1
|
Néel E, Chiritoiu-Butnaru M, Fargues W, Denus M, Colladant M, Filaquier A, Stewart SE, Lehmann S, Zurzolo C, Rubinsztein DC, Marin P, Parmentier ML, Villeneuve J. The endolysosomal system in conventional and unconventional protein secretion. J Cell Biol 2024; 223:e202404152. [PMID: 39133205 PMCID: PMC11318669 DOI: 10.1083/jcb.202404152] [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: 05/02/2024] [Revised: 07/12/2024] [Accepted: 07/26/2024] [Indexed: 08/13/2024] Open
Abstract
Most secreted proteins are transported through the "conventional" endoplasmic reticulum-Golgi apparatus exocytic route for their delivery to the cell surface and release into the extracellular space. Nonetheless, formative discoveries have underscored the existence of alternative or "unconventional" secretory routes, which play a crucial role in exporting a diverse array of cytosolic proteins outside the cell in response to intrinsic demands, external cues, and environmental changes. In this context, lysosomes emerge as dynamic organelles positioned at the crossroads of multiple intracellular trafficking pathways, endowed with the capacity to fuse with the plasma membrane and recognized for their key role in both conventional and unconventional protein secretion. The recent recognition of lysosomal transport and exocytosis in the unconventional secretion of cargo proteins provides new and promising insights into our understanding of numerous physiological processes.
Collapse
Affiliation(s)
- Eloïse Néel
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM , Montpellier, France
| | | | - William Fargues
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM , Montpellier, France
| | - Morgane Denus
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM , Montpellier, France
| | - Maëlle Colladant
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM , Montpellier, France
| | - Aurore Filaquier
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM , Montpellier, France
| | - Sarah E Stewart
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Australia
| | - Sylvain Lehmann
- Laboratoire de Biochimie-Protéomique Clinique-Plateforme de Protéomique Clinique, Université de Montpellier, Institute for Regenerative Medicine and Biotherapy Centre Hospitalier Universitaire de Montpellier, Institute for Neurosciences of Montpellier INSERM , Montpellier, France
| | - Chiara Zurzolo
- Unité de Trafic Membranaire et Pathogenèse, Institut Pasteur, UMR3691 CNRS , Paris, France
| | - David C Rubinsztein
- Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
- UK Dementia Research Institute , Cambridge, UK
| | - Philippe Marin
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM , Montpellier, France
| | - Marie-Laure Parmentier
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM , Montpellier, France
| | - Julien Villeneuve
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM , Montpellier, France
| |
Collapse
|
2
|
Sun Y, Tao X, Han Y, Lin X, Tian R, Wang H, Chang P, Sun Q, Ge L, Zhang M. A dual role of ERGIC-localized Rabs in TMED10-mediated unconventional protein secretion. Nat Cell Biol 2024; 26:1077-1092. [PMID: 38926505 DOI: 10.1038/s41556-024-01445-4] [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: 11/22/2023] [Accepted: 05/23/2024] [Indexed: 06/28/2024]
Abstract
Cargo translocation across membranes is a crucial aspect of secretion. In conventional secretion signal peptide-equipped proteins enter the endoplasmic reticulum (ER), whereas a subset of cargo lacking signal peptides translocate into the ER-Golgi intermediate compartment (ERGIC) in a process called unconventional protein secretion (UcPS). The regulatory events at the ERGIC in UcPS are unclear. Here we reveal the involvement of ERGIC-localized small GTPases, Rab1 (Rab1A and Rab1B) and Rab2A, in regulating UcPS cargo transport via TMED10 on the ERGIC. Rab1 enhances TMED10 translocator activity, promoting cargo translocation into the ERGIC, whereas Rab2A, in collaboration with KIF5B, regulates ERGIC compartmentalization, establishing a UcPS-specific compartment. This study highlights the pivotal role of ERGIC-localized Rabs in governing cargo translocation and specifying the ERGIC's function in UcPS.
Collapse
Affiliation(s)
- Yuxin Sun
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, China
| | - Xuan Tao
- State Key Laboratory of Membrane Biology, School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Yaping Han
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, China
| | - Xubo Lin
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, Beihang University, Beijing, China
| | - Rui Tian
- Department of Biochemistry and Department of Cardiology of Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Haodong Wang
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, China
| | - Pei Chang
- State Key Laboratory of Membrane Biology, School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Qiming Sun
- Department of Biochemistry and Department of Cardiology of Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Liang Ge
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, China.
| | - Min Zhang
- State Key Laboratory of Membrane Biology, School of Pharmaceutical Sciences, Tsinghua University, Beijing, China.
| |
Collapse
|
3
|
Yuan SHC, Wu CC, Wang YC, Chan XY, Chu HW, Yang Y, Liu HP. AGR2-mediated unconventional secretion of 14-3-3ε and α-actinin-4, responsive to ER stress and autophagy, drives chemotaxis in canine mammary tumor cells. Cell Mol Biol Lett 2024; 29:84. [PMID: 38822246 PMCID: PMC11140979 DOI: 10.1186/s11658-024-00601-w] [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: 12/25/2023] [Accepted: 05/21/2024] [Indexed: 06/02/2024] Open
Abstract
BACKGROUND Canine mammary tumors (CMTs) in intact female dogs provide a natural model for investigating metastatic human cancers. Our prior research identified elevated expression of Anterior Gradient 2 (AGR2), a protein disulfide isomerase (PDI) primarily found in the endoplasmic reticulum (ER), in CMT tissues, highly associated with CMT progression. We further demonstrated that increased AGR2 expression actively influences the extracellular microenvironment, promoting chemotaxis in CMT cells. Unraveling the underlying mechanisms is crucial for assessing the potential of therapeutically targeting AGR2 as a strategy to inhibit a pro-metastatic microenvironment and impede tumor metastasis. METHODS To identify the AGR2-modulated secretome, we employed proteomics analysis of the conditioned media (CM) from two CMT cell lines ectopically expressing AGR2, compared with corresponding vector-expressing controls. AGR2-regulated release of 14-3-3ε (gene: YWHAE) and α-actinin 4 (gene: ACTN4) was validated through ectopic expression, knockdown, and knockout of the AGR2 gene in CMT cells. Extracellular vesicles derived from CMT cells were isolated using either differential ultracentrifugation or size exclusion chromatography. The roles of 14-3-3ε and α-actinin 4 in the chemotaxis driven by the AGR2-modulated CM were investigated through gene knockdown, antibody-mediated interference, and recombinant protein supplement. Furthermore, the clinical relevance of the release of 14-3-3ε and α-actinin 4 was assessed using CMT tissue-immersed saline and sera from CMT-afflicted dogs. RESULTS Proteomics analysis of the AGR2-modulated secretome revealed increased abundance in 14-3-3ε and α-actinin 4. Ectopic expression of AGR2 significantly increased the release of 14-3-3ε and α-actinin 4 in the CM. Conversely, knockdown or knockout of AGR2 expression remarkably reduced their release. Silencing 14-3-3ε or α-actinin 4 expression diminished the chemotaxis driven by AGR2-modulated CM. Furthermore, AGR2 controls the release of 14-3-3ε and α-actinin 4 primarily via non-vesicular routes, responding to the endoplasmic reticulum (ER) stress and autophagy activation. Knockout of AGR2 resulted in increased α-actinin 4 accumulation and impaired 14-3-3ε translocation in autophagosomes. Depletion of extracellular 14-3-3ε or α-actinin 4 reduced the chemotaxis driven by AGR2-modulated CM, whereas supplement with recombinant 14-3-3ε in the CM enhanced the CM-driven chemotaxis. Notably, elevated levels of 14-3-3ε or α-actinin 4 were observed in CMT tissue-immersed saline compared with paired non-tumor samples and in the sera of CMT dogs compared with healthy dogs. CONCLUSION This study elucidates AGR2's pivotal role in orchestrating unconventional secretion of 14-3-3ε and α-actinin 4 from CMT cells, thereby contributing to paracrine-mediated chemotaxis. The insight into the intricate interplay between AGR2-involved ER stress, autophagy, and unconventional secretion provides a foundation for refining strategies aimed at impeding metastasis in both canine mammary tumors and potentially human cancers.
Collapse
Affiliation(s)
- Stephen Hsien-Chi Yuan
- Department of Veterinary Medicine, College of Veterinary Medicine, National Chung Hsing University, Taichung, 40227, Taiwan
| | - Chih-Ching Wu
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Otolaryngology-Head and Neck Surgery, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Molecular Medicine Research Center, Chang Gung University, Taoyuan, Taiwan
- Department of Medical Biotechnology and Laboratory Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yu-Chih Wang
- Graduate Institute of Veterinary Pathology, College of Veterinary Medicine, National Chung Hsing University, Taichung, 40227, Taiwan
| | - Xiu-Ya Chan
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Hao-Wei Chu
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Youngsen Yang
- Department of Oncology, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Hao-Ping Liu
- Department of Veterinary Medicine, College of Veterinary Medicine, National Chung Hsing University, Taichung, 40227, Taiwan.
- Biotechnology Center, National Chung Hsing University, Taichung, 40227, Taiwan.
| |
Collapse
|
4
|
Zheng N, Liu S, Chen J, Xu Y, Cao W, Lin J, Lu G, Zhang G. SARS-CoV-2 NSP2 as a Potential Delivery Vehicle for Proteins. Mol Pharm 2024; 21:1149-1159. [PMID: 38288708 DOI: 10.1021/acs.molpharmaceut.3c00680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2024]
Abstract
The development of biomolecule delivery systems is essential for the treatment of various diseases such as cancer, immunological diseases, and metabolic disorders. For the first time, we found that SARS-CoV-2-encoded nonstructural protein 2 (NSP2) can be secreted from the cells, where it is synthesized. Brefeldin A and H89, inhibitors of ER/Golgi secretion pathways, did not inhibit NSP2 secretion. NSP2 is likely secreted via an unconventional secretory pathway. Moreover, both secreted and purified NSP2 proteins were able to traverse the plasma membrane barrier and enter both immortalized human umbilical vein endothelial cells and tumor cell lines. After entry, the NSP2 protein was localized in only the cytoplasm. Cytochalasin D, a potent inhibitor of actin polymerization, inhibited the entry of NSP2. NSP2 can carry other molecules into cells. Burkholderia lethal factor 1, a monomeric toxin from the intracellular pathogen Burkholderia pseudomallei, has demonstrated antitumor activity by targeting host eukaryotic initiation translation factor 4A. An NSP2-BLF1 fusion protein was translocated across the cellular membranes of Huh7 cells and mediated cell killing. By using different approaches, including protein purification, chemical inhibition, and cell imaging, we confirm that NSP2 is able to deliver heterologous proteins into cells. NSP2 can act as a potential delivery vehicle for proteins.
Collapse
Affiliation(s)
- Ningze Zheng
- Institute of Human Virology, Department of Pathogen Biology and Biosecurity, and Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Shurui Liu
- Institute of Human Virology, Department of Pathogen Biology and Biosecurity, and Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Jianheng Chen
- Institute of Human Virology, Department of Pathogen Biology and Biosecurity, and Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Yue Xu
- Institute of Human Virology, Department of Pathogen Biology and Biosecurity, and Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Wenyin Cao
- Institute of Human Virology, Department of Pathogen Biology and Biosecurity, and Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Jinyi Lin
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Guang Lu
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Guigen Zhang
- Institute of Human Virology, Department of Pathogen Biology and Biosecurity, and Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| |
Collapse
|
5
|
Denus M, Fargues W, Filaquier A, Néel É, Marin P, Parmentier ML, Villeneuve J. [Unconventional protein secretion - new perspectives in protein trafficking]. Med Sci (Paris) 2024; 40:267-274. [PMID: 38520102 DOI: 10.1051/medsci/2024013] [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] [Indexed: 03/25/2024] Open
Abstract
The characterization of the structural and functional organization of eukaryotic cells has revealed the membrane compartments and machinery required for vesicular protein transport. Most proteins essential for intercellular communication contain an N-terminal signal sequence enabling them to be incorporated into the biosynthetic or conventional secretory pathway, in which proteins are sequentially transported through the endoplasmic reticulum (ER) and the Golgi apparatus. However, major research studies have shown the existence of alternative secretory routes that are independent of the ER-Golgi and designated as unconventional secretory pathways. These pathways involve a large number of players that may divert specific compartments from their primary function in favor of secretory roles. The comprehensive description of these processes is therefore of utmost importance to unveil how proteins secreted through these alternative pathways control cell homeostasis or contribute to disease development.
Collapse
Affiliation(s)
- Morgane Denus
- Institut de génomique fonctionnelle,Université de Montpellier, CNRS UMR 5 203. Inserm U1191, Montpellier, France
| | - William Fargues
- Institut de génomique fonctionnelle,Université de Montpellier, CNRS UMR 5 203. Inserm U1191, Montpellier, France
| | - Aurore Filaquier
- Institut de génomique fonctionnelle,Université de Montpellier, CNRS UMR 5 203. Inserm U1191, Montpellier, France
| | - Éloïse Néel
- Institut de génomique fonctionnelle,Université de Montpellier, CNRS UMR 5 203. Inserm U1191, Montpellier, France
| | - Philippe Marin
- Institut de génomique fonctionnelle,Université de Montpellier, CNRS UMR 5 203. Inserm U1191, Montpellier, France
| | - Marie-Laure Parmentier
- Institut de génomique fonctionnelle,Université de Montpellier, CNRS UMR 5 203. Inserm U1191, Montpellier, France
| | - Julien Villeneuve
- Institut de génomique fonctionnelle,Université de Montpellier, CNRS UMR 5 203. Inserm U1191, Montpellier, France
| |
Collapse
|
6
|
Zhang S, Williams KJ, Verlande-Ferrero A, Chan AP, Su GB, Kershaw EE, Cox JE, Maschek JA, Shapira SN, Christofk HR, de Aguiar Vallim TQ, Masri S, Villanueva CJ. Acute activation of adipocyte lipolysis reveals dynamic lipid remodeling of the hepatic lipidome. J Lipid Res 2024; 65:100434. [PMID: 37640283 PMCID: PMC10839691 DOI: 10.1016/j.jlr.2023.100434] [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: 06/07/2023] [Revised: 07/27/2023] [Accepted: 08/16/2023] [Indexed: 08/31/2023] Open
Abstract
Adipose tissue is the site of long-term energy storage. During the fasting state, exercise, and cold exposure, the white adipose tissue mobilizes energy for peripheral tissues through lipolysis. The mobilization of lipids from white adipose tissue to the liver can lead to excess triglyceride accumulation and fatty liver disease. Although the white adipose tissue is known to release free fatty acids, a comprehensive analysis of lipids mobilized from white adipocytes in vivo has not been completed. In these studies, we provide a comprehensive quantitative analysis of the adipocyte-secreted lipidome and show that there is interorgan crosstalk with liver. Our analysis identifies multiple lipid classes released by adipocytes in response to activation of lipolysis. Time-dependent analysis of the serum lipidome showed that free fatty acids increase within 30 min of β3-adrenergic receptor activation and subsequently decrease, followed by a rise in serum triglycerides, liver triglycerides, and several ceramide species. The triglyceride composition of liver is enriched for linoleic acid despite higher concentrations of palmitate in the blood. To further validate that these findings were a specific consequence of lipolysis, we generated mice with conditional deletion of adipose tissue triglyceride lipase exclusively in adipocytes. This loss of in vivo adipocyte lipolysis prevented the rise in serum free fatty acids and hepatic triglycerides. Furthermore, conditioned media from adipocytes promotes lipid remodeling in hepatocytes with concomitant changes in genes/pathways mediating lipid utilization. Together, these data highlight critical role of adipocyte lipolysis in interorgan crosstalk between adipocytes and liver.
