1
|
Delgado T, Emerson J, Hong M, Keillor JW, Johnson GVW. Pharmacological Inhibition of Astrocytic Transglutaminase 2 Facilitates the Expression of a Neurosupportive Astrocyte Reactive Phenotype in Association with Increased Histone Acetylation. Biomolecules 2024; 14:1594. [PMID: 39766301 PMCID: PMC11673777 DOI: 10.3390/biom14121594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/02/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Astrocytes play critical roles in supporting structural and metabolic homeostasis in the central nervous system (CNS). CNS injury leads to the development of a range of reactive phenotypes in astrocytes whose molecular determinants are poorly understood. Finding ways to modulate astrocytic injury responses and leverage a pro-recovery phenotype holds promise in treating CNS injury. Recently, it has been demonstrated that ablation of astrocytic transglutaminase 2 (TG2) shifts reactive astrocytes towards a phenotype that improves neuronal injury outcomes both in vitro and in vivo. Additionally, in an in vivo mouse model, pharmacological inhibition of TG2 with the irreversible inhibitor VA4 phenocopied the neurosupportive effects of TG2 deletion in astrocytes. In this study, we extended our comparisons of VA4 treatment and TG2 deletion to provide insights into the mechanisms by which TG2 attenuates neurosupportive astrocytic function after injury. Using a neuron-astrocyte co-culture model, we found that VA4 treatment improves the ability of astrocytes to support neurite outgrowth on an injury-relevant matrix, as we previously showed for astrocytic TG2 deletion. We hypothesize that TG2 mediates its influence on astrocytic phenotype through transcriptional regulation, and our previous RNA sequencing suggests that TG2 is primarily transcriptionally repressive in astrocytes, although it can facilitate both up- and downregulation of gene expression. Therefore, we asked whether VA4 inhibition could alter TG2's interaction with Zbtb7a, a transcription factor that we previously identified as a functionally relevant TG2 nuclear interactor. We found that VA4 significantly decreased the interaction of TG2 and Zbtb7a. Additionally, we assessed the effect of TG2 deletion and VA4 treatment on transcriptionally permissive histone acetylation and found significantly greater acetylation in both experimental groups. Consistent with these findings, our present proteomic analysis further supports the predominant transcriptionally repressive role of TG2 in astrocytes. Our proteomic data additionally unveiled pronounced changes in lipid and antioxidant metabolism in astrocytes with TG2 deletion or inhibition, which likely contribute to the enhanced neurosupportive function of these astrocytes.
Collapse
Affiliation(s)
- Thomas Delgado
- Department of Anesthesiology and Perioperative Medicine, University of Rochester, 601 Elmwood Ave, Box 604, Rochester, NY 14620, USA; (T.D.); (J.E.); (M.H.)
| | - Jacen Emerson
- Department of Anesthesiology and Perioperative Medicine, University of Rochester, 601 Elmwood Ave, Box 604, Rochester, NY 14620, USA; (T.D.); (J.E.); (M.H.)
| | - Matthew Hong
- Department of Anesthesiology and Perioperative Medicine, University of Rochester, 601 Elmwood Ave, Box 604, Rochester, NY 14620, USA; (T.D.); (J.E.); (M.H.)
| | - Jeffrey W. Keillor
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON K1N6N5, Canada;
| | - Gail V. W. Johnson
- Department of Anesthesiology and Perioperative Medicine, University of Rochester, 601 Elmwood Ave, Box 604, Rochester, NY 14620, USA; (T.D.); (J.E.); (M.H.)
| |
Collapse
|
2
|
Delgado T, Emerson J, Hong M, Keillor JW, Johnson GVW. Pharmacological inhibition of astrocytic transglutaminase 2 facilitates the expression of a neurosupportive astrocyte reactive phenotype in association with increased histone acetylation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.15.589192. [PMID: 38659783 PMCID: PMC11042235 DOI: 10.1101/2024.04.15.589192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Astrocytes play critical roles in supporting structural and metabolic homeostasis in the central nervous system (CNS). CNS injury leads to the development of a range of reactive phenotypes in astrocytes whose molecular determinants are poorly understood. Finding ways to modulate astrocytic injury responses and leverage a pro-recovery phenotype holds promise in treating CNS injury. Recently, it has been demonstrated that ablation of astrocytic transglutaminase 2 (TG2) modulates the phenotype of reactive astrocytes in a way that improves neuronal injury outcomes both in vitro and in vivo. In an in vivo mouse model, pharmacological inhibition of TG2 with the irreversible inhibitor VA4 phenocopies the neurosupportive effects of TG2 deletion in astrocytes. In this study, we provide insights into the mechanisms by which TG2 deletion or inhibition result in a more neurosupportive astrocytic phenotype. Using a neuron-astrocyte co-culture model, we show that VA4 treatment improves the ability of astrocytes to support neurite outgrowth on an injury-relevant matrix. To better understand how pharmacologically altering TG2 affects its ability to regulate reactive astrocyte phenotypes, we assessed how VA4 inhibition impacts TG2's interaction with Zbtb7a, a transcription factor we have previously identified as a functionally relevant TG2 nuclear interactor. The results of these studies demonstrate that VA4 significantly decreases the interaction of TG2 and Zbtb7a. TG2's interactions with Zbtb7a, as well as a wide range of other transcription factors and chromatin regulatory proteins, suggest that TG2 may act as an epigenetic regulator to modulate gene expression. To begin to understand if TG2-mediated epigenetic modification may impact astrocytic phenotypes in our models, we interrogated the effect of TG2 deletion and VA4 treatment on histone acetylation and found significantly greater acetylation in both experimental groups. Consistent with these findings, previous RNA-sequencing and our present proteomic analysis also supported a predominant transcriptionally suppressive role of TG2 in astrocytes. Our proteomic data additionally unveiled pronounced changes in lipid and antioxidant metabolism in astrocytes with TG2 deletion or inhibition, which likely contribute to the enhanced neurosupportive function of these astrocytes.
Collapse
Affiliation(s)
- Thomas Delgado
- 601 Elmwood Ave, box 604, Department of Anesthesiology and Perioperative Medicine, University of Rochester, Rochester, NY, 14620, USA
| | - Jacen Emerson
- 601 Elmwood Ave, box 604, Department of Anesthesiology and Perioperative Medicine, University of Rochester, Rochester, NY, 14620, USA
| | - Matthew Hong
- 601 Elmwood Ave, box 604, Department of Anesthesiology and Perioperative Medicine, University of Rochester, Rochester, NY, 14620, USA
| | - Jeffrey W. Keillor
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON K1N6N5, Canada
| | - Gail VW Johnson
- 601 Elmwood Ave, box 604, Department of Anesthesiology and Perioperative Medicine, University of Rochester, Rochester, NY, 14620, USA
| |
Collapse
|
3
|
Delgado T, Emerson J, Hong M, Keillor JW, Johnson GVW. Pharmacological inhibition of astrocytic transglutaminase 2 facilitates the expression of a neurosupportive astrocyte reactive phenotype in association with increased histone acetylation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.02.06.527263. [PMID: 36798305 PMCID: PMC9934526 DOI: 10.1101/2023.02.06.527263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Astrocytes play critical roles in supporting structural and metabolic homeostasis in the central nervous system (CNS). CNS injury leads to the development of a range of reactive phenotypes in astrocytes whose molecular determinants are poorly understood. Finding ways to modulate astrocytic injury responses and leverage a pro-recovery phenotype holds promise in treating CNS injury. Recently, it has been demonstrated that ablation of astrocytic transglutaminase 2 (TG2) modulates the phenotype of reactive astrocytes in a way that improves neuronal injury outcomes both in vitro and in vivo. In an in vivo mouse model, pharmacological inhibition of TG2 with the irreversible inhibitor VA4 phenocopies the neurosupportive effects of TG2 deletion in astrocytes. In this study, we provide insights into the mechanisms by which TG2 deletion or inhibition result in a more neurosupportive astrocytic phenotype. Using a neuron-astrocyte co-culture model, we show that VA4 treatment improves the ability of astrocytes to support neurite outgrowth on an injury-relevant matrix. To better understand how pharmacologically altering TG2 affects its ability to regulate reactive astrocyte phenotypes, we assessed how VA4 inhibition impacts TG2's interaction with Zbtb7a, a transcription factor we have previously identified as a functionally relevant TG2 nuclear interactor. The results of these studies demonstrate that VA4 significantly decreases the interaction of TG2 and Zbtb7a. TG2's interactions with Zbtb7a, as well as a wide range of other transcription factors and chromatin regulatory proteins, suggest that TG2 may act as an epigenetic regulator to modulate gene expression. To begin to understand if TG2-mediated epigenetic modification may impact astrocytic phenotypes in our models, we interrogated the effect of TG2 deletion and VA4 treatment on histone acetylation and found significantly greater acetylation in both experimental groups. Consistent with these findings, previous RNA-sequencing and our present proteomic analysis also supported a predominant transcriptionally suppressive role of TG2 in astrocytes. Our proteomic data additionally unveiled pronounced changes in lipid and antioxidant metabolism in astrocytes with TG2 deletion or inhibition, which likely contribute to the enhanced neurosupportive function of these astrocytes.
