1
|
Gostomczyk K, Drozd M, Marsool Marsool MD, Pandey A, Tugas K, Chacon J, Tayyab H, Ullah A, Borowczak J, Szylberg Ł. Biomarkers for the detection of circulating tumor cells. Exp Cell Res 2025; 448:114555. [PMID: 40228709 DOI: 10.1016/j.yexcr.2025.114555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 04/05/2025] [Accepted: 04/09/2025] [Indexed: 04/16/2025]
Abstract
Circulating tumor cells (CTCs) have emerged as a key biomarker in cancer detection and prognosis, and their molecular profiling is gaining importance in precision oncology. Liquid biopsies, which allow the extraction of CTCs, circulating tumor DNA (ctDNA) or cell-free DNA (cfDNA), have measurable advantages over traditional tissue biopsies, especially when molecular material is difficult to obtain. However, this method is not without limitations. Difficulties in differentiating between primary and metastatic lesions, uncertain predictive values and the complexity of the biomarkers used can prove challenging. Recently, high cell heterogeneity has been identified as the main obstacle to achieving high diagnostic accuracy. Because not all cells undergo epithelial-mesenchymal transition (EMT) at the same time, there is a large population of hybrid CTCs that express both epithelial and mesenchymal markers. Since traditional diagnostic tools primarily detect epithelial markers, they are often unable to detect cells with a hybrid phenotype; therefore, additional markers may be required to avoid false negatives. In this review, we summarize recent reports on emerging CTCs markers, with particular emphasis on their use in cancer diagnosis. Most of them, including vimentin, TWIST1, SNAI1, ZEB1, cadherins, CD44, TGM2, PD-L1 and GATA, hold promise for the detection of CTCs, but are also implicated in cancer progression, metastasis, and therapeutic resistance. Therefore, understanding the nature and drivers of epithelial-mesenchymal plasticity (EMP) is critical to advancing our knowledge in this field.
Collapse
Affiliation(s)
- Karol Gostomczyk
- Department of Obstetrics, Gynaecology and Oncology, Collegium Medicum Nicolaus Copernicus University, Bydgoszcz, Poland; Department of Tumor Pathology and Pathomorphology, Oncology Center - Prof. Franciszek Łukaszczyk Memorial Hospital, Bydgoszcz, Poland; Department of Pathology, Dr Jan Biziel Memorial University Hospital, Bydgoszcz, Poland.
| | - Magdalena Drozd
- Department of Obstetrics, Gynaecology and Oncology, Collegium Medicum Nicolaus Copernicus University, Bydgoszcz, Poland; Department of Pathology, Dr Jan Biziel Memorial University Hospital, Bydgoszcz, Poland
| | | | - Anju Pandey
- Memorial Sloan Kettering Cancer Center, New York, USA
| | | | - Jose Chacon
- American University of Integrative Sciences, Saint Martin, Cole Bay, Barbados
| | | | - Ashraf Ullah
- Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Jędrzej Borowczak
- Department of Clinical Oncology, Oncology Center - Prof. Franciszek Łukaszczyk Memorial Hospital, Bydgoszcz, Poland
| | - Łukasz Szylberg
- Department of Obstetrics, Gynaecology and Oncology, Collegium Medicum Nicolaus Copernicus University, Bydgoszcz, Poland; Department of Tumor Pathology and Pathomorphology, Oncology Center - Prof. Franciszek Łukaszczyk Memorial Hospital, Bydgoszcz, Poland; Department of Pathology, Dr Jan Biziel Memorial University Hospital, Bydgoszcz, Poland
| |
Collapse
|
2
|
Libring S, Reinhart-King CA. Premetastatic niche mechanics and organotropism in breast cancer. NPJ BIOLOGICAL PHYSICS AND MECHANICS 2025; 2:11. [PMID: 40191104 PMCID: PMC11968405 DOI: 10.1038/s44341-025-00015-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 02/14/2025] [Indexed: 04/09/2025]
Abstract
Numerous physical and mechanical changes occur in the premetastatic niche. Here, we review the mechanics of the premetastatic niche and how the altered extracellular matrix and cancer cell mechanics may play a role in organotropism in breast cancer. Future research into premetastatic niche development and organotropic cell behavior should address physical alterations and biomechanical effects to the same rigor that biochemical alterations are studied.