Collapse
Affiliation(s)
- Sicheng Zhang
- Department of Integrative Biology and Physiology, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Kevin J Williams
- UCLA Lipidomics Lab, Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Amandine Verlande-Ferrero
- Department of Biological Chemistry, Center for Epigenetics and Metabolism, Chao Family Comprehensive Cancer Center, University of California, Irvine (UCI), Irvine, CA, USA
| | - Alvin P Chan
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Gino B Su
- UCLA Lipidomics Lab, Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Erin E Kershaw
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, PA, USA
| | - James E Cox
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - John Alan Maschek
- Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, UT, USA
| | - Suzanne N Shapira
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Heather R Christofk
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, USA; Molecular Biology Institute, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Thomas Q de Aguiar Vallim
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, USA; Molecular Biology Institute, University of California, Los Angeles (UCLA), Los Angeles, CA, USA; Division of Cardiology, Department of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Selma Masri
- Department of Biological Chemistry, Center for Epigenetics and Metabolism, Chao Family Comprehensive Cancer Center, University of California, Irvine (UCI), Irvine, CA, USA
| | - Claudio J Villanueva
- Department of Integrative Biology and Physiology, University of California, Los Angeles (UCLA), Los Angeles, CA, USA; Molecular Biology Institute, University of California, Los Angeles (UCLA), Los Angeles, CA, USA.
| |
Collapse
|
7
|
Ekim Kocabey A, Schneiter R. Human lipocalins bind and export fatty acids through the secretory pathway of yeast cells. Front Microbiol 2024; 14:1309024. [PMID: 38328584 PMCID: PMC10849133 DOI: 10.3389/fmicb.2023.1309024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 12/12/2023] [Indexed: 02/09/2024] Open
Abstract
The activation of fatty acids to their acyl-CoA derivatives is a crucial step for their integration into more complex lipids or their degradation via beta-oxidation. Yeast cells employ five distinct acyl-CoA synthases to facilitate this ATP-dependent activation of acyl chains. Notably, mutant cells that are deficient in two of these fatty acid-activating (FAA) enzymes, namely, Faa1 and Faa4, do not take up free fatty acids but rather export them out of the cell. This unique fatty acid export pathway depends on small, secreted pathogenesis-related yeast proteins (Pry). In this study, we investigate whether the expression of human fatty acid-binding proteins, including Albumin, fatty acid-binding protein 4 (Fabp4), and three distinct lipocalins (ApoD, Lcn1, and Obp2a), could promote fatty acid secretion in yeast. To optimize the expression and secretion of these proteins, we systematically examined various signal sequences in both low-copy and high-copy number plasmids. Our findings reveal that directing these fatty-acid binding proteins into the secretory pathway effectively promotes fatty acid secretion from a sensitized quadruple mutant model strain (faa1∆ faa4∆ pry1∆ pry3∆). Furthermore, the level of fatty acid secretion exhibited a positive correlation with the efficiency of protein secretion. Importantly, the expression of all human lipid-binding proteins rescued Pry-dependent fatty acid secretion, resulting in the secretion of both long-chain saturated and unsaturated fatty acids. These results not only affirm the in vitro binding capabilities of lipocalins to fatty acids but also present a novel avenue for enhancing the secretion of valuable lipidic compounds. Given the growing interest in utilizing yeast as a cellular factory for producing poorly soluble compounds and the potential of lipocalins as platforms for engineering substrate-binding specificity, our model is considered as a powerful tool for promoting the secretion of high-value lipid-based molecules.
Collapse
Affiliation(s)
| | - Roger Schneiter
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
8
|
Huang Y, Hertzel AV, Fish SR, Halley CL, Bohm EK, Martinez HM, Durfee CC, Sanders MA, Harris RS, Niedernhofer LJ, Bernlohr DA. TP53/p53 Facilitates Stress-Induced Exosome and Protein Secretion by Adipocytes. Diabetes 2023; 72:1560-1573. [PMID: 37347719 PMCID: PMC10588298 DOI: 10.2337/db22-1027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 06/06/2023] [Indexed: 06/24/2023]
Abstract
Besides the secretion of fatty acids, lipolytic stimulation of adipocytes results in the secretion of triglyceride-rich extracellular vesicles and some free proteins (e.g., fatty acid binding protein 4) that, in sum, affect adipose homeostasis as well as the development of metabolic disease. At the mechanistic level, lipolytic signals activate p53 in an adipose triglyceride lipase-dependent manner, and pharmacologic inhibition of p53 attenuates adipocyte-derived extracellular vesicle (AdEV) protein and FABP4 secretion. Mass spectrometry analyses of the lipolytic secretome identified proteins involved in glucose and fatty acid metabolism, translation, chaperone activities, and redox control. Consistent with a role for p53 in adipocyte protein secretion, activation of p53 by the MDM2 antagonist nutlin potentiated AdEV particles and non-AdEV protein secretion from cultured 3T3-L1 or OP9 adipocytes while the levels of FABP4 and AdEV proteins were significantly reduced in serum from p53-/- mice compared with wild-type controls. The genotoxin doxorubicin increased AdEV protein and FABP4 secretion in a p53-dependent manner and DNA repair-depleted ERCC1-/Δ-haploinsufficient mice expressed elevated p53 in adipose depots, along with significantly increased serum FABP4. In sum, these data suggest that lipolytic signals, and cellular stressors such as DNA damage, facilitate AdEV protein and FABP4 secretion by adipocytes in a p53-dependent manner. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Yimao Huang
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN
| | - Ann V. Hertzel
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN
| | - Shayla R. Fish
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN
| | - Catherine L. Halley
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN
| | - Ellie K. Bohm
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN
| | - Hector Martell Martinez
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN
| | - Cameron C. Durfee
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN
| | - Mark A. Sanders
- University Imaging Center, University of Minnesota, Minneapolis, MN
| | - Reuben S. Harris
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN
| | - Laura J. Niedernhofer
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN
| | - David A. Bernlohr
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN
- Institute for Diabetes, Obesity and Metabolism, University of Minnesota, Minneapolis, MN
| |
Collapse
|
9
|
Abstract
Fatty acid-binding proteins (FABPs) are small lipid-binding proteins abundantly expressed in tissues that are highly active in fatty acid (FA) metabolism. Ten mammalian FABPs have been identified, with tissue-specific expression patterns and highly conserved tertiary structures. FABPs were initially studied as intracellular FA transport proteins. Further investigation has demonstrated their participation in lipid metabolism, both directly and via regulation of gene expression, and in signaling within their cells of expression. There is also evidence that they may be secreted and have functional impact via the circulation. It has also been shown that the FABP ligand binding repertoire extends beyond long-chain FAs and that their functional properties also involve participation in systemic metabolism. This article reviews the present understanding of FABP functions and their apparent roles in disease, particularly metabolic and inflammation-related disorders and cancers.
Collapse
Affiliation(s)
- Judith Storch
- Department of Nutritional Sciences and Rutgers Center for Lipid Research, Rutgers University, New Brunswick, New Jersey, United States;
| | - Betina Corsico
- Instituto de Investigaciones Bioquímicas de La Plata, CONICET-UNLP, Facultad de Ciencias Médicas, La Plata, Argentina;
| |
Collapse
|
10
|
Inouye KE, Prentice KJ, Lee A, Wang ZB, Dominguez-Gonzalez C, Chen MX, Riveros JK, Burak MF, Lee GY, Hotamışlıgil GS. Endothelial-derived FABP4 constitutes the majority of basal circulating hormone and regulates lipolysis-driven insulin secretion. JCI Insight 2023; 8:e164642. [PMID: 37279064 PMCID: PMC10443803 DOI: 10.1172/jci.insight.164642] [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: 08/22/2022] [Accepted: 05/31/2023] [Indexed: 06/07/2023] Open
Abstract
Fatty acid binding protein 4 (FABP4) is a lipid chaperone secreted from adipocytes upon stimulation of lipolysis. Circulating FABP4 levels strongly correlate with obesity and metabolic pathologies in experimental models and humans. While adipocytes have been presumed to be the major source of hormonal FABP4, this question has not been addressed definitively in vivo. We generated mice with Fabp4 deletion in cells known to express the gene - adipocytes (Adipo-KO), endothelial cells (Endo-KO), myeloid cells (Myeloid-KO), and the whole body (Total-KO) - to examine the contribution of these cell types to basal and stimulated plasma FABP4 levels. Unexpectedly, baseline plasma FABP4 was not significantly reduced in Adipo-KO mice, whereas Endo-KO mice showed ~87% reduction versus WT controls. In contrast, Adipo-KO mice exhibited ~62% decreased induction of FABP4 responses to lipolysis, while Endo-KO mice showed only mildly decreased induction, indicating that adipocytes are the main source of increases in FABP4 during lipolysis. We did not detect any myeloid contribution to circulating FABP4. Surprisingly, despite the nearly intact induction of FABP4, Endo-KO mice showed blunted lipolysis-induced insulin secretion, identical to Total-KO mice. We conclude that the endothelium is the major source of baseline hormonal FABP4 and is required for the insulin response to lipolysis.
Collapse
Affiliation(s)
- Karen E. Inouye
- Sabri Ülker Center for Metabolic Research, Harvard T.H. Chan School of Public Health, Department of Molecular Metabolism, Boston, Massachusetts, USA
| | - Kacey J. Prentice
- Sabri Ülker Center for Metabolic Research, Harvard T.H. Chan School of Public Health, Department of Molecular Metabolism, Boston, Massachusetts, USA
| | - Alexandra Lee
- Sabri Ülker Center for Metabolic Research, Harvard T.H. Chan School of Public Health, Department of Molecular Metabolism, Boston, Massachusetts, USA
| | - Zeqiu B. Wang
- Sabri Ülker Center for Metabolic Research, Harvard T.H. Chan School of Public Health, Department of Molecular Metabolism, Boston, Massachusetts, USA
| | - Carla Dominguez-Gonzalez
- Sabri Ülker Center for Metabolic Research, Harvard T.H. Chan School of Public Health, Department of Molecular Metabolism, Boston, Massachusetts, USA
| | - Mu Xian Chen
- Sabri Ülker Center for Metabolic Research, Harvard T.H. Chan School of Public Health, Department of Molecular Metabolism, Boston, Massachusetts, USA
| | - Jillian K. Riveros
- Sabri Ülker Center for Metabolic Research, Harvard T.H. Chan School of Public Health, Department of Molecular Metabolism, Boston, Massachusetts, USA
| | - M. Furkan Burak
- Sabri Ülker Center for Metabolic Research, Harvard T.H. Chan School of Public Health, Department of Molecular Metabolism, Boston, Massachusetts, USA
| | - Grace Y. Lee
- Sabri Ülker Center for Metabolic Research, Harvard T.H. Chan School of Public Health, Department of Molecular Metabolism, Boston, Massachusetts, USA
| | - Gökhan S. Hotamışlıgil
- Sabri Ülker Center for Metabolic Research, Harvard T.H. Chan School of Public Health, Department of Molecular Metabolism, Boston, Massachusetts, USA
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
| |
Collapse
|
11
|
Yabut KCB, Isoherranen N. Impact of Intracellular Lipid Binding Proteins on Endogenous and Xenobiotic Ligand Metabolism and Disposition. Drug Metab Dispos 2023; 51:700-717. [PMID: 37012074 PMCID: PMC10197203 DOI: 10.1124/dmd.122.001010] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 03/16/2023] [Accepted: 02/10/2023] [Indexed: 04/05/2023] Open
Abstract
The family of intracellular lipid binding proteins (iLBPs) is comprised of 16 members of structurally related binding proteins that have ubiquitous tissue expression in humans. iLBPs collectively bind diverse essential endogenous lipids and xenobiotics. iLBPs solubilize and traffic lipophilic ligands through the aqueous milieu of the cell. Their expression is correlated with increased rates of ligand uptake into tissues and altered ligand metabolism. The importance of iLBPs in maintaining lipid homeostasis is well established. Fatty acid binding proteins (FABPs) make up the majority of iLBPs and are expressed in major organs relevant to xenobiotic absorption, distribution, and metabolism. FABPs bind a variety of xenobiotics including nonsteroidal anti-inflammatory drugs, psychoactive cannabinoids, benzodiazepines, antinociceptives, and peroxisome proliferators. FABP function is also associated with metabolic disease, making FABPs currently a target for drug development. Yet the potential contribution of FABP binding to distribution of xenobiotics into tissues and the mechanistic impact iLBPs may have on xenobiotic metabolism are largely undefined. This review examines the tissue-specific expression and functions of iLBPs, the ligand binding characteristics of iLBPs, their known endogenous and xenobiotic ligands, methods for measuring ligand binding, and mechanisms of ligand delivery from iLBPs to membranes and enzymes. Current knowledge of the importance of iLBPs in affecting disposition of xenobiotics is collectively described. SIGNIFICANCE STATEMENT: The data reviewed here show that FABPs bind many drugs and suggest that binding of drugs to FABPs in various tissues will affect drug distribution into tissues. The extensive work and findings with endogenous ligands suggest that FABPs may also alter the metabolism and transport of drugs. This review illustrates the potential significance of this understudied area.