Collapse
Affiliation(s)
- Thomas Delgado
- 601 Elmwood Ave, box 604, Department of Anesthesiology and Perioperative Medicine, University of Rochester, Rochester, NY, 14620, USA
| | - Jacen Emerson
- 601 Elmwood Ave, box 604, Department of Anesthesiology and Perioperative Medicine, University of Rochester, Rochester, NY, 14620, USA
| | - Matthew Hong
- 601 Elmwood Ave, box 604, Department of Anesthesiology and Perioperative Medicine, University of Rochester, Rochester, NY, 14620, USA
| | - Jeffrey W. Keillor
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON K1N6N5, Canada
| | - Gail VW Johnson
- 601 Elmwood Ave, box 604, Department of Anesthesiology and Perioperative Medicine, University of Rochester, Rochester, NY, 14620, USA
| |
Collapse
|
4
|
Yao Z, Fan Y, Lin L, Kellems RE, Xia Y. Tissue transglutaminase: a multifunctional and multisite regulator in health and disease. Physiol Rev 2024; 104:281-325. [PMID: 37712623 DOI: 10.1152/physrev.00003.2023] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 09/07/2023] [Accepted: 09/10/2023] [Indexed: 09/16/2023] Open
Abstract
Tissue transglutaminase (TG2) is a widely distributed multifunctional protein involved in a broad range of cellular and metabolic functions carried out in a variety of cellular compartments. In addition to transamidation, TG2 also functions as a Gα signaling protein, a protein disulfide isomerase (PDI), a protein kinase, and a scaffolding protein. In the nucleus, TG2 modifies histones and transcription factors. The PDI function catalyzes the trimerization and activation of heat shock factor-1 in the nucleus and regulates the oxidation state of several mitochondrial complexes. Cytosolic TG2 modifies proteins by the addition of serotonin or other primary amines and in this way affects cell signaling. Modification of protein-bound glutamines reduces ubiquitin-dependent proteasomal degradation. At the cell membrane, TG2 is associated with G protein-coupled receptors (GPCRs), where it functions in transmembrane signaling. TG2 is also found in the extracellular space, where it functions in protein cross-linking and extracellular matrix stabilization. Of particular importance in transglutaminase research are recent findings concerning the role of TG2 in gene expression, protein homeostasis, cell signaling, autoimmunity, inflammation, and hypoxia. Thus, TG2 performs a multitude of functions in multiple cellular compartments, making it one of the most versatile cellular proteins. Additional evidence links TG2 with multiple human diseases including preeclampsia, hypertension, cardiovascular disease, organ fibrosis, cancer, neurodegenerative diseases, and celiac disease. In conclusion, TG2 provides a multifunctional and multisite response to physiological stress.
Collapse
Affiliation(s)
- Zhouzhou Yao
- National Medical Metabolomics International Collaborative Research Center, Central South University, Changsha, Hunan, People's Republic of China
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Yuhua Fan
- National Medical Metabolomics International Collaborative Research Center, Central South University, Changsha, Hunan, People's Republic of China
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Lizhen Lin
- National Medical Metabolomics International Collaborative Research Center, Central South University, Changsha, Hunan, People's Republic of China
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Rodney E Kellems
- Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School at Houston, Houston, Texas, United States
| | - Yang Xia
- National Medical Metabolomics International Collaborative Research Center, Central South University, Changsha, Hunan, People's Republic of China
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| |
Collapse
|
5
|
Lu J, Huang G, Chang X, Wei B, Sun Y, Yang Z, Zhao Y, Zhao Z, Dong G, Chen J. Effects of Serotonin on Cell Viability, Permeability of Bovine Mammary Gland Epithelial Cells and Their Transcriptome Analysis. Int J Mol Sci 2023; 24:11388. [PMID: 37511146 PMCID: PMC10379418 DOI: 10.3390/ijms241411388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/10/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
Serotonin (5-HT) has been reported to play an important role in mammary gland involution that is defined as the process through which the gland returns to a nonlactating state. However, the overall picture of the regulatory mechanisms of 5-HT and the effects of serotonylation on mammary gland involution still need to be further investigated. The current study aimed to investigate the effects of 5-HT on global gene expression profiles of bovine mammary epithelial cells (MAC-T) and to preliminarily examine whether the serotonylation involved in the mammary gland involution by using Monodansylcadaverine (MDC), a competitive inhibitor of transglutaminase 2. Results showed that a high concentration of 5-HT decreased viability and transepithelial electrical resistance (TEER) in MAC-T cells. Transcriptome analysis indicated that 2477 genes were differentially expressed in MAC-T cells treated with 200 μg/mL of 5-HT compared with the control group, and the Notch, p53, and PI3K-Akt signaling pathways were enriched. MDC influenced 5-HT-induced MAC-T cell death, fatty acid synthesis, and the formation and disruption of tight junctions. Overall, a high concentration of 5-HT is able to accelerate mammary gland involution, which may be regulated through the Notch, p53, and PI3K-Akt signaling pathways. Serotonylation is involved in bovine mammary gland involution.
Collapse
Affiliation(s)
- Jie Lu
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China
- Chongqing Key Laboratory of Herbivore Science, Chongqing 400715, China
| | - Guohao Huang
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China
- Chongqing Key Laboratory of Herbivore Science, Chongqing 400715, China
| | - Xuan Chang
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China
- Chongqing Key Laboratory of Herbivore Science, Chongqing 400715, China
| | - Bingni Wei
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China
- Chongqing Key Laboratory of Herbivore Science, Chongqing 400715, China
| | - Yawang Sun
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China
- Chongqing Key Laboratory of Herbivore Science, Chongqing 400715, China
| | - Zhengguo Yang
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China
- Chongqing Key Laboratory of Herbivore Science, Chongqing 400715, China
| | - Yongju Zhao
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China
- Chongqing Key Laboratory of Herbivore Science, Chongqing 400715, China
| | - Zhongquan Zhao
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China
- Chongqing Key Laboratory of Herbivore Science, Chongqing 400715, China
| | - Guozhong Dong
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China
- Chongqing Key Laboratory of Herbivore Science, Chongqing 400715, China
| | - Juncai Chen
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China
- Chongqing Key Laboratory of Herbivore Science, Chongqing 400715, China
| |
Collapse
|
6
|
Atobatele AG, Tonoli E, Vadakekolathu J, Savoca MP, Barr M, Kataria Y, Rossanese M, Burhan I, McArdle S, Caccamo D, Verderio EAM. Canonical and truncated transglutaminase-2 regulate mucin-1 expression and androgen independency in prostate cancer cell lines. Cell Death Dis 2023; 14:317. [PMID: 37160910 PMCID: PMC10170068 DOI: 10.1038/s41419-023-05818-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 04/13/2023] [Accepted: 04/19/2023] [Indexed: 05/11/2023]
Abstract
Androgen independency is associated with poor prostate cancer (PCa) survival. Here we report that silencing of transglutaminase-2 (TG2) expression by CRISPR-Cas9 is associated with upregulation of androgen receptor (AR) transcription in PCa cell lines. Knockout of TG2 reversed the migratory potential and anchorage independency of PC3 and DU145 cells and revealed a reduced level of mucin-1 (MUC1) RNA transcript through unbiased multi-omics profiling, which was restored by selective add-back of the truncated TG2 isoform (TGM2_v2). Silencing of AR resulted into increased MUC1 in TG2KO PC3 cells showing that TG2 affects transcriptional regulation of MUC1 via repressing AR expression. Treatment of PC3 WT cell line with TG2 inhibitor ZDON led to a significant increase in AR expression and decrease in MUC1. ZDON also blocked the formation of MUC1-multimers labelled with TG amine-donor substrates in reducing conditions, revealing for the first time a role for TG2, which we show to be externalised via extracellular vesicles, in MUC1 stabilisation via calcium-dependent transamidation. A specific antibody towards TGM2_v2 revealed its restricted nuclear location compared to the canonical long form of TG2 (TGM2_v1), which is predominantly cytosolic, suggesting that this form contributes to the previously suggested TG2-mediated NF-κB activation and AR transcriptional repression. As TGM2_v2 transcription was increased in biopsies of early-stage prostate adenocarcinoma (PRAD) patients compared to subjects presenting inflammatory prostatitis, and total TG2 protein expression significantly increased in PRAD versus normal tissue, the role of TG2 and its truncated form as a prostate malignancy marker is suggested. In conclusion, this investigation has provided the first unbiased discovery of a novel pathway mediated by TG2 via MUC1, which is shown to contribute to androgen insensitivity and malignancy of PCa cells and be upregulated in PCa biopsies, with potential relevance to cancer immune evasion.