Collapse
Affiliation(s)
- Sarah Libring
- Department of Bioengineering, Rice University, Houston, TX USA
| | | |
Collapse
|
3
|
Shu ST, Chen L, Gonzalez-Areizaga G, Smithgall TE. Constitutive activation of the Src-family kinases Fgr and Hck enhances the tumor burden of acute myeloid leukemia cells in immunocompromised mice. Sci Rep 2025; 15:174. [PMID: 39747387 PMCID: PMC11697302 DOI: 10.1038/s41598-024-83740-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 12/17/2024] [Indexed: 01/04/2025] Open
Abstract
Overexpression of the myeloid Src-family kinases Fgr and Hck has been linked to the development of acute myeloid leukemia (AML). Here we characterized the contribution of active forms of these kinases to AML cell cytokine dependence, inhibitor sensitivity, and AML cell engraftment in vivo. The human TF-1 erythroleukemia cell line was used as a model system as it does not express endogenous Hck or Fgr. To induce constitutive kinase activity, Hck and Fgr were fused to the coiled-coil (CC) oligomerization domain of the breakpoint cluster region protein associated with the Bcr-Abl tyrosine kinase in chronic myeloid leukemia. Expression of CC-Hck or CC-Fgr transformed TF-1 cells to a granulocyte-macrophage colony-stimulating factor (GM-CSF)-independent phenotype that correlated with enhanced phosphorylation of the kinase domain activation loop. Both CC-Hck and CC-Fgr cell populations became sensitized to growth arrest by Src-family kinase inhibitors previously shown to suppress the growth of bone marrow cells from AML patients in vitro and decrease AML cell engraftment in immunocompromised mice. Methionine substitution of the 'gatekeeper' residue (Thr338) also stimulated Hck and Fgr kinase activity and transformed TF-1 cells to GM-CSF independence without CC fusion. TF-1 cells expressing either active form of Hck or Fgr engrafted immunocompromised mice faster and developed more extensive tumors compared to mice engrafted with the parent cell line, resulting in shorter survival. Expression of wild-type Hck also significantly enhanced bone marrow engraftment without an activating mutation. Reverse phase protein array analysis linked active Hck and Fgr to the mammalian target of rapamycin complex-1/p70 S6 ribosomal protein (mTORC-1/S6) kinase and focal adhesion kinase (Fak) signaling pathways. Combining Hck and Fgr inhibitors with existing mTORC-1/S6 kinase or Fak inhibitors may improve clinical responses and reduce the potential for acquired resistance.
Collapse
Affiliation(s)
- Sherry T Shu
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Suite 523, Bridgeside Point II, 450 Technology Drive, Pittsburgh, PA, 15219, USA
| | - Li Chen
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Suite 523, Bridgeside Point II, 450 Technology Drive, Pittsburgh, PA, 15219, USA
| | - Giancarlo Gonzalez-Areizaga
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Suite 523, Bridgeside Point II, 450 Technology Drive, Pittsburgh, PA, 15219, USA
| | - Thomas E Smithgall
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Suite 523, Bridgeside Point II, 450 Technology Drive, Pittsburgh, PA, 15219, USA.
| |
Collapse
|
4
|
Blaheta RA, Han J, Oppermann E, Bechstein WO, Burkhard K, Haferkamp A, Rieger MA, Malkomes P. Transglutaminase 2 promotes epithelial-to-mesenchymal transition by regulating the expression of matrix metalloproteinase 7 in colorectal cancer cells via the MEK/ERK signaling pathway. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167538. [PMID: 39389321 DOI: 10.1016/j.bbadis.2024.167538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 09/19/2024] [Accepted: 10/04/2024] [Indexed: 10/12/2024]
Abstract
Tissue transglutaminase 2 (TGM2) and matrix metalloproteinase 7 (MMP7) are suggested to be involved in cancer development and progression, however, their specific role in colon cancer remains elusive. The present study investigated whether TGM2 and MMP7 influence epithelial-mesenchymal-transition (EMT) processes of colon cancer cells. TGM2 was either overexpressed or knocked down in SW480 and HCT-116 cells, and MMP7 expression and activity analyzed. Conversely, MMP7 was silenced and its correlation with TGM2