Collapse
Affiliation(s)
- King Clyde B Yabut
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington
| | - Nina Isoherranen
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington
| |
Collapse
|
12
|
Prentice KJ, Lee A, Cedillo P, Inouye KE, Ertunc ME, Riveros JK, Lee GY, Hotamisligil GS. Sympathetic tone dictates the impact of lipolysis on FABP4 secretion. J Lipid Res 2023; 64:100386. [PMID: 37172691 PMCID: PMC10248869 DOI: 10.1016/j.jlr.2023.100386] [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: 10/20/2022] [Revised: 04/19/2023] [Accepted: 05/04/2023] [Indexed: 05/15/2023] Open
Abstract
Levels of circulating fatty acid binding protein 4 (FABP4) protein are strongly associated with obesity and metabolic disease in both mice and humans, and secretion is stimulated by β-adrenergic stimulation both in vivo and in vitro. Previously, lipolysis-induced FABP4 secretion was found to be significantly reduced upon pharmacological inhibition of adipose triglyceride lipase (ATGL) and was absent from adipose tissue explants from mice specifically lacking ATGL in their adipocytes (ATGLAdpKO). Here, we find that upon activation of β-adrenergic receptors in vivo, ATGLAdpKO mice unexpectedly exhibited significantly higher levels of circulating FABP4 as compared with ATGLfl/fl controls, despite no corresponding induction of lipolysis. We generated an additional model with adipocyte-specific deletion of both FABP4 and ATGL (ATGL/FABP4AdpKO) to evaluate the cellular source of this circulating FABP4. In these animals, there was no evidence of lipolysis-induced FABP4 secretion, indicating that the source of elevated FABP4 levels in ATGLAdpKO mice was indeed from the adipocytes. ATGLAdpKO mice exhibited significantly elevated corticosterone levels, which positively correlated with plasma FABP4 levels. Pharmacological inhibition of sympathetic signaling during lipolysis using hexamethonium or housing mice at thermoneutrality to chronically reduce sympathetic tone significantly reduced FABP4 secretion in ATGLAdpKO mice compared with controls. Therefore, activity of a key enzymatic step of lipolysis mediated by ATGL, per se, is not required for in vivo stimulation of FABP4 secretion from adipocytes, which can be induced through sympathetic signaling.
Collapse
Affiliation(s)
- Kacey J Prentice
- Department of Molecular Metabolism; Sabri Ülker Center for Metabolic Research, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Alexandra Lee
- Department of Molecular Metabolism; Sabri Ülker Center for Metabolic Research, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Paulina Cedillo
- Department of Molecular Metabolism; Sabri Ülker Center for Metabolic Research, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Karen E Inouye
- Department of Molecular Metabolism; Sabri Ülker Center for Metabolic Research, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Meric Erikci Ertunc
- Department of Molecular Metabolism; Sabri Ülker Center for Metabolic Research, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Jillian K Riveros
- Department of Molecular Metabolism; Sabri Ülker Center for Metabolic Research, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Grace Yankun Lee
- Department of Molecular Metabolism; Sabri Ülker Center for Metabolic Research, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Gökhan S Hotamisligil
- Department of Molecular Metabolism; Sabri Ülker Center for Metabolic Research, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| |
Collapse
|
13
|
Wagh AR, Sulakshane P, Glickman MH. Alzheimer's disease-associated mutant ubiquitin (UBB +1) is secreted through an autophagosome-like vesicle-mediated unconventional pathway. BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2023; 1866:194936. [PMID: 37075976 DOI: 10.1016/j.bbagrm.2023.194936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 03/28/2023] [Accepted: 04/13/2023] [Indexed: 04/21/2023]
Abstract
Misfolded protein aggregation at both intracellular and extracellular milieus is thought to be the major etiology of Alzheimer's disease (AD). UBB+1, a frameshift variant of the ubiquitin B gene (UBB) results in a folded ubiquitin domain fused to a flexible unstructured extension. Accumulation of UBB+1 in extracellular plaques in the brains of AD patients undoubtedly suggests a role of the ubiquitin-proteasome system in AD. However, the exact mechanism of extracellular secretion of UBB+1 remains unknown. In an attempt to understand the molecular mechanism of UBB+1 secretion, we performed a survey of secretory pathways and identified the involvement of unconventional autophagosome-mediated UBB+1 secretion. Expression of UBB+1 was sufficient to stimulate LC3B/Atg8 conversion from LC3B-I to LC3B-II, which indicates initiation of the autophagy pathway. Furthermore, deficiency of ATG5 - a key player in autophagosome formation - inhibited UBB+1 secretion. Based on immunofluorescence 3D structured illumination (SIM) microscopy and co-immunoprecipitation, we provide evidence that UBB+1 is associated with the secretory autophagosome marker, SEC22B, while HSP90 possibly acts as a carrier. Using LC-MS/MS and mutagenesis we found that in cells, UBB+1 is ubiquitinated on lysine 11, 29, and 48, however, this ubiquitination does not contribute to its secretion. By contrast, proteasome or lysosome inhibition slightly enhanced secretion. Taken together, this study suggests that by ridding cells of UBB+1, secretory autophagosomes may alleviate the cellular stress associated with UBB+1, yet simultaneously mediate the spreading of a mutant specie with disordered characteristics to the extracellular milieu.
Collapse
Affiliation(s)
- Ajay R Wagh
- The Faculty of Biology, Technion Israel Institute of Technology, Haifa 32000, Israel
| | - Prasad Sulakshane
- The Faculty of Biology, Technion Israel Institute of Technology, Haifa 32000, Israel
| | - Michael H Glickman
- The Faculty of Biology, Technion Israel Institute of Technology, Haifa 32000, Israel.
| |
Collapse
|
14
|
Asami S, Tsutsui Y, Yamamoto S, Miyajima M. T-cell deficiency induces deficits in social behavior and dyslipidemia in mice. Biochem Biophys Res Commun 2023; 648:81-86. [PMID: 36739817 DOI: 10.1016/j.bbrc.2023.01.059] [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: 12/14/2022] [Accepted: 01/19/2023] [Indexed: 01/22/2023]
Abstract
Patients with neuropsychiatric disorders often exhibit an altered metabolic status. However, the underlying factors that induce behavioral and metabolic dysfunctions remain poorly understood. Therefore, we investigated whether behavioral and metabolic alterations could be induced in immunodeficient conditions. We found that T-cell-deficient Cd3e-/- mice exhibit deficits in social behavior associated with dyslipidemia. Cd3e-/- mice exhibited abnormal social novelty preference, but normal anxiety-like behavior. We also detected decreases in the concentrations of plasma triglyceride and the lipid transporter molecule fatty acid-binding protein 2. Furthermore, the adoptive transfer of T-cells to Cd3e-/- mice ameliorated the deficits in social behavior and recovered plasma triglyceride concentration. Thus, we found that T-cell disruption can induce defects in social behavior and systemic lipid homeostasis in mice. Given these findings, we believe that Cd3e-/- mice represent a useful tool for investigating the mechanisms of causal relationships among immune dysfunction, behavior, and metabolism.
Collapse
Affiliation(s)
- Shohei Asami
- Laboratory for Mucosal Immunity, RIKEN Center for Integrative Medical Sciences (IMS), 1-7-22, Suehirocho, Tsurumi-ku, Yokohama, 230-0045, Japan; Division for Molecular Biology, Research Institute for Biomedical Sciences (RIBS), Tokyo University of Science, 2669, Yamazaki, Noda, 278-0022, Japan
| | - Yumi Tsutsui
- Laboratory for Mucosal Immunity, RIKEN Center for Integrative Medical Sciences (IMS), 1-7-22, Suehirocho, Tsurumi-ku, Yokohama, 230-0045, Japan
| | - Sachiko Yamamoto
- Laboratory for Mucosal Immunity, RIKEN Center for Integrative Medical Sciences (IMS), 1-7-22, Suehirocho, Tsurumi-ku, Yokohama, 230-0045, Japan
| | - Michio Miyajima
- Laboratory for Mucosal Immunity, RIKEN Center for Integrative Medical Sciences (IMS), 1-7-22, Suehirocho, Tsurumi-ku, Yokohama, 230-0045, Japan.
| |
Collapse
|
15
|
Kreissl FK, Banki MA, Droujinine IA. Molecular methods to study protein trafficking between organs. Proteomics 2023; 23:e2100331. [PMID: 36478633 DOI: 10.1002/pmic.202100331] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 11/16/2022] [Accepted: 11/21/2022] [Indexed: 12/13/2022]
Abstract
Interorgan communication networks are key regulators of organismal homeostasis, and their dysregulation is associated with a variety of pathologies. While mass spectrometry proteomics identifies circulating proteins and can correlate their abundance with disease phenotypes, the tissues of origin and destinations of these secreted proteins remain largely unknown. In vitro approaches to study protein secretion are valuable, however, they may not mimic the complexity of in vivo environments. More recently, the development of engineered promiscuous BirA* biotin ligase derivatives has enabled tissue-specific tagging of cellular secreted proteomes in vivo. The use of biotin as a molecular tag provides information on the tissue of origin and destination, and enables the enrichment of low-abundance hormone proteins. Therefore, promiscuous protein biotinylation is a valuable tool to study protein secretion in vivo.
Collapse
Affiliation(s)
- Felix K Kreissl
- Department of Immunology and Microbiology, Scripps Research, La Jolla, California, USA
| | - Michael A Banki
- Department of Molecular Medicine, Scripps Research, La Jolla, California, USA
| | - Ilia A Droujinine
- Department of Molecular Medicine, Scripps Research, La Jolla, California, USA
| |
Collapse
|
16
|
Scheyer A, Yasmin F, Naskar S, Patel S. Endocannabinoids at the synapse and beyond: implications for neuropsychiatric disease pathophysiology and treatment. Neuropsychopharmacology 2023; 48:37-53. [PMID: 36100658 PMCID: PMC9700791 DOI: 10.1038/s41386-022-01438-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 08/10/2022] [Accepted: 08/18/2022] [Indexed: 11/09/2022]
Abstract
Endocannabinoids (eCBs) are lipid neuromodulators that suppress neurotransmitter release, reduce postsynaptic excitability, activate astrocyte signaling, and control cellular respiration. Here, we describe canonical and emerging eCB signaling modes and aim to link adaptations in these signaling systems to pathological states. Adaptations in eCB signaling systems have been identified in a variety of biobehavioral and physiological process relevant to neuropsychiatric disease states including stress-related disorders, epilepsy, developmental disorders, obesity, and substance use disorders. These insights have enhanced our understanding of the pathophysiology of neurological and psychiatric disorders and are contributing to the ongoing development of eCB-targeting therapeutics. We suggest future studies aimed at illuminating how adaptations in canonical as well as emerging cellular and synaptic modes of eCB signaling contribute to disease pathophysiology or resilience could further advance these novel treatment approaches.
Collapse
Affiliation(s)
| | - Farhana Yasmin
- Northwestern Center for Psychiatric Neuroscience, Chicago, IL, USA
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Saptarnab Naskar
- Northwestern Center for Psychiatric Neuroscience, Chicago, IL, USA
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Sachin Patel
- Northwestern Center for Psychiatric Neuroscience, Chicago, IL, USA.
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA.
| |
Collapse
|
17
|
Kaczocha M, Haj-Dahmane S. Mechanisms of endocannabinoid transport in the brain. Br J Pharmacol 2022; 179:4300-4310. [PMID: 33786823 PMCID: PMC8481389 DOI: 10.1111/bph.15469] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/22/2021] [Accepted: 03/24/2021] [Indexed: 12/14/2022] Open
Abstract
The endocannabinoids 2-arachidonoylglycerol (2-AG) and anandamide are among the best studied lipid messengers in the brain. By activating cannabinoid receptors in the CNS, endocannabinoids tune synaptic function, thereby influencing a variety of physiological and behavioural processes. Extensive research conducted over the last few decades has considerably enhanced our understanding of the molecular mechanisms and physiological functions of the endocannabinoid system. It is now well-established that endocannabinoids are synthesized by postsynaptic neurons and serve as retrograde messengers that suppress neurotransmitter release at central synapses. While the detailed mechanisms by which endocannabinoids gate synaptic function and behavioural processes are relatively well characterized, the mechanisms governing endocannabinoid transport at central synapses remain ill defined. Recently, several studies have begun to unravel the mechanisms governing intracellular and intercellular endocannabinoid transport. In this review, we will focus on new advances in the mechanisms of intracellular and synaptic endocannabinoid transport in the CNS. LINKED ARTICLES: This article is part of a themed issue on New discoveries and perspectives in mental and pain disorders. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v179.17/issuetoc.
Collapse
Affiliation(s)
- Martin Kaczocha
- Department of Anesthesiology, Stony Brook University, Stony Brook, New York, USA
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, USA
| | - Samir Haj-Dahmane
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, New York, USA
- Neuroscience Program, University at Buffalo, Buffalo, New York, USA
| |
Collapse
|
18
|
Ahat E, Bui S, Zhang J, da Veiga Leprevost F, Sharkey L, Reid W, Nesvizhskii AI, Paulson HL, Wang Y. GRASP55 regulates the unconventional secretion and aggregation of mutant huntingtin. J Biol Chem 2022; 298:102219. [PMID: 35780830 PMCID: PMC9352920 DOI: 10.1016/j.jbc.2022.102219] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 06/19/2022] [Accepted: 06/20/2022] [Indexed: 11/25/2022] Open
Abstract
Recent studies demonstrated that the Golgi reassembly stacking proteins (GRASPs), especially GRASP55, regulate Golgi-independent unconventional secretion of certain cytosolic and transmembrane cargoes; however, the underlying mechanism remains unknown. Here, we surveyed several neurodegenerative disease-related proteins, including mutant huntingtin (Htt-Q74), superoxide dismutase 1 (SOD1), tau, and TAR DNA-binding protein 43 (TDP-43), for unconventional secretion; our results show that Htt-Q74 is most robustly secreted in a GRASP55-dependent manner. Using Htt-Q74 as a model system, we demonstrate that unconventional secretion of Htt is GRASP55 and autophagy dependent and is enhanced under stress conditions such as starvation and endoplasmic reticulum stress. Mechanistically, we show that GRASP55 facilitates Htt secretion by tethering autophagosomes to lysosomes to promote autophagosome maturation and subsequent lysosome secretion and by stabilizing p23/TMED10, a channel for translocation of cytoplasmic proteins into the lumen of the endoplasmic reticulum-Golgi intermediate compartment. Moreover, we found that GRASP55 levels are upregulated by various stresses to facilitate unconventional secretion, whereas inhibition of Htt-Q74 secretion by GRASP55 KO enhances Htt aggregation and toxicity. Finally, comprehensive secretomic analysis identified novel cytosolic cargoes secreted by the same unconventional pathway, including transgelin (TAGLN), multifunctional protein ADE2 (PAICS), and peroxiredoxin-1 (PRDX1). In conclusion, this study defines the pathway of GRASP55-mediated unconventional protein secretion and provides important insights into the progression of Huntington's disease.