Collapse
Affiliation(s)
- Adeola Grace Atobatele
- School of Science and Technology, Centre for Health, Ageing and Understanding of Disease, Nottingham Trent University, Nottingham, NG11 8NS, UK
- Department of Biological and Biomedical Sciences, Science Centre, School of Health, Science and Wellbeing, Staffordshire University, Leek Road, Stoke-on-Trent, ST4 2DF, UK
| | - Elisa Tonoli
- School of Science and Technology, Centre for Health, Ageing and Understanding of Disease, Nottingham Trent University, Nottingham, NG11 8NS, UK
| | - Jayakumar Vadakekolathu
- John van Geest Cancer Research Centre, Nottingham Trent University, Clifton Lane, Nottingham, NG11 8NS, UK
| | - Maria Pia Savoca
- School of Science and Technology, Centre for Health, Ageing and Understanding of Disease, Nottingham Trent University, Nottingham, NG11 8NS, UK
| | - Melissa Barr
- School of Science and Technology, Centre for Health, Ageing and Understanding of Disease, Nottingham Trent University, Nottingham, NG11 8NS, UK
| | - Yukti Kataria
- School of Science and Technology, Centre for Health, Ageing and Understanding of Disease, Nottingham Trent University, Nottingham, NG11 8NS, UK
| | - Marta Rossanese
- Department of Human and Paediatric Pathology, Polyclinic Hospital University, Via C. Valeria 1, 98125, Messina, Italy
| | - Izhar Burhan
- School of Science and Technology, Centre for Health, Ageing and Understanding of Disease, Nottingham Trent University, Nottingham, NG11 8NS, UK
| | - Stephanie McArdle
- John van Geest Cancer Research Centre, Nottingham Trent University, Clifton Lane, Nottingham, NG11 8NS, UK
| | - Daniela Caccamo
- Department of Biomedical Sciences, Dental Sciences & Morpho-Functional Imaging, Polyclinic Hospital University, Via C. Valeria 1, 98125, Messina, Italy
| | - Elisabetta A M Verderio
- School of Science and Technology, Centre for Health, Ageing and Understanding of Disease, Nottingham Trent University, Nottingham, NG11 8NS, UK.
- Biological Sciences Department (BiGeA), University of Bologna, Bologna, 40126, Italy.
| |
Collapse
|
7
|
Emerson J, Delgado T, Girardi P, Johnson GVW. Deletion of Transglutaminase 2 from Mouse Astrocytes Significantly Improves Their Ability to Promote Neurite Outgrowth on an Inhibitory Matrix. Int J Mol Sci 2023; 24:6058. [PMID: 37047031 PMCID: PMC10094709 DOI: 10.3390/ijms24076058] [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: 02/03/2023] [Revised: 03/01/2023] [Accepted: 03/16/2023] [Indexed: 04/14/2023] Open
Abstract
Astrocytes are the primary support cells of the central nervous system (CNS) that help maintain the energetic requirements and homeostatic environment of neurons. CNS injury causes astrocytes to take on reactive phenotypes with an altered overall function that can range from supportive to harmful for recovering neurons. The characterization of reactive astrocyte populations is a rapidly developing field, and the underlying factors and signaling pathways governing which type of reactive phenotype that astrocytes take on are poorly understood. Our previous studies suggest that transglutaminase 2 (TG2) has an important role in determining the astrocytic response to injury. Selectively deleting TG2 from astrocytes improves functional outcomes after CNS injury and causes widespread changes in gene regulation, which is associated with its nuclear localization. To begin to understand how TG2 impacts astrocytic function, we used a neuron-astrocyte co-culture paradigm to compare the effects of TG2-/- and wild-type (WT) mouse astrocytes on neurite outgrowth and synapse formation. Neurons were grown on a control substrate or an injury-simulating matrix comprised of inhibitory chondroitin sulfate proteoglycans (CSPGs). Compared to WT astrocytes, TG2-/- astrocytes supported neurite outgrowth to a significantly greater extent only on the CSPG matrix, while synapse formation assays showed mixed results depending on the pre- and post-synaptic markers analyzed. We hypothesize that TG2 regulates the supportive functions of astrocytes in injury conditions by modulating gene expression through interactions with transcription factors and transcription complexes. Based on the results of a previous yeast two-hybrid screen for TG2 interactors, we further investigated the interaction of TG2 with Zbtb7a, a ubiquitously expressed transcription factor. Co-immunoprecipitation and colocalization analyses confirmed the interaction of TG2 and Zbtb7a in the nucleus of astrocytes. Overexpression or knockdown of Zbtb7a levels in WT and TG2-/- astrocytes revealed that Zbtb7a robustly influenced astrocytic morphology and the ability of astrocytes to support neuronal outgrowth, which was significantly modulated by the presence of TG2. These findings support our hypothesis that astrocytic TG2 acts as a transcriptional regulator to influence astrocytic function, with greater influence under injury conditions that increase its expression, and Zbtb7a likely contributes to the overall effects observed with astrocytic TG2 deletion.
Collapse
Affiliation(s)
| | | | - Peter Girardi
- Department of Anesthesiology and Perioperative Medicine, University of Rochester, 601 Elmwood Ave., Box 604, Rochester, NY 14620, USA
| | - Gail V. W. Johnson
- Department of Anesthesiology and Perioperative Medicine, University of Rochester, 601 Elmwood Ave., Box 604, Rochester, NY 14620, USA
| |
Collapse
|
8
|
Steiner CA, Cartwright IM, Taylor CT, Colgan SP. Hypoxia-inducible factor as a bridge between healthy barrier function, wound healing, and fibrosis. Am J Physiol Cell Physiol 2022; 323:C866-C878. [PMID: 35912990 PMCID: PMC9467472 DOI: 10.1152/ajpcell.00227.2022] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/07/2022] [Accepted: 07/23/2022] [Indexed: 11/22/2022]
Abstract
The healthy mammalian intestine is lined by a single layer of epithelial cells. These cells provide a selectively permeable barrier to luminal contents and normally do so in an efficient and effective manner. Barrier function in the healthy mucosa is provided via several mechanisms including epithelial junctional complexes, mucus production, as well as mucosal-derived antimicrobial proteins. As tissue metabolism is central to the maintenance of homeostasis in the mucosa, intestinal [Formula: see text] levels are uniquely low due to counter-current blood flow and the presence of the microbiota, resulting in the stabilization of the transcription factor hypoxia-inducible factor (HIF). Ongoing studies have revealed that HIF molds normal intestinal metabolism and is central to the coordination of barrier regulation during both homeostasis and active disease. During acute inflammation, HIF is central to controlling the rapid restitution of the epithelium consistent with normal wound healing responses. In contrast, HIF may also contribute to the fibrostenotic response associated with chronic, nonresolving inflammation. As such, HIF may function as a double-edged sword in the overall course of the inflammatory response. Here, we review recent literature on the contribution of HIF to mucosal barrier function, wound healing, and fibrosis.
Collapse
Affiliation(s)
- Calen A Steiner
- Division of Gastroenterology and Hepatology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- Department of Medicine and the Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, Colorado
| | - Ian M Cartwright
- Division of Gastroenterology and Hepatology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- Department of Medicine and the Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, Colorado
- Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, Colorado
| | - Cormac T Taylor
- School of Medicine, Conway Institute and Systems Biology Ireland, University College Dublin, Dublin, Ireland
| | - Sean P Colgan
- Division of Gastroenterology and Hepatology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- Department of Medicine and the Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, Colorado
- Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, Colorado
| |
Collapse
|
9
|
Chen S, Ma J, Chi J, Zhang B, Zheng X, Chen J, Liu J. Roles and potential clinical implications of tissue transglutaminase in cardiovascular diseases. Pharmacol Res 2022; 177:106085. [PMID: 35033646 DOI: 10.1016/j.phrs.2022.106085] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 12/28/2021] [Accepted: 01/11/2022] [Indexed: 02/06/2023]
Abstract
Cardiovascular disease (CVD)-related mortality and morbidity are among the most critical disease burdens worldwide. CVDs encompass many diseases and involve complex pathogenesis and pathological changes. While research on these diseases has advanced significantly, treatments and their efficacy remain rather limited. New therapeutic strategies and targets must, therefore, be explored. Tissue transglutaminase (TG2) is pivotal to the pathological development of CVDs, including participating in the cross-linking of extracellular proteins, activation of fibroblasts, hypertrophy and apoptosis of cardiomyocytes, proliferation and migration of smooth muscle cells (SMCs), and inflammatory reactions. Regulating TG2 activity and expression could ensure remarkable improvements in disorders like heart failure (HF), pulmonary hypertension (PH), hypertension, and coronary atherosclerosis. In this review, we summarize recent advances in TG2: we discuss its role and mechanisms in the progression of various CVDs and its potential as a diagnostic and therapeutic target.
Collapse
Affiliation(s)
- Shiqi Chen
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jingwei Ma
- Department of Immunology, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430030, China
| | - Jiangyang Chi
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Bingxia Zhang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiaojuan Zheng
- Department of Biochemistry and Molecular Biology, Medical School of Southeast University, Nanjing, Jiangsu 210003, China
| | - Jie Chen
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Junwei Liu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
10
|
Keillor JW, Johnson GVW. Transglutaminase 2 as a therapeutic target for neurological conditions. Expert Opin Ther Targets 2021; 25:721-731. [PMID: 34607527 DOI: 10.1080/14728222.2021.1989410] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 10/01/2021] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Transglutaminase 2 (TG2) has been implicated in numerous neurological conditions, including neurodegenerative diseases, multiple sclerosis, and CNS injury. Early studies on the role of TG2 in neurodegenerative conditions focused on its ability to 'crosslink' proteins into insoluble aggregates. However, more recent studies have suggested that this is unlikely to be the primary mechanism by which TG2 contributes to the pathogenic processes. Although the specific mechanisms by which TG2 is involved in neurological conditions have not been clearly defined, TG2 regulates numerous cellular processes through which it could contribute to a specific disease. Given the fact that TG2 is a stress-induced gene and elevated in disease or injury conditions, TG2 inhibitors may be useful neurotherapeutics. AREAS COVERED Overview of TG2 and different TG2 inhibitors. A brief review of TG2 in neurodegenerative diseases, multiple sclerosis and CNS injury and inhibitors that have been tested in different models. Database search: https://pubmed.ncbi.nlm.nih.gov prior to 1 July 2021. EXPERT OPINION Currently, it appears unlikely that inhibiting TG2 in the context of neurodegenerative diseases would be therapeutically advantageous. However, for multiple sclerosis and CNS injuries, TG2 inhibitors may have the potential to be therapeutically useful and thus there is rationale for their further development.
Collapse
Affiliation(s)
- Jeffrey W Keillor
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Gail V W Johnson
- Department of Anesthesiology and Perioperative Medicine, University of Rochester, Rochester, NY, USA
| |
Collapse
|
11
|
Maffei B, Laverrière M, Wu Y, Triboulet S, Perrinet S, Duchateau M, Matondo M, Hollis RL, Gourley C, Rupp J, Keillor JW, Subtil A. Infection-driven activation of transglutaminase 2 boosts glucose uptake and hexosamine biosynthesis in epithelial cells. EMBO J 2020; 39:e102166. [PMID: 32134139 DOI: 10.15252/embj.2019102166] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 01/21/2020] [Accepted: 01/31/2020] [Indexed: 12/16/2022] Open
Abstract
Transglutaminase 2 (TG2) is a ubiquitously expressed enzyme with transamidating activity. We report here that both expression and activity of TG2 are enhanced in mammalian epithelial cells infected with the obligate intracellular bacteria Chlamydia trachomatis. Genetic or pharmacological inhibition of TG2 impairs bacterial development. We show that TG2 increases glucose import by up-regulating the transcription of the glucose transporter genes GLUT-1 and GLUT-3. Furthermore, TG2 activation drives one specific glucose-dependent pathway in the host, i.e., hexosamine biosynthesis. Mechanistically, we identify the glucosamine:fructose-6-phosphate amidotransferase (GFPT) among the substrates of TG2. GFPT modification by TG2 increases its enzymatic activity, resulting in higher levels of UDP-N-acetylglucosamine biosynthesis and protein O-GlcNAcylation. The correlation between TG2 transamidating activity and O-GlcNAcylation is disrupted in infected cells because host hexosamine biosynthesis is being exploited by the bacteria, in particular to assist their division. In conclusion, our work establishes TG2 as a key player in controlling glucose-derived metabolic pathways in mammalian cells, themselves hijacked by C. trachomatis to sustain their own metabolic needs.
Collapse
Affiliation(s)
- Benoit Maffei
- Unité de Biologie cellulaire de l'infection microbienne, CNRS UMR3691, Institut Pasteur, Paris, France.,Collège Doctoral, Sorbonne Université, Paris, France
| | - Marc Laverrière
- Unité de Biologie cellulaire de l'infection microbienne, CNRS UMR3691, Institut Pasteur, Paris, France
| | - Yongzheng Wu
- Unité de Biologie cellulaire de l'infection microbienne, CNRS UMR3691, Institut Pasteur, Paris, France
| | - Sébastien Triboulet
- Unité de Biologie cellulaire de l'infection microbienne, CNRS UMR3691, Institut Pasteur, Paris, France
| | - Stéphanie Perrinet
- Unité de Biologie cellulaire de l'infection microbienne, CNRS UMR3691, Institut Pasteur, Paris, France
| | - Magalie Duchateau
- Plateforme Protéomique, Unité de Spectrométrie de Masse pour la Biologie, USR 2000 CNRS, Institut Pasteur, Paris, France
| | - Mariette Matondo
- Plateforme Protéomique, Unité de Spectrométrie de Masse pour la Biologie, USR 2000 CNRS, Institut Pasteur, Paris, France
| | - Robert L Hollis
- Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Edinburgh Centre, MRC IGMM, University of Edinburgh, Edinburgh, UK
| | - Charlie Gourley
- Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Edinburgh Centre, MRC IGMM, University of Edinburgh, Edinburgh, UK
| | - Jan Rupp
- Department of Infectious Diseases and Microbiology, University of Lübeck, Lübeck, Germany
| | - Jeffrey W Keillor
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Agathe Subtil
- Unité de Biologie cellulaire de l'infection microbienne, CNRS UMR3691, Institut Pasteur, Paris, France
| |
Collapse
|
12
|
Liu B, Wang D, Luo E, Hou J, Qiao Y, Yan G, Wang Q, Tang C. Role of TG2-Mediated SERCA2 Serotonylation on Hypoxic Pulmonary Vein Remodeling. Front Pharmacol 2020; 10:1611. [PMID: 32116663 PMCID: PMC7026497 DOI: 10.3389/fphar.2019.01611] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 12/10/2019] [Indexed: 12/12/2022] Open
Abstract
Sarco-endoplasmic reticulum Ca2+ ATPase (SERCA) pumps take up Ca2+ from the cytoplasm to maintain the balance of intracellular Ca2+. A decline in expression or activity of SERCA results in persistent store-operated calcium entry (SOCE). In cardiomyocytes as well as vascular smooth muscle cells (SMCs), SERCA2 acts as an important regulator of calcium cycling. The purpose of this study is to identify and better understand the role of transglutaminases2 (TG2) as a key factor involved in SERCA2 serotonination (s-SERCA2) and to elucidate the underlying mechanism of action. Human pulmonary venous smooth muscle cell in normal pulmonary lobe were isolated and cultured in vitro. Establishment of hypoxic pulmonary hypertension model in wild type and TG2 knockout mice. SERCA2 serotonylation was analyzed by co-(immunoprecipitation) IP when the TG2 gene silenced or overexpressed under normoxia and hypoxia in vivo and in vitro. Intracellular calcium ion was measured by using Fluo-4AM probe under normoxia and hypoxia. Real-time (RT)-PCR and Western blot analyzed expression of TG2, TRPC1, and TRPC6 under normoxia and hypoxia. Bioactivity of cells were analyzed by using Cell Counting Kit (CCK)-8, flow cytometry, wound healing, RT-PCR, and Western blot under PST-2744 and cyclopiazonic acid. We confirmed that 1) hypoxia enhanced the expression and activity of TG2, and 2) hypoxia increased the basal intracellular Ca2+ concentration ([Ca2+]i) and SOCE through activating TRPC6 on human pulmonary vein smooth muscle cells (hPVSMC). Then, we investigated the effects of overexpression and downregulation of the TG2 gene on the activity of SERCA2, s-SERCA2, basal [Ca2+]i, and SOCE under normoxia and hypoxia in vitro, and investigated the activity of SERCA2 and s-SERCA2 in vivo, respectively. We confirmed that SERCA2 serotonylation inhibited the activity of SERCA2 and increased the Ca2+ influx, and that hypoxia induced TG2-mediated SERCA2 serotonylation both in vivo and in vitro. Furthermore, we investigated the effect of TG2 activity on the biological behavior of hPVSMC by using an inhibitor and agonist of SERCA2, respectively. Finally, we confirmed that chronic hypoxia cannot increase vessel wall thickness, the right ventricular systolic pressure (RVSP), and right ventricular hypertrophy index (RVHI) of vascular smooth muscle-specific Tgm2−/− mice. These results indicated that hypoxia promoted TG2-mediated SERCA2 serotonylation, thereby leading to inhibition of SERCA2 activity, which further increased the calcium influx through the TRPC6 channel. Furthermore, tissue-specific conditional TG2 knockout mice prevents the development of pulmonary hypertension caused by hypoxia. In summary, we uncovered a new target (TG2) for treatment of chronic hypoxic pulmonary hypertension (CHPH).
Collapse
Affiliation(s)
- Bo Liu
- Department of Cardiology, Zhongda Hospital, Southeast University, Nanjing, China
| | - Dong Wang
- Department of Cardiology, Zhongda Hospital, Southeast University, Nanjing, China
| | - Erfei Luo
- Department of Cardiology, Zhongda Hospital, Southeast University, Nanjing, China
| | - Jiantong Hou
- Department of Cardiology, Zhongda Hospital, Southeast University, Nanjing, China
| | - Yong Qiao
- Department of Cardiology, Zhongda Hospital, Southeast University, Nanjing, China
| | - Gaoliang Yan
- Department of Cardiology, Zhongda Hospital, Southeast University, Nanjing, China
| | - Qingjie Wang
- Department of Cardiology, Changzhou No. 2 People's Hospital, Nanjing Medical University, Changzhou, China
| | - Chengchun Tang
- Department of Cardiology, Zhongda Hospital, Southeast University, Nanjing, China
| |
Collapse
|
13
|
D'Eletto M, Rossin F, Fedorova O, Farrace MG, Piacentini M. Transglutaminase type 2 in the regulation of proteostasis. Biol Chem 2019; 400:125-140. [PMID: 29908126 DOI: 10.1515/hsz-2018-0217] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 05/15/2018] [Indexed: 12/21/2022]
Abstract
The maintenance of protein homeostasis (proteostasis) is a fundamental aspect of cell physiology that is essential for the survival of organisms under a variety of environmental and/or intracellular stress conditions. Acute and/or persistent stress exceeding the capacity of the intracellular homeostatic systems results in protein aggregation and/or damaged organelles that leads to pathological cellular states often resulting in cell death. These events are continuously suppressed by a complex macromolecular machinery that uses different intracellular pathways to maintain the proteome integrity in the various subcellular compartments ensuring a healthy cellular life span. Recent findings have highlighted the role of the multifunctional enzyme type 2 transglutaminase (TG2) as a key player in the regulation of intracellular pathways, such as autophagy/mitophagy, exosomes formation and chaperones function, which form the basis of proteostasis regulation under conditions of cellular stress. Here, we review the role of TG2 in these stress response pathways and how its various enzymatic activities might contributes to the proteostasis control.
Collapse
Affiliation(s)
- Manuela D'Eletto
- Department of Biology, University of Rome 'Tor Vergata', Via della Ricerca Scientifica, I-00133 Rome, Italy
| | - Federica Rossin
- Department of Biology, University of Rome 'Tor Vergata', Via della Ricerca Scientifica, I-00133 Rome, Italy
| | - Olga Fedorova
- Institute of Cytology, 194064 Saint-Petersburg, Russia
| | - Maria Grazia Farrace
- Department of Biology, University of Rome 'Tor Vergata', Via della Ricerca Scientifica, I-00133 Rome, Italy
| | - Mauro Piacentini
- Department of Biology, University of Rome 'Tor Vergata', Via della Ricerca Scientifica, I-00133 Rome, Italy.,National Institute for Infectious Diseases I.R.C.C.S. 'Lazzaro Spallanzani', I-00149 Rome, Italy
| |
Collapse
|
14
|
Prieto K, Cao Y, Mohamed E, Trillo-Tinoco J, Sierra RA, Urueña C, Sandoval TA, Fiorentino S, Rodriguez PC, Barreto A. Polyphenol-rich extract induces apoptosis with immunogenic markers in melanoma cells through the ER stress-associated kinase PERK. Cell Death Discov 2019; 5:134. [PMID: 31531232 PMCID: PMC6733947 DOI: 10.1038/s41420-019-0214-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 07/17/2019] [Accepted: 07/25/2019] [Indexed: 12/14/2022] Open
Abstract
Polyphenols elicit antitumor activities, in part, through the induction of anti- or pro-oxidant effects in cancer cells which promote priming of protective anti-tumor immunity. We recently characterized a polyphenol-rich extract from Caesalpinia spinosa (P2Et) that stimulates in vivo antitumor responses against breast and melanoma tumor models via the promotion of immunogenic cancer cell death (ICD). However, the primary mediators whereby P2Et promotes ICD remained unknown. Here, we sought to elucidate the role that severe endoplasmic reticulum (ER) stress plays in mediating P2Et-induced apoptosis and ICD in murine melanoma cells. Our findings demonstrate a substantial selective induction of specific ER-stress mediators in B16-F10 melanoma cells treated with P2Et. While knockout of the ER stress-associated PKR-like ER kinase (PERK) prevented induction of apoptosis and expression of ICD markers in P2Et-treated cells, deletion of X-box binding protein 1 (Xbp1) did not. P2Et-driven activation of PERK in melanoma cells was found to promote ER-calcium release, disrupt mitochondrial membrane potential, and trigger upregulation of ICD drivers, surface calreticulin expression, and extracellular release of ATP and HMGB1. Notably, calcium release inhibition, but not targeting of PERK-driven integrated stress responses, prevented P2Et-induced apoptosis. Collectively, these results underline the central role of PERK-directed calcium release in mediating the antitumor and immunogenic actions of P2Et in melanoma cells.
Collapse
Affiliation(s)
- Karol Prieto
- Grupo de Inmunobiología y Biología Celular, Departamento de Microbiología, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Yu Cao
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL USA
| | - Eslam Mohamed
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL USA
| | - Jimena Trillo-Tinoco
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL USA
| | - Rosa A. Sierra
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL USA
| | - Claudia Urueña
- Grupo de Inmunobiología y Biología Celular, Departamento de Microbiología, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Tito Alejandro Sandoval
- Grupo de Inmunobiología y Biología Celular, Departamento de Microbiología, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Susana Fiorentino
- Grupo de Inmunobiología y Biología Celular, Departamento de Microbiología, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Paulo C. Rodriguez
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL USA
| | - Alfonso Barreto
- Grupo de Inmunobiología y Biología Celular, Departamento de Microbiología, Pontificia Universidad Javeriana, Bogotá, Colombia
| |
Collapse
|
15
|
Ji C, Tang M, Harrison J, Paciorkowski A, Johnson GVW. Nuclear transglutaminase 2 directly regulates expression of cathepsin S in rat cortical neurons. Eur J Neurosci 2018; 48:3043-3051. [PMID: 30239049 DOI: 10.1111/ejn.14159] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 08/14/2018] [Accepted: 08/28/2018] [Indexed: 01/06/2023]
Abstract
Transglutaminase 2 (TG2) is a protein that modulates neuronal survival processes. Although TG2 is primarily cytosolic, data have suggested the nuclear localization of TG2 is strongly associated with neuronal viability. Depletion of TG2 in neurons results in neurite retraction and loss of viability, which is likely due to a dysregulation in gene expression. To begin to understand how TG2 regulates neuronal gene expression, chromatin immunoprecipitation was performed in neurons with TG2 overexpression. The resulting genomic DNA was recovered and sequenced. Bioinformatics analyses revealed that a signature DNA motif was enriched in the TG2 immunoprecipitated genomic DNA. In particular, this motif strongly mapped to a region proximate to the gene Ctss (cathepsin S). Knockdown of TG2 resulted in a significant increase in cathepsin S expression, which preceded the loss of neuronal viability. This is the first demonstration that TG2 directly associates with genomic DNA and regulates gene expression in neurons. Given that expression of cathepsin S is increased in neurological disease states, our data suggest that TG2 may play a role in promoting neuron health in part by repressing the expression of cathepsin S. Overall these data provide new insights into the function of nuclear TG2 in neurons.
Collapse
Affiliation(s)
- Changyi Ji
- Department of Anesthesiology and Perioperative Medicine, University of Rochester, Rochester, New York
| | - Maoping Tang
- Department of Anesthesiology and Perioperative Medicine, University of Rochester, Rochester, New York
| | - Jarreau Harrison
- Department of Pharmacology and Physiology, University of Rochester, Rochester, New York
| | - Alex Paciorkowski
- Departments of Neurology, Pediatrics, Biomedical Genetics, and Neuroscience, University of Rochester Medical Center, Rochester, New York.,Del Monte Institute for Neuroscience, University of Rochester, Rochester, New York
| | - Gail V W Johnson
- Department of Anesthesiology and Perioperative Medicine, University of Rochester, Rochester, New York.,Department of Pharmacology and Physiology, University of Rochester, Rochester, New York.,Del Monte Institute for Neuroscience, University of Rochester, Rochester, New York
| |
Collapse
|
16
|
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
|
17
|
Subcellular localization patterns of transglutaminase 2 in astrocytes and neurons are differentially altered by hypoxia. Neuroreport 2018; 28:1208-1214. [PMID: 28957946 DOI: 10.1097/wnr.0000000000000895] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The multifunctional protein transglutaminase 2 (TG2) has been widely implicated as a modulator of cellular viability. Specifically, TG2 expression is beneficial to neuronal survival following an ischemic injury, whereas the opposite is true in astrocytes. Furthermore, its role in mediating cell death and survival processes has been suggested to be dependent on its subcellular localization. Therefore, the aim of this study was to examine the subcellular localization patterns of neuronal and astrocytic TG2 in ischemia-relevant conditions. We found that nuclear levels of TG2 were significantly increased in neurons, but reduced in astrocytes, in response to hypoxia. In addition, there were no changes in extracellular TG2 in astrocytes exposed to hypoxia. Thus, these findings demonstrate a difference in the subcellular localization pattern of TG2 in neurons and astrocytes in ischemia-relevant conditions and provide further avenues for investigation into the role of TG2 in mediating cellular viability.
Collapse
|
18
|
Quinn BR, Yunes-Medina L, Johnson GVW. Transglutaminase 2: Friend or foe? The discordant role in neurons and astrocytes. J Neurosci Res 2018; 96:1150-1158. [PMID: 29570839 DOI: 10.1002/jnr.24239] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 03/01/2018] [Accepted: 03/12/2018] [Indexed: 12/19/2022]
Abstract
Members of the transglutaminase family catalyze the formation of isopeptide bonds between a polypeptide-bound glutamine and a low molecular weight amine (e.g., spermidine) or the ɛ-amino group of a polypeptide-bound lysine. Transglutaminase 2 (TG2), a prominent member of this family, is unique because in addition to being a transamidating enzyme, it exhibits numerous other activities. As a result, TG2 plays a role in many physiological processes, and its function is highly cell type specific and relies upon a number of factors, including conformation, cellular compartment location, and local concentrations of Ca2+ and guanine nucleotides. TG2 is the most abundant transglutaminase in the central nervous system (CNS) and plays a pivotal role in the CNS injury response. How TG2 affects the cell in response to an insult is strikingly different in astrocytes and neurons. In neurons, TG2 supports survival. Overexpression of TG2 in primary neurons protects against oxygen and glucose deprivation (OGD)-induced cell death and in vivo results in a reduction in infarct volume subsequent to a stroke. Knockdown of TG2 in primary neurons results in a loss of viability. In contrast, deletion of TG2 from astrocytes results in increased survival following OGD and improved ability to protect neurons from injury. Here, a brief overview of TG2 is provided, followed by a discussion of the role of TG2 in transcriptional regulation, cellular dynamics, and cell death. The differing roles TG2 plays in neurons and astrocytes are highlighted and compared to how TG2 functions in other cell types.
Collapse
Affiliation(s)
- Breandan R Quinn
- Department of Pharmacology and Physiology, University of Rochester, Rochester, New York
| | - Laura Yunes-Medina
- Department of Neuroscience, University of Rochester, Rochester, New York
| | - Gail V W Johnson
- Department of Pharmacology and Physiology, University of Rochester, Rochester, New York.,Department of Neuroscience, University of Rochester, Rochester, New York.,Department of Anesthesiology and Perioperative Medicine, University of Rochester, Rochester, New York
| |
Collapse
|
19
|
Tripathy D, Vignoli B, Ramesh N, Polanco MJ, Coutelier M, Stephen CD, Canossa M, Monin ML, Aeschlimann P, Turberville S, Aeschlimann D, Schmahmann JD, Hadjivassiliou M, Durr A, Pandey UB, Pennuto M, Basso M. Mutations in TGM6 induce the unfolded protein response in SCA35. Hum Mol Genet 2018; 26:3749-3762. [PMID: 28934387 DOI: 10.1093/hmg/ddx259] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 06/30/2017] [Indexed: 12/23/2022] Open
Abstract
Spinocerebellar ataxia type 35 (SCA35) is a rare autosomal-dominant neurodegenerative disease caused by mutations in the TGM6 gene, which codes for transglutaminase 6 (TG6). Mutations in TG6 induce cerebellar degeneration by an unknown mechanism. We identified seven patients bearing new mutations in TGM6. To gain insights into the molecular basis of mutant TG6-induced neurotoxicity, we analyzed all the seven new TG6 mutants and the five TG6 mutants previously linked to SCA35. We found that the wild-type (TG6-WT) protein mainly localized to the nucleus and perinuclear area, whereas five TG6 mutations showed nuclear depletion, increased accumulation in the perinuclear area, insolubility and loss of enzymatic function. Aberrant accumulation of these TG6 mutants in the perinuclear area led to activation of the unfolded protein response (UPR), suggesting that specific TG6 mutants elicit an endoplasmic reticulum stress response. Mutations associated with activation of the UPR caused death of primary neurons and reduced the survival of novel Drosophila melanogaster models of SCA35. These results indicate that mutations differently impacting on TG6 function cause neuronal dysfunction and death through diverse mechanisms and highlight the UPR as a potential therapeutic target for patient treatment.
Collapse
Affiliation(s)
- Debasmita Tripathy
- Laboratory of Transcriptional Neurobiology, Centre for Integrative Biology (CIBIO), University of Trento, Trento (TN), Italy
| | - Beatrice Vignoli
- Laboratory of Neural Stem Cells and Neurogenesis, Centre for Integrative Biology (CIBIO), University of Trento, Trento (TN), Italy
| | - Nandini Ramesh
- Division of Child Neurology, Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, USA.,Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Maria Jose Polanco
- Dulbecco Telethon Institute Lab of Neurodegenerative Diseases, Centre for Integrative Biology (CIBIO), University of Trento, Trento (TN), Italy
| | - Marie Coutelier
- INSERM U 1127, Centre National de la Recherche Scientifique UMR 7225, UMRS 1127, Université Pierre et Marie Curie (Paris 06), Sorbonne Universités, Institut du Cerveau et de la Moelle Epinière, 75013 Paris, France
| | - Christopher D Stephen
- Ataxia Unit, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Marco Canossa
- Laboratory of Neural Stem Cells and Neurogenesis, Centre for Integrative Biology (CIBIO), University of Trento, Trento (TN), Italy
| | - Marie-Lorraine Monin
- INSERM U 1127, Centre National de la Recherche Scientifique UMR 7225, UMRS 1127, Université Pierre et Marie Curie (Paris 06), Sorbonne Universités, Institut du Cerveau et de la Moelle Epinière, 75013 Paris, France
| | - Pascale Aeschlimann
- Matrix Biology & Tissue Repair Research Unit, College of Biomedical and Life Sciences, School of Dentistry, Cardiff University, Cardiff, UK
| | - Shannon Turberville
- Matrix Biology & Tissue Repair Research Unit, College of Biomedical and Life Sciences, School of Dentistry, Cardiff University, Cardiff, UK
| | - Daniel Aeschlimann
- Matrix Biology & Tissue Repair Research Unit, College of Biomedical and Life Sciences, School of Dentistry, Cardiff University, Cardiff, UK
| | - Jeremy D Schmahmann
- Ataxia Unit, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Marios Hadjivassiliou
- Academic Department of Neurosciences, Royal Hallamshire Hospital, University of Sheffield, Sheffield, UK
| | - Alexandra Durr
- INSERM U 1127, Centre National de la Recherche Scientifique UMR 7225, UMRS 1127, Université Pierre et Marie Curie (Paris 06), Sorbonne Universités, Institut du Cerveau et de la Moelle Epinière, 75013 Paris, France
| | - Udai B Pandey
- Division of Child Neurology, Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Maria Pennuto
- Dulbecco Telethon Institute Lab of Neurodegenerative Diseases, Centre for Integrative Biology (CIBIO), University of Trento, Trento (TN), Italy
| | - Manuela Basso
- Laboratory of Transcriptional Neurobiology, Centre for Integrative Biology (CIBIO), University of Trento, Trento (TN), Italy
| |
Collapse
|
20
|
Yunes-Medina L, Paciorkowski A, Nuzbrokh Y, Johnson GVW. Depletion of transglutaminase 2 in neurons alters expression of extracellular matrix and signal transduction genes and compromises cell viability. Mol Cell Neurosci 2018; 86:72-80. [PMID: 29197584 PMCID: PMC5736014 DOI: 10.1016/j.mcn.2017.11.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 10/20/2017] [Accepted: 11/21/2017] [Indexed: 12/22/2022] Open
Abstract
The protein transglutaminase 2 (TG2) has been implicated as a modulator of neuronal viability. TG2's role in mediating cell survival processes has been suggested to involve its ability to alter transcriptional events. The goal of this study was to examine the role of TG2 in neuronal survival and to begin to delineate the pathways it regulates. We show that depletion of TG2 significantly compromises the viability of neurons in the absence of any stressors. RNA sequencing revealed that depletion of TG2 dysregulated the expression of 86 genes with 59 of these being upregulated. The genes that were upregulated by TG2 knockdown were primarily involved in extracellular matrix function, cell signaling and cytoskeleton integrity pathways. Finally, depletion of TG2 significantly reduced neurite length. These findings suggest for the first time that TG2 plays a crucial role in mediating neuronal survival through its regulation of genes involved in neurite length and maintenance.
Collapse
Affiliation(s)
- Laura Yunes-Medina
- Department of Neuroscience, University of Rochester, 601 Elmwood Ave, Box 603, Rochester, NY 14642, United States.
| | - Alex Paciorkowski
- Department of Neuroscience, University of Rochester, 601 Elmwood Ave, Box 603, Rochester, NY 14642, United States; Department of Neurology, University of Rochester, 601 Elmwood Ave, Box 603, Rochester, NY 14642, United States; Department of Pediatrics, University of Rochester Medical Center, 601 Elmwood Ave, Box 603, Rochester, NY 14642, United States; Department Biomedical Genetics, University of Rochester Medical Center, 601 Elmwood Ave, Box 604, Rochester, NY 14642, United States.
| | - Yan Nuzbrokh
- Department of Anesthesiology and Perioperative Medicine, University of Rochester Medical Center, 601 Elmwood Ave, Box 604, Rochester, NY 14642, United States.
| | - Gail V W Johnson
- Department of Anesthesiology and Perioperative Medicine, University of Rochester Medical Center, 601 Elmwood Ave, Box 604, Rochester, NY 14642, United States; Department of Pharmacology and Physiology, University of Rochester Medical Center, 601 Elmwood Ave, Box 604, Rochester, NY 14642, United States.
| |
Collapse
|
21
|
Meshram DD, Pike CV, Coussons PJ. Inhibition of Transglutaminase 2 activity increases cisplatin cytotoxicity in a model of human hepatocarcinoma chemotherapy. Eur J Pharmacol 2017; 815:332-342. [DOI: 10.1016/j.ejphar.2017.09.035] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 09/18/2017] [Accepted: 09/20/2017] [Indexed: 02/07/2023]
|
22
|
Lingan JV, Alanzalon RE, Porter GA. Preventing permeability transition pore opening increases mitochondrial maturation, myocyte differentiation and cardiac function in the neonatal mouse heart. Pediatr Res 2017; 81:932-941. [PMID: 28141792 DOI: 10.1038/pr.2017.19] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 12/15/2016] [Indexed: 12/15/2022]
Abstract
BACKGROUND In embryonic myocytes, closure of the mitochondrial permeability transition pore (PTP) drives mitochondrial maturation and cardiac myocyte differentiation. Since neonatal cardiac myocytes remain relatively immature, we hypothesized that inducing PTP closure at this age, by inhibiting the PTP regulator, cyclophilin D (CyPD), genetically or with Cyclosporin A (CsA) and NIM811, would increase cardiac function by increasing mitochondrial maturation and myocyte differentiation. METHODS Cultured neonatal myocytes or neonatal mice were treated for 5 d with vehicle, CsA or NIM811. Mitochondrial function and structure were measured in vitro. Myocyte differentiation was assessed by immunolabeling for contractile proteins. Cardiac function was determined using echocardiography. RESULTS The probability of PTP opening was high in WT neonatal myocytes. Treatment with CsA or NIM811 in vitro increased mitochondrial structural complexity and membrane potential, decreased reactive oxygen species levels, and increased myocyte differentiation. WT mice treated with either CsA or NIM811 in vivo for the first 5 d of life had higher ejection fractions. Deleting CyPD had similar effects as CsA and NIM811 on all parameters. CONCLUSIONS It may be feasible to inhibit the PTP using available drugs to increase mitochondrial maturation, myocyte differentiation, and cardiac function in neonates.
Collapse
Affiliation(s)
- Jayson V Lingan
- Current affiliation: Department of Pediatrics, Benefits Health System, Great Falls, Montana.,Department of Pediatrics, University of Rochester Medical Center, Rochester, New York
| | - Ryan E Alanzalon
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York
| | - George A Porter
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York.,Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, New York.,Aab Cardiovascular Research Institute, University of Rochester Medical Center, Rochester, New York
| |
Collapse
|
23
|
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
|
24
|
Gundemir S, Monteagudo A, Akbar A, Keillor JW, Johnson GVW. The complex role of transglutaminase 2 in glioblastoma proliferation. Neuro Oncol 2017; 19:208-218. [PMID: 27591334 PMCID: PMC5464277 DOI: 10.1093/neuonc/now157] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Glioblastomas (GBMs) are a heterogeneous group of primary brain tumors. These tumors are resistant to therapeutic interventions and invariably recur after surgical resection. The multifunctional protein transglutaminase 2 (TG2) has been shown to promote cell survival in a number of different tumors. There is also evidence that TG2 may be a pro-survival factor in GBMs. However, the roles that TG2 plays in facilitating GBM survival and proliferation have not yet been clearly delineated . METHODS The functions of TG2 are often cell- and context-specific. Therefore, in this study we examined the ability of TG2 to facilitate GBM proliferation using colony formation assays and 5-ethynyl-2'-deoxyuridine (EdU) incorporation in several different GBM cell lines as well as neurospheres derived from patient tumors representing the 3 major subtypes of GBM tumors (mesenchymal, proneural, and classical) and maintained in the absence of serum. TG2 knockdown or selective TG2 inhibitors were used to modulate TG2 expression and activity. RESULTS We show that TG2 plays differential roles in the proliferative process depending on the cell type. In most, but not all, GBM models TG2 plays a crucial role in the proliferative process, and some but not all TG2 inhibitors were highly effective at reducing proliferation in a large subset of the GBM models. CONCLUSION Our results show that TG2 plays an important-but notoriously context-specific-role in GBM cell biology. Nonetheless, as future studies unravel the genetic "fingerprints" that make TG2 inhibitors effective, this information could be exploited to develop TG2 inhibitors into personalized GBM therapies.
Collapse
Affiliation(s)
- Soner Gundemir
- Department of Anesthesiology, University of Rochester, Rochester, New York; Department of Pharmacology and Physiology, University of Rochester, Rochester, New York; Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Ontario, Canada
| | - Alina Monteagudo
- Department of Anesthesiology, University of Rochester, Rochester, New York; Department of Pharmacology and Physiology, University of Rochester, Rochester, New York; Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Ontario, Canada
| | - Abdullah Akbar
- Department of Anesthesiology, University of Rochester, Rochester, New York; Department of Pharmacology and Physiology, University of Rochester, Rochester, New York; Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Ontario, Canada
| | - Jeffrey W Keillor
- Department of Anesthesiology, University of Rochester, Rochester, New York; Department of Pharmacology and Physiology, University of Rochester, Rochester, New York; Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Ontario, Canada
| | - Gail V W Johnson
- Department of Anesthesiology, University of Rochester, Rochester, New York; Department of Pharmacology and Physiology, University of Rochester, Rochester, New York; Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
25
|
Identification of DNAJA1 as a novel interacting partner and a substrate of human transglutaminase 2. Biochem J 2016; 473:3889-3901. [PMID: 27551108 DOI: 10.1042/bcj20160440] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 08/22/2016] [Indexed: 11/17/2022]
Abstract
Transglutaminase 2 (TG2) is a ubiquitously expressed multifunctional member of the transglutaminase enzyme family. It has been implicated to have roles in many physiological and pathological processes such as differentiation, apoptosis, signal transduction, adhesion and migration, wound healing and inflammation. Previous studies revealed that TG2 has various intra- and extra-cellular interacting partners, which contribute to these processes. In the present study, we identified a molecular co-chaperone, DNAJA1, as a novel interacting partner of human TG2 using a GST pull-down assay and subsequent mass spectrometry analysis, and further confirmed this interaction via ELISA and surface plasmon resonance measurements. Interaction studies were also performed with domain variants of TG2 and results suggest that the catalytic core domain of TG2 is essential for the TG2-DNAJA1 interaction. Cross-linking activity was not essential for the interaction since DNAJA1 was also found to interact with the catalytically inactive form of TG2. Furthermore, we have showed that DNAJA1 interacts with the open form of TG2 and regulates its transamidation activity under both in vitro and in situ conditions. We also found that DNAJA1 is a glutamine donor substrate of TG2. Since DNAJA1 and TG2 are reported to regulate common pathological conditions such as neurodegenerative disorders and cancer, the findings in the present paper open up possibilities to explore molecular mechanisms behind TG2-regulated functions.
Collapse
|
26
|
Fisher ML, Adhikary G, Xu W, Kerr C, Keillor JW, Eckert RL. Type II transglutaminase stimulates epidermal cancer stem cell epithelial-mesenchymal transition. Oncotarget 2016; 6:20525-39. [PMID: 25971211 PMCID: PMC4653023 DOI: 10.18632/oncotarget.3890] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 04/25/2015] [Indexed: 12/12/2022] Open
Abstract
Type II transglutaminase (TG2) is a multifunctional protein that has recently been implicated as having a role in ECS cell survival. In the present study we investigate the role of TG2 in regulating epithelial mesenchymal transition (EMT) in ECS cells. Our studies show that TG2 knockdown or treatment with TG2 inhibitor, results in a reduced EMT marker expression, and reduced cell migration and invasion. TG2 has several activities, but the most prominent are its transamidase and GTP binding activity. Analysis of a series of TG2 mutants reveals that TG2 GTP binding activity, but not the transamidase activity, is required for expression of EMT markers (Twist, Snail, Slug, vimentin, fibronectin, N-cadherin and HIF-1α), and increased ECS cell invasion and migration. This coupled with reduced expression of E-cadherin. Additional studies indicate that NF&#ξ03BA;B signaling, which has been implicated as mediating TG2 impact on EMT in breast cancer cells, is not involved in TG2 regulation of EMT in skin cancer. These studies suggest that TG2 is required for maintenance of ECS cell EMT, invasion and migration, and suggests that inhibiting TG2 GTP binding/G-protein related activity may reduce skin cancer tumor survival.
Collapse
Affiliation(s)
- Matthew L Fisher
- Departments of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Gautam Adhikary
- Departments of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Wen Xu
- Departments of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Candace Kerr
- Departments of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Jeffrey W Keillor
- Department of Chemistry, University of Ottawa, Ottawa, Ontario, Canada
| | - Richard L Eckert
- Departments of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland, USA.,Dermatology, University of Maryland School of Medicine, Baltimore, Maryland, USA.,Reproductive Biology, University of Maryland School of Medicine, Baltimore, Maryland, USA.,Marlene and Stewart Greenebaum Cancer, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
27
|
Singh G, Zhang J, Ma Y, Cerione RA, Antonyak MA. The Different Conformational States of Tissue Transglutaminase Have Opposing Affects on Cell Viability. J Biol Chem 2016; 291:9119-32. [PMID: 26893378 PMCID: PMC4861479 DOI: 10.1074/jbc.m115.699108] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 02/11/2016] [Indexed: 02/06/2023] Open
Abstract
Tissue transglutaminase (tTG) is an acyltransferase/GTP-binding protein that contributes to the development of various diseases. In human cancer cells, tTG activates signaling pathways that promote cell growth and survival, whereas in other disorders (i.e. neurodegeneration), overexpression of tTG enhances cell death. Therefore, it is important to understand how tTG is differentially regulated and functioning to promote diametrically distinct cellular outcomes. Previous structural studies revealed that tTG adopts either a nucleotide-bound closed conformation or a transamidation-competent open conformation. Here we provide evidence showing that these different conformational states determine whether tTG promotes, or is detrimental to, cell survival, with the open conformation of the protein being responsible for inducing cell death. First, we demonstrate that a nucleotide binding-defective form of tTG, which has previously been shown to induce cell death, assumes an open conformation in solution as assessed by an enhanced sensitivity to trypsin digestion and by small angle x-ray scattering (SAXS) analysis. We next identify two pairs of intramolecular hydrogen bonds that, based on existing x-ray structures, are predicted to form between the most C-terminal β-barrel domain and the catalytic core domain of tTG. By disrupting these hydrogen bonds, we are able to generate forms of tTG that constitutively assume an open conformation and induce apoptosis. These findings provide important insights into how tTG participates in the pathogenesis of neurodegenerative diseases, particularly with regard to the actions of a C-terminal truncated form of tTG (TG-Short) that has been linked to such disorders and induces apoptosis by assuming an open-like conformation.
Collapse
Affiliation(s)
| | | | - Yilun Ma
- From the Department of Molecular Medicine and
| | - Richard A Cerione
- From the Department of Molecular Medicine and the Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853
| | | |
Collapse
|
28
|
Currò M, Gangemi C, Gugliandolo A, Risitano R, Ferlazzo N, Ientile R, Caccamo D. Transglutaminase 2 is involved in homocysteine-induced activation of human THP-1 monocytes. Free Radic Res 2015; 49:299-308. [PMID: 25547897 DOI: 10.3109/10715762.2014.1002495] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Aberrant transglutaminase 2 (TG2) expression and protein cross-linking activity have been associated with several chronic neurodegenerative disorders in which inflammatory processes triggered by activated microglia and monocytes play a key role, such as Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and multiple sclerosis. Interestingly, mild-to-moderate hyperhomocysteinemia (HHcy), corresponding to increased plasma homocysteine (Hcy) concentrations in the range 16-60 μM, have recently been associated with the above-cited diseases. Using THP-1 monocytes, here we investigated the role of TG2 in cell response to mildly elevated Hcy concentrations. A five-day incubation with Hcy (∼25 μM) increased reactive oxygen species, peroxide lipids, as well as 8-hydroxyguanosine levels by twofold, and decreased the endogenous cell antioxidant defenses, that is reduced glutathione, by 50% in Hcy-exposed cultures compared with controls (p < 0.01). Hcy-induced oxidative stress was associated with increases in TG2 expression and activity, as well as nuclear factor kappa B activation. Notably, the latter was reduced in the presence of the TG-specific inhibitor R283. Hcy exposure also significantly increased the mRNA levels of tumor necrosis factor alpha, interleukin (IL)-6, and IL-1β, as well as the level of Hcy-inducible endoplasmic reticulum (ER) stress protein, a marker of ER stress, in Hcy-exposed cultures compared with controls. Notably, these effects were dramatically reduced by R283. These preliminary findings indicate that TG2 plays a key role in Hcy-induced activation of THP-1 monocytes, involving oxidative as well as ER stress and inflammation. This underlines the potential of TG2 inhibition in the therapeutic management of inflammatory processes contributing to neurodegenerative disorders associated with mild HHcy.
Collapse
Affiliation(s)
- M Currò
- Department of Biomedical Sciences and Morphofunctional Imaging, University of Messina , Messina , Italy
| | | | | | | | | | | | | |
Collapse
|
29
|
Tissue transglutaminase promotes serotonin-induced AKT signaling and mitogenesis in pulmonary vascular smooth muscle cells. Cell Signal 2014; 26:2818-25. [PMID: 25218191 DOI: 10.1016/j.cellsig.2014.09.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2014] [Accepted: 09/02/2014] [Indexed: 01/06/2023]
Abstract
Tissue transglutaminase 2 (TG2) is a multifunctional enzyme that cross-links proteins with monoamines such as serotonin (5-hydroxytryptamine, 5-HT) via a transglutamidation reaction, and is associated with pathophysiologic vascular responses. 5-HT is a mitogen for pulmonary artery smooth muscle cells (PASMCs) that has been linked to pulmonary vascular remodeling underlying pulmonary hypertension development. We previously reported that 5-HT-induced PASMC proliferation is inhibited by the TG2 inhibitor monodansylcadaverine (MDC); however, the mechanisms are poorly understood. In the present study we hypothesized that TG2 contributes to 5-HT-induced signaling pathways of PASMCs. Pre-treatment of bovine distal PASMCs with varying concentrations of the inhibitor MDC led to differential inhibition of 5-HT-stimulated AKT and ROCK activation, while p-P38 was unaffected. Concentration response studies showed significant inhibition of AKT activation at 50 μM MDC, along with inhibition of the AKT downstream targets mTOR, p-S6 kinase and p-S6. Furthermore, TG2 depletion by siRNA led to reduced 5-HT-induced AKT activation. Immunoprecipitation studies showed that 5-HT treatment led to increased levels of serotonylated AKT and increased TG2-AKT complex formations which were inhibited by MDC. Overexpression of TG2 point mutant cDNAs in PASMCs showed that the TG2 C277V transamidation mutant blunted 5-HT-induced AKT activation and 5-HT-induced PASMC mitogenesis. Finally, 5-HT-induced AKT activation was blunted in SERT genetic knock-out rat cells, but not in their wild-type counterpart. The SERT inhibitor imipramine similarly blocked AKT activation. These results indicate that TG2 contributes to 5-HT-induced distal PASMC proliferation via promotion of AKT signaling, likely via its serotonylation. Taken together, these results provide new insight into how TG2 may participate in vascular smooth muscle remodeling.
Collapse
|
30
|
New biomarkers to predict the evolution of in situ breast cancers. BIOMED RESEARCH INTERNATIONAL 2014; 2014:159765. [PMID: 25243117 PMCID: PMC4160633 DOI: 10.1155/2014/159765] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 07/21/2014] [Accepted: 07/23/2014] [Indexed: 11/17/2022]
Abstract
BACKGROUND Genomic studies have shown that gene expression profiles are similar in in situ (CIS) and invasive breast cancers, suggesting that several biofunctional modifications of the transformation process occur before or during the development of CIS lesion. METHODS We investigated 3 biomarkers in 44 patients with CIS: TG2 (transglutaminase 2), HJURP (Holliday junction recognition protein), and HIF-1α (hypoxia inducible factor-1 alpha). RESULTS TG2 was more highly expressed than the other two markers and significantly more so in stromal than in tumor cells. HIF-1α evaluation showed a higher expression in both tumor and stromal cells in patients with relapsed G3 tumors, indicating a potential role of this marker in CIS evolution. A greater than sevenfold higher risk of relapse (P = 0.050) was observed in patients highly expressing HJURP in stroma and a tenfold higher recurrence risk (P = 0.026) was seen in those with a higher stromal HIF-1α expression. An important increase in risk accuracy (AUC 0.80) was obtained when HIF-1α and HJURP were evaluated together. CONCLUSIONS Despite the limited number of relapsed patients, we formulated some hypotheses on the factors responsible for malignant evolution and recurrence which are now being tested in a large case series with a longer follow-up.
Collapse
|
31
|
Transglutaminase 2 ablation leads to mitophagy impairment associated with a metabolic shift towards aerobic glycolysis. Cell Death Differ 2014; 22:408-18. [PMID: 25060553 DOI: 10.1038/cdd.2014.106] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 06/11/2014] [Accepted: 06/13/2014] [Indexed: 01/07/2023] Open
Abstract
Macroautophagy selectively degrades dysfunctional mitochondria by a process known as mitophagy. Here we demonstrate the involvement of transglutaminase 2 (TG2) in the turnover and degradation of damaged mitochondria. In TG2-ablated cells we observed the presence of a large number of fragmented mitochondria that display decreased membrane potential, downregulation of IF1 along with increased Drp1 and PINK1 levels, two key proteins regulating the mitochondrial fission. Of note, we demonstrate that in healthy mitochondria, TG2 interacts with the dynamic proteins Drp1 and Fis1; interestingly, their interaction is largely reduced upon induction of the fission process by carbonyl cyanide m-chlorophenyl hydrazine (CCCP). In keeping with these findings, mitochondria lacking TG2 are more susceptible to CCCP treatment. As a consequence of accumulation of damaged mitochondria, cells lacking TG2 increased their aerobic glycolysis and became sensitive to the glycolytic inhibitor 2-deoxy-D-glucose (2-DG). In contrast, TG2-proficient cells are more resistant to 2-DG-induced apoptosis as the caspase 3 is inactivated through the enzyme's crosslinking activity. The data presented in this study show that TG2 plays a key role in cellular dynamics and consequently influences the energetic metabolism.
Collapse
|
32
|
Eckert RL, Kaartinen MT, Nurminskaya M, Belkin AM, Colak G, Johnson GVW, Mehta K. Transglutaminase regulation of cell function. Physiol Rev 2014; 94:383-417. [PMID: 24692352 DOI: 10.1152/physrev.00019.2013] [Citation(s) in RCA: 330] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Transglutaminases (TGs) are multifunctional proteins having enzymatic and scaffolding functions that participate in regulation of cell fate in a wide range of cellular systems and are implicated to have roles in development of disease. This review highlights the mechanism of action of these proteins with respect to their structure, impact on cell differentiation and survival, role in cancer development and progression, and function in signal transduction. We also discuss the mechanisms whereby TG level is controlled and how TGs control downstream targets. The studies described herein begin to clarify the physiological roles of TGs in both normal biology and disease states.
Collapse
|