Helen Diller Comprehensive Cancer Center, University of California, San Francisco, CA 94158, USA
| | - Gabriela K Fragiadakis
- CoLabs, University of California, San Francisco, CA 94143, USA.,Bakar ImmunoX Initiative, University of California, San Francisco, CA 94143, USA.,Department of Medicine, Division of Rheumatology, University of California, San Francisco, CA 94143, USA
| | - Frank McCormick
- Helen Diller Comprehensive Cancer Center, University of California, San Francisco, CA 94158, USA
| | - Charalambos Andreadis
- Department of Medicine, University of California, San Francisco, CA 94158, USA.,Helen Diller Comprehensive Cancer Center, University of California, San Francisco, CA 94158, USA
| |
Collapse
|