Collapse
Affiliation(s)
- Erpan Ahat
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Sarah Bui
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Jianchao Zhang
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Lisa Sharkey
- Department of Neurology, University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| | - Whitney Reid
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Alexey I. Nesvizhskii
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, Michigan, USA,Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, USA
| | - Henry L. Paulson
- Department of Neurology, University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| | - Yanzhuang Wang
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA; Department of Neurology, University of Michigan School of Medicine, Ann Arbor, Michigan, USA.
| |
Collapse
|
19
|
Iglesia RP, Prado MB, Alves RN, Escobar MIM, Fernandes CFDL, Fortes ACDS, Souza MCDS, Boccacino JM, Cangiano G, Soares SR, de Araújo JPA, Tiek DM, Goenka A, Song X, Keady JR, Hu B, Cheng SY, Lopes MH. Unconventional Protein Secretion in Brain Tumors Biology: Enlightening the Mechanisms for Tumor Survival and Progression. Front Cell Dev Biol 2022; 10:907423. [PMID: 35784465 PMCID: PMC9242006 DOI: 10.3389/fcell.2022.907423] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 05/26/2022] [Indexed: 11/28/2022] Open
Abstract
Non-canonical secretion pathways, collectively known as unconventional protein secretion (UPS), are alternative secretory mechanisms usually associated with stress-inducing conditions. UPS allows proteins that lack a signal peptide to be secreted, avoiding the conventional endoplasmic reticulum-Golgi complex secretory pathway. Molecules that generally rely on the canonical pathway to be secreted may also use the Golgi bypass, one of the unconventional routes, to reach the extracellular space. UPS studies have been increasingly growing in the literature, including its implication in the biology of several diseases. Intercellular communication between brain tumor cells and the tumor microenvironment is orchestrated by various molecules, including canonical and non-canonical secreted proteins that modulate tumor growth, proliferation, and invasion. Adult brain tumors such as gliomas, which are aggressive and fatal cancers with a dismal prognosis, could exploit UPS mechanisms to communicate with their microenvironment. Herein, we provide functional insights into the UPS machinery in the context of tumor biology, with a particular focus on the secreted proteins by alternative routes as key regulators in the maintenance of brain tumors.
Collapse
Affiliation(s)
- Rebeca Piatniczka Iglesia
- Laboratory of Neurobiology and Stem Cells, Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil,The Robert H. Lurie Comprehensive Cancer Center, The Ken and Ruth Davee Department of Neurology, Lou and Jean Malnati Brain Tumor Institute at Northwestern Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Mariana Brandão Prado
- Laboratory of Neurobiology and Stem Cells, Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Rodrigo Nunes Alves
- Laboratory of Neurobiology and Stem Cells, Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Maria Isabel Melo Escobar
- Laboratory of Neurobiology and Stem Cells, Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Camila Felix de Lima Fernandes
- Laboratory of Neurobiology and Stem Cells, Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Ailine Cibele dos Santos Fortes
- Laboratory of Neurobiology and Stem Cells, Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Maria Clara da Silva Souza
- Laboratory of Neurobiology and Stem Cells, Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Jacqueline Marcia Boccacino
- Laboratory of Neurobiology and Stem Cells, Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Giovanni Cangiano
- Laboratory of Neurobiology and Stem Cells, Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Samuel Ribeiro Soares
- Laboratory of Neurobiology and Stem Cells, Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - João Pedro Alves de Araújo
- Laboratory of Neurobiology and Stem Cells, Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Deanna Marie Tiek
- The Robert H. Lurie Comprehensive Cancer Center, The Ken and Ruth Davee Department of Neurology, Lou and Jean Malnati Brain Tumor Institute at Northwestern Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Anshika Goenka
- The Robert H. Lurie Comprehensive Cancer Center, The Ken and Ruth Davee Department of Neurology, Lou and Jean Malnati Brain Tumor Institute at Northwestern Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Xiao Song
- The Robert H. Lurie Comprehensive Cancer Center, The Ken and Ruth Davee Department of Neurology, Lou and Jean Malnati Brain Tumor Institute at Northwestern Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Jack Ryan Keady
- The Robert H. Lurie Comprehensive Cancer Center, The Ken and Ruth Davee Department of Neurology, Lou and Jean Malnati Brain Tumor Institute at Northwestern Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Bo Hu
- The Robert H. Lurie Comprehensive Cancer Center, The Ken and Ruth Davee Department of Neurology, Lou and Jean Malnati Brain Tumor Institute at Northwestern Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Shi Yuan Cheng
- The Robert H. Lurie Comprehensive Cancer Center, The Ken and Ruth Davee Department of Neurology, Lou and Jean Malnati Brain Tumor Institute at Northwestern Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Marilene Hohmuth Lopes
- Laboratory of Neurobiology and Stem Cells, Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil,*Correspondence: Marilene Hohmuth Lopes,
| |
Collapse
|
20
|
Noh SH, Kim YJ, Lee MG. Autophagy-Related Pathways in Vesicular Unconventional Protein Secretion. Front Cell Dev Biol 2022; 10:892450. [PMID: 35774225 PMCID: PMC9237382 DOI: 10.3389/fcell.2022.892450] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 05/26/2022] [Indexed: 12/14/2022] Open
Abstract
Cellular proteins directed to the plasma membrane or released into the extracellular space can undergo a number of different pathways. Whereas the molecular mechanisms that underlie conventional ER-to-Golgi trafficking are well established, those associated with the unconventional protein secretion (UPS) pathways remain largely elusive. A pathway with an emerging role in UPS is autophagy. Although originally known as a degradative process for maintaining intracellular homeostasis, recent studies suggest that autophagy has diverse biological roles besides its disposal function and that it is mechanistically involved in the UPS of various secretory cargos including both leaderless soluble and Golgi-bypassing transmembrane proteins. Here, we summarize current knowledge of the autophagy-related UPS pathways, describing and comparing diverse features in the autophagy-related UPS cargos and autophagy machineries utilized in UPS. Additionally, we also suggest potential directions that further research in this field can take.
Collapse
Affiliation(s)
- Shin Hye Noh
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Ye Jin Kim
- Department of Pharmacology, Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Min Goo Lee
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea
- Department of Pharmacology, Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
21
|
Extracellular vimentin mimics VEGF and is a target for anti-angiogenic immunotherapy. Nat Commun 2022; 13:2842. [PMID: 35606362 PMCID: PMC9126915 DOI: 10.1038/s41467-022-30063-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 04/14/2022] [Indexed: 12/12/2022] Open
Abstract
Anti-angiogenic cancer therapies possess immune-stimulatory properties by counteracting pro-angiogenic molecular mechanisms. We report that tumor endothelial cells ubiquitously overexpress and secrete the intermediate filament protein vimentin through type III unconventional secretion mechanisms. Extracellular vimentin is pro-angiogenic and functionally mimics VEGF action, while concomitantly acting as inhibitor of leukocyte-endothelial interactions. Antibody targeting of extracellular vimentin shows inhibition of angiogenesis in vitro and in vivo. Effective and safe inhibition of angiogenesis and tumor growth in several preclinical and clinical studies is demonstrated using a vaccination strategy against extracellular vimentin. Targeting vimentin induces a pro-inflammatory condition in the tumor, exemplified by induction of the endothelial adhesion molecule ICAM1, suppression of PD-L1, and altered immune cell profiles. Our findings show that extracellular vimentin contributes to immune suppression and functions as a vascular immune checkpoint molecule. Targeting of extracellular vimentin presents therefore an anti-angiogenic immunotherapy strategy against cancer. The pro-tumorigenic effects of vimentin have been attributed to intracellular functions in tumour cells so far. Here, the authors show that tumour endothelial cells can secrete vimentin as a pro-angiogenic factor and that targeting of vimentin can be used as an immunotherapeutic strategy.
Collapse
|
22
|
Maricchiolo E, Panfili E, Pompa A, De Marchis F, Bellucci M, Pallotta MT. Unconventional Pathways of Protein Secretion: Mammals vs. Plants. Front Cell Dev Biol 2022; 10:895853. [PMID: 35573696 PMCID: PMC9096121 DOI: 10.3389/fcell.2022.895853] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 04/13/2022] [Indexed: 01/08/2023] Open
Abstract
In eukaryotes, many proteins contain an N-terminal signal peptide that allows their translocation into the endoplasmic reticulum followed by secretion outside the cell according to the classical secretory system. However, an increasing number of secreted proteins lacking the signal peptide sequence are emerging. These proteins, secreted in several alternative ways collectively known as unconventional protein secretion (UPS) pathways, exert extracellular functions including cell signaling, immune modulation, as well as moonlighting activities different from their well-described intracellular functions. Pathways for UPS include direct transfer across the plasma membrane, secretion from endosomal/multivesicular body-related components, release within plasma membrane-derived microvesicles, or use of elements of autophagy. In this review we describe the mammals and plants UPS pathways identified so far highlighting commonalities and differences.
Collapse
Affiliation(s)
- Elisa Maricchiolo
- Section of Biological and Biotechnological Sciences, Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Eleonora Panfili
- Section of Pharmacology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Andrea Pompa
- Section of Biological and Biotechnological Sciences, Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Francesca De Marchis
- Institute of Biosciences and Bioresources, National Research Council of Italy, Perugia, Italy
| | - Michele Bellucci
- Institute of Biosciences and Bioresources, National Research Council of Italy, Perugia, Italy
- *Correspondence: Michele Bellucci, ; Maria Teresa Pallotta,
| | - Maria Teresa Pallotta
- Section of Pharmacology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
- *Correspondence: Michele Bellucci, ; Maria Teresa Pallotta,
| |
Collapse
|
23
|
Filaquier A, Marin P, Parmentier ML, Villeneuve J. Roads and hubs of unconventional protein secretion. Curr Opin Cell Biol 2022; 75:102072. [PMID: 35305454 DOI: 10.1016/j.ceb.2022.02.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 02/07/2022] [Accepted: 02/14/2022] [Indexed: 12/11/2022]
Abstract
In eukaryotes, there is now compelling evidence that in addition to the conventional endoplasmic reticulum-Golgi secretory pathway, there are additional routes for the export of cytoplasmic proteins with a critical role in numerous physio-pathological conditions. These alternative secretory pathways or unconventional protein secretion (UPS) start now to be molecularly dissected, and while UPS landscape appears to be governed by a striking diversity and heterogeneity of mechanisms, common principles are emerging. We review here the role of key molecular determinants as well as the role of central hubs for UPS, highlighting the plasticity and dynamic properties of membrane-bound compartments. We also describe recent findings that position UPS as an integral component of adaptive responses to cope with particular cellular needs and stresses.
Collapse
Affiliation(s)
- Aurore Filaquier
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Philippe Marin
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Marie-Laure Parmentier
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Julien Villeneuve
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France.
| |
Collapse
|
24
|
Grochowska KM, Andres‐Alonso M, Karpova A, Kreutz MR. The needs of a synapse—How local organelles serve synaptic proteostasis. EMBO J 2022; 41:e110057. [PMID: 35285533 PMCID: PMC8982616 DOI: 10.15252/embj.2021110057] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/24/2021] [Accepted: 02/10/2022] [Indexed: 12/12/2022] Open
Abstract
Synaptic function crucially relies on the constant supply and removal of neuronal membranes. The morphological complexity of neurons poses a significant challenge for neuronal protein transport since the machineries for protein synthesis and degradation are mainly localized in the cell soma. In response to this unique challenge, local micro‐secretory systems have evolved that are adapted to the requirements of neuronal membrane protein proteostasis. However, our knowledge of how neuronal proteins are synthesized, trafficked to membranes, and eventually replaced and degraded remains scarce. Here, we review recent insights into membrane trafficking at synaptic sites and into the contribution of local organelles and micro‐secretory pathways to synaptic function. We describe the role of endoplasmic reticulum specializations in neurons, Golgi‐related organelles, and protein complexes like retromer in the synthesis and trafficking of synaptic transmembrane proteins. We discuss the contribution of autophagy and of proteasome‐mediated and endo‐lysosomal degradation to presynaptic proteostasis and synaptic function, as well as nondegradative roles of autophagosomes and lysosomes in signaling and synapse remodeling. We conclude that the complexity of neuronal cyto‐architecture necessitates long‐distance protein transport that combines degradation with signaling functions.
Collapse
Affiliation(s)
- Katarzyna M Grochowska
- Leibniz Group “Dendritic Organelles and Synaptic Function” Center for Molecular Neurobiology ZMNH University Medical Center Hamburg‐Eppendorf Hamburg Germany
- Research Group Neuroplasticity Leibniz Institute for Neurobiology Magdeburg Germany
| | - Maria Andres‐Alonso
- Leibniz Group “Dendritic Organelles and Synaptic Function” Center for Molecular Neurobiology ZMNH University Medical Center Hamburg‐Eppendorf Hamburg Germany
- Research Group Neuroplasticity Leibniz Institute for Neurobiology Magdeburg Germany
| | - Anna Karpova
- Research Group Neuroplasticity Leibniz Institute for Neurobiology Magdeburg Germany
- Center for Behavioral Brain Sciences Otto von Guericke University Magdeburg Germany
| | - Michael R Kreutz
- Leibniz Group “Dendritic Organelles and Synaptic Function” Center for Molecular Neurobiology ZMNH University Medical Center Hamburg‐Eppendorf Hamburg Germany
- Research Group Neuroplasticity Leibniz Institute for Neurobiology Magdeburg Germany
- Center for Behavioral Brain Sciences Otto von Guericke University Magdeburg Germany
- German Center for Neurodegenerative Diseases (DZNE) Magdeburg Germany
| |
Collapse
|
25
|
Li Y, Li Z, Ngandiri DA, Llerins Perez M, Wolf A, Wang Y. The Molecular Brakes of Adipose Tissue Lipolysis. Front Physiol 2022; 13:826314. [PMID: 35283787 PMCID: PMC8907745 DOI: 10.3389/fphys.2022.826314] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/10/2022] [Indexed: 12/11/2022] Open
Abstract
Adaptation to changes in energy availability is pivotal for the survival of animals. Adipose tissue, the body’s largest reservoir of energy and a major source of metabolic fuel, exerts a buffering function for fluctuations in nutrient availability. This functional plasticity ranges from energy storage in the form of triglycerides during periods of excess energy intake to energy mobilization via lipolysis in the form of free fatty acids for other organs during states of energy demands. The subtle balance between energy storage and mobilization is important for whole-body energy homeostasis; its disruption has been implicated as contributing to the development of insulin resistance, type 2 diabetes and cancer cachexia. As a result, adipocyte lipolysis is tightly regulated by complex regulatory mechanisms involving lipases and hormonal and biochemical signals that have opposing effects. In thermogenic brown and brite adipocytes, lipolysis stimulation is the canonical way for the activation of non-shivering thermogenesis. Lipolysis proceeds in an orderly and delicately regulated manner, with stimulation through cell-surface receptors via neurotransmitters, hormones, and autocrine/paracrine factors that activate various intracellular signal transduction pathways and increase kinase activity. The subsequent phosphorylation of perilipins, lipases, and cofactors initiates the translocation of key lipases from the cytoplasm to lipid droplets and enables protein-protein interactions to assemble the lipolytic machinery on the scaffolding perilipins at the surface of lipid droplets. Although activation of lipolysis has been well studied, the feedback fine-tuning is less well appreciated. This review focuses on the molecular brakes of lipolysis and discusses some of the divergent fine-tuning strategies in the negative feedback regulation of lipolysis, including delicate negative feedback loops, intermediary lipid metabolites-mediated allosteric regulation and dynamic protein–protein interactions. As aberrant adipocyte lipolysis is involved in various metabolic diseases and releasing the brakes on lipolysis in thermogenic adipocytes may activate thermogenesis, targeting adipocyte lipolysis is thus of therapeutic interest.
Collapse
|
26
|
Ho LWC, Chan CKW, Han R, Lau YFY, Li H, Ho YP, Zhuang X, Choi CHJ. Mammalian Cells Exocytose Alkylated Gold Nanoparticles via Extracellular Vesicles. ACS NANO 2022; 16:2032-2045. [PMID: 35137580 DOI: 10.1021/acsnano.1c07418] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Understanding the exocytosis of nanoparticles (NPs) from cells is valuable because it informs design rules of NPs that support desirable cellular retention for nanomedicine applications, but investigations into the mechanism for the exocytosis of NPs remain scarce. We elucidate the mechanism for the exocytosis of dodecyl-terminated, polyethylene glycol-coated gold NPs (termed "dodecyl-PEG-AuNP"). The Au core enables ultrastructural differentiation of the exocytosed NPs from the nearby extracellular vesicles (EVs). The PEG shell prevents interparticle agglomeration or aggregation that disfavors exocytosis. The minute amounts of alkyl chains on the PEG shell not only promote cellular uptake but also improve exocytosis by up to 4-fold higher probability and upregulate exocytosis- and vesicle-related genes. After entering Kera-308 keratinocytes and trafficking to multivesicular bodies and lysosomes, these NPs exit the cell predominantly via unconventional exocytosis, accompanied by enhanced secretion of sub-100 nm, CD81-enriched exosomes. The pathway for NP exocytosis and subpopulation of EVs that are secreted alongside the exocytosed NPs depends on dodecyl loading. This work provides insights into dissecting the mechanism of NP exocytosis and its relationship with EV secretion.
Collapse
|
27
|
Ren Y, Zhao H, Yin C, Lan X, Wu L, Du X, Griffiths HR, Gao D. Adipokines, Hepatokines and Myokines: Focus on Their Role and Molecular Mechanisms in Adipose Tissue Inflammation. Front Endocrinol (Lausanne) 2022; 13:873699. [PMID: 35909571 PMCID: PMC9329830 DOI: 10.3389/fendo.2022.873699] [Citation(s) in RCA: 65] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 06/17/2022] [Indexed: 11/18/2022] Open
Abstract
Chronic low-grade inflammation in adipose tissue (AT) is a hallmark of obesity and contributes to various metabolic disorders, such as type 2 diabetes and cardiovascular diseases. Inflammation in ATs is characterized by macrophage infiltration and the activation of inflammatory pathways mediated by NF-κB, JNK, and NLRP3 inflammasomes. Adipokines, hepatokines and myokines - proteins secreted from AT, the liver and skeletal muscle play regulatory roles in AT inflammation via endocrine, paracrine, and autocrine pathways. For example, obesity is associated with elevated levels of pro-inflammatory adipokines (e.g., leptin, resistin, chemerin, progranulin, RBP4, WISP1, FABP4, PAI-1, Follistatin-like1, MCP-1, SPARC, SPARCL1, and SAA) and reduced levels of anti-inflammatory adipokines such as adiponectin, omentin, ZAG, SFRP5, CTRP3, vaspin, and IL-10. Moreover, some hepatokines (Fetuin A, DPP4, FGF21, GDF15, and MANF) and myokines (irisin, IL-6, and DEL-1) also play pro- or anti-inflammatory roles in AT inflammation. This review aims to provide an updated understanding of these organokines and their role in AT inflammation and related metabolic abnormalities. It serves to highlight the molecular mechanisms underlying the effects of these organokines and their clinical significance. Insights into the roles and mechanisms of these organokines could provide novel and potential therapeutic targets for obesity-induced inflammation.
Collapse
Affiliation(s)
- Yakun Ren
- Institute of Molecular and Translational Medicine, Xian Jiaotong University Health Science Center, Xi’an, China
| | - Hao Zhao
- School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Chunyan Yin
- Department of Pediatrics, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Xi Lan
- Institute of Molecular and Translational Medicine, Xian Jiaotong University Health Science Center, Xi’an, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Litao Wu
- Institute of Molecular and Translational Medicine, Xian Jiaotong University Health Science Center, Xi’an, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Xiaojuan Du
- Institute of Molecular and Translational Medicine, Xian Jiaotong University Health Science Center, Xi’an, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Helen R. Griffiths
- Swansea University Medical School, Swansea University, Swansea, United Kingdom
| | - Dan Gao
- Institute of Molecular and Translational Medicine, Xian Jiaotong University Health Science Center, Xi’an, China
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Center, Xi’an, China
- *Correspondence: Dan Gao,
| |
Collapse
|
28
|
Prentice KJ, Saksi J, Robertson LT, Lee GY, Inouye KE, Eguchi K, Lee A, Cakici O, Otterbeck E, Cedillo P, Achenbach P, Ziegler AG, Calay ES, Engin F, Hotamisligil GS. A hormone complex of FABP4 and nucleoside kinases regulates islet function. Nature 2021; 600:720-726. [PMID: 34880500 DOI: 10.1038/s41586-021-04137-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 10/14/2021] [Indexed: 11/09/2022]
Abstract
The liberation of energy stores from adipocytes is critical to support survival in times of energy deficit; however, uncontrolled or chronic lipolysis associated with insulin resistance and/or insulin insufficiency disrupts metabolic homeostasis1,2. Coupled to lipolysis is the release of a recently identified hormone, fatty-acid-binding protein 4 (FABP4)3. Although circulating FABP4 levels have been strongly associated with cardiometabolic diseases in both preclinical models and humans4-7, no mechanism of action has yet been described8-10. Here we show that hormonal FABP4 forms a functional hormone complex with adenosine kinase (ADK) and nucleoside diphosphate kinase (NDPK) to regulate extracellular ATP and ADP levels. We identify a substantial effect of this hormone on beta cells and given the central role of beta-cell function in both the control of lipolysis and development of diabetes, postulate that hormonal FABP4 is a key regulator of an adipose-beta-cell endocrine axis. Antibody-mediated targeting of this hormone complex improves metabolic outcomes, enhances beta-cell function and preserves beta-cell integrity to prevent both type 1 and type 2 diabetes. Thus, the FABP4-ADK-NDPK complex, Fabkin, represents a previously unknown hormone and mechanism of action that integrates energy status with the function of metabolic organs, and represents a promising target against metabolic disease.
Collapse
Affiliation(s)
- Kacey J Prentice
- Sabri Ülker Center for Metabolic Research, Harvard T. H. Chan School of Public Health, Department of Molecular Metabolism, Boston, MA, USA
| | - Jani Saksi
- Sabri Ülker Center for Metabolic Research, Harvard T. H. Chan School of Public Health, Department of Molecular Metabolism, Boston, MA, USA
| | - Lauren T Robertson
- Sabri Ülker Center for Metabolic Research, Harvard T. H. Chan School of Public Health, Department of Molecular Metabolism, Boston, MA, USA
| | - Grace Y Lee
- Sabri Ülker Center for Metabolic Research, Harvard T. H. Chan School of Public Health, Department of Molecular Metabolism, Boston, MA, USA
| | - Karen E Inouye
- Sabri Ülker Center for Metabolic Research, Harvard T. H. Chan School of Public Health, Department of Molecular Metabolism, Boston, MA, USA
| | - Kosei Eguchi
- Sabri Ülker Center for Metabolic Research, Harvard T. H. Chan School of Public Health, Department of Molecular Metabolism, Boston, MA, USA
| | - Alexandra Lee
- Sabri Ülker Center for Metabolic Research, Harvard T. H. Chan School of Public Health, Department of Molecular Metabolism, Boston, MA, USA
| | - Ozgur Cakici
- Sabri Ülker Center for Metabolic Research, Harvard T. H. Chan School of Public Health, Department of Molecular Metabolism, Boston, MA, USA
| | - Emily Otterbeck
- Sabri Ülker Center for Metabolic Research, Harvard T. H. Chan School of Public Health, Department of Molecular Metabolism, Boston, MA, USA
| | - Paulina Cedillo
- Sabri Ülker Center for Metabolic Research, Harvard T. H. Chan School of Public Health, Department of Molecular Metabolism, Boston, MA, USA
| | - Peter Achenbach
- Institute of Diabetes Research, Helmholtz Zentrum Munchen, German Research Center for Environmental Health, Munich-Neuherberg, Germany
| | - Anette-Gabriele Ziegler
- Institute of Diabetes Research, Helmholtz Zentrum Munchen, German Research Center for Environmental Health, Munich-Neuherberg, Germany
| | - Ediz S Calay
- Sabri Ülker Center for Metabolic Research, Harvard T. H. Chan School of Public Health, Department of Molecular Metabolism, Boston, MA, USA
| | - Feyza Engin
- Sabri Ülker Center for Metabolic Research, Harvard T. H. Chan School of Public Health, Department of Molecular Metabolism, Boston, MA, USA.,Departments of Biomolecular Chemistry and Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Gökhan S Hotamisligil
- Sabri Ülker Center for Metabolic Research, Harvard T. H. Chan School of Public Health, Department of Molecular Metabolism, Boston, MA, USA. .,Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| |
Collapse
|
29
|
Liang X, Jiao Y, Gong X, Gu H, Nuermaimaiti N, Meng X, Liu D, Guan Y. Staufen1 unwinds the secondary structure and facilitates the translation of fatty acid binding protein 4 mRNA during adipogenesis. Adipocyte 2021; 10:350-360. [PMID: 34224297 PMCID: PMC8259723 DOI: 10.1080/21623945.2021.1948165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
Adipogenesis is regulated by genetic interactions, in which post-transcriptional regulation plays an important role. Staufen double-stranded RNA binding protein 1 (Staufen1 or STAU1) plays diverse roles in RNA processing and adipogenesis. Previously, we found that the downregulation of STAU1 affects the expression of fatty acid-binding protein 4 (FABP4) at the protein level but not at the mRNA level. This study aimed to determine the mechanism underlying the regulation of FABP4 expression by STAU1, explaining the inconsistency between FABP4 mRNA and protein levels. We used RNA interference, photoactivatable ribonucleoside enhanced cross-linking and immunoprecipitation, and an adeno-associated virus to examine the functions of STAU1 in adipogenesis. Our results indicate that STAU1 binds to the coding sequences of FABP4, thereby regulating the translation of FABP4 mRNA by unwinding the double-stranded structure. Furthermore, STAU1 mediates adipogenesis by regulating the secretion of free fatty acids. However, STAU1 knockdown decreases the fat weight/body weight ratio but does not affect the plasma triglyceride levels. These findings describe the mechanisms involved in STAU1-mediated regulation of FABP4 expression at the translational level during adipogenesis.
Collapse
Affiliation(s)
- Xiaodi Liang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Biochemistry and Molecular Biology, Preclinical Medicine College, Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Yi Jiao
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Biochemistry and Molecular Biology, Preclinical Medicine College, Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Xueli Gong
- Department of Pathophysiology, Preclinical Medicine College, Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Hao Gu
- Department of Laparoscopic Surgery, First Affiliated Hospital, Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Nuerbiye Nuermaimaiti
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Biochemistry and Molecular Biology, Preclinical Medicine College, Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Xuanyu Meng
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Biochemistry and Molecular Biology, Preclinical Medicine College, Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Dihui Liu
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Biochemistry and Molecular Biology, Preclinical Medicine College, Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Yaqun Guan
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Biochemistry and Molecular Biology, Preclinical Medicine College, Xinjiang Medical University, Urumqi, Xinjiang, China
| |
Collapse
|
30
|
Maeda FY, van Haaren JJ, Langley DB, Christ D, Andrews NW, Song W. Surface-associated antigen induces permeabilization of primary mouse B-cells and lysosome exocytosis facilitating antigen uptake and presentation to T-cells. eLife 2021; 10:66984. [PMID: 34704555 PMCID: PMC8589448 DOI: 10.7554/elife.66984] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 10/26/2021] [Indexed: 12/13/2022] Open
Abstract
B-cell receptor (BCR)-mediated antigen internalization and presentation are essential for humoral memory immune responses. Antigen encountered by B-cells is often tightly associated with the surface of pathogens and/or antigen-presenting cells. Internalization of such antigens requires myosin-mediated traction forces and extracellular release of lysosomal enzymes, but the mechanism triggering lysosomal exocytosis is unknown. Here, we show that BCR-mediated recognition of antigen tethered to beads, to planar lipid-bilayers or expressed on cell surfaces causes localized plasma membrane (PM) permeabilization, a process that requires BCR signaling and non-muscle myosin II activity. B-cell permeabilization triggers PM repair responses involving lysosomal exocytosis, and B-cells permeabilized by surface-associated antigen internalize more antigen than cells that remain intact. Higher affinity antigens cause more B-cell permeabilization and lysosomal exocytosis and are more efficiently presented to T-cells. Thus, PM permeabilization by surface-associated antigen triggers a lysosome-mediated B-cell resealing response, providing the extracellular hydrolases that facilitate antigen internalization and presentation.
Collapse
Affiliation(s)
- Fernando Y Maeda
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, United States
| | - Jurriaan Jh van Haaren
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, United States
| | - David B Langley
- Immunology Division, Garvan Institute of Medical Research, Darlinghurst, Australia
| | - Daniel Christ
- Immunology, Garvan Institute of Medical Research, Darlinghurst/Sydney, Australia
| | - Norma W Andrews
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, United States
| | - Wenxia Song
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, United States
| |
Collapse
|
31
|
Novel Biomolecules in the Pathogenesis of Gestational Diabetes Mellitus. Int J Mol Sci 2021; 22:ijms222111578. [PMID: 34769010 PMCID: PMC8584125 DOI: 10.3390/ijms222111578] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/16/2021] [Accepted: 10/22/2021] [Indexed: 12/12/2022] Open
Abstract
Gestational diabetes mellitus (GDM) is one of the most common metabolic diseases in pregnant women. Its early diagnosis seems to have a significant impact on the developing fetus, the course of delivery, and the neonatal period. It may also affect the later stages of child development and subsequent complications in the mother. Therefore, the crux of the matter is to find a biopredictor capable of singling out women at risk of developing GDM as early as the very start of pregnancy. Apart from the well-known molecules with a proven and clear-cut role in the pathogenesis of GDM, e.g., adiponectin and leptin, a potential role of newer biomolecules is also emphasized. Less popular and less known factors with different mechanisms of action include: galectins, growth differentiation factor-15, chemerin, omentin-1, osteocalcin, resistin, visfatin, vaspin, irisin, apelin, fatty acid-binding protein 4 (FABP4), fibroblast growth factor 21, and lipocalin-2. The aim of this review is to present the potential and significance of these 13 less known biomolecules in the pathogenesis of GDM. It seems that high levels of FABP4, low levels of irisin, and high levels of under-carboxylated osteocalcin in the serum of pregnant women can be used as predictive markers in the diagnosis of GDM. Hopefully, future clinical trials will be able to determine which biomolecules have the most potential to predict GDM.
Collapse
|
32
|
Wu M, Zhang M, Zhang Y, Li Z, Li X, Liu Z, Liu H, Li X. Relationship between lysosomal dyshomeostasis and progression of diabetic kidney disease. Cell Death Dis 2021; 12:958. [PMID: 34663802 PMCID: PMC8523726 DOI: 10.1038/s41419-021-04271-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 10/04/2021] [Indexed: 12/11/2022]
Abstract
Lysosomes are organelles involved in cell metabolism, waste degradation, and cellular material circulation. They play a key role in the maintenance of cellular physiological homeostasis. Compared with the lysosomal content of other organs, that of the kidney is abundant, and lysosomal abnormalities are associated with the occurrence and development of certain renal diseases. Lysosomal structure and function in intrinsic renal cells are impaired in diabetic kidney disease (DKD). Promoting lysosomal biosynthesis and/or restoring lysosomal function can repair damaged podocytes and proximal tubular epithelial cells, and delay the progression of DKD. Lysosomal homeostasis maintenance may be advantageous in alleviating DKD. Here, we systematically reviewed the latest advances in the relationship between lysosomal dyshomeostasis and progression of DKD based on recent literature to further elucidate the mechanism of renal injury in diabetes mellitus and to highlight the application potential of lysosomal homeostasis maintenance as a new prevention and treatment strategy for DKD. However, research on screening effective interventions for lysosomal dyshomeostasis is still in its infancy, and thus should be the focus of future research studies. The screening out of cell-specific lysosomal function regulation targets according to the different stages of DKD, so as to realize the controllable targeted regulation of cell lysosomal function during DKD, is the key to the successful clinical development of this therapeutic strategy.
Collapse
Affiliation(s)
- Man Wu
- Institute of Nephrology, and Key Laboratory of Prevention and Management of Chronic kidney Disease of Zhanjiang City, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524001, China
| | - Minjie Zhang
- Institute of Nephrology, and Key Laboratory of Prevention and Management of Chronic kidney Disease of Zhanjiang City, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524001, China
| | - Yaozhi Zhang
- Institute of Nephrology, and Key Laboratory of Prevention and Management of Chronic kidney Disease of Zhanjiang City, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524001, China
| | - Zixian Li
- Institute of Nephrology, and Key Laboratory of Prevention and Management of Chronic kidney Disease of Zhanjiang City, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524001, China
| | - Xingyu Li
- Institute of Nephrology, and Key Laboratory of Prevention and Management of Chronic kidney Disease of Zhanjiang City, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524001, China
| | - Zejian Liu
- Institute of Nephrology, and Key Laboratory of Prevention and Management of Chronic kidney Disease of Zhanjiang City, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524001, China
| | - Huafeng Liu
- Institute of Nephrology, and Key Laboratory of Prevention and Management of Chronic kidney Disease of Zhanjiang City, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524001, China.
| | - Xiaoyu Li
- Institute of Nephrology, and Key Laboratory of Prevention and Management of Chronic kidney Disease of Zhanjiang City, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524001, China.
| |
Collapse
|
33
|
Hassan Z, Kumar ND, Reggiori F, Khan G. How Viruses Hijack and Modify the Secretory Transport Pathway. Cells 2021; 10:2535. [PMID: 34685515 PMCID: PMC8534161 DOI: 10.3390/cells10102535] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/28/2021] [Accepted: 09/06/2021] [Indexed: 12/23/2022] Open
Abstract
Eukaryotic cells contain dynamic membrane-bound organelles that are constantly remodeled in response to physiological and environmental cues. Key organelles are the endoplasmic reticulum, the Golgi apparatus and the plasma membrane, which are interconnected by vesicular traffic through the secretory transport route. Numerous viruses, especially enveloped viruses, use and modify compartments of the secretory pathway to promote their replication, assembly and cell egression by hijacking the host cell machinery. In some cases, the subversion mechanism has been uncovered. In this review, we summarize our current understanding of how the secretory pathway is subverted and exploited by viruses belonging to Picornaviridae, Coronaviridae, Flaviviridae,Poxviridae, Parvoviridae and Herpesviridae families.
Collapse
Affiliation(s)
- Zubaida Hassan
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates;
- Department of Microbiology, School of Life Sciences, Modibbo Adama University, Yola PMB 2076, Nigeria
| | - Nilima Dinesh Kumar
- Department of Biomedical Sciences of Cells and Systems, University of Groningen, University Medical Center Groningen, 9713 AV Groningen, The Netherlands; (N.D.K.); (F.R.)
- Department of Medical Microbiology and Infection Prevention, University of Groningen, University Medical Center Groningen, 9713 AV Groningen, The Netherlands
| | - Fulvio Reggiori
- Department of Biomedical Sciences of Cells and Systems, University of Groningen, University Medical Center Groningen, 9713 AV Groningen, The Netherlands; (N.D.K.); (F.R.)
| | - Gulfaraz Khan
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates;
| |
Collapse
|
34
|
Chen MT, Huang JS, Gao DD, Li YX, Wang HY. Combined treatment with FABP4 inhibitor ameliorates rosiglitazone-induced liver steatosis in obese diabetic db/db mice. Basic Clin Pharmacol Toxicol 2021; 129:173-182. [PMID: 34128319 DOI: 10.1111/bcpt.13621] [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: 03/04/2021] [Accepted: 06/06/2021] [Indexed: 11/28/2022]
Abstract
Rosiglitazone has been reported to exert dual effects on liver steatosis, and it could exacerbate liver steatosis in obese animal models, which was suggested to be closely related to the elevated hepatic expression of FABP4. This study aimed to investigate whether combined treatment with FABP4 inhibitor I-9 could alleviate rosiglitazone-induced liver steatosis in obese diabetic db/db mice. Male C57BL/KsJ-db/db mice were orally treated with rosiglitazone, rosiglitazone combined with I-9 daily for 8 weeks. The liver steatosis was evaluated by triglyceride content and H&E staining. The expression of hepatic lipogenic genes or proteins in liver tissue or in FFA-treated hepatocytes and PMA-stimulated macrophages were determined by real-time quantitative polymerase chain reaction (RT-qPCR) or western blotting. Results showed that combined treatment with I-9 decreased rosiglitazone-induced increase in serum FABP4 level and expression of lipogenic genes in liver, especially FABP4, and ameliorated liver steatosis in db/db mice. Rosiglitazone-induced intracellular TG accumulation and increased expression of FABP4 in the cultured hepatocytes and macrophages were also suppressed by combined treatment. We concluded that combined treatment with FABP4 inhibitor I-9 could ameliorate rosiglitazone-exacerbated elevated serum FABP4 level and ectopic liver fat accumulation in obese diabetic db/db mice without affecting its anti-diabetic efficacy.
Collapse
Affiliation(s)
- Meng-Ting Chen
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,School of Pharmacy, University of Chinese Academy of Sciences, Beijing, China
| | - Jun-Shang Huang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,School of Pharmacy, University of Chinese Academy of Sciences, Beijing, China
| | - Ding-Ding Gao
- School of Pharmacy, Fudan University, Shanghai, China
| | - Ying-Xia Li
- School of Pharmacy, Fudan University, Shanghai, China
| | - He-Yao Wang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,School of Pharmacy, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
35
|
Barroso IG, Cardoso C, Ferreira C, Terra WR. Transcriptomic and proteomic analysis of the underlying mechanisms of digestion of triacylglycerols and phosphatides and absorption and fate of fatty acids along the midgut of Musca domestica. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY D-GENOMICS & PROTEOMICS 2021; 39:100826. [PMID: 33839527 DOI: 10.1016/j.cbd.2021.100826] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 03/23/2021] [Accepted: 03/23/2021] [Indexed: 11/24/2022]
Abstract
Most dietary lipids are triacylglycerols (TAGs) and phosphatides that are digested by TAG lipases and phospholipases (PLIPs), respectively, originating fatty acids (FA). The genome of Musca domestica has genes coding for phospholipases A1 (1PLIP), A2 (2PLIP), B (BPLIP), and acid lipases (ALIP), as for proteins involved in activation, binding, and metabolism of FA, which expression in the larval midgut was evaluated by RNA-seq. Some of the codified proteins were identified in midgut microvillar-enriched membrane by proteomics. 1PLIPs are the most expressed PLIPs, mainly in anterior midgut whereas 2PLIPs, and BPLIP in middle and posterior midgut, and ALIPs between middle and posterior regions. Absorption of FAs is putatively accomplished by proteins involved in FA activation (acyl-CoA synthetases) found in microvillar-enriched membrane preparations. Furthermore, FA uptake could be enhanced by proteins that bind FAs (FA-binding proteins) and its activated form (acyl-CoA binding proteins) mainly expressed in posterior midgut. Activated FAs could have different fates: synthesis of diacylglycerol (DAG) and TAG through monoacylglycerol and glycerol-3-phosphate pathways; synthesis of phosphatides; energy source by β-oxidation. Most genes coding for enzymes of those routes is expressed mainly at the end of posterior midgut. Data suggest that phosphatides are digested in anterior midgut by Md1PLIPs, releasing lysophosphatides that emulsify fats to be digested by MdALIPs in the middle and posterior midgut. Most resulting FAs is absorbed in the posterior midgut, where they follow the synthesis of DAG, TAG, and phosphatides or are oxidized along the midgut, mainly in highly metabolic middle and posterior midgut regions.
Collapse
Affiliation(s)
- Ignacio G Barroso
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Av. Prof. Lineu Prestes 748, 05508-000 São Paulo, Brazil
| | - Christiane Cardoso
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Av. Prof. Lineu Prestes 748, 05508-000 São Paulo, Brazil
| | - Clelia Ferreira
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Av. Prof. Lineu Prestes 748, 05508-000 São Paulo, Brazil
| | - Walter R Terra
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Av. Prof. Lineu Prestes 748, 05508-000 São Paulo, Brazil.
| |
Collapse
|
36
|
Lee CH, Lui DTW, Lam KSL. Adipocyte Fatty Acid-Binding Protein, Cardiovascular Diseases and Mortality. Front Immunol 2021; 12:589206. [PMID: 33815359 PMCID: PMC8017191 DOI: 10.3389/fimmu.2021.589206] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 03/04/2021] [Indexed: 12/21/2022] Open
Abstract
It has been increasingly recognized that inflammation plays an important role in the pathogenesis of cardiovascular disease (CVD). In obesity, adipose tissue inflammation, especially in the visceral fat depots, contributes to systemic inflammation and promotes the development of atherosclerosis. Adipocyte fatty acid-binding protein (AFABP), a lipid chaperone abundantly secreted from the adipocytes and macrophages, is one of the key players mediating this adipose-vascular cross-talk, in part via its interaction with c-Jun NH2-terminal kinase (JNK) and activator protein-1 (AP-1) to form a positive feedback loop, and perpetuate inflammatory responses. In mice, selective JNK inactivation in the adipose tissue significantly reduced the expression of AFABP in their adipose tissue, as well as circulating AFABP levels. Importantly, fat transplant experiments showed that adipose-specific JNK inactivation in the visceral fat was sufficient to protect mice with apoE deficiency from atherosclerosis, with the beneficial effects attenuated by the continuous infusion of recombinant AFABP, supporting the role of AFABP as the link between visceral fat inflammation and atherosclerosis. In humans, raised circulating AFABP levels are associated with incident metabolic syndrome, type 2 diabetes and CVD, as well as non-alcoholic steatohepatitis, diabetic nephropathy and adverse renal outcomes, all being conditions closely related to inflammation and enhanced CV mortality. Collectively, these clinical data have provided support to AFABP as an important adipokine linking obesity, inflammation and CVD. This review will discuss recent findings on the role of AFABP in CVD and mortality, the possible underlying mechanisms, and pharmacological inhibition of AFABP as a potential strategy to combat CVD.
Collapse
Affiliation(s)
- Chi-Ho Lee
- Department of Medicine, University of Hong Kong, Hong Kong, Hong Kong.,State Key Laboratory of Pharmaceutical Biotechnology, University of Hong Kong, Hong Kong, Hong Kong
| | - David T W Lui
- Department of Medicine, University of Hong Kong, Hong Kong, Hong Kong
| | - Karen S L Lam
- Department of Medicine, University of Hong Kong, Hong Kong, Hong Kong.,State Key Laboratory of Pharmaceutical Biotechnology, University of Hong Kong, Hong Kong, Hong Kong
| |
Collapse
|
37
|
Padmanabhan S, Manjithaya R. Facets of Autophagy Based Unconventional Protein Secretion-The Road Less Traveled. Front Mol Biosci 2020; 7:586483. [PMID: 33363205 PMCID: PMC7755989 DOI: 10.3389/fmolb.2020.586483] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 09/07/2020] [Indexed: 12/28/2022] Open
Abstract
Unconventional protein secretion (UCPS) of leaderless proteins bypasses the conventional endoplasmic reticulum (ER)-Golgi route. The proportion of UCPS in the secretome varies tremendously across eukaryotes. Interestingly, macroautophagy, an intracellular recycling process that is generally involved in cargo degradation, also participates in UCPS. This emerging field of secretory mode of autophagy is underexplored and has several unanswered questions regarding the composition of players, cargo, and the mechanisms that drive it. As secretomes vary considerably across cell types and physiological conditions, the contribution of secretory autophagy in healthy and pathophysiological states remain to be elucidated. Recent studies have begun to shed light on this enigmatic process.
Collapse
Affiliation(s)
- Sreedevi Padmanabhan
- Autophagy Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, India
| | - Ravi Manjithaya
- Autophagy Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, India.,Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, India
| |
Collapse
|
38
|
Cohen MJ, Chirico WJ, Lipke PN. Through the back door: Unconventional protein secretion. Cell Surf 2020; 6:100045. [PMID: 33225116 PMCID: PMC7666356 DOI: 10.1016/j.tcsw.2020.100045] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 09/08/2020] [Accepted: 09/08/2020] [Indexed: 12/19/2022] Open
Abstract
Proteins are secreted from eukaryotic cells by several mechanisms besides the well-characterized classical secretory system. Proteins destined to enter the classical secretory system contain a signal peptide for translocation into the endoplasmic reticulum. However, many proteins lacking a signal peptide are secreted nonetheless. Contrary to conventional belief, these proteins are not just released as a result of membrane damage leading to cell leakage, but are actively packaged for secretion in alternative pathways. They are called unconventionally secreted proteins, and the best-characterized are from fungi and mammals. These proteins have extracellular functions including cell signaling, immune modulation, as well as moonlighting activities different from their well-described intracellular functions. Among the pathways for unconventional secretion are direct transfer across the plasma membrane, release within plasma membrane-derived microvesicles, use of elements of autophagy, or secretion from endosomal/multivesicular body-related components. We review the fungal and metazoan unconventional secretory pathways and their regulation, and propose experimental criteria to identify their mode of secretion.
Collapse
Affiliation(s)
- Michael J. Cohen
- The Graduate Center of the City University of New York, United States
- Biology Department, Brooklyn College of the City University of New York, United States
| | - William J. Chirico
- Department of Cell Biology, Molecular and Cellular Biology Program, SUNY Downstate Medical Center, United States
| | - Peter N. Lipke
- The Graduate Center of the City University of New York, United States
- Biology Department, Brooklyn College of the City University of New York, United States
| |
Collapse
|
39
|
Adipocyte lipolysis: from molecular mechanisms of regulation to disease and therapeutics. Biochem J 2020; 477:985-1008. [PMID: 32168372 DOI: 10.1042/bcj20190468] [Citation(s) in RCA: 114] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 02/19/2020] [Accepted: 02/26/2020] [Indexed: 12/20/2022]
Abstract
Fatty acids (FAs) are stored safely in the form of triacylglycerol (TAG) in lipid droplet (LD) organelles by professional storage cells called adipocytes. These lipids are mobilized during adipocyte lipolysis, the fundamental process of hydrolyzing TAG to FAs for internal or systemic energy use. Our understanding of adipocyte lipolysis has greatly increased over the past 50 years from a basic enzymatic process to a dynamic regulatory one, involving the assembly and disassembly of protein complexes on the surface of LDs. These dynamic interactions are regulated by hormonal signals such as catecholamines and insulin which have opposing effects on lipolysis. Upon stimulation, patatin-like phospholipase domain containing 2 (PNPLA2)/adipocyte triglyceride lipase (ATGL), the rate limiting enzyme for TAG hydrolysis, is activated by the interaction with its co-activator, alpha/beta hydrolase domain-containing protein 5 (ABHD5), which is normally bound to perilipin 1 (PLIN1). Recently identified negative regulators of lipolysis include G0/G1 switch gene 2 (G0S2) and PNPLA3 which interact with PNPLA2 and ABHD5, respectively. This review focuses on the dynamic protein-protein interactions involved in lipolysis and discusses some of the emerging concepts in the control of lipolysis that include allosteric regulation and protein turnover. Furthermore, recent research demonstrates that many of the proteins involved in adipocyte lipolysis are multifunctional enzymes and that lipolysis can mediate homeostatic metabolic signals at both the cellular and whole-body level to promote inter-organ communication. Finally, adipocyte lipolysis is involved in various diseases such as cancer, type 2 diabetes and fatty liver disease, and targeting adipocyte lipolysis is of therapeutic interest.
Collapse
|
40
|
Dimou S, Martzoukou O, Dionysopoulou M, Bouris V, Amillis S, Diallinas G. Translocation of nutrient transporters to cell membrane via Golgi bypass in Aspergillus nidulans. EMBO Rep 2020; 21:e49929. [PMID: 32452614 DOI: 10.15252/embr.201949929] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 04/15/2020] [Accepted: 04/24/2020] [Indexed: 02/06/2023] Open
Abstract
Nutrient transporters, being polytopic membrane proteins, are believed, but not formally shown, to traffic from their site of synthesis, the ER, to the plasma membrane through Golgi-dependent vesicular trafficking. Here, we develop a novel genetic system to investigate the trafficking of a neosynthesized model transporter, the well-studied UapA purine transporter of Aspergillus nidulans. We show that sorting of neosynthesized UapA to the plasma membrane (PM) bypasses the Golgi and does not necessitate key Rab GTPases, AP adaptors, microtubules or endosomes. UapA PM localization is found to be dependent on functional COPII vesicles, actin polymerization, clathrin heavy chain and the PM t-SNARE SsoA. Actin polymerization proved to primarily affect COPII vesicle formation, whereas the essential role of ClaH seems indirect and less clear. We provide evidence that other evolutionary and functionally distinct transporters of A. nidulans also follow the herein identified Golgi-independent trafficking route of UapA. Importantly, our findings suggest that specific membrane cargoes drive the formation of distinct COPII subpopulations that bypass the Golgi to be sorted non-polarly to the PM, and thus serving house-keeping cell functions.
Collapse
Affiliation(s)
- Sofia Dimou
- Department of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Olga Martzoukou
- Department of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Vangelis Bouris
- Department of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Sotiris Amillis
- Department of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - George Diallinas
- Department of Biology, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
41
|
Han X, Caron JM, Brooks PC. Cryptic collagen elements as signaling hubs in the regulation of tumor growth and metastasis. J Cell Physiol 2020; 235:9005-9020. [PMID: 32400053 DOI: 10.1002/jcp.29752] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 03/26/2020] [Accepted: 04/17/2020] [Indexed: 12/23/2022]
Abstract
Structural remodeling of the extracellular matrix is a well-established process associated with tumor growth and metastasis. Tumor and stromal cells that compose the tumor mass function cooperatively to promote the malignant phenotype in part by physically interacting with intact and structurally altered matrix proteins. To this end, collagen represents the most abundant component of the extracellular matrix and is known to control the behavior of histologically distinct tumor types as well as a diversity of stromal cells. Although a significant molecular understanding has been established concerning how cellular interactions with intact collagen govern signaling pathways that control tumor progression, considerably less is known concerning how interactions with cryptic or hidden regions within remodeled collagen may selectively alter signaling cascades, or whether inhibition of these cryptic signaling pathways may represent clinically effective therapeutic strategies. Here, we review the emerging evidence concerning the possible mechanisms for the selective generation of cryptic or hidden elements within collagen and their potential cell surface receptors that may facilitate signal transduction. We discuss the concept that cellular communication links between cell surface receptors and these cryptic collagen elements may serve as functional signaling hubs that coordinate multiple signaling pathways operating within both tumor and stromal cells. Finally, we provide examples to help illustrate the possibility that direct targeting of these unique cryptic signaling hubs may lead to the development of more effective therapeutic strategies to control tumor growth and metastasis.
Collapse
Affiliation(s)
- XiangHua Han
- Maine Medical Center Research Institute, Center for Molecular Medicine, Scarborough, Maine
| | - Jennifer M Caron
- Maine Medical Center Research Institute, Center for Molecular Medicine, Scarborough, Maine
| | - Peter C Brooks
- Maine Medical Center Research Institute, Center for Molecular Medicine, Scarborough, Maine
| |
Collapse
|
42
|
Zhang M, Liu L, Lin X, Wang Y, Li Y, Guo Q, Li S, Sun Y, Tao X, Zhang D, Lv X, Zheng L, Ge L. A Translocation Pathway for Vesicle-Mediated Unconventional Protein Secretion. Cell 2020; 181:637-652.e15. [PMID: 32272059 DOI: 10.1016/j.cell.2020.03.031] [Citation(s) in RCA: 125] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 01/22/2020] [Accepted: 03/11/2020] [Indexed: 12/13/2022]
Abstract
Many cytosolic proteins lacking a signal peptide, called leaderless cargoes, are secreted through unconventional secretion. Vesicle trafficking is a major pathway involved. It is unclear how leaderless cargoes enter into the vesicle. Here, we find a translocation pathway regulating vesicle entry and secretion of leaderless cargoes. We identify TMED10 as a protein channel for the vesicle entry and secretion of many leaderless cargoes. The interaction of TMED10 C-terminal region with a motif in the cargo accounts for the selective release of the cargoes. In an in vitro reconstitution assay, TMED10 directly mediates the membrane translocation of leaderless cargoes into the liposome, which is dependent on protein unfolding and enhanced by HSP90s. In the cell, TMED10 localizes on the endoplasmic reticulum (ER)-Golgi intermediate compartment and directs the entry of cargoes into this compartment. Furthermore, cargo induces the formation of TMED10 homo-oligomers which may act as a protein channel for cargo translocation.
Collapse
Affiliation(s)
- Min Zhang
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Lei Liu
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Xubo Lin
- Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing 100191, China
| | - Yang Wang
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Ying Li
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Qing Guo
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Shulin Li
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yuxin Sun
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Xuan Tao
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Di Zhang
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Xiachen Lv
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Li Zheng
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Liang Ge
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
43
|
Dou HX, Wang T, Su HX, Gao DD, Xu YC, Li YX, Wang HY. Exogenous FABP4 interferes with differentiation, promotes lipolysis and inflammation in adipocytes. Endocrine 2020; 67:587-596. [PMID: 31845180 DOI: 10.1007/s12020-019-02157-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Accepted: 12/09/2019] [Indexed: 12/14/2022]
Abstract
PURPOSE Fatty acid binding protein 4 (FABP4) has been demonstrated to be secreted from adipocytes in an unconventional pathway associated with lipolysis. Circulating FABP4 is elevated in metabolic disorders and has been shown to affect various peripheral cells such as pancreatic β-cells, hepatocytes and macrophages, but its effects on adipocytes remains unclear. The aim of this study was to investigate the effects of exogenous FABP4 (eFABP4) on adipocyte differentiation and function. METHODS 3T3-L1 pre-adipocytes or mature adipocytes were treated with recombinant FABP4 in the absence or presence of FABP4 inhibitor I-9/p38 MAPK inhibitor SB203580; Meanwhile male C57BL/6J mice were subcutaneously injected twice a day with recombinant FABP4 (0.35 mg/kg) with or without I-9 (50 mg/kg) for 2 weeks. The effects of eFABP4 on differentiation, lipolysis and inflammation were determined by triglyceride measurement or lipolysis assay, western blotting, or RT-qPCR analysis. RESULTS eFABP4 treatment significantly reduced intracellular triglyceride content and decreased expression of adipogenic markers peroxisome proliferator-activated receptor gamma (PPARγ), CCAAT/enhancer binding protein alpha (C/EBPα), intracellular FABP4, and adiponectin in 3T3-L1 cells. Besides, eFABP4 promoted lipolysis and inflammation in differentiated 3T3-L1 adipocytes as well as in adipose tissue of eFABP4-treated C57BL/6J mice, with elevated gene expression of monocyte chemoattractant protein (MCP)-1, tumor necrosis factor (TNF)-α, and elevated protein expression of adipose triglyceride lipase (ATGL), phosphorylation of hormone-sensitive lipase (HSL) (Ser-660), p38, and nuclear factor-kappa B (NF-κB). The pro-inflammatory and pro-lipolytic effects of eFABP4 could be reversed by SB203580/I-9. CONCLUSIONS These findings indicate that eFABP4 interferes with adipocyte differentiation, induces p38/HSL mediated lipolysis and p38/NF-κB mediated inflammation in adipocytes in vitro and in vivo.
Collapse
Affiliation(s)
- Hui-Xia Dou
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Ting Wang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Hai-Xia Su
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Ding-Ding Gao
- School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Ye-Chun Xu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Ying-Xia Li
- School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - He-Yao Wang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, 100049, Beijing, China.
| |
Collapse
|
44
|
Walenna NF, Kurihara Y, Chou B, Ishii K, Soejima T, Hiromatsu K. Chlamydia pneumoniae infection-induced endoplasmic reticulum stress causes fatty acid-binding protein 4 secretion in murine adipocytes. J Biol Chem 2020; 295:2713-2723. [PMID: 31992597 DOI: 10.1074/jbc.ra119.010683] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 01/23/2020] [Indexed: 12/28/2022] Open
Abstract
Fatty acid-binding protein 4 (FABP4) is predominantly expressed in adipocytes and macrophages and regulates metabolic and inflammatory pathways. FABP4 is secreted from adipocytes during lipolysis, and elevated circulating FABP4 levels are associated with obesity, metabolic disease, and cardiac dysfunction. We previously reported that the bacterial respiratory pathogen Chlamydia pneumoniae infects murine adipocytes and exploits host FABP4 to mobilize fat and replicate within adipocytes. However, whether C. pneumoniae induces FABP4 secretion from adipocytes has not been determined. Here, we show that FABP4 is actively secreted by murine adipocytes upon C. pneumoniae infection. Chemical inhibition of lipase activity and genetic deficiency of hormone-sensitive lipase blocked FABP4 secretion from C. pneumoniae-infected adipocytes. Mechanistically, C. pneumoniae infection induced endoplasmic reticulum (ER) stress and the unfolded protein response (UPR), resulting in elevated levels of mitochondrial reactive oxygen species and cytosolic Ca2+ Of note, exposure to a mitochondrial reactive oxygen species-specific scavenger, MitoTEMPO, reduced FABP4 release from C. pneumoniae-infected adipocytes. Furthermore, treatment with azoramide, which protects cells against ER stress, decreased FABP4 release from C. pneumoniae-infected adipocytes. Using gene silencing of CHOP (C/EBP homologous protein), a central regulator of ER stress, we further validated the role of C. pneumoniae infection-induced ER stress/UPR in promoting FABP4 secretion. Overall, these results indicate that C. pneumoniae infection robustly induces FABP4 secretion from adipocytes by stimulating ER stress/UPR. Our findings shed additional light on the etiological link between C. pneumoniae infection and metabolic syndrome.
Collapse
Affiliation(s)
- Nirwana Fitriani Walenna
- Department of Microbiology & Immunology, Fukuoka University Faculty of Medicine, Fukuoka 814-0180, Japan; Department of Bacteriology, Graduate School of Medical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan
| | - Yusuke Kurihara
- Department of Microbiology & Immunology, Fukuoka University Faculty of Medicine, Fukuoka 814-0180, Japan
| | - Bin Chou
- Department of Microbiology & Immunology, Fukuoka University Faculty of Medicine, Fukuoka 814-0180, Japan
| | - Kazunari Ishii
- Department of Microbiology & Immunology, Fukuoka University Faculty of Medicine, Fukuoka 814-0180, Japan
| | - Toshinori Soejima
- Department of Microbiology & Immunology, Fukuoka University Faculty of Medicine, Fukuoka 814-0180, Japan
| | - Kenji Hiromatsu
- Department of Microbiology & Immunology, Fukuoka University Faculty of Medicine, Fukuoka 814-0180, Japan.
| |
Collapse
|
45
|
Gao L, Mei S, Zhang S, Qin Q, Li H, Liao Y, Fan H, Liu Z, Zhu H. Cardio-renal Exosomes in Myocardial Infarction Serum Regulate Proangiogenic Paracrine Signaling in Adipose Mesenchymal Stem Cells. Am J Cancer Res 2020; 10:1060-1073. [PMID: 31938051 PMCID: PMC6956822 DOI: 10.7150/thno.37678] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 10/21/2019] [Indexed: 12/22/2022] Open
Abstract
Rationale: Mesenchymal stem cells (MSCs) play important roles in tissue repair and regeneration. However, the molecular mechanisms underlying MSCs activation remain largely unknown, thus hindering their clinical translation. Exosomes are small vesicles that act as intercellular messengers, and their potential for stem cell activation in pathological conditions has not been fully characterized yet. Here, we aim to investigate whether serum exosomes are involved in the remote activation of MSCs after myocardial infarction (MI). Methods: We established MI mouse model by ligating the left anterior descending branch of the coronary artery. Afterwards, serum exosomes were isolated from control (Con Exo) and MI mice (MI Exo) by differential centrifugation. Exosomes were characterized through transmission electron microscopy and nanoparticle tracking analysis. The cell proliferation rate was evaluated by CCK-8 and EdU incorporation assays. Exosomal miRNA and protein levels were assessed using qRT-PCR and western blotting, respectively. VEGF levels in the supernatant and serum were quantified by ELISA. Matrigel plug and tube formation assays were used to evaluate angiogenesis. To explore miR-1956 roles, overexpression and knock-down experiments were performed using mimic and inhibitor, respectively. Finally, miR-1956 target genes were confirmed using the luciferase reporter assay. Results: Both types of exosomes exhibited typical characteristics and could be internalized by adipose-derived MSCs (ADMSCs). MI Exo enhanced ADMSCs proliferation through the activation of ERK1/2. Gain- and loss-of-function studies allowed the validation of miR-1956 (enriched in MI Exo) as the functional messenger that stimulates ADMSCs-mediated angiogenesis and paracrine VEGF signaling, by downregulating Notch-1. Finally, we found that the ischemic myocardium and kidney may be the main sources that release serum exosomes after MI. Conclusions: Cardio-renal exosomes deliver miR-1956 and activate paracrine proangiogenic VEGF signaling in ADMSCs after MI; this process also involves Notch-1, which functions as the core mediator.
Collapse
|
46
|
Bassaganyas L, Popa SJ, Horlbeck M, Puri C, Stewart SE, Campelo F, Ashok A, Butnaru CM, Brouwers N, Heydari K, Ripoche J, Weissman J, Rubinsztein DC, Schekman R, Malhotra V, Moreau K, Villeneuve J. New factors for protein transport identified by a genome-wide CRISPRi screen in mammalian cells. J Cell Biol 2019; 218:3861-3879. [PMID: 31488582 PMCID: PMC6829651 DOI: 10.1083/jcb.201902028] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 06/16/2019] [Accepted: 08/12/2019] [Indexed: 12/11/2022] Open
Abstract
Protein and membrane trafficking pathways are critical for cell and tissue homeostasis. Traditional genetic and biochemical approaches have shed light on basic principles underlying these processes. However, the list of factors required for secretory pathway function remains incomplete, and mechanisms involved in their adaptation poorly understood. Here, we present a powerful strategy based on a pooled genome-wide CRISPRi screen that allowed the identification of new factors involved in protein transport. Two newly identified factors, TTC17 and CCDC157, localized along the secretory pathway and were found to interact with resident proteins of ER-Golgi membranes. In addition, we uncovered that upon TTC17 knockdown, the polarized organization of Golgi cisternae was altered, creating glycosylation defects, and that CCDC157 is an important factor for the fusion of transport carriers to Golgi membranes. In conclusion, our work identified and characterized new actors in the mechanisms of protein transport and secretion and opens stimulating perspectives for the use of our platform in physiological and pathological contexts.
Collapse
Affiliation(s)
- Laia Bassaganyas
- Department of Medical Genetics, National Institute for Health Research Cambridge Biomedical Research Centre, and Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
| | - Stephanie J Popa
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Max Horlbeck
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA
| | - Claudia Puri
- Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| | - Sarah E Stewart
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Felix Campelo
- Institut de Ciencies Fotoniques, Barcelona Institute of Science and Technology, Castelldefels, Spain
| | - Anupama Ashok
- Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Cristian M Butnaru
- Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain
- Department of Photonic Investigations, Center of Advanced Laser Technologies, National Institute for Laser, Plasma and Radiation Physics, Magurele, Romania
| | - Nathalie Brouwers
- Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain
| | | | - Jean Ripoche
- Institut National de la Sante et de la Recherche Medicale U1026, Université de Bordeaux, Bordeaux, France
| | - Jonathan Weissman
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA
| | - David C Rubinsztein
- Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
- UK Dementia Research Institute, Cambridge, UK
| | - Randy Schekman
- Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA
| | - Vivek Malhotra
- Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Kevin Moreau
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Julien Villeneuve
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| |
Collapse
|
47
|
Xu H, Diolintzi A, Storch J. Fatty acid-binding proteins: functional understanding and diagnostic implications. Curr Opin Clin Nutr Metab Care 2019; 22:407-412. [PMID: 31503024 PMCID: PMC9940447 DOI: 10.1097/mco.0000000000000600] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW Fatty acid-binding proteins (FABPs) are a family of small, abundant proteins with highly tissue-specific expression patterns whose different functions remain incompletely understood. The purpose of this review is to summarize recent findings regarding FABP functions and mechanisms of action, including their potential utilization as serum markers of tissue-specific metabolic diseases. RECENT FINDINGS FABPs are important not only in their tissues of origin but also appear to influence the metabolism and function of tissues distal to their sites of expression. This may be secondary to metabolic changes in their primary tissues, and/or a result of FABP secretion from these tissues leading to effects on distal sites. Their levels in the circulation are increasingly explored as potential biomarkers for tissue-specific disease prognosis and progression. SUMMARY The nine fatty acid-binding members of the FABP family have unique tissue-specific functions and important secondary effects on tissues in which they are not expressed. For many of the FABPs, circulating levels may be indicative of disease processes related to their primary tissues, and may influence physiological function in distal tissues.
Collapse
Affiliation(s)
- Heli Xu
- Department of Nutritional Sciences, Rutgers University, New Brunswick,
- Rutgers Center for Lipid Research, New Jersey, USA
| | - Anastasia Diolintzi
- Department of Kinesiology and Health, New Jersey, USA
- Rutgers Center for Lipid Research, New Jersey, USA
| | - Judith Storch
- Department of Nutritional Sciences, Rutgers University, New Brunswick,
- Rutgers Center for Lipid Research, New Jersey, USA
| |
Collapse
|
48
|
Scheja L, Heeren J. The endocrine function of adipose tissues in health and cardiometabolic disease. Nat Rev Endocrinol 2019; 15:507-524. [PMID: 31296970 DOI: 10.1038/s41574-019-0230-6] [Citation(s) in RCA: 347] [Impact Index Per Article: 69.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/17/2019] [Indexed: 12/16/2022]
Abstract
In addition to their role in glucose and lipid metabolism, adipocytes respond differentially to physiological cues or metabolic stress by releasing endocrine factors that regulate diverse processes, such as energy expenditure, appetite control, glucose homeostasis, insulin sensitivity, inflammation and tissue repair. Both energy-storing white adipocytes and thermogenic brown and beige adipocytes secrete hormones, which can be peptides (adipokines), lipids (lipokines) and exosomal microRNAs. Some of these factors have defined targets; for example, adiponectin and leptin signal through their respective receptors that are expressed in multiple organs. For other adipocyte hormones, receptors are more promiscuous or remain to be identified. Furthermore, many of these hormones are also produced by other organs and tissues, which makes defining the endocrine contribution of adipose tissues a challenge. In this Review, we discuss the functional role of adipose tissue-derived endocrine hormones for metabolic adaptations to the environment and we highlight how these factors contribute to the development of cardiometabolic diseases. We also cover how this knowledge can be translated into human therapies. In addition, we discuss recent findings that emphasize the endocrine role of white versus thermogenic adipocytes in conditions of health and disease.
Collapse
Affiliation(s)
- Ludger Scheja
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Joerg Heeren
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
49
|
Josephrajan A, Hertzel AV, Bohm EK, McBurney MW, Imai SI, Mashek DG, Kim DH, Bernlohr DA. Unconventional Secretion of Adipocyte Fatty Acid Binding Protein 4 Is Mediated By Autophagic Proteins in a Sirtuin-1-Dependent Manner. Diabetes 2019; 68:1767-1777. [PMID: 31171562 PMCID: PMC6702637 DOI: 10.2337/db18-1367] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Accepted: 05/29/2019] [Indexed: 12/11/2022]
Abstract
Fatty acid binding protein 4 (FABP4) is a leaderless lipid carrier protein primarily expressed by adipocytes and macrophages that not only functions intracellularly but is also secreted. The secretion is mediated via unconventional mechanism(s), and in a variety of species, metabolic dysfunction is correlated with elevated circulating FABP4 levels. In diabetic animals, neutralizing antibodies targeting serum FABP4 increase insulin sensitivity and attenuate hepatic glucose output, suggesting the functional importance of circulating FABP4. Using animal and cell-based models, we show that FABP4 is secreted from white, but not brown, adipose tissue in response to lipolytic stimulation in a sirtuin-1 (SIRT1)-dependent manner via a mechanism that requires some, but not all, autophagic components. Silencing of early autophagic genes such as Ulk1/2, Fip200, or Beclin-1 or chemical inhibition of ULK1/2 or VPS34 attenuated secretion, while Atg5 knockdown potentiated FABP4 release. Genetic knockout of Sirt1 diminished secretion, and serum FABP4 levels were undetectable in Sirt1 knockout mice. In addition, blocking SIRT1 by EX527 attenuated secretion while activating SIRT1 by resveratrol-potentiated secretion. These studies suggest that FABP4 secretion from adipocytes is regulated by SIRT1 and requires early autophagic components.
Collapse
Affiliation(s)
- Ajeetha Josephrajan
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN
| | - Ann V Hertzel
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN
| | - Ellie K Bohm
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN
| | - Michael W McBurney
- Department of Biochemistry, Microbiology and Immunology, The University of Ottawa, Ottawa, Ontario, Canada
| | - Shin-Ichiro Imai
- Department of Developmental Biology, Washington University in St. Louis, St. Louis, MO
| | - Douglas G Mashek
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN
| | - Do-Hyung Kim
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN
| | - David A Bernlohr
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN
- Department of Developmental Biology, Washington University in St. Louis, St. Louis, MO
| |
Collapse
|
50
|
Prentice KJ, Saksi J, Hotamisligil GS. Adipokine FABP4 integrates energy stores and counterregulatory metabolic responses. J Lipid Res 2019; 60:734-740. [PMID: 30705117 PMCID: PMC6446704 DOI: 10.1194/jlr.s091793] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Indexed: 12/15/2022] Open
Abstract
Although counterregulatory hormones and mediators of the fight-or-flight responses are well defined at many levels, how energy stores per se are integrated into this system remains an enigmatic question. Recent years have seen the adipose tissue become a central focus for mediating intracellular signaling and communication through the release of a variety of bioactive lipids and substrates, as well as various adipokines. A critical integration node among these mediators and responses is controlled by FA binding protein 4 (FABP4), also known as adipocyte protein 2 (aP2), which is highly expressed in adipose tissue and functions as a lipid chaperone protein. Recently, it was demonstrated that FABP4 is a secreted hormone that has roles in maintaining glucose homeostasis, representing a key juncture facilitating communication between energy-storage systems and distant organs to respond to life-threatening situations. However, chronic engagement of FABP4 under conditions of immunometabolic stress, such as obesity, exacerbates a number of immunometabolic diseases, including diabetes, asthma, cancer, and atherosclerosis. In both preclinical mouse models and humans, levels of circulating FABP4 have been correlated with metabolic disease incidence, and reducing FABP4 levels or activity is associated with improved metabolic health. In this review, we will discuss the intriguing emerging biology of this protein, including potential therapeutic options for targeting circulating FABP4.
Collapse
Affiliation(s)
- Kacey J Prentice
- Sabri Ülker Center for Metabolic Research Harvard T. H. Chan School of Public Health, Boston, MA; Department of Genetics and Complex Diseases Harvard T. H. Chan School of Public Health, Boston, MA
| | - Jani Saksi
- Sabri Ülker Center for Metabolic Research Harvard T. H. Chan School of Public Health, Boston, MA; Department of Genetics and Complex Diseases Harvard T. H. Chan School of Public Health, Boston, MA
| | - Gökhan S Hotamisligil
- Sabri Ülker Center for Metabolic Research Harvard T. H. Chan School of Public Health, Boston, MA; Department of Genetics and Complex Diseases Harvard T. H. Chan School of Public Health, Boston, MA; Broad Institute of Harvard and MIT Cambridge, MA.
| |
Collapse
|