1
|
Chen J, Ma J, Qi D, Wang Y, Sun X, Yang J, Sun W, Luan C, Shan Q, Liu W, Ma X. Inhibition of transglutaminase 2 inhibits ionizing radiation-induced cellular senescence in skin keratinocytes in vitro. IUBMB Life 2024; 76:1252-1263. [PMID: 39139071 DOI: 10.1002/iub.2906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 06/28/2024] [Indexed: 08/15/2024]
Abstract
Senescent cells are typically characterized by a stable proliferation arrested in dividing cells accompanied with a senescence-associated secretory phenotype (SASP). Skin cellular senescence is the primary cause of skin aging, whereas the lack of identified skin senescence markers limits our understanding of the mechanisms involved in skin aging. Recent studies have revealed that intracellular calcium signaling has emerged as a key player in regulating cellular senescence and aging. However, the implication and roles of calcium signaling in skin keratinocyte senescence remain only partially understood. In this study, we developed a model for skin keratinocyte senescence using ionizing radiation (I/R) stimulation and found that the calcium-associated gene transglutaminase 2 (TGM2) was significantly induced compared with normal control. Interestingly, inhibition of TGM2 was found to delay skin keratinocyte senescence by suppressing I/R-promoted intracellular calcium signaling, accumulation of reactive oxygen species (ROS), DNA damage, as well as NF-κB-mediated SASP secretion. Taken together, our findings demonstrate that inhibition of TGM2 contributes to bypassing I/R-induced skin keratinocyte senescence and sheds light on novel strategies against skin stresses caused by I/R.
Collapse
Affiliation(s)
- Juping Chen
- Laboratory of Intensive Care, Department of Intensive Care, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
- Department of Dermatology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Jiang Ma
- Laboratory of Intensive Care, Department of Intensive Care, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
- Department of Dermatology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Dandan Qi
- Department of Dermatology, Nanxiang Branch of Ruijin Hospital, Shanghai, China
| | - Yuxuan Wang
- Laboratory of Intensive Care, Department of Intensive Care, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
- Department of Dermatology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Xiaoming Sun
- Laboratory of Intensive Care, Department of Intensive Care, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
- Department of Dermatology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Jinghui Yang
- Laboratory of Intensive Care, Department of Intensive Care, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
- Department of Dermatology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Wentao Sun
- Laboratory of Intensive Care, Department of Intensive Care, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Changjiao Luan
- Department of Lung, The Third People's Hospital of Yangzhou, Yangzhou, China
| | - Qing Shan
- Department of Geriatrics, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Weili Liu
- Laboratory of Intensive Care, Department of Intensive Care, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Xingjie Ma
- Laboratory of Intensive Care, Department of Intensive Care, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| |
Collapse
|
2
|
Kang J, Jeon HY, Lee J, Bae S, Park GY, Min KJ, Joo J, Lee AJ, Kim HJ, Im CY, Kim EB, Lee JH, Hwang JS, Lee S, Lee JY, Navals P, Keillor JW, Ha KS, Song M. Structurally Minimalized and Druglike TGase2 Inhibitors Based on 7-Aminoquinoline-5,8-dione Scaffolds for the Treatment of Diabetic Retinopathy. J Med Chem 2024; 67:19716-19735. [PMID: 39445793 DOI: 10.1021/acs.jmedchem.4c02081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Diabetic retinopathy is a disease that can cause vision loss leading to blindness in people with diabetes. Improved methods to treat and prevent vision loss in diabetic patients are in high demand owing to limited current treatment procedures. Herein, we report a new class of transglutaminase 2 (TGase2) inhibitors for the treatment of diabetic retinopathy based on 7-aminoquinoline-5,8-dione derivatives. 7-Amino-2-phenylquinoline-5,8-dione 11 and 7-amino-2-{4-[(1-methylpiperidin-4-yl)oxy]phenyl}quinoline-5,8-dione 23 exhibited potent inhibitory activities against TGase2 in a fibrinogen array-based on-chip TGase2 activity assay and in an in situ assay in human retinal microvascular endothelial cells, with IC50 values of 5.88 and 1.12 μM in vitro, and 0.09 and 0.07 μM in situ, respectively. Pharmacokinetically favorable 7-amino-2-{4-[(1-isopropylpiperidin-4-yl)oxy] phenyl}quinoline-5,8-dione 22 inhibited vascular leakage in the retinas of streptozotocin-induced diabetic mice via oral administration. Results from the AL5 kinetic assay and a molecular docking study suggest that the inhibitors may bind to TGase2 remote from the active site.
Collapse
Affiliation(s)
- Jihee Kang
- New Drug Discovery Center (NDDC), Daegu Gyeongbuk Medical Innovation Foundation (K-MEDI hub), 80 Cheombok-ro Dong-gu, Daegu 41061, Korea
| | - Hye-Yoon Jeon
- Department of Molecular and Cellular Biochemistry, Kangwon National University School of Medicine, Chuncheon, Kangwon-do 24341, Korea
- Scripps Korea Antibody Institute, Kangwon National University Chuncheon Campus, Chuncheon, Kangwon-do 24341, Korea
| | - Jieon Lee
- New Drug Discovery Center (NDDC), Daegu Gyeongbuk Medical Innovation Foundation (K-MEDI hub), 80 Cheombok-ro Dong-gu, Daegu 41061, Korea
| | - Seri Bae
- New Drug Discovery Center (NDDC), Daegu Gyeongbuk Medical Innovation Foundation (K-MEDI hub), 80 Cheombok-ro Dong-gu, Daegu 41061, Korea
| | - Ga Young Park
- New Drug Discovery Center (NDDC), Daegu Gyeongbuk Medical Innovation Foundation (K-MEDI hub), 80 Cheombok-ro Dong-gu, Daegu 41061, Korea
| | - Kyoung-Jin Min
- New Drug Discovery Center (NDDC), Daegu Gyeongbuk Medical Innovation Foundation (K-MEDI hub), 80 Cheombok-ro Dong-gu, Daegu 41061, Korea
| | - Jeongmin Joo
- New Drug Discovery Center (NDDC), Daegu Gyeongbuk Medical Innovation Foundation (K-MEDI hub), 80 Cheombok-ro Dong-gu, Daegu 41061, Korea
| | - Ah-Jun Lee
- Department of Molecular and Cellular Biochemistry, Kangwon National University School of Medicine, Chuncheon, Kangwon-do 24341, Korea
| | - Hyo-Ji Kim
- New Drug Discovery Center (NDDC), Daegu Gyeongbuk Medical Innovation Foundation (K-MEDI hub), 80 Cheombok-ro Dong-gu, Daegu 41061, Korea
| | - Chun Young Im
- New Drug Discovery Center (NDDC), Daegu Gyeongbuk Medical Innovation Foundation (K-MEDI hub), 80 Cheombok-ro Dong-gu, Daegu 41061, Korea
| | - Eun-Bin Kim
- Department of Molecular and Cellular Biochemistry, Kangwon National University School of Medicine, Chuncheon, Kangwon-do 24341, Korea
| | - Ji Hun Lee
- New Drug Discovery Center (NDDC), Daegu Gyeongbuk Medical Innovation Foundation (K-MEDI hub), 80 Cheombok-ro Dong-gu, Daegu 41061, Korea
| | - Ji Sun Hwang
- New Drug Discovery Center (NDDC), Daegu Gyeongbuk Medical Innovation Foundation (K-MEDI hub), 80 Cheombok-ro Dong-gu, Daegu 41061, Korea
| | - Seungju Lee
- New Drug Discovery Center (NDDC), Daegu Gyeongbuk Medical Innovation Foundation (K-MEDI hub), 80 Cheombok-ro Dong-gu, Daegu 41061, Korea
| | - Jee-Young Lee
- New Drug Discovery Center (NDDC), Daegu Gyeongbuk Medical Innovation Foundation (K-MEDI hub), 80 Cheombok-ro Dong-gu, Daegu 41061, Korea
| | - Pauline Navals
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Jeffrey W Keillor
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Kwon-Soo Ha
- Department of Molecular and Cellular Biochemistry, Kangwon National University School of Medicine, Chuncheon, Kangwon-do 24341, Korea
| | - Minsoo Song
- New Drug Discovery Center (NDDC), Daegu Gyeongbuk Medical Innovation Foundation (K-MEDI hub), 80 Cheombok-ro Dong-gu, Daegu 41061, Korea
| |
Collapse
|
3
|
Meling MT, Kleppa L, Besser HA, Khosla C, du Pré MF, Sollid LM. Enterocyte-Derived and Catalytically Active Transglutaminase 2 in the Gut Lumen of Mice: Implications for Celiac Disease. Gastroenterology 2024; 167:1026-1028.e4. [PMID: 38825048 DOI: 10.1053/j.gastro.2024.05.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 04/29/2024] [Accepted: 05/24/2024] [Indexed: 06/04/2024]
Affiliation(s)
- Maureen T Meling
- Norwegian Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Liv Kleppa
- Norwegian Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Harrison A Besser
- Department of Chemistry, Stanford University, Stanford, California; Stanford Medical Scientist Training Program, Stanford University School of Medicine, Stanford, California
| | - Chaitan Khosla
- Department of Chemistry, Stanford University, Stanford, California; Department of Chemical Engineering, Stanford University, Stanford, California; Sarafan ChEM-H, Stanford University, Stanford, California
| | - M Fleur du Pré
- Norwegian Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Ludvig M Sollid
- Norwegian Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
4
|
Freeberg MAT, Thatcher TH, Camus SV, Huang L, Atkinson J, Narrow W, Haak J, Dylag AM, Cowart LA, Johnson TS, Sime PJ. Transglutaminase 2 knockout mice are protected from bleomycin-induced lung fibrosis with preserved lung function and reduced metabolic derangements. Physiol Rep 2024; 12:e16012. [PMID: 38959068 PMCID: PMC11189770 DOI: 10.14814/phy2.16012] [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: 12/14/2023] [Revised: 02/27/2024] [Accepted: 02/27/2024] [Indexed: 07/05/2024] Open
Abstract
Pulmonary fibrosis is an interstitial scarring disease of the lung characterized by poor prognosis and limited treatment options. Tissue transglutaminase 2 (TG2) is believed to promote lung fibrosis by crosslinking extracellular matrix components and activating latent TGFβ. This study assessed physiologic pulmonary function and metabolic alterations in the mouse bleomycin model with TG2 genetic deletion. TG2-deficient mice demonstrated attenuated the fibrosis and preservation of lung function, with significant reduction in elastance and increases in compliance and inspiratory capacity compared to control mice treated with bleomycin. Bleomycin induced metabolic changes in the mouse lung that were consistent with increased aerobic glycolysis, including increased expression of lactate dehydrogenase A and increased production of lactate, as well as increased glutamine, glutamate, and aspartate. TG2-deficient mice treated with bleomycin exhibited similar metabolic changes but with reduced magnitude. Our results demonstrate that TG2 is required for a typical fibrosis response to injury. In the absence of TG2, the fibrotic response is biochemically similar to wild-type, but lesions are smaller and lung function is preserved. We also show for the first time that profibrotic pathways of tissue stiffening and metabolic reprogramming are interconnected, and that metabolic disruptions in fibrosis go beyond glycolysis.
Collapse
Affiliation(s)
- Margaret A. T. Freeberg
- Division of Pulmonary Disease and Critical Care MedicineVirginia Commonwealth UniversityRichmondVirginiaUSA
- Division of Pulmonary and Critical Care MedicineUniversity of RochesterRochesterNew YorkUSA
| | - Thomas H. Thatcher
- Division of Pulmonary Disease and Critical Care MedicineVirginia Commonwealth UniversityRichmondVirginiaUSA
- Division of Pulmonary and Critical Care MedicineUniversity of RochesterRochesterNew YorkUSA
| | - Sarah V. Camus
- Division of Pulmonary Disease and Critical Care MedicineVirginia Commonwealth UniversityRichmondVirginiaUSA
| | - Linghong Huang
- UCB Pharma SASloughBerkshireUK
- Present address:
Mestag TherapeuticsCambridgeUK
| | | | - Wade Narrow
- Division of Pulmonary and Critical Care MedicineUniversity of RochesterRochesterNew YorkUSA
- Present address:
Department of SurgeryUniversity of RochesterRochesterNew YorkUSA
| | - Jeannie Haak
- Department of Pediatrics, Division of NeonatologyUniversity of Rochester Medical CenterRochesterNew YorkUSA
| | - Andrew M. Dylag
- Department of Pediatrics, Division of NeonatologyUniversity of Rochester Medical CenterRochesterNew YorkUSA
| | - L. Ashley Cowart
- Department of Biochemistry and Molecular BiologyVirginia Commonwealth UniversityRichmondVirginiaUSA
| | - Timothy S. Johnson
- UCB Pharma SASloughBerkshireUK
- Present address:
Mestag TherapeuticsCambridgeUK
| | - Patricia J. Sime
- Division of Pulmonary Disease and Critical Care MedicineVirginia Commonwealth UniversityRichmondVirginiaUSA
- Division of Pulmonary and Critical Care MedicineUniversity of RochesterRochesterNew YorkUSA
| |
Collapse
|
5
|
Kawaguchi S, Moukette B, Sepúlveda MN, Hayasaka T, Aonuma T, Haskell AK, Mah J, Liangpunsakul S, Tang Y, Conway SJ, Kim IM. SPRR1A is a key downstream effector of MiR-150 during both maladaptive cardiac remodeling in mice and human cardiac fibroblast activation. Cell Death Dis 2023; 14:446. [PMID: 37468478 PMCID: PMC10356860 DOI: 10.1038/s41419-023-05982-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 07/10/2023] [Accepted: 07/11/2023] [Indexed: 07/21/2023]
Abstract
MicroRNA-150 (miR-150) is conserved between rodents and humans, is significantly downregulated during heart failure (HF), and correlates with patient outcomes. We previously reported that miR-150 is protective during myocardial infarction (MI) in part by decreasing cardiomyocyte (CM) apoptosis and that proapoptotic small proline-rich protein 1a (Sprr1a) is a direct CM target of miR-150. We also showed that Sprr1a knockdown in mice improves cardiac dysfunction and fibrosis post-MI and that Sprr1a is upregulated in pathological mouse cardiac fibroblasts (CFs) from ischemic myocardium. However, the direct functional relationship between miR-150 and SPRR1A during both post-MI remodeling in mice and human CF (HCF) activation was not established. Here, using a novel miR-150 knockout;Sprr1a-hypomorphic (Sprr1ahypo/hypo) mouse model, we demonstrate that Sprr1a knockdown blunts adverse post-MI effects caused by miR-150 loss. Moreover, HCF studies reveal that SPRR1A is upregulated in hypoxia/reoxygenation-treated HCFs and is downregulated in HCFs exposed to the cardioprotective β-blocker carvedilol, which is inversely associated with miR-150 expression. Significantly, we show that the protective roles of miR-150 in HCFs are directly mediated by functional repression of profibrotic SPRR1A. These findings delineate a pivotal functional interaction between miR-150 and SPRR1A as a novel regulatory mechanism pertinent to CF activation and ischemic HF.
Collapse
Affiliation(s)
- Satoshi Kawaguchi
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Emergency Medicine, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Bruno Moukette
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
- Internal Medicine Research Unit, Pfizer Inc., Cambridge, MA, USA
| | - Marisa N Sepúlveda
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Taiki Hayasaka
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Tatsuya Aonuma
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
- Division of Cardiology, Nephrology, Pulmonology, and Neurology, Department of Internal Medicine, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Angela K Haskell
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jessica Mah
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Suthat Liangpunsakul
- Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yaoliang Tang
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Simon J Conway
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Il-Man Kim
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA.
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA.
- Krannert Cardiovascular Research Center, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
6
|
Yiu TW, Holman SR, Kaidonis X, Graham RM, Iismaa SE. Transglutaminase 2 Facilitates Murine Wound Healing in a Strain-Dependent Manner. Int J Mol Sci 2023; 24:11475. [PMID: 37511238 PMCID: PMC10380275 DOI: 10.3390/ijms241411475] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/12/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Transglutaminase 2 (TG2) plays a role in cellular processes that are relevant to wound healing, but to date no studies of wound healing in TG2 knockout mice have been reported. Here, using 129T2/SvEmsJ (129)- or C57BL/6 (B6)-backcrossed TG2 knockout mice, we show that TG2 facilitates murine wound healing in a strain-dependent manner. Early healing of in vivo cutaneous wounds and closure of in vitro scratch wounds in murine embryonic fibroblast (MEF) monolayers were delayed in 129, but not B6, TG2 knockouts, relative to their wild-type counterparts, with wound closure in 129 being faster than in B6 wild-types. A single dose of exogenous recombinant wild-type TG2 to 129 TG2-/- mice or MEFs immediately post-wounding accelerated wound closure. Neutrophil and monocyte recruitment to 129 cutaneous wounds was not affected by Tgm2 deletion up to 5 days post-wounding. Tgm2 mRNA and TG2 protein abundance were higher in 129 than in B6 wild-types and increased in abundance following cutaneous and scratch wounding. Tgm1 and factor XIIA (F13A) mRNA abundance increased post-wounding, but there was no compensation by TG family members in TG2-/- relative to TG2+/+ mice in either strain before or after wounding. 129 TG2+/+ MEF adhesion was greater and spreading was faster than that of B6 TG2+/+ MEFs, and was dependent on syndecan binding in the presence, but not absence, of RGD inhibition of integrin binding. Adhesion and spreading of 129, but not B6, TG2-/- MEFs was impaired relative to their wild-type counterparts and was accelerated by exogenous addition or transfection of TG2 protein or cDNA, respectively, and was independent of the transamidase or GTP-binding activity of TG2. Rho-family GTPase activation, central to cytoskeletal organization, was altered in 129 TG2-/- MEFs, with delayed RhoA and earlier Rac1 activation than in TG2+/+ MEFs. These findings indicate that the rate of wound healing is different between 129 and B6 mouse strains, correlating with TG2 abundance, and although not essential for wound healing, TG2 facilitates integrin- and syndecan-mediated RhoA- and Rac1-activation in fibroblasts to promote efficient wound contraction.
Collapse
Affiliation(s)
- Ting W. Yiu
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; (T.W.Y.); (S.R.H.); (X.K.)
| | - Sara R. Holman
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; (T.W.Y.); (S.R.H.); (X.K.)
| | - Xenia Kaidonis
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; (T.W.Y.); (S.R.H.); (X.K.)
| | - Robert M. Graham
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; (T.W.Y.); (S.R.H.); (X.K.)
- School of Clinical Medicine, UNSW Medicine and Health, University of New South Wales Sydney, Kensington, NSW 2052, Australia
| | - Siiri E. Iismaa
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; (T.W.Y.); (S.R.H.); (X.K.)
- School of Clinical Medicine, UNSW Medicine and Health, University of New South Wales Sydney, Kensington, NSW 2052, Australia
| |
Collapse
|
7
|
Hou A, Mohamed Ali S, Png E, Hunziker W, Tong L. Transglutaminase-2 is critical for corneal epithelial barrier function via positive regulation of Claudin-1. Ocul Surf 2023; 28:155-164. [PMID: 37037393 DOI: 10.1016/j.jtos.2023.04.002] [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: 12/22/2022] [Revised: 02/18/2023] [Accepted: 04/06/2023] [Indexed: 04/12/2023]
Abstract
PURPOSE Transglutaminase (TG)-2 is a ubiquitous multi-functional protein expressed in all living cells. The purpose of the current study was to investigate the role of TG-2 in corneal barrier function and its potential regulation of epithelial junctional proteins and transcription factors. METHODS Corneal barrier function to ions in TG-2-/- and TG-2+/+ mice was assessed by Ussing chamber assay. Hypo-osmolar water or FITC-dextran was applied on top of mouse eyes to evaluate the corneal barrier function to water and macromolecules. Western blots, qPCR and immunofluorescent staining were used to investigate the expression of tight junction proteins in TG-2-/- and TG-2+/+ mouse corneas, and also in TG-2 knockdown human corneal epithelial cells. RESULTS Corneal explants from TG-2-/- mice had a lower trans-epithelial electrical resistance compared to TG-2+/+ mice. When challenged by hypo-osmolar water, the central corneal thickness of TG-2-/- mice increased faster, and these mice had a faster rise of fluorescence in the anterior chamber after ocular exposure to FITC-dextran, compared to TG-2+/+. Claudin-1 protein and transcript levels were reduced in the cornea of TG-2-/- mice and in TG-2 knockdown human corneal epithelial cells. Slug which previously reported suppressing Claudin-1 transcription, was increased at both protein and transcript level in TG-2 knockdown cells. TG-2 and Claudin-1 protein levels were unchanged in shRNA and shTG cells after MG132 treatment, while Slug accumulated in treated cells. CONCLUSION TG-2 may positively regulate Claudin-1 through repressing Slug at transcript level, and thus it is critical for normal corneal barrier function.
Collapse
Affiliation(s)
- Aihua Hou
- Ocular Surface Research Group, Singapore Eye Research Institute, 169856, Singapore; Eye-Academic Clinical Programme, Duke-NUS Medical School, 169857, Singapore
| | - Safiah Mohamed Ali
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| | - Evelyn Png
- Ocular Surface Research Group, Singapore Eye Research Institute, 169856, Singapore
| | - Walter Hunziker
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore; SERI-IMCB Program in Retinal Angiogenic Diseases, Singapore Eye Research Institute, 169856, Singapore; Department of Physiology, National University of Singapore, 117593, Singapore
| | - Louis Tong
- Ocular Surface Research Group, Singapore Eye Research Institute, 169856, Singapore; Eye-Academic Clinical Programme, Duke-NUS Medical School, 169857, Singapore; Corneal and External Eye Disease Service, Singapore National Eye Centre, 168751, Singapore; Yong Loo Lin School of Medicine, National University of Singapore, 119228, Singapore.
| |
Collapse
|
8
|
Tissue transglutaminase exacerbates renal fibrosis via alternative activation of monocyte-derived macrophages. Cell Death Dis 2023; 14:136. [PMID: 36864028 PMCID: PMC9981766 DOI: 10.1038/s41419-023-05622-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 01/24/2023] [Accepted: 01/25/2023] [Indexed: 03/04/2023]
Abstract
Macrophages are important components in modulating homeostatic and inflammatory responses and are generally categorized into two broad but distinct subsets: classical activated (M1) and alternatively activated (M2) depending on the microenvironment. Fibrosis is a chronic inflammatory disease exacerbated by M2 macrophages, although the detailed mechanism by which M2 macrophage polarization is regulated remains unclear. These polarization mechanisms have little in common between mice and humans, making it difficult to adapt research results obtained in mice to human diseases. Tissue transglutaminase (TG2) is a known marker common to mouse and human M2 macrophages and is a multifunctional enzyme responsible for crosslinking reactions. Here we sought to identify the role of TG2 in macrophage polarization and fibrosis. In IL-4-treated macrophages derived from mouse bone marrow and human monocyte cells, the expression of TG2 was increased with enhancement of M2 macrophage markers, whereas knockout or inhibitor treatment of TG2 markedly suppressed M2 macrophage polarization. In the renal fibrosis model, accumulation of M2 macrophages in fibrotic kidney was significantly reduced in TG2 knockout or inhibitor-administrated mice, along with the resolution of fibrosis. Bone marrow transplantation using TG2-knockout mice revealed that TG2 is involved in M2 polarization of infiltrating macrophages derived from circulating monocytes and exacerbates renal fibrosis. Furthermore, the suppression of renal fibrosis in TG2-knockout mice was abolished by transplantation of wild-type bone marrow or by renal subcapsular injection of IL4-treated macrophages derived from bone marrow of wild-type, but not TG2 knockout. Transcriptome analysis of downstream targets involved in M2 macrophages polarization revealed that ALOX15 expression was enhanced by TG2 activation and promoted M2 macrophage polarization. Furthermore, the increase in the abundance of ALOX15-expressing macrophages in fibrotic kidney was dramatically suppressed in TG2-knockout mice. These findings demonstrated that TG2 activity exacerbates renal fibrosis by polarization of M2 macrophages from monocytes via ALOX15.
Collapse
|
9
|
Resolution of Eczema with Multivalent Peptides. JID INNOVATIONS 2022; 2:100142. [PMID: 36039327 PMCID: PMC9418603 DOI: 10.1016/j.xjidi.2022.100142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 06/08/2022] [Accepted: 06/10/2022] [Indexed: 11/24/2022] Open
|
10
|
Lockridge O, Schopfer LM. Naturally Occurring Epsilon Gamma Glutamyl Lysine Isopeptide Crosslinks in Human Neuroblastoma SH-SY5Y Cells. ACS OMEGA 2022; 7:21978-21986. [PMID: 35785306 PMCID: PMC9245130 DOI: 10.1021/acsomega.2c02502] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 06/07/2022] [Indexed: 05/05/2023]
Abstract
Zero-length isopeptide crosslinks between the side chains of glutamine and lysine are the product of transglutaminase activity. It is generally accepted that transglutaminase activity is dormant under physiological conditions because the calcium concentration inside cells is too low to activate transglutaminase to an open conformation with access to the catalytic triad. Traditional assays for transglutaminase activity measure incorporation of biotin pentylamine or of radiolabeled putrescine in the presence of added calcium. In this report, we identified naturally occurring isopeptide crosslinked proteins using the following steps: immunopurification of tryptic peptides by binding to anti-isopeptide antibody 81D1C2, separation of immunopurified peptides by liquid chromatography-tandem mass spectrometry, Protein Prospector database searches of mass spectrometry data for isopeptide crosslinked peptides, and manual evaluation of candidate crosslinked peptide pairs. The most labor intense step was manual evaluation. We developed criteria for accepting and rejecting candidate crosslinked peptides and showed examples of MS/MS spectra that confirm or invalidate a possible crosslink. The SH-SY5Y cells that we examined for crosslinked proteins had not been exposed to calcium and had been lysed in the presence of ethylenediaminetetraacetic acid. This precaution allows us to claim that the crosslinks we found inside the cells occurred naturally under physiological conditions. The quantity of crosslinks was very low, and the crosslinked proteins were mostly low abundance proteins. In conclusion, intracellular transglutaminase crosslinking/transamidase activity is very low but detectable. The low level of intracellular crosslinked proteins is consistent with tight regulation of transglutaminase activity.
Collapse
|
11
|
Al-U'datt DGF, Tranchant CC, Al-Dwairi A, AlQudah M, Al-Shboul O, Hiram R, Allen BG, Jaradat S, Alqbelat J, Abu-Zaiton AS. Implications of enigmatic transglutaminase 2 (TG2) in cardiac diseases and therapeutic developments. Biochem Pharmacol 2022; 201:115104. [PMID: 35617996 DOI: 10.1016/j.bcp.2022.115104] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 05/18/2022] [Accepted: 05/18/2022] [Indexed: 01/07/2023]
Abstract
Cardiac diseases are the leading cause of mortality and morbidity worldwide. Mounting evidence suggests that transglutaminases (TGs), tissue TG (TG2) in particular, are involved in numerous molecular responses underlying the pathogenesis of cardiac diseases. The TG family has several intra- and extracellular functions in the human body, including collagen cross-linking, angiogenesis, cell growth, differentiation, migration, adhesion as well as survival. TGs are thiol- and calcium-dependent acyl transferases that catalyze the formation of a covalent bond between the γ-carboxamide group of a glutamine residue and an amine group, thus increasing the stability, rigidity, and stiffness of the myocardial extracellular matrix (ECM). Excessive accumulation of cross-linked collagen leads to increase myocardial stiffness and fibrosis. Beyond TG2 extracellular protein cross-linking action, mounting evidence suggests that this pleiotropic TG isozyme may also promote fibrotic diseases through cell survival and profibrotic pathway activation at the signaling, transcriptional and translational levels. Due to its multiple functions and localizations, TG2 fulfils critical yet incompletely understood roles in myocardial fibrosis and associated heart diseases, such as cardiac hypertrophy, heart failure, and age-related myocardial stiffness under several conditions. This review summarizes current knowledge and existing gaps regarding the ECM-dependent and ECM-independent roles of TG2 and highlights the therapeutic prospects of targeting TG2 to treat cardiac diseases.
Collapse
Affiliation(s)
- Doa'a G F Al-U'datt
- Department of Physiology and Biochemistry, Faculty of Medicine, Jordan University of Science and Technology, Irbid, 22110, Jordan.
| | - Carole C Tranchant
- School of Food Science, Nutrition and Family Studies, Faculty of Health Sciences and Community Services, Université de Moncton, New Brunswick, Canada
| | - Ahmed Al-Dwairi
- Department of Physiology and Biochemistry, Faculty of Medicine, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Mohammad AlQudah
- Department of Physiology and Biochemistry, Faculty of Medicine, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Othman Al-Shboul
- Department of Physiology and Biochemistry, Faculty of Medicine, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Roddy Hiram
- Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada; Department of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Bruce G Allen
- Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada; Department of Medicine, Université de Montréal, Montreal, Quebec, Canada; Department of Pharmacology and Physiology, Université de Montréal, Montreal, Quebec, Canada; Department of Biochemistry and Molecular Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Saied Jaradat
- Princess Haya Biotechnology Center, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Jenan Alqbelat
- Department of Physiology and Biochemistry, Faculty of Medicine, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Ahmed S Abu-Zaiton
- Department of Biological Sciences, Al al-bayt University, Al-Mafraq, Jordan
| |
Collapse
|
12
|
Lénárt K, Bankó C, Ujlaki G, Póliska S, Kis G, Csősz É, Antal M, Bacso Z, Bai P, Fésüs L, Mádi A. Tissue Transglutaminase Knock-Out Preadipocytes and Beige Cells of Epididymal Fat Origin Possess Decreased Mitochondrial Functions Required for Thermogenesis. Int J Mol Sci 2022; 23:5175. [PMID: 35563567 PMCID: PMC9105016 DOI: 10.3390/ijms23095175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/29/2022] [Accepted: 04/29/2022] [Indexed: 11/24/2022] Open
Abstract
Beige adipocytes with thermogenic function are activated during cold exposure in white adipose tissue through the process of browning. These cells, similar to brown adipocytes, dissipate stored chemical energy in the form of heat with the help of uncoupling protein 1 (UCP1). Recently, we have shown that tissue transglutaminase (TG2) knock-out mice have decreased cold tolerance in parallel with lower utilization of their epididymal adipose tissue and reduced browning. To learn more about the thermogenic function of this fat depot, we isolated preadipocytes from the epididymal adipose tissue of wild-type and TG2 knock-out mice and differentiated them in the beige direction. Although differentiation of TG2 knock-out preadipocytes is phenotypically similar to the wild-type cells, the mitochondria of the knock-out beige cells have multiple impairments including an altered electron transport system generating lower electrochemical potential difference, reduced oxygen consumption, lower UCP1 protein content, and a higher portion of fragmented mitochondria. Most of these differences are present in preadipocytes as well, and the differentiation process cannot overcome the functional disadvantages completely. TG2 knock-out beige adipocytes produce more iodothyronine deiodinase 3 (DIO3) which may inactivate thyroid hormones required for the establishment of optimal mitochondrial function. The TG2 knock-out preadipocytes and beige cells are both hypometabolic as compared with the wild-type controls which may also be explained by the lower expression of solute carrier proteins SLC25A45, SLC25A47, and SLC25A42 which transport acylcarnitine, Co-A, and amino acids into the mitochondrial matrix. As a consequence, the mitochondria in TG2 knock-out beige adipocytes probably cannot reach the energy-producing threshold required for normal thermogenic functions, which may contribute to the decreased cold tolerance of TG2 knock-out mice.
Collapse
Affiliation(s)
- Kinga Lénárt
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Egyetem ter 1., H-4032 Debrecen, Hungary; (K.L.); (S.P.); (É.C.); (L.F.)
- Doctoral School of Molecular Cell and Immune Biology, University of Debrecen, Egyetem ter 1., H-4032 Debrecen, Hungary;
| | - Csaba Bankó
- Doctoral School of Molecular Cell and Immune Biology, University of Debrecen, Egyetem ter 1., H-4032 Debrecen, Hungary;
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Egyetem ter 1., H-4032 Debrecen, Hungary;
| | - Gyula Ujlaki
- NKFIH-DE Lendület Laboratory of Cellular Metabolism, Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Egyetem ter 1., H-4032 Debrecen, Hungary; (G.U.); (P.B.)
| | - Szilárd Póliska
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Egyetem ter 1., H-4032 Debrecen, Hungary; (K.L.); (S.P.); (É.C.); (L.F.)
| | - Gréta Kis
- Department of Anatomy, Histology Embryology, Faculty of Medicine, University of Debrecen, Egyetem ter 1., H-4032 Debrecen, Hungary; (G.K.); (M.A.)
| | - Éva Csősz
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Egyetem ter 1., H-4032 Debrecen, Hungary; (K.L.); (S.P.); (É.C.); (L.F.)
| | - Miklós Antal
- Department of Anatomy, Histology Embryology, Faculty of Medicine, University of Debrecen, Egyetem ter 1., H-4032 Debrecen, Hungary; (G.K.); (M.A.)
| | - Zsolt Bacso
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Egyetem ter 1., H-4032 Debrecen, Hungary;
| | - Péter Bai
- NKFIH-DE Lendület Laboratory of Cellular Metabolism, Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Egyetem ter 1., H-4032 Debrecen, Hungary; (G.U.); (P.B.)
- Research Center for Molecular Medicine, Faculty of Medicine, University of Debrecen, Egyetem ter 1., H-4032 Debrecen, Hungary
| | - László Fésüs
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Egyetem ter 1., H-4032 Debrecen, Hungary; (K.L.); (S.P.); (É.C.); (L.F.)
| | - András Mádi
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Egyetem ter 1., H-4032 Debrecen, Hungary; (K.L.); (S.P.); (É.C.); (L.F.)
| |
Collapse
|
13
|
Lockhart-Cairns MP, Cain SA, Dajani R, Steer R, Thomson J, Alanazi YF, Kielty CM, Baldock C. Latent TGFβ complexes are transglutaminase cross-linked to fibrillin to facilitate TGFβ activation. Matrix Biol 2022; 107:24-39. [PMID: 35122964 PMCID: PMC8932414 DOI: 10.1016/j.matbio.2022.01.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 01/21/2022] [Accepted: 01/25/2022] [Indexed: 11/24/2022]
Abstract
TGFβ is regulated via the formation latent complexes in the extracellular matrix. Fibrillin-1 is a substrate for transglutaminase-2 which forms a covalent link between fibrillin-1 and latent TGFβ complexes. Formation of the cross-link increases TGFβ activation in cell-based assays. Fibrillin may direct the latent TGFβ complexes to the cell surface for activation. The structure of the cross-linked LTBP1-fibrillin complex has a perpendicular arrangement to enable bridging long-range interactions between the matrix and cell surface.
TGFβ superfamily members are potent growth factors in the extracellular matrix with essential roles in all aspects of cellular behaviour. Latent TGFβ binding proteins (LTBPs) are co-expressed with TGFβ, essential for correct folding and secretion of the growth factor, to form large latent complexes. These large latent complexes bind extracellular proteins such as fibrillin for sequestration of TGFβ in the matrix, essential for normal tissue function, and dysregulated TGFβ signalling is a hallmark of many fibrillinopathies. Transglutaminase-2 (TG2) cross-linking of LTBPs is known to play a role in TGFβ activation but the underlying molecular mechanisms are not resolved. Here we show that fibrillin is a matrix substrate for TG2 and that TG2 cross-linked complexes can be formed between fibrillin and LTBP-1 and -3, and their latent TGFβ complexes. The structure of the fibrillin-LTBP1 complex shows that the two elongated proteins interact in a perpendicular arrangement which would allow them to form distal interactions between the matrix and the cell surface. Formation of the cross-link with fibrillin does not change the interaction between latent TGFβ and integrin αVβ6 but does increase TGFβ activation in cell-based assays. The activating effect may be due to direction of the latent complexes to the cell surface by fibrillin, as competition with heparan sulphate can ameliorate the activating effect. Together, these data support that TGFβ activation can be enhanced by covalent tethering of LTBPs to the matrix via fibrillin.
Collapse
Affiliation(s)
- Michael P Lockhart-Cairns
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT, UK
| | - Stuart A Cain
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT, UK
| | - Rana Dajani
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT, UK
| | - Ruth Steer
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT, UK
| | - Jennifer Thomson
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT, UK
| | - Yasmene F Alanazi
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT, UK
| | - Cay M Kielty
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT, UK
| | - Clair Baldock
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT, UK.
| |
Collapse
|
14
|
Wang Z, Moresco P, Yan R, Li J, Gao Y, Biasci D, Yao M, Pearson J, Hechtman JF, Janowitz T, Zaidi RM, Weiss MJ, Fearon DT. Carcinomas assemble a filamentous CXCL12-keratin-19 coating that suppresses T cell-mediated immune attack. Proc Natl Acad Sci U S A 2022; 119:e2119463119. [PMID: 35046049 PMCID: PMC8794816 DOI: 10.1073/pnas.2119463119] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 12/13/2021] [Indexed: 12/26/2022] Open
Abstract
Cancer immunotherapy frequently fails because most carcinomas have few T cells, suggesting that cancers can suppress T cell infiltration. Here, we show that cancer cells of human pancreatic ductal adenocarcinoma (PDA), colorectal cancer, and breast cancer are coated with transglutaminase-2 (TGM2)-dependent covalent CXCL12-keratin-19 (KRT19) heterodimers that are organized as filamentous networks. Since a dimeric form of CXCL12 suppresses the motility of human T cells, we determined whether this polymeric CXCL12-KRT19 coating mediated T cell exclusion. Mouse tumors containing control PDA cells exhibited the CXCL12-KRT19 coating, excluded T cells, and did not respond to treatment with anti-PD-1 antibody. Tumors containing PDA cells not expressing either KRT19 or TGM2 lacked the CXCL12-KRT19 coating, were infiltrated with activated CD8+ T cells, and growth was suppressed with anti-PD-1 antibody treatment. Thus, carcinomas assemble a CXCL12-KRT19 coating to evade cancer immune attack.
Collapse
Affiliation(s)
- Zhikai Wang
- Cancer Center, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| | - Philip Moresco
- Cancer Center, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
- Graduate Program in Genetics, Stony Brook University, Stony Brook, NY 11794
- Medical Scientist Training Program, Stony Brook University Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794
| | - Ran Yan
- Cancer Center, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
- School of Biological Sciences, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| | - Jiayun Li
- Cancer Center, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| | - Ya Gao
- Cancer Center, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| | - Daniele Biasci
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, United Kingdom
| | - Min Yao
- Cancer Center, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| | - Jordan Pearson
- Cancer Center, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
- Graduate Program in Genetics, Stony Brook University, Stony Brook, NY 11794
- Medical Scientist Training Program, Stony Brook University Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794
| | - Jaclyn F Hechtman
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Tobias Janowitz
- Cancer Center, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
- Northwell Health Cancer Institute, Northwell Health, Lake Success, NY 11042
| | - Raza M Zaidi
- Northwell Health Cancer Institute, Northwell Health, Lake Success, NY 11042
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Northwell Health, Hempstead, NY 11549
| | - Matthew J Weiss
- Northwell Health Cancer Institute, Northwell Health, Lake Success, NY 11042
| | - Douglas T Fearon
- Cancer Center, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724;
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065
| |
Collapse
|
15
|
Elahi A, Emerson J, Rudlong J, Keillor JW, Salois G, Visca A, Girardi P, Johnson GV, Pröschel C. Deletion or Inhibition of Astrocytic Transglutaminase 2 Promotes Functional Recovery after Spinal Cord Injury. Cells 2021; 10:2942. [PMID: 34831164 PMCID: PMC8616117 DOI: 10.3390/cells10112942] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/20/2021] [Accepted: 10/21/2021] [Indexed: 01/23/2023] Open
Abstract
Following CNS injury, astrocytes become "reactive" and exhibit pro-regenerative or harmful properties. However, the molecular mechanisms that cause astrocytes to adopt either phenotype are not well understood. Transglutaminase 2 (TG2) plays a key role in regulating the response of astrocytes to insults. Here, we used mice in which TG2 was specifically deleted in astrocytes (Gfap-Cre+/- TG2fl/fl, referred to here as TG2-A-cKO) in a spinal cord contusion injury (SCI) model. Deletion of TG2 from astrocytes resulted in a significant improvement in motor function following SCI. GFAP and NG2 immunoreactivity, as well as number of SOX9 positive cells, were significantly reduced in TG2-A-cKO mice. RNA-seq analysis of spinal cords from TG2-A-cKO and control mice 3 days post-injury identified thirty-seven differentially expressed genes, all of which were increased in TG2-A-cKO mice. Pathway analysis revealed a prevalence for fatty acid metabolism, lipid storage and energy pathways, which play essential roles in neuron-astrocyte metabolic coupling. Excitingly, treatment of wild type mice with the selective TG2 inhibitor VA4 significantly improved functional recovery after SCI, similar to what was observed using the genetic model. These findings indicate the use of TG2 inhibitors as a novel strategy for the treatment of SCI and other CNS injuries.
Collapse
Affiliation(s)
- Anissa Elahi
- Department of Biomedical Genetics, University of Rochester, Rochester, NY 14620, USA; (A.E.); (G.S.); (A.V.); (C.P.)
| | - Jacen Emerson
- Department of Anesthesiology and Perioperative Medicine, University of Rochester, Rochester, NY 14620, USA; (J.E.); (J.R.); (P.G.)
| | - Jacob Rudlong
- Department of Anesthesiology and Perioperative Medicine, University of Rochester, Rochester, NY 14620, USA; (J.E.); (J.R.); (P.G.)
| | - Jeffrey W. Keillor
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada;
| | - Garrick Salois
- Department of Biomedical Genetics, University of Rochester, Rochester, NY 14620, USA; (A.E.); (G.S.); (A.V.); (C.P.)
| | - Adam Visca
- Department of Biomedical Genetics, University of Rochester, Rochester, NY 14620, USA; (A.E.); (G.S.); (A.V.); (C.P.)
| | - Peter Girardi
- Department of Anesthesiology and Perioperative Medicine, University of Rochester, Rochester, NY 14620, USA; (J.E.); (J.R.); (P.G.)
| | - Gail V.W. Johnson
- Department of Anesthesiology and Perioperative Medicine, University of Rochester, Rochester, NY 14620, USA; (J.E.); (J.R.); (P.G.)
| | - Christoph Pröschel
- Department of Biomedical Genetics, University of Rochester, Rochester, NY 14620, USA; (A.E.); (G.S.); (A.V.); (C.P.)
| |
Collapse
|
16
|
Transglutaminase 2 moderates the expansion of mouse abdominal aortic aneurysms. JVS Vasc Sci 2021; 2:95-109. [PMID: 34617062 PMCID: PMC8489235 DOI: 10.1016/j.jvssci.2021.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 04/01/2021] [Indexed: 11/23/2022] Open
Abstract
Objective Previously published work has indicated that transcripts encoding transglutaminase 2 (TG2) increase markedly in a rat model of abdominal aortic aneurysm. This study determines whether TG2 and the related TG, factor XIII-A (FXIII-A), protect against aortic aneurysm development in mice. Methods C57BL/6J wild-type, Tgm2 -/- knockout, F13a1 -/- knockout, and Tgm2 -/- /F13a1 -/- double knockout mice were subjected to laparotomy and periaortic application of CaCl2. Results Tgm2 -/- mice showed slightly greater aortic dilatation at 6 weeks after treatment when compared with wild type. However, vessels from Tgm2 -/- mice, but not wild-type mice, continued to dilate up to 6 months after injury and by 24 weeks, a greater number of Tgm2 -/- mice had developed aneurysms (16/17 vs 10/19; P = .008). Laparotomy resulted in a high death rate in F13a1 -/- knockout mice, more frequently from cardiac complications than from hemorrhage, but among F13a1 -/- mice that survived for 6 weeks after CaCl2 treatment, abdominal aortic aneurysm diameter was unaltered relative to wild-type mice. Laparotomy resulted in a higher death rate among Tgm2 -/- /F13a1 -/- double knockout mice, owing to an increased frequency of delayed bleeding. Surprisingly, Tgm2 -/- /F13a1 -/- double knockout mice showed a trend toward decreased dilatation of the aorta 6 weeks after injury, and this finding was replicated in Tgm2 -/- /F13a1 -/- mice subjected to carotid artery injury. Levels of transcripts encoding TG2 were not increased in the aortas of injured wild-type or F13a1 -/- knockout mice relative to uninjured mice, although changes in the levels of other transcripts accorded with previous descriptions of the CaCl2 aneurysm model in mice. Conclusions Knockout of Tgm2, but not F13a1 exacerbates aortic dilatation, suggesting that TG2 confers protection. However, levels of TG2 messenger RNA are not acutely elevated after injury. FXIII-A plays a role in preventing postoperative damage after laparotomy, confirming previous reports that it prevents distal organ damage after trauma. TG2 promotes wound healing after surgery and, in its absence, the bleeding diathesis associated with FXIII-A deficiency is further exposed.
Collapse
|
17
|
Sima LE, Chen S, Cardenas H, Zhao G, Wang Y, Ivan C, Huang H, Zhang B, Matei D. Loss of host tissue transglutaminase boosts antitumor T cell immunity by altering STAT1/STAT3 phosphorylation in ovarian cancer. J Immunother Cancer 2021; 9:jitc-2021-002682. [PMID: 34593619 PMCID: PMC8487211 DOI: 10.1136/jitc-2021-002682] [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] [Accepted: 09/01/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Tissue transglutaminase (TG2), an enzyme overexpressed in cancer cells, promotes metastasis and resistance to chemotherapy. Its distinct effects in cancer versus the host compartments have not been elucidated. METHODS Here, by using a TG2-/- syngeneic ovarian cancer mouse model, we assessed the effects of TG2 deficiency in the host tissues on antitumor immunity and tumor progression. Multicolor flow cytometry was used to phenotype immune cell populations in the peritoneal environment. Cancer cells recovered from malignant ascites were characterized by RNA sequencing, proliferation, and apoptosis assays. RESULTS We observed that host TG2 loss delayed tumor growth and ascites accumulation and caused increased infiltration of CD8+ T cells and decreased numbers of myeloid cells in the peritoneal fluid. Tumor antigen-specific CD8+ T cell cytotoxic responses were enhanced in ascites from TG2-/- versus TG2+/+ mice and CD8+ T cell depletion caused accelerated ascites accumulation in TG2-/- mice. CD8+ T cells from tumor-bearing TG2-/- mice displayed an effector T cell phenotype, differentiated toward effector memory (Tem). Mechanistically, absence of TG2 augmented signals promoting T cell activation, such as increased cytokine-induced STAT1 and attenuated STAT3 phosphorylation in T cells. Additionally, immune-suppressive myeloid cell populations were reduced in the peritoneal milieu of TG2-/- tumor-bearing mice. In response to the more robust immune response caused by loss of TG2, cancer cells growing intraperitoneally exhibited an interferon-γ(IFN-γ) responsive gene signature and underwent apoptosis. In human specimens, stromal, not tumor, TG2 expression correlated indirectly with numbers of tumor-infiltrating lymphocytes. CONCLUSIONS Collectively, our data demonstrate decreased tumor burden, increased activation and effector function of T cells, and loss of immunosuppressive signals in the tumor microenvironment of TG2-/- mice. We propose that TG2 acts as an attenuator of antitumor T cell immunity and is a new immunomodulatory target.
Collapse
Affiliation(s)
- Livia Elena Sima
- Department of Obstetrics and Gynecology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.,Department of Molecular Cell Biology, Institute of Biochemistry of the Romanian Academy, Bucharest, Romania
| | - Siqi Chen
- Department of Medicine; Hematology/Oncology Division, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Horacio Cardenas
- Department of Obstetrics and Gynecology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Guangyuan Zhao
- Department of Obstetrics and Gynecology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Yinu Wang
- Department of Obstetrics and Gynecology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Cristina Ivan
- Department of Experimental Therapeutics, Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Hao Huang
- Department of Obstetrics and Gynecology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Bin Zhang
- Department of Medicine; Hematology/Oncology Division, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA .,Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Daniela Matei
- Department of Obstetrics and Gynecology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA .,Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.,Jesse Brown Veterans Affairs Medical Center, Chicago, IL, USA
| |
Collapse
|
18
|
Aonuma T, Moukette B, Kawaguchi S, Barupala NP, Sepulveda MN, Corr C, Tang Y, Liangpunsakul S, Payne RM, Willis MS, Kim IM. Cardiomyocyte microRNA-150 confers cardiac protection and directly represses pro-apoptotic small proline-rich protein 1A. JCI Insight 2021; 6:e150405. [PMID: 34403363 PMCID: PMC8492334 DOI: 10.1172/jci.insight.150405] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 08/11/2021] [Indexed: 11/17/2022] Open
Abstract
MicroRNA-150 (miR-150) is downregulated in patients with multiple cardiovascular diseases and in diverse mouse models of heart failure (HF). miR-150 is significantly associated with HF severity and outcome in humans. We previously reported that miR-150 is activated by β-blocker carvedilol (Carv) and plays a protective role in the heart using a systemic miR-150 KO mouse model. However, mechanisms that regulate cell-specific miR-150 expression and function in HF are unknown. Here, we demonstrate that potentially novel conditional cardiomyocyte–specific (CM-specific) miR-150 KO (miR-150 cKO) in mice worsens maladaptive cardiac remodeling after myocardial infarction (MI). Genome-wide transcriptomic analysis in miR-150 cKO mouse hearts identifies small proline–rich protein 1a (Sprr1a) as a potentially novel target of miR-150. Our studies further reveal that Sprr1a expression is upregulated in CMs isolated from ischemic myocardium and subjected to simulated ischemia/reperfusion, while its expression is downregulated in hearts and CMs by Carv. We also show that left ventricular SPRR1A is upregulated in patients with HF and that Sprr1a knockdown in mice prevents maladaptive post-MI remodeling. Lastly, protective roles of CM miR-150 are, in part, attributed to the direct and functional repression of proapoptotic Sprr1a. Our findings suggest a crucial role for the miR-150/SPRR1A axis in regulating CM function post-MI.
Collapse
Affiliation(s)
- Tatsuya Aonuma
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, United States of America
| | - Bruno Moukette
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, United States of America
| | - Satoshi Kawaguchi
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, United States of America
| | - Nipuni P Barupala
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, United States of America
| | - Marisa N Sepulveda
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, United States of America
| | - Christopher Corr
- Department of Medicine, Indiana University School of Medicine, Indianapolis, United States of America
| | - Yaoliang Tang
- Department of Medicine, Augusta University, Augusta, United States of America
| | - Suthat Liangpunsakul
- Department of Medicine, Indiana University School of Medicine, Indianapolis, United States of America
| | - R Mark Payne
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, United States of America
| | - Monte S Willis
- Department of Medicine, Indiana University School of Medicine, Indianapolis, United States of America
| | - Il-Man Kim
- Indiana University School of Medicine, Indianapolis, United States of America
| |
Collapse
|
19
|
Kim HJ, Lee JH, Cho SY, Jeon JH, Kim IG. Transglutaminase 2 mediates transcriptional regulation through BAF250a polyamination. Genes Genomics 2021; 43:333-342. [PMID: 33555506 DOI: 10.1007/s13258-021-01055-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 01/22/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Transglutaminase 2 (TG2) mediates protein modifications by crosslinking or by incorporating polyamine in response to oxidative or DNA-damaging stress, thereby regulating apoptosis, extracellular matrix formation, and inflammation. The regulation of transcriptional activity by TG2-mediated histone serotonylation or by Sp1 crosslinking may also contribute to cellular stress responses. OBJECTIVE In this study, we attempted to identify TG2-interacting proteins to better understand the role of TG2 in transcriptional regulation. METHODS Using a yeast two-hybrid assay to screen a HeLa cell cDNA library, we found that TG2 bound BAF250a, a core subunit of the cBAF chromatin remodeling complex, through an interaction between the TG2 barrel 1 and BAF250a C-terminal domains. RESULTS TG2 was pulled down with a GST-BAF250a C-term fusion protein. Moreover, TG2 and BAF250a were co-fractionated using P11 chromatography, and co-immunoprecipitated. A transamidation reaction showed that TG2 mediated incorporation of polyamine into BAF250a. In glucocorticoid response-element reporter-expressing cells, TG2 overexpression increased the luciferase reporter activity in a transamidation-dependent manner. In addition, a comparison of genome-wide gene expression between wild-type and TG2-deficient primary hepatocytes in response to dexamethasone treatment showed that TG2 further enhanced or suppressed the expression of dexamethasone-regulated genes that were identified by a gene ontology enrichment analysis. CONCLUSION Thus, our results indicate that TG2 regulates transcriptional activity through BAF250a polyamination.
Collapse
Affiliation(s)
- Hyo-Jun Kim
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Korea
| | - Jin-Haeng Lee
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Korea
| | - Sung-Yup Cho
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Ju-Hong Jeon
- Institute of Human-Environment Interface Biology, Seoul National University College of Medicine, Seoul, Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - In-Gyu Kim
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Korea.
- Institute of Human-Environment Interface Biology, Seoul National University College of Medicine, Seoul, Korea.
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|
20
|
Transglutaminase 2 as a Marker for Inflammation and Therapeutic Target in Sepsis. Int J Mol Sci 2021; 22:ijms22041897. [PMID: 33672962 PMCID: PMC7918628 DOI: 10.3390/ijms22041897] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/08/2021] [Accepted: 02/11/2021] [Indexed: 02/07/2023] Open
Abstract
Sepsis results in lethal organ malfunction due to dysregulated host response to infection, which is a condition with increasing prevalence worldwide. Transglutaminase 2 (TG2) is a crosslinking enzyme that forms a covalent bond between lysine and glutamine. TG2 plays important roles in diverse cellular processes, including extracellular matrix stabilization, cytoskeletal function, cell motility, adhesion, signal transduction, apoptosis, and cell survival. We have shown that the co-culture of Candida albicans and hepatocytes activates and induces the translocation of TG2 into the nucleus. In addition, the expression and activation of TG2 in liver macrophages was dramatically induced in the lipopolysaccharide-injected and cecal ligation puncture-operated mouse models of sepsis. Based on these findings and recently published research, we have reviewed the current understanding of the relationship between TG2 and sepsis. Following the genetic and pharmacological inhibition of TG2, we also assessed the evidence regarding the use of TG2 as a potential marker and therapeutic target in inflammation and sepsis.
Collapse
|
21
|
Chiou B, Gao C, Giera S, Folts CJ, Kishore P, Yu D, Oak HC, Jiang R, Piao X. Cell type-specific evaluation of ADGRG1/GPR56 function in developmental central nervous system myelination. Glia 2020; 69:413-423. [PMID: 32902916 DOI: 10.1002/glia.23906] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 08/14/2020] [Accepted: 08/16/2020] [Indexed: 12/13/2022]
Abstract
Myelination of axons in the central nervous system (CNS) is a concerted effort between many cell types, resulting in significant cross-talk and communication among cells. Adhesion G protein-coupled receptor ADGRG1 (GPR56) is expressed in all major glial cells and regulates a wide variety of physiological processes by mediating cell-cell and cell-matrix communications. Previous literature has demonstrated the requirement of ADGRG1 in oligodendrocyte precursor cells (OPCs) during developmental myelination. However, it is unknown if ADGRG1 is responsible for myelin formation in a cell-type-specific manner. To that end, here we profiled myelin status in response to deletion of Adgrg1 specifically in OPCs, microglia, astrocytes, and neurons. Interestingly, we find that knocking out Adgrg1 in OPCs significantly decreases OPC proliferation and reduced number of myelinated axons. However, deleting Adgrg1 in microglia, astrocytes, and neurons does not impact developmental myelination. These data support an autonomous functional role for Adgrg1 in OPCs related to myelination.
Collapse
Affiliation(s)
- Brian Chiou
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco, California, USA
| | - Chuang Gao
- Department of Medicine, Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Department of Neurosurgery, General Hospital of Tianjin Medical University, Tianjin, China
| | - Stefanie Giera
- Department of Medicine, Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Sanofi S.A., Framingham, Massachusetts, USA
| | - Christopher J Folts
- Department of Medicine, Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Vertex Pharmaceuticals, Boston, Massachusetts, USA
| | - Priya Kishore
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco, California, USA
| | - Diankun Yu
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco, California, USA
| | - Hayeon C Oak
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco, California, USA.,Department of Medicine, Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Rongcai Jiang
- Department of Neurosurgery, General Hospital of Tianjin Medical University, Tianjin, China
| | - Xianhua Piao
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco, California, USA.,Department of Medicine, Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Newborn Brain Research Institute, University of California at San Francisco, San Francisco, California, USA.,Department of Pediatrics, University of California at San Francisco, San Francisco, California, USA
| |
Collapse
|
22
|
Watanabe Y, Okuya K, Takada Y, Kinoshita M, Yokoi S, Chisada S, Kamei Y, Tatsukawa H, Yamamoto N, Abe H, Hashimoto H, Hitomi K. Gene disruption of medaka (Oryzias latipes) orthologue for mammalian tissue-type transglutaminase (TG2) causes movement retardation. J Biochem 2020; 168:213-222. [PMID: 32251518 DOI: 10.1093/jb/mvaa038] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 03/19/2020] [Indexed: 12/20/2022] Open
Abstract
Transglutaminases are an enzyme family that catalyses protein cross-linking essential for several biological functions. In the previous studies, we characterized the orthologues of the mammalian transglutaminase family in medaka (Oryzias latipes), an established fish model. Among the human isozymes, tissue-type transglutaminase (TG2) has multiple functions that are involved in several biological phenomena. In this study, we established medaka mutants deficient for the orthologue of human TG2 using the CRISPR/Cas9 and transcription activator-like effector nucleases systems. Although apparent morphological changes in the phenotype were not observed, movement retardation was found in the mutant fish when evaluated by a tank-diving test. Furthermore, comparative immunohistochemistry analysis using in this fish model revealed that orthologue of human TG2 was expressed at the periventricular layer of the optic tectum. Our findings provide novel insight for the relationship between tissue-type transglutaminase and the nervous system and the associated behaviour.
Collapse
Affiliation(s)
- Yuko Watanabe
- Graduate School of Pharmaceutical Sciences, Nagoya University, Chikusa, Nagoya 4648601, Japan
| | - Kazuho Okuya
- Graduate School of Pharmaceutical Sciences, Nagoya University, Chikusa, Nagoya 4648601, Japan
| | - Yuki Takada
- Graduate School of Pharmaceutical Sciences, Nagoya University, Chikusa, Nagoya 4648601, Japan
| | - Masato Kinoshita
- Graduate School of Agriculture, Kyoto University, Sakyo-ku, Kyoto 606-8502, Japan
| | - Saori Yokoi
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo 0600812, Japan
| | - Shinichi Chisada
- Kyorin University School of Medicine, Mitaka, Tokyo 1818611, Japan
| | - Yasuhiro Kamei
- National Institute for Basic Biology, Okazaki 4448585, Japan
| | - Hideki Tatsukawa
- Graduate School of Pharmaceutical Sciences, Nagoya University, Chikusa, Nagoya 4648601, Japan
| | - Naoyuki Yamamoto
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 4648601, Japan
| | - Hideki Abe
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 4648601, Japan
| | - Hisashi Hashimoto
- Graduate School of Science, Nagoya University, Nagoya 4648602, Japan
| | - Kiyotaka Hitomi
- Graduate School of Pharmaceutical Sciences, Nagoya University, Chikusa, Nagoya 4648601, Japan
| |
Collapse
|
23
|
Chermnykh ES, Alpeeva EV, Vorotelyak EA. Transglutaminase 3: The Involvement in Epithelial Differentiation and Cancer. Cells 2020; 9:cells9091996. [PMID: 32872587 PMCID: PMC7563467 DOI: 10.3390/cells9091996] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 08/21/2020] [Accepted: 08/26/2020] [Indexed: 12/14/2022] Open
Abstract
Transglutaminases (TGMs) contribute to the formation of rigid, insoluble macromolecular complexes, which are essential for the epidermis and hair follicles to perform protective and barrier functions against the environment. During differentiation, epidermal keratinocytes undergo structural alterations being transformed into cornified cells, which constitute a highly tough outermost layer of the epidermis, the stratum corneum. Similar processes occur during the hardening of the hair follicle and the hair shaft, which is provided by the enzymatic cross-linking of the structural proteins and keratin intermediate filaments. TGM3, also known as epidermal TGM, is one of the pivotal enzymes responsible for the formation of protein polymers in the epidermis and the hair follicle. Numerous studies have shown that TGM3 is extensively involved in epidermal and hair follicle physiology and pathology. However, the roles of TGM3, its substrates, and its importance for the integument system are not fully understood. Here, we summarize the main advances that have recently been achieved in TGM3 analyses in skin and hair follicle biology and also in understanding the functional role of TGM3 in human tumor pathology as well as the reliability of its prognostic clinical usage as a cancer diagnosis biomarker. This review also focuses on human and murine hair follicle abnormalities connected with TGM3 mutations.
Collapse
|
24
|
Suzuki AS, Yagi R, Kimura MY, Iwamura C, Shinoda K, Onodera A, Hirahara K, Tumes DJ, Koyama-Nasu R, Iismaa SE, Graham RM, Motohashi S, Nakayama T. Essential Role for CD30-Transglutaminase 2 Axis in Memory Th1 and Th17 Cell Generation. Front Immunol 2020; 11:1536. [PMID: 32793209 PMCID: PMC7385138 DOI: 10.3389/fimmu.2020.01536] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 06/11/2020] [Indexed: 12/24/2022] Open
Abstract
Memory helper T (Th) cells are crucial for secondary immune responses against infectious microorganisms but also drive the pathogenesis of chronic inflammatory diseases. Therefore, it is of fundamental importance to understand how memory T cells are generated. However, the molecular mechanisms governing memory Th cell generation remain incompletely understood. Here, we identified CD30 as a molecule heterogeneously expressed on effector Th1 and Th17 cells, and CD30hi effector Th1 and Th17 cells preferentially generated memory Th1 and Th17 cells. We found that CD30 mediated signal induced Transglutaminase-2 (TG2) expression, and that the TG2 expression in effector Th cells is essential for memory Th cell generation. In fact, Cd30-deficiency resulted in the impaired generation of memory Th1 and Th17 cells, which can be rescued by overexpression of TG2. Furthermore, transglutaminase-2 (Tgm2)-deficient CD4 T cells failed to become memory Th cells. As a result, T cells from Tgm2-deficient mice displayed impaired antigen-specific antibody production and attenuated Th17-mediated allergic responses. Our data indicate that CD30-induced TG2 expression in effector Th cells is essential for the generation of memory Th1 and Th17 cells, and that CD30 can be a marker for precursors of memory Th1 and Th17 cells.
Collapse
Affiliation(s)
- Akane S Suzuki
- Department of Medical Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Ryoji Yagi
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Motoko Y Kimura
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Chiaki Iwamura
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Kenta Shinoda
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Atsushi Onodera
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan.,Institute for Global Prominent Research, Chiba University, Chiba, Japan
| | - Kiyoshi Hirahara
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Damon J Tumes
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan.,Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
| | - Ryo Koyama-Nasu
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Siiri E Iismaa
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, NSW, Australia
| | - Robert M Graham
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, NSW, Australia
| | - Shinichiro Motohashi
- Department of Medical Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Toshinori Nakayama
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| |
Collapse
|
25
|
Transglutaminase 2 Depletion Attenuates α-Synuclein Mediated Toxicity in Mice. Neuroscience 2020; 441:58-64. [PMID: 32502569 PMCID: PMC8024061 DOI: 10.1016/j.neuroscience.2020.05.047] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Revised: 05/22/2020] [Accepted: 05/26/2020] [Indexed: 02/04/2023]
Abstract
α-Synuclein (α-Syn) is a key pathogenic protein in α-synucleinopathies including Parkinson disease (PD) and Dementia with Lewy Bodies. The aggregation of α-Syn is believed to be deleterious and a critical step leading to neuronal dysfunction and death. One of the factors that may contribute to the initial steps of this aggregation is crosslinking through transglutaminase 2 (TG2). We previously demonstrated that overexpression of TG2 exacerbates α-Syn toxicity in mice and yeast by increasing the higher-order species of α-Syn. Herein, we investigated whether deletion of the TG2 encoding gene could mitigate the toxicity of α-Syn in vivo. Compared with α-Syn transgenic (SynTg) mice, TG2 null /α-Syn transgenic mice (TG2KO/SynTg) exhibited a reduced amount of phosphorylated α-Syn aggregates and fewer proteinase K-resistant α-Syn aggregates in sections of brain tissue. Neuritic processes that are depleted in SynTg mice compared to wild-type mice were preserved in double TG2KO/SynTg mice. Additionally, the neuroinflammatory reaction to α-Syn was attenuated in TG2KO/SynTg animals. These neuropathological markers of diminished α-Syn toxicity in the absence of TG2 were associated with better motor performance on the rotarod and balance beam. These results suggest that deleting TG2 reduces the toxicity of α-Syn in vivo and improves the behavioral performance of SynTg mice. Accordingly, these findings collectively support pharmacological inhibition of TG2 as a potential disease modifying therapeutic strategy for α-synucleinopathies.
Collapse
|
26
|
Lénárt K, Pap A, Pórszász R, V. Oláh A, Fésüs L, Mádi A. Transglutaminase 2 Has Metabolic and Vascular Regulatory Functions Revealed by In Vivo Activation of Alpha1-Adrenergic Receptor. Int J Mol Sci 2020; 21:E3865. [PMID: 32485850 PMCID: PMC7312910 DOI: 10.3390/ijms21113865] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 05/20/2020] [Accepted: 05/27/2020] [Indexed: 12/14/2022] Open
Abstract
The multifunctional tissue transglutaminase has been demonstrated to act as α1-adrenergic receptor-coupled G protein with GTPase activity in several cell types. To explore further the pathophysiological significance of this function we investigated the in vivo effects of the α1-adrenergic receptor agonist phenylephrine comparing responses in wild type and TG2-/- mice. Injection of phenylephrine, but not a beta3-adrenergic agonist (CL-316,243), resulted in the long-term decline of the respiratory exchange ratio and lower lactate concentration in TG2-/- mice indicating they preferred to utilize fatty acids instead of glucose as fuels. Measurement of tail blood pressure revealed that the vasoconstrictive effect of phenylephrine was milder in TG2-/- mice leading to lower levels of lactate dehydrogenase (LDH) isoenzymes in blood. LDH isoenzyme patterns indicated more damage in lung, liver, kidney, skeletal, and cardiac muscle of wild type mice; the latter was confirmed by a higher level of heart-specific CK-MB. Our data suggest that TG2 as an α1-adrenergic receptor-coupled G protein has important regulatory functions in alpha1-adrenergic receptor-mediated metabolic processes and vascular functions.
Collapse
Affiliation(s)
- Kinga Lénárt
- Department of Biochemistry and Molecular Biology, University of Debrecen, H-4032 Debrecen, Hungary; (K.L.); (A.P.); (L.F.)
- Doctoral School of Molecular Cell and Immune Biology, University of Debrecen, H-4032 Debrecen, Hungary
| | - Attila Pap
- Department of Biochemistry and Molecular Biology, University of Debrecen, H-4032 Debrecen, Hungary; (K.L.); (A.P.); (L.F.)
| | - Róbert Pórszász
- Department of Pharmacology and Pharmacotherapy, University of Debrecen, H-4032 Debrecen, Hungary;
| | - Anna V. Oláh
- Department of Laboratory Medicine, University of Debrecen, H-4032 Debrecen, Hungary;
| | - László Fésüs
- Department of Biochemistry and Molecular Biology, University of Debrecen, H-4032 Debrecen, Hungary; (K.L.); (A.P.); (L.F.)
| | - András Mádi
- Department of Biochemistry and Molecular Biology, University of Debrecen, H-4032 Debrecen, Hungary; (K.L.); (A.P.); (L.F.)
| |
Collapse
|
27
|
A Precision Strategy to Cure Renal Cell Carcinoma by Targeting Transglutaminase 2. Int J Mol Sci 2020; 21:ijms21072493. [PMID: 32260198 PMCID: PMC7177245 DOI: 10.3390/ijms21072493] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 03/29/2020] [Accepted: 04/01/2020] [Indexed: 12/12/2022] Open
Abstract
In a recent report, no significance of transglutaminase 2 (TGase 2) was noted in the analyses of expression differences between normal and clear cell renal cell carcinoma (ccRCC), although we found that knock down of TGase 2 induced significant p53-mediated cell death in ccRCC. Generally, to find effective therapeutic targets, we need to identify targets that belong specifically to a cancer phenotype that can be differentiated from a normal phenotype. Here, we offer precise reasons why TGase 2 may be the first therapeutic target for ccRCC, according to several lines of evidence. TGase 2 is negatively regulated by von Hippel-Lindau tumor suppressor protein (pVHL) and positively regulated by hypoxia-inducible factor 1-α (HIF-1α) in renal cell carcinoma (RCC). Therefore, most of ccRCC presents high level expression of TGase 2 because over 90% of ccRCC showed VHL inactivity through mutation and methylation. Cell death, angiogenesis and drug resistance were specifically regulated by TGase 2 through p53 depletion in ccRCC because over 90% of ccRCC express wild type p53, which is a cell death inducer as well as a HIF-1α suppressor. Although there have been no detailed studies of the physiological role of TGase 2 in multi-omics analyses of ccRCC, a life-long study of the physiological roles of TGase 2 led to the discovery of the first target as well as the first therapeutic treatment for ccRCC in the clinical field.
Collapse
|
28
|
Su T, Qin XY, Furutani Y, Yu W, Kojima S. Imaging of the ex vivo transglutaminase activity in liver macrophages of sepsis mice. Anal Biochem 2020; 597:113654. [PMID: 32142762 DOI: 10.1016/j.ab.2020.113654] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 02/19/2020] [Accepted: 02/25/2020] [Indexed: 12/13/2022]
Abstract
Sepsis is the leading cause of death in hospitalized patients and is characterized by a dysregulated inflammatory response to infection and multiple organ failure, including the liver. Transglutaminase 2 (TG2) is a multifunctional enzyme that exhibits transamidase, GTPase, and integrin-binding activities and has opposing roles in the regulation of cell growth, differentiation, and apoptosis. TG2 plays both pathogenic and protective roles in liver diseases, revealing the need to examine the activities of TG2. Here, we introduced an ex vivo imaging approach to examine the in vivo transamidase activity of TG2 based on the combination of intraperitoneal injection of 5-biotinamidopentylamine (5BAPA), a biotinylated substrate for TG2, and fluorescent streptavidin staining in frozen liver sections. Increased 5BAPA signals was observed in the livers of lipopolysaccharide (LPS) and cecal ligation and puncture (CLP)-induced sepsis mice. Pharmacological inhibition of TG2 activity ameliorated LPS-induced liver injury. 5BAPA signals were observed in TG2-expressing and F4/80-positive midzonal macrophages, providing direct evidence that activated macrophages are the major cellular source of active TG2 in the livers of sepsis mice. Further studies focusing on the activation of 5BAPA-stained midzonal macrophages may improve understanding of the molecular pathophysiology and the development of therapeutic strategies for sepsis.
Collapse
Affiliation(s)
- Ting Su
- Department of Intensive Care Unit, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, 210008, China; Liver Cancer Prevention Research Unit, RIKEN Cluster for Pioneering Research, Wako, Saitama, 351-0198, Japan
| | - Xian-Yang Qin
- Liver Cancer Prevention Research Unit, RIKEN Cluster for Pioneering Research, Wako, Saitama, 351-0198, Japan.
| | - Yutaka Furutani
- Liver Cancer Prevention Research Unit, RIKEN Cluster for Pioneering Research, Wako, Saitama, 351-0198, Japan
| | - Wenkui Yu
- Department of Intensive Care Unit, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, 210008, China
| | - Soichi Kojima
- Liver Cancer Prevention Research Unit, RIKEN Cluster for Pioneering Research, Wako, Saitama, 351-0198, Japan
| |
Collapse
|
29
|
Piro MC, Ventura A, Smirnov A, Saggini A, Lena AM, Mauriello A, Bianchi L, Melino G, Candi E. Transglutaminase 3 Reduces the Severity of Psoriasis in Imiquimod-Treated Mouse Skin. Int J Mol Sci 2020; 21:ijms21051566. [PMID: 32106600 PMCID: PMC7084269 DOI: 10.3390/ijms21051566] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 02/21/2020] [Accepted: 02/24/2020] [Indexed: 12/26/2022] Open
Abstract
Four transglutaminase (TG) isoforms have been detected in epidermal keratinocytes: TG1, TG2, TG3, and TG5. Except for TG1 and TG3, their contribution to keratinocyte development and structure remains undefined. In this paper, we focused on the roles of TG2 and TG3 in imiquimod-induced psoriasis in mouse skin. We evaluated the severity of psoriasis markers in the skin of imiquimod-treated TG3 null and TG2 null mice. Our results showed that compromised TG3KO mouse skin was more responsive than WT or TG2KO mouse skin to the action of the pro-inflammatory drug imiquimod.
Collapse
Affiliation(s)
- Maria Cristina Piro
- Department of Experimental Medicine, TOR, University of Rome “Tor Vergata”, 00133 Rome, Italy; (M.C.P.); (A.S.); (A.S.); (A.M.L.); (A.M.)
| | - Alessandra Ventura
- Dermatology Unit, Department of Biotechnological and Applied Clinical Science, University of L’Aquila, IT-67100 L’Aquila, Italy;
| | - Artem Smirnov
- Department of Experimental Medicine, TOR, University of Rome “Tor Vergata”, 00133 Rome, Italy; (M.C.P.); (A.S.); (A.S.); (A.M.L.); (A.M.)
| | - Andrea Saggini
- Department of Experimental Medicine, TOR, University of Rome “Tor Vergata”, 00133 Rome, Italy; (M.C.P.); (A.S.); (A.S.); (A.M.L.); (A.M.)
| | - Anna Maria Lena
- Department of Experimental Medicine, TOR, University of Rome “Tor Vergata”, 00133 Rome, Italy; (M.C.P.); (A.S.); (A.S.); (A.M.L.); (A.M.)
| | - Alessandro Mauriello
- Department of Experimental Medicine, TOR, University of Rome “Tor Vergata”, 00133 Rome, Italy; (M.C.P.); (A.S.); (A.S.); (A.M.L.); (A.M.)
| | - Luca Bianchi
- Dermatology Unit, Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy;
| | - Gerry Melino
- Department of Experimental Medicine, TOR, University of Rome “Tor Vergata”, 00133 Rome, Italy; (M.C.P.); (A.S.); (A.S.); (A.M.L.); (A.M.)
- Medical Research Council, University of Cambridge, Cambridge CB21QP, UK
| | - Eleonora Candi
- Department of Experimental Medicine, TOR, University of Rome “Tor Vergata”, 00133 Rome, Italy; (M.C.P.); (A.S.); (A.S.); (A.M.L.); (A.M.)
- IDI-IRCCS, Biochemistry laboratory, 00167 Rome, Italy
- Correspondence: ; Tel.: +39-06-72596976
| |
Collapse
|
30
|
Zhuang R, Khosla C. Substrates, inhibitors, and probes of mammalian transglutaminase 2. Anal Biochem 2020; 591:113560. [PMID: 31874171 PMCID: PMC6948143 DOI: 10.1016/j.ab.2019.113560] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 12/15/2019] [Accepted: 12/20/2019] [Indexed: 02/07/2023]
Abstract
Transglutaminase 2 (TG2) is a ubiquitous but enigmatic mammalian protein to which a number of biological functions have been ascribed but not definitively proven. As a member of the transglutaminase family, TG2 can catalyze deamidation or alternatively transamidation of selected Gln residues in proteins and peptides. It is also known to harbor other enzymatic properties, including protein disulfide isomerase, GTP-dependent signal transduction, and ATP dependent protein kinase activity. Given its multifunctional chemistry, it is unsurprising that a long list of proteins from the mammalian proteome have been identified as substrates and/or binding partners; however, the biological relevance of none of these protein-protein interactions has been clarified as yet. Remarkably, the most definitive insights into the biology of TG2 stem from its pathophysiological role in gluten peptide deamidation in celiac disease. Meanwhile our understanding of TG2 chemistry has been leveraged to engineer a spectrum of inhibitors and other molecular probes of TG2 biology in vivo. This review summarizes our current knowledge of the enzymology and regulation of human TG2 with a focus on its physiological substrates as well as tool molecules whose engineering was inspired by their identities.
Collapse
Affiliation(s)
- Ruize Zhuang
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| | - Chaitan Khosla
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA; Department of Chemistry, Stanford University, Stanford, CA, USA; Stanford ChEM-H, Stanford University, Stanford, CA, USA.
| |
Collapse
|
31
|
Rudlong J, Cheng A, Johnson GVW. The role of transglutaminase 2 in mediating glial cell function and pathophysiology in the central nervous system. Anal Biochem 2019; 591:113556. [PMID: 31866289 DOI: 10.1016/j.ab.2019.113556] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 12/04/2019] [Accepted: 12/18/2019] [Indexed: 12/13/2022]
Abstract
The ubiquitously expressed transglutaminase 2 (TG2) has diverse functions in virtually all cell types, with its role depending not only on cell type, but also on specific subcellular localization. In the central nervous system (CNS) different types of glial cells, such as astrocytes, microglia, and oligodendrocytes and their precursor cells (OPCs), play pivotal supportive functions. This review is focused on what is currently known about the role of TG2 in each type of glial cell, in the context of normal function and pathophysiology. For example, astrocytic TG2 facilitates their migration and proliferation, but hinders their ability to protect neurons after CNS injury. The review also examines the interactions between glial cell types, and how TG2 in one cell type may affect another, as well as implications for specific TG2 populations as therapeutic targets in CNS pathology.
Collapse
Affiliation(s)
- Jacob Rudlong
- Department of Anesthesiology and Perioperative Medicine and the Neuroscience Graduate Program, University of Rochester, Rochester, NY, 14620, USA
| | - Anson Cheng
- Department of Anesthesiology and Perioperative Medicine and the Neuroscience Graduate Program, University of Rochester, Rochester, NY, 14620, USA
| | - Gail V W Johnson
- Department of Anesthesiology and Perioperative Medicine and the Neuroscience Graduate Program, University of Rochester, Rochester, NY, 14620, USA.
| |
Collapse
|
32
|
Griffin KJ, Newell LM, Simpson KR, Beckers CML, Drinkhill MJ, Standeven KF, Cheah LT, Iismaa SE, Grant PJ, Jackson CL, Pease RJ. Transglutaminase 2 limits the extravasation and the resultant myocardial fibrosis associated with factor XIII-A deficiency. Atherosclerosis 2019; 294:1-9. [PMID: 31874419 PMCID: PMC7024992 DOI: 10.1016/j.atherosclerosis.2019.12.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 11/15/2019] [Accepted: 12/13/2019] [Indexed: 12/12/2022]
Abstract
Background and aims Transglutaminase (TG) 2 and Factor (F) XIII-A have both been implicated in cardiovascular protection and repair. This study was designed to differentiate between two competing hypotheses: that TG2 and FXIII-A mediate these functions in mice by fulfilling separate roles, or that they act redundantly in this respect. Methods Atherosclerosis was assessed in brachiocephalic artery plaques of fat-fed mixed strain apolipoprotein (Apo)e deficient mice that lacked either or both transglutaminases. Cardiac fibrosis was assessed both in the mixed strain mice and also in C57BL/6J Apoe expressing mice lacking either or both transglutaminases. Results No difference was found in the density of buried fibrous caps within brachiocephalic plaques from mice expressing or lacking these transglutaminases. Cardiac fibrosis developed in both Apoe/F13a1 double knockout and F13a1 single knockout mice, but not in Tgm2 knockout mice. However, concomitant Tgm2 knockout markedly increased fibrosis, as apparent in both Apoe/Tgm2/F13a1 knockout and Tgm2/F13a1 knockout mice. Amongst F13a1 knockout and Tgm2/F13a1 knockout mice, the extent of fibrosis correlated with hemosiderin deposition, suggesting that TG2 limits the extravasation of blood in the myocardium, which in turn reduces the pro-fibrotic stimulus. The resulting fibrosis was interstitial in nature and caused only minor changes in cardiac function. Conclusions These studies confirm that FXIII-A and TG2 fulfil different roles in the mouse myocardium. FXIII-A protects against vascular leakage while TG2 contributes to the stability or repair of the vasculature. The protective function of TG2 must be considered when designing clinical anti-fibrotic therapies based upon FXIII-A or TG2 inhibition. Double transglutaminase 2 and Factor XIII-A knockout exacerbates cardiac fibrosis. Double knockout does not promote the growth of, or destabilise, brachiocephalic plaques. FXIII-A in resident cardiac macrophages does not protect against cardiac fibrosis. FXIII-A in inflammatory macrophages may contribute to protection against fibrosis. Transglutaminase 2 and Factor XIII-A protect against extravasation of blood.
Collapse
Affiliation(s)
- Kathryn J Griffin
- Discovery and Translational Science Division, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, LS2 9JT, UK.
| | - Laura M Newell
- Bristol Heart Institute, University of Bristol, Bristol, BS2 8HW, UK
| | - Kingsley R Simpson
- Discovery and Translational Science Division, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, LS2 9JT, UK
| | - Cora M L Beckers
- Discovery and Translational Science Division, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, LS2 9JT, UK
| | - Mark J Drinkhill
- Discovery and Translational Science Division, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, LS2 9JT, UK
| | - Kristina F Standeven
- Discovery and Translational Science Division, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, LS2 9JT, UK
| | - Lih T Cheah
- Discovery and Translational Science Division, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, LS2 9JT, UK
| | - Siiri E Iismaa
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, 2010, Australia
| | - Peter J Grant
- Discovery and Translational Science Division, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, LS2 9JT, UK
| | | | - Richard J Pease
- Discovery and Translational Science Division, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, LS2 9JT, UK
| |
Collapse
|
33
|
O'Brien JJ, O'Callaghan JP, Miller DB, Chalgeri S, Wennogle LP, Davis RE, Snyder GL, Hendrick JP. Inhibition of calcium-calmodulin-dependent phosphodiesterase (PDE1) suppresses inflammatory responses. Mol Cell Neurosci 2019; 102:103449. [PMID: 31770590 PMCID: PMC7783477 DOI: 10.1016/j.mcn.2019.103449] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 10/01/2019] [Accepted: 11/21/2019] [Indexed: 11/08/2022] Open
Abstract
A novel, potent, and highly specific inhibitor of calcium-calmodulin-dependent phosphodiesterases (PDE) of the PDE1 family, ITI-214, was used to investigate the role of PDE1 in inflammatory responses. ITI-214 dose-dependently suppressed lipopolysaccharide (LPS)-induced gene expression of pro-inflammatory cytokines in an immortalized murine microglial cell line, BV2 cells. RNA profiling (RNA-Seq) was used to analyze the impact of ITI-214 on the BV2 cell transcriptome in the absence and the presence of LPS. ITI-214 was found to regulate classes of genes that are involved in inflammation and cell migration responses to LPS exposure. The gene expression changes seen with ITI-214 treatment were distinct from those elicited by inhibitors of other PDEs with anti-inflammatory activity (e.g., a PDE4 inhibitor), indicating a distinct mechanism of action for PDE1. Functionally, ITI-214 inhibited ADP-induced migration of BV2 cells through a P2Y12-receptor-dependent pathway, possibly due to increases in the extent of cAMP and VASP phosphorylation downstream of receptor activation. Importantly, this effect was recapitulated in P2 rat microglial cells in vitro, indicating that these pathways are active in native microglial cells. These studies are the first to demonstrate that inhibition of PDE1 exerts anti-inflammatory effects through effects on microglia signaling pathways. The ability of PDE1 inhibitors to prevent or dampen excessive inflammatory responses of BV2 cells and microglia provides a basis for exploring their therapeutic utility in the treatment of neurodegenerative diseases associated with increased inflammation and microglia proliferation such as Parkinson's disease and Alzheimer's disease.
Collapse
Affiliation(s)
- Jennifer J O'Brien
- Intra-Cellular Therapies, Inc., The Alexandria Center for Life Sciences, 430 East 29th St Suite 900, New York, NY 10016, United States of America
| | - James P O'Callaghan
- Centers for Disease Control and Prevention, National Institute for Occupational Safety and Health, Morgantown, WV 26505, United States of America
| | - Diane B Miller
- Centers for Disease Control and Prevention, National Institute for Occupational Safety and Health, Morgantown, WV 26505, United States of America
| | - Suman Chalgeri
- Intra-Cellular Therapies, Inc., The Alexandria Center for Life Sciences, 430 East 29th St Suite 900, New York, NY 10016, United States of America
| | - Lawrence P Wennogle
- Intra-Cellular Therapies, Inc., The Alexandria Center for Life Sciences, 430 East 29th St Suite 900, New York, NY 10016, United States of America
| | - Robert E Davis
- Intra-Cellular Therapies, Inc., The Alexandria Center for Life Sciences, 430 East 29th St Suite 900, New York, NY 10016, United States of America
| | - Gretchen L Snyder
- Intra-Cellular Therapies, Inc., The Alexandria Center for Life Sciences, 430 East 29th St Suite 900, New York, NY 10016, United States of America.
| | - Joseph P Hendrick
- Intra-Cellular Therapies, Inc., The Alexandria Center for Life Sciences, 430 East 29th St Suite 900, New York, NY 10016, United States of America
| |
Collapse
|
34
|
Serrano RL, Yu W, Graham RM, Bryan RL, Terkeltaub R. A vascular smooth muscle cell X-box binding protein 1 and transglutaminase 2 regulatory circuit limits neointimal hyperplasia. PLoS One 2019; 14:e0212235. [PMID: 30943188 PMCID: PMC6447169 DOI: 10.1371/journal.pone.0212235] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 01/29/2019] [Indexed: 12/11/2022] Open
Abstract
Neointimal hyperplasia, stimulated by injury and certain vascular diseases, promotes artery obstruction and tissue ischemia. In vascular smooth muscle cell (VSMCs), multiple modulators of protein handling machinery regulate intimal hyperplasia. These include elements of the VSMC unfolded protein response to endoplasmic reticulum stress (UPRER), and transglutaminase 2 (TG2), which catalyzes post-translational protein modification. Previous results for deficiency of UPRER-specific mediator XBP1, and of TG2, have been significant, but in multiple instances contradictory, for effects on cultured VSMC function, and, using multiple models, for neointimal hyperplasia in vivo. Here, we engineered VSMC-specific deficiency of XBP1, and studied cultured VSMCs, and neointimal hyperplasia in response to carotid artery ligation in vivo. Intimal area almost doubled in Xbp1fl/fl SM22α-CRE+ mice 21 days post-ligation. Cultured murine Xbp1 deficient VSMCs migrated more in response to platelet derived growth factor (PDGF) than control VSMCs, and had an increased level of inositol-requiring enzyme 1α (Ire1α), a PDGF receptor-binding UPRER transmembrane endonuclease whose substrates include XBP1. Cultured XBP1-deficient VSMCs demonstrated decreased levels of TG2 protein, in association with increased TG2 polyubiquitination, but with increased TG transamidation catalytic activity. Moreover, IRE1α, and TG2-specific transamidation cross-links were increased in carotid artery neointima in Xbp1fl/fl SM22α-CRE+ mice. Cultured TG2-deficient VSMCs had decreased XBP1 associated with increased IRE1α, and increased migration in response to PDGF. Neointimal hyperplasia also was significantly increased in Tgm2fl/fl SM22α-CRE+ mice at 21 days after carotid ligation. In conclusion, a VSMC regulatory circuit between XBP1 and TG2 limits neointimal hyperplasia in response to carotid ligation.
Collapse
Affiliation(s)
- Ramon L. Serrano
- Department of Medicine, Veterans Affairs Healthcare System, University of California San Diego, California, United States of America
| | - Weifang Yu
- Department of Medicine, Veterans Affairs Healthcare System, University of California San Diego, California, United States of America
| | - Robert M. Graham
- Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia
| | - Ru Liu- Bryan
- Department of Medicine, Veterans Affairs Healthcare System, University of California San Diego, California, United States of America
| | - Robert Terkeltaub
- Department of Medicine, Veterans Affairs Healthcare System, University of California San Diego, California, United States of America
| |
Collapse
|
35
|
Furini G, Verderio EAM. Spotlight on the Transglutaminase 2-Heparan Sulfate Interaction. Med Sci (Basel) 2019; 7:E5. [PMID: 30621228 PMCID: PMC6359630 DOI: 10.3390/medsci7010005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 12/17/2018] [Accepted: 12/18/2018] [Indexed: 02/07/2023] Open
Abstract
Heparan sulfate proteoglycans (HSPGs), syndecan-4 (Sdc4) especially, have been suggested as potential partners of transglutaminase-2 (TG2) in kidney and cardiac fibrosis, metastatic cancer, neurodegeneration and coeliac disease. The proposed role for HSPGs in the trafficking of TG2 at the cell surface and in the extracellular matrix (ECM) has been linked to the fibrogenic action of TG2 in experimental models of kidney fibrosis. As the TG2-HSPG interaction is largely mediated by the heparan sulfate (HS) chains of proteoglycans, in the past few years a number of studies have investigated the affinity of TG2 for HS, and the TG2 heparin binding site has been mapped with alternative outlooks. In this review, we aim to provide a compendium of the main literature available on the interaction of TG2 with HS, with reference to the pathological processes in which extracellular TG2 plays a role.
Collapse
Affiliation(s)
- Giulia Furini
- School of Science and Technology, Nottingham Trent University, Nottingham NG11 8NS, UK.
| | - Elisabetta A M Verderio
- School of Science and Technology, Nottingham Trent University, Nottingham NG11 8NS, UK.
- BiGeA, University of Bologna, 40126 Bologna, Italy.
| |
Collapse
|
36
|
Poole LG, Pant A, Baker KS, Kopec AK, Cline-Fedewa HM, Iismaa SE, Flick MJ, Luyendyk JP. Chronic liver injury drives non-traditional intrahepatic fibrin(ogen) crosslinking via tissue transglutaminase. J Thromb Haemost 2019; 17:113-125. [PMID: 30415489 PMCID: PMC6322974 DOI: 10.1111/jth.14330] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Indexed: 12/25/2022]
Abstract
Essentials Fibrin clots are often implicated in the progression of liver fibrosis. Liver fibrosis was induced in transgenic mice with defects in clot formation or stabilization. Liver fibrosis and fibrin(ogen) deposition do not require fibrin polymerization or factor XIIIa. Fibrin(ogen) is an in vivo substrate of tissue transglutaminase in experimental liver fibrosis. SUMMARY: Background Intravascular fibrin clots and extravascular fibrin deposits are often implicated in the progression of liver fibrosis. However, evidence supporting a pathological role of fibrin in hepatic fibrosis is indirect and based largely on studies using anticoagulant drugs that inhibit activation of the coagulation protease thrombin, which has other downstream targets that promote fibrosis. Therefore, the goal of this study was to determine the precise role of fibrin deposits in experimental hepatic fibrosis. Methods Liver fibrosis was induced in mice expressing mutant fibrinogen insensitive to thrombin-mediated proteolysis (i.e. locked in the monomeric form), termed FibAEK mice, and factor XIII A2 subunit-deficient (FXIII-/- ) mice. Female wild-type mice, FXIII-/- mice and homozygous FibAEK mice were challenged with carbon tetrachloride (CCl4 ) twice weekly for 4 weeks or 6 weeks (1 mL kg-1 , intraperitoneal). Results Hepatic injury and fibrosis induced by CCl4 challenge were unaffected by FXIII deficiency or inhibition of thrombin-catalyzed fibrin polymer formation (in FibAEK mice). Surprisingly, hepatic deposition of crosslinked fibrin(ogen) was not reduced in CCl4 -challenged FXIII-/- mice or FibAEK mice as compared with wild-type mice. Rather, deposition of crosslinked hepatic fibrin(ogen) following CCl4 challenge was dramatically reduced in tissue transglutaminase-2 (TGM2)-deficient (TGM2-/- ) mice. However, the reduction in crosslinked fibrin(ogen) in TGM2-/- mice did not affect CCl4 -induced liver fibrosis. Conclusions These results indicate that neither traditional fibrin clots, formed by the thrombin-activated FXIII pathway nor atypical TGM2-crosslinked fibrin(ogen) contribute to experimental CCl4 -induced liver fibrosis. Collectively, the results indicate that liver fibrosis occurs independently of intrahepatic fibrin(ogen) deposition.
Collapse
Affiliation(s)
- L G Poole
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, USA
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, MI, USA
| | - A Pant
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, USA
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, MI, USA
| | - K S Baker
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, USA
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| | - A K Kopec
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, USA
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, MI, USA
| | - H M Cline-Fedewa
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, MI, USA
| | - S E Iismaa
- Division of Molecular Cardiology and Biophysics, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia
| | - M J Flick
- Cancer and Blood Diseases Institute, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - J P Luyendyk
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, USA
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, MI, USA
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
37
|
Affiliation(s)
- Laszlo Lorand
- Department of Cell and Molecular BiologyNorthwestern University Feinberg School of Medicine Chicago Illinois USA
| | - Siiri E. Iismaa
- Molecular Cardiology and Biophysics DivisionVictor Chang Cardiac Research Institute Darlinghurst New South Wales Australia
- St Vincent's Clinical SchoolUniversity of New South Wales Kensington New South Wales Australia
| |
Collapse
|
38
|
Kim SY. New Insights into Development of Transglutaminase 2 Inhibitors as Pharmaceutical Lead Compounds. Med Sci (Basel) 2018; 6:medsci6040087. [PMID: 30297644 PMCID: PMC6313797 DOI: 10.3390/medsci6040087] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 09/28/2018] [Accepted: 10/05/2018] [Indexed: 12/18/2022] Open
Abstract
Transglutaminase 2 (EC 2.3.2.13; TG2 or TGase 2) plays important roles in the pathogenesis of many diseases, including cancers, neurodegeneration, and inflammatory disorders. Under normal conditions, however, mice lacking TGase 2 exhibit no obvious abnormal phenotype. TGase 2 expression is induced by chemical, physical, and viral stresses through tissue-protective signaling pathways. After stress dissipates, expression is normalized by feedback mechanisms. Dysregulation of TGase 2 expression under pathologic conditions, however, can potentiate pathogenesis and aggravate disease severity. Consistent with this, TGase 2 knockout mice exhibit reversal of disease phenotypes in neurodegenerative and chronic inflammatory disease models. Accordingly, TGase 2 is considered to be a potential therapeutic target. Based on structure–activity relationship assays performed over the past few decades, TGase 2 inhibitors have been developed that target the enzyme’s active site, but clinically applicable inhibitors are not yet available. The recently described the small molecule GK921, which lacks a group that can react with the active site of TGase 2, and efficiently inhibits the enzyme’s activity. Mechanistic studies revealed that GK921 binds at an allosteric binding site in the N-terminus of TGase 2 (amino acids (a.a.) 81–116), triggering a conformational change that inactivates the enzyme. Because the binding site of GK921 overlaps with the p53-binding site of TGase 2, the drug induces apoptosis in renal cell carcinoma by stabilizing p53. In this review, we discuss the possibility of developing TGase 2 inhibitors that target the allosteric binding site of TGase 2.
Collapse
Affiliation(s)
- Soo-Youl Kim
- Tumor Microenvironment Research Branch, Division of Cancer Biology, Research Institute, National Cancer Center, Goyang 10408, Korea.
| |
Collapse
|
39
|
Katt WP, Blobel NJ, Komarova S, Antonyak MA, Nakano I, Cerione RA. A small molecule regulator of tissue transglutaminase conformation inhibits the malignant phenotype of cancer cells. Oncotarget 2018; 9:34379-34397. [PMID: 30344949 PMCID: PMC6188150 DOI: 10.18632/oncotarget.26193] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 09/15/2018] [Indexed: 12/26/2022] Open
Abstract
The protein crosslinking enzyme tissue transglutaminase (tTG) is an acyltransferase which catalyzes transamidation reactions between two proteins, or between a protein and a polyamine. It is frequently overexpressed in several different types of human cancer cells, where it has been shown to contribute to their growth, survival, and invasiveness. tTG is capable of adopting two distinct conformational states: a protein crosslinking active (“open”) state, and a GTP-bound, crosslinking inactive (“closed”) state. We have previously shown that the ectopic expression of mutant forms of tTG, which constitutively adopt the open conformation, are toxic to cells. This raises the possibility that strategies directed toward causing tTG to maintain an open state could potentially provide a therapeutic benefit for cancers in which tTG is highly expressed. Here, we report the identification of a small molecule, TTGM 5826, which stabilizes the open conformation of tTG. Treatment of breast and brain cancer cell lines, as well as glioma stem cells, with this molecule broadly inhibits their transformed phenotypes. Thus, TTGM 5826 represents the lead compound for a new class of small molecules that promote the toxicity of cancer cells by stabilizing the open state of tTG.
Collapse
Affiliation(s)
- William P Katt
- Department of Molecular Medicine, Cornell University, Ithaca, NY, USA
| | - Nicolas J Blobel
- Department of Molecular Medicine, Cornell University, Ithaca, NY, USA
| | - Svetlana Komarova
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Marc A Antonyak
- Department of Molecular Medicine, Cornell University, Ithaca, NY, USA
| | - Ichiro Nakano
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Richard A Cerione
- Department of Molecular Medicine, Cornell University, Ithaca, NY, USA.,Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, USA
| |
Collapse
|
40
|
Mižíková I, Pfeffer T, Nardiello C, Surate Solaligue DE, Steenbock H, Tatsukawa H, Silva DM, Vadász I, Herold S, Pease RJ, Iismaa SE, Hitomi K, Seeger W, Brinckmann J, Morty RE. Targeting transglutaminase 2 partially restores extracellular matrix structure but not alveolar architecture in experimental bronchopulmonary dysplasia. FEBS J 2018; 285:3056-3076. [PMID: 29935061 DOI: 10.1111/febs.14596] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 06/12/2018] [Accepted: 06/21/2018] [Indexed: 12/21/2022]
Abstract
The generation, maturation and remodelling of the extracellular matrix (ECM) are essential for the formation of alveoli during lung development. Alveoli formation is disturbed in preterm infants that develop bronchopulmonary dysplasia (BPD), where collagen fibres are malformed, and perturbations to lung ECM structures may underlie BPD pathogenesis. Malformed ECM structures might result from abnormal protein cross-linking, in part attributable to the increased expression and activity of transglutaminase 2 (TGM2) that have been noted in affected patient lungs, as well as in hyperoxia-based BPD animal models. The objective of the present study was to assess whether TGM2 plays a causal role in normal and aberrant lung alveolarization. Targeted deletion of Tgm2 in C57BL/6J mice increased septal thickness and reduced gas-exchange surface area in otherwise normally developing lungs. During aberrant lung alveolarization that occurred under hyperoxic conditions, collagen structures in Tgm2-/- mice were partially protected from the impact of hyperoxia, where normal dihydroxylysinonorleucine and hydroxylysylpiridinoline collagen cross-link abundance was restored; however, the lung alveolar architecture remained abnormal. Inhibition of transglutaminases (including TGM2) with cysteamine appreciably reduced transglutaminase activity in vivo, as assessed by Nε -(γ-l-glutamyl)-l-lysine abundance and TGM catalytic activity, and restored normal dihydroxylysinonorleucine and hydroxylysylpiridinoline collagen cross-link abundance under pathological conditions. Furthermore, a moderate improvement in alveoli size and gas-exchange surface density was noted in cysteamine-treated mouse lungs in which BPD was modelled. These data indicate that TGM2 plays a role in normal lung alveolarization, and contributes to the formation of aberrant ECM structures during disordered lung alveolarization.
Collapse
Affiliation(s)
- Ivana Mižíková
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Giessen, Germany, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Germany
| | - Tilman Pfeffer
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Giessen, Germany, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Germany
| | - Claudio Nardiello
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Giessen, Germany, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Germany
| | - David E Surate Solaligue
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Giessen, Germany, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Germany
| | - Heiko Steenbock
- Institute of Virology and Cell Biology, University of Lübeck, Germany
| | - Hideki Tatsukawa
- Graduate School of Pharmaceutical Sciences, Nagoya University, Japan
| | - Diogo M Silva
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Giessen, Germany, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Germany
| | - István Vadász
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Germany
| | - Susanne Herold
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Germany
| | - Richard J Pease
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, UK
| | - Siiri E Iismaa
- Victor Chang Cardiac Research Institute, Darlinghurst, Australia
| | - Kiyotaka Hitomi
- Graduate School of Pharmaceutical Sciences, Nagoya University, Japan
| | - Werner Seeger
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Giessen, Germany, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Germany
| | - Jürgen Brinckmann
- Institute of Virology and Cell Biology, University of Lübeck, Germany.,Department of Dermatology, University of Lübeck, Germany
| | - Rory E Morty
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Giessen, Germany, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Germany
| |
Collapse
|
41
|
Monteagudo A, Feola J, Natola H, Ji C, Pröschel C, Johnson GVW. Depletion of astrocytic transglutaminase 2 improves injury outcomes. Mol Cell Neurosci 2018; 92:128-136. [PMID: 29969654 DOI: 10.1016/j.mcn.2018.06.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 06/26/2018] [Accepted: 06/27/2018] [Indexed: 01/12/2023] Open
Abstract
Astrocytes play an indispensable role in maintaining a healthy, functional neural network in the central nervous system (CNS). A primary function of CNS astrocytes is to support the survival and function of neurons. In response to injury, astrocytes take on a reactive phenotype, which alters their molecular functions. Reactive astrocytes have been reported to be both beneficial and harmful to the CNS recovery process subsequent to injury. Understanding the molecular processes and regulatory proteins that determine the extent to which an astrocyte hinders or supports neuronal survival is important within the context of CNS repair. One protein that plays a role in modulating cellular survival is transglutaminase 2 (TG2). Global deletion of TG2 results in beneficial outcomes subsequent to in vivo ischemic brain injury. Ex vivo studies have also implicated TG2 as a negative regulator of astrocyte viability subsequent to injury. In this study we show that knocking down TG2 in astrocytes significantly increases their ability to protect neurons from oxygen glucose deprivation (OGD)/reperfusion injury. To begin to understand how deletion of TG2 in astrocytes improves their ability to protect neurons from injury, we performed transcriptome analysis of wild type and TG2-/- astrocytes. TG2 deletion resulted in alterations in genes involved in extracellular matrix remodeling, cell adhesion and axon growth/guidance. In addition, the majority of genes that showed increases in the TG2-/- astrocytes had predicted cJun/AP-1 binding motifs in their promoters. Furthermore, phospho-cJun levels were robustly elevated in TG2-/- astrocytes, a finding which was consistent with the increase in expression of AP-1 responsive genes. These in vitro data were subsequently extended into an in vivo model to determine whether the absence of astrocytic TG2 improves outcomes after CNS injury. Our results show that, following a spinal cord injury, scar formation is significantly attenuated in mice with astrocyte-specific TG2 deletion compared to mice expressing normal TG2 levels. Taken together, these data indicate that TG2 plays a pivotal role in mediating reactive astrocyte properties following CNS injury. Further, the data suggest that limiting the AP-1 mediated pro-survival injury response may be a contributing factor to that the detrimental effects of astrocytic TG2.
Collapse
Affiliation(s)
- Alina Monteagudo
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY 14642, USA
| | - Julianne Feola
- Department of Biomedical Genetics, University of Rochester, Rochester, NY 14642, USA
| | - Heather Natola
- Department of Biomedical Genetics, University of Rochester, Rochester, NY 14642, USA
| | - Changyi Ji
- Department of Anesthesiology and Perioperative Medicine, University of Rochester, Rochester, NY 14642, USA
| | - Christoph Pröschel
- Department of Biomedical Genetics, University of Rochester, Rochester, NY 14642, USA; Stem Cell and Regenerative Medicine Institute, University of Rochester, Rochester, NY 14642, USA
| | - Gail V W Johnson
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY 14642, USA; Department of Biomedical Genetics, University of Rochester, Rochester, NY 14642, USA; Department of Anesthesiology and Perioperative Medicine, University of Rochester, Rochester, NY 14642, USA.
| |
Collapse
|
42
|
Raychaudhuri U, Millar JC, Clark AF. Knockout of tissue transglutaminase ameliorates TGFβ2-induced ocular hypertension: A novel therapeutic target for glaucoma? Exp Eye Res 2018. [DOI: 10.1016/j.exer.2018.03.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
43
|
Giera S, Luo R, Ying Y, Ackerman SD, Jeong SJ, Stoveken HM, Folts CJ, Welsh CA, Tall GG, Stevens B, Monk KR, Piao X. Microglial transglutaminase-2 drives myelination and myelin repair via GPR56/ADGRG1 in oligodendrocyte precursor cells. eLife 2018; 7:33385. [PMID: 29809138 PMCID: PMC5980231 DOI: 10.7554/elife.33385] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 05/18/2018] [Indexed: 12/29/2022] Open
Abstract
In the central nervous system (CNS), myelin formation and repair are regulated by oligodendrocyte (OL) lineage cells, which sense and integrate signals from their environment, including from other glial cells and the extracellular matrix (ECM). The signaling pathways that coordinate this complex communication, however, remain poorly understood. The adhesion G protein-coupled receptor ADGRG1 (also known as GPR56) is an evolutionarily conserved regulator of OL development in humans, mice, and zebrafish, although its activating ligand for OL lineage cells is unknown. Here, we report that microglia-derived transglutaminase-2 (TG2) signals to ADGRG1 on OL precursor cells (OPCs) in the presence of the ECM protein laminin and that TG2/laminin-dependent activation of ADGRG1 promotes OPC proliferation. Signaling by TG2/laminin to ADGRG1 on OPCs additionally improves remyelination in two murine models of demyelination. These findings identify a novel glia-to-glia signaling pathway that promotes myelin formation and repair, and suggest new strategies to enhance remyelination.
Collapse
Affiliation(s)
- Stefanie Giera
- Division of Newborn Medicine, Department of Medicine, Children's Hospital and Harvard Medical School, Boston, United States.,Department of Neurology, F. M. Kirby Neurobiology Center, Children's Hospital and Harvard Medical School, Boston, United States
| | - Rong Luo
- Division of Newborn Medicine, Department of Medicine, Children's Hospital and Harvard Medical School, Boston, United States.,Department of Neurology, F. M. Kirby Neurobiology Center, Children's Hospital and Harvard Medical School, Boston, United States
| | - Yanqin Ying
- Division of Newborn Medicine, Department of Medicine, Children's Hospital and Harvard Medical School, Boston, United States.,Department of Neurology, F. M. Kirby Neurobiology Center, Children's Hospital and Harvard Medical School, Boston, United States.,Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Sarah D Ackerman
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, United States
| | - Sung-Jin Jeong
- Division of Newborn Medicine, Department of Medicine, Children's Hospital and Harvard Medical School, Boston, United States.,Department of Neurology, F. M. Kirby Neurobiology Center, Children's Hospital and Harvard Medical School, Boston, United States.,Department of Neural Development and Diseases, Korea Brain Research Institute (KBRI), Daegu, South Korea
| | - Hannah M Stoveken
- Department of Pharmacology, University of Michigan Medical Center, Ann Arbor, United States
| | - Christopher J Folts
- Division of Newborn Medicine, Department of Medicine, Children's Hospital and Harvard Medical School, Boston, United States.,Department of Neurology, F. M. Kirby Neurobiology Center, Children's Hospital and Harvard Medical School, Boston, United States
| | - Christina A Welsh
- Department of Neurology, F. M. Kirby Neurobiology Center, Children's Hospital and Harvard Medical School, Boston, United States
| | - Gregory G Tall
- Department of Pharmacology, University of Michigan Medical Center, Ann Arbor, United States
| | - Beth Stevens
- Department of Neurology, F. M. Kirby Neurobiology Center, Children's Hospital and Harvard Medical School, Boston, United States
| | - Kelly R Monk
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, United States
| | - Xianhua Piao
- Division of Newborn Medicine, Department of Medicine, Children's Hospital and Harvard Medical School, Boston, United States.,Department of Neurology, F. M. Kirby Neurobiology Center, Children's Hospital and Harvard Medical School, Boston, United States
| |
Collapse
|
44
|
Spotlight on the transglutaminase 2 gene: a focus on genomic and transcriptional aspects. Biochem J 2018; 475:1643-1667. [PMID: 29764956 DOI: 10.1042/bcj20170601] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 04/10/2018] [Accepted: 04/12/2018] [Indexed: 01/16/2023]
Abstract
The type 2 isoenzyme is the most widely expressed transglutaminase in mammals displaying several intra- and extracellular activities depending on its location (protein modification, modulation of gene expression, membrane signalling and stabilization of cellular interactions with the extracellular matrix) in relation to cell death, survival and differentiation. In contrast with the appreciable knowledge about the regulation of the enzymatic activities, much less is known concerning its inducible expression, which is altered in inflammatory and neoplastic diseases. In this context, we first summarize the gene's basic features including single-nucleotide polymorphism characterization, epigenetic DNA methylation and identification of regulatory regions and of transcription factor-binding sites at the gene promoter, which could concur to direct gene expression. Further aspects related to alternative splicing events and to ncRNAs (microRNAs and lncRNAs) are involved in the modulation of its expression. Notably, this important gene displays transcriptional variants relevant for the protein's function with the occurrence of at least seven transcripts which support the synthesis of five isoforms with modified catalytic activities. The different expression of the TG2 (type 2 transglutaminase) variants might be useful for dictating the multiple biological features of the protein and their alterations in pathology, as well as from a therapeutic perspective.
Collapse
|
45
|
Kárpáti S, Sárdy M, Németh K, Mayer B, Smyth N, Paulsson M, Traupe H. Transglutaminases in autoimmune and inherited skin diseases: The phenomena of epitope spreading and functional compensation. Exp Dermatol 2018; 27:807-814. [PMID: 28940785 DOI: 10.1111/exd.13449] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/13/2017] [Indexed: 02/06/2023]
Abstract
Transglutaminases (TGs) are structurally and functionally related enzymes that modify the post-translational structure and activity of proteins or peptides, and thus are able to turn on or switch off their function. Depending on location and activities, TGs are able to modify the signalling, the function and the fate of cells and extracellular connective tissues. Besides mouse models, human diseases enable us to appreciate the function of various TGs. In this study, skin diseases induced by genetic damages or autoimmune targeting of these enzymes will be discussed. TG1, TG3 and TG5 contribute to the cutaneous barrier and thus to the integrity and function of epidermis. TGM1 mutations related to autosomal recessive ichthyosis subtypes, TGM5 mutations to a mild epidermolysis bullosa phenotype and as novelty TGM3 mutation to uncombable hair syndrome will be discussed. Autoimmunity to TG2, TG3 and TG6 may develop in a few of those genetically determined individuals who lost tolerance to gluten, and manifest as coeliac disease, dermatitis herpetiformis or gluten-dependent neurological symptoms, respectively. These gluten responder diseases commonly occur in combination. In autoimmune diseases, the epitope spreading is remarkable, while in some inherited pathologies, a unique compensation of the lost enzyme function is noted.
Collapse
Affiliation(s)
- Sarolta Kárpáti
- Dermatology, Venereology and Dermatooncology, Semmelweis University, Budapest, Hungary
| | - Miklós Sárdy
- Dermatology, Venereology and Dermatooncology, Semmelweis University, Budapest, Hungary
| | - Krisztián Németh
- Dermatology, Venereology and Dermatooncology, Semmelweis University, Budapest, Hungary
| | - Balázs Mayer
- Dermatology, Venereology and Dermatooncology, Semmelweis University, Budapest, Hungary
| | - Neil Smyth
- Biological Sciences, University of Southampton, Southampton, UK
| | - Mats Paulsson
- Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany
| | - Heiko Traupe
- Department of Dermatology, University of Münster, Münster, Germany
| |
Collapse
|
46
|
β 2 -adrenoceptor-induced modulation of transglutaminase 2 transamidase activity in cardiomyoblasts. Eur J Pharmacol 2017; 813:105-121. [DOI: 10.1016/j.ejphar.2017.07.043] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 07/24/2017] [Accepted: 07/24/2017] [Indexed: 12/12/2022]
|
47
|
Shibata T, Kawabata SI. Pluripotency and a secretion mechanism of Drosophila transglutaminase. J Biochem 2017; 163:165-176. [DOI: 10.1093/jb/mvx059] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 07/14/2017] [Indexed: 01/13/2023] Open
Affiliation(s)
- Toshio Shibata
- Institute for Advanced Study, Kyushu University, Fukuoka 819-0395, Japan
- Department of Biology, Faculty of Science, Kyushu University, Motooka 744, Fukuoka 819-0395, Japan
| | - Shun-ichiro Kawabata
- Department of Biology, Faculty of Science, Kyushu University, Motooka 744, Fukuoka 819-0395, Japan
| |
Collapse
|
48
|
Hoac B, Nelea V, Jiang W, Kaartinen MT, McKee MD. Mineralization-inhibiting effects of transglutaminase-crosslinked polymeric osteopontin. Bone 2017; 101:37-48. [PMID: 28428079 DOI: 10.1016/j.bone.2017.04.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 03/21/2017] [Accepted: 04/16/2017] [Indexed: 01/12/2023]
Abstract
Osteopontin (OPN) belongs to the SIBLING family (Small, Integrin-Binding LIgand N-linked Glycoproteins) of mineral-binding matrix proteins found in bones and teeth. OPN is a well-known inhibitor of matrix mineralization, and enzymatic modification of OPN can affect this inhibitory function. In bone, OPN exists both as a monomer and as a high-molecular-weight polymer - the latter is formed by transglutaminase-mediated crosslinking of glutamine and lysine residues in OPN to create homotypic protein assemblies. OPN can be covalently crosslinked by transglutaminase 2 (TG2) and Factor XIII-A. Polymeric OPN has increased binding to collagen and promotes osteoblast adhesion, but despite these initial observations, its role in mineralization is not clear. In this study, we investigated the effect of polymerized OPN on mineralization using a hydroxyapatite crystal growth assay and mineralizing MC3T3-E1 osteoblast cultures. In the cultures, endogenous polymeric OPN was detected after mineralization occurred. In cell-free conditions, TG2 was used to crosslink bovine OPN into its polymeric form, and atomic force microscopy and dynamic light scattering revealed variably-sized, large branched aggregates ranging across hundreds of nanometers. These OPN polymers inhibited the growth of hydroxyapatite crystals in solution at concentrations similar to monomeric OPN, although the crosslinking slightly reduced its inhibitory potency. When added to MC3T3-E1 osteoblast cultures, this exogenous polymeric OPN essentially did not inhibit mineralization when given during the later mineralization stages of culture; however, cultures treated early and then continuously with polymeric OPN throughout both the matrix assembly and mineral deposition stages showed reduced mineralization. Immunoblotting of protein extracts from these continuously treated cultures revealed exogenous OPN polymers incorporated into mature matrix that had not yet mineralized. These results suggest that in bone, the increased size and branched structure of crosslinked inhibitory polymeric OPN near the mineralization front could hinder it from accessing focal mineralization sites in the dense collagen-rich matrix, suggesting that OPN-crosslinking into polymers may represent a way to fine-tune the inhibitory potency of OPN on bone mineralization.
Collapse
Affiliation(s)
- Betty Hoac
- Faculty of Dentistry, McGill University, Montreal, QC, Canada
| | - Valentin Nelea
- Faculty of Dentistry, McGill University, Montreal, QC, Canada; Department of Anatomy and Cell Biology, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Wenge Jiang
- Faculty of Dentistry, McGill University, Montreal, QC, Canada
| | - Mari T Kaartinen
- Faculty of Dentistry, McGill University, Montreal, QC, Canada; Division of Experimental Medicine, Department of Medicine, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Marc D McKee
- Faculty of Dentistry, McGill University, Montreal, QC, Canada; Department of Anatomy and Cell Biology, Faculty of Medicine, McGill University, Montreal, QC, Canada.
| |
Collapse
|
49
|
Shrestha R, Shrestha R, Qin XY, Kuo TF, Oshima Y, Iwatani S, Teraoka R, Fujii K, Hara M, Li M, Takahashi-Nakaguchi A, Chibana H, Lu J, Cai M, Kajiwara S, Kojima S. Fungus-derived hydroxyl radicals kill hepatic cells by enhancing nuclear transglutaminase. Sci Rep 2017; 7:4746. [PMID: 28684792 PMCID: PMC5500562 DOI: 10.1038/s41598-017-04630-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 05/18/2017] [Indexed: 02/08/2023] Open
Abstract
We previously reported the importance of induced nuclear transglutaminase (TG) 2 activity, which results in hepatic cell death, in ethanol-induced liver injury. Here, we show that co-incubation of either human hepatic cells or mouse primary hepatocytes derived from wild-type but not TG2-/- mice with pathogenic fungi Candida albicans and C. glabrata, but not baker's yeast Saccharomyces cerevisiae, induced cell death in host cells by enhancing cellular, particularly nuclear, TG activity. Further pharmacological and genetic approaches demonstrated that this phenomenon was mediated partly by the production of reactive oxygen species (ROS) such as hydroxyl radicals, as detected by a fluorescent probe and electron spin resonance. A ROS scavenger, N-acetyl cysteine, blocked enhanced TG activity primarily in the nuclei and inhibited cell death. In contrast, deletion of C. glabrata nox-1, which encodes a ROS-generating enzyme, resulted in a strain that failed to induce the same phenomena. A similar induction of hepatic ROS and TG activities was observed in C. albicans-infected mice. An antioxidant corn peptide fraction inhibited these phenomena in hepatic cells. These results address the impact of ROS-generating pathogens in inducing nuclear TG2-related liver injuries, which provides novel therapeutic targets for preventing and curing alcoholic liver disease.
Collapse
Affiliation(s)
- Ronak Shrestha
- Micro-Signaling Regulation Technology Unit, RIKEN Center for Life Science Technologies, Wako, Saitama, Japan
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Kanagawa, Japan
| | - Rajan Shrestha
- Micro-Signaling Regulation Technology Unit, RIKEN Center for Life Science Technologies, Wako, Saitama, Japan
| | - Xian-Yang Qin
- Micro-Signaling Regulation Technology Unit, RIKEN Center for Life Science Technologies, Wako, Saitama, Japan
| | - Ting-Fang Kuo
- Micro-Signaling Regulation Technology Unit, RIKEN Center for Life Science Technologies, Wako, Saitama, Japan
| | - Yugo Oshima
- Condensed Molecular Materials Laboratory, RIKEN, Wako, Saitama, Japan
| | - Shun Iwatani
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Kanagawa, Japan
| | - Ryutaro Teraoka
- Micro-Signaling Regulation Technology Unit, RIKEN Center for Life Science Technologies, Wako, Saitama, Japan
| | - Keisuke Fujii
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Kanagawa, Japan
| | - Mitsuko Hara
- Micro-Signaling Regulation Technology Unit, RIKEN Center for Life Science Technologies, Wako, Saitama, Japan
| | - Mengqian Li
- Micro-Signaling Regulation Technology Unit, RIKEN Center for Life Science Technologies, Wako, Saitama, Japan
| | | | - Hiroji Chibana
- Medical Mycology Research Center, Chiba University, Chiba, Chiba, Japan
| | - Jun Lu
- China National Research Institute of Food and Fermentation Industries, Beijing, China
| | - Muyi Cai
- China National Research Institute of Food and Fermentation Industries, Beijing, China
| | - Susumu Kajiwara
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Kanagawa, Japan.
| | - Soichi Kojima
- Micro-Signaling Regulation Technology Unit, RIKEN Center for Life Science Technologies, Wako, Saitama, Japan.
| |
Collapse
|
50
|
Transglutaminase 2 modulation of NF-κB signaling in astrocytes is independent of its ability to mediate astrocytic viability in ischemic injury. Brain Res 2017; 1668:1-11. [PMID: 28522262 DOI: 10.1016/j.brainres.2017.05.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 04/14/2017] [Accepted: 05/10/2017] [Indexed: 12/20/2022]
Abstract
Transglutaminase 2 (TG2) is a multifunctional protein that can contribute to cell death and cell survival processes in a variety of disease contexts. Within the brain, TG2 has been shown to promote cell death in ischemic injury when expressed in astrocytes (Colak and Johnson, 2012). However, the specific functions and characteristics of astrocytic TG2 that mediate this effect are largely unknown. Therefore, the goal of this study was to investigate the role of astrocytic TG2 in mediating cellular viability processes in the context of ischemic injury, with a specific focus on its contributions to intracellular signaling cascades. We show that, in response to oxygen/glucose deprivation (OGD), acute lentiviral-mediated knockdown of TG2, as well as inhibition with an irreversible TG2 inhibitor, enhances cell survival. We also show that TG2 depletion increases nuclear factor-κB (NF-κB) signaling, whereas inhibition reduces NF-κB activity. Despite its clear contribution to NF-κB signaling, however, TG2 modulation of NF-κB signaling is not likely to be a major contributor to its ability to mediate astrocytic viability in this context. Overall, the results of this study provide insight into the role of TG2 in astrocytes and suggest possible avenues for future study of the relationship between astrocytic TG2 and ischemic injury.
Collapse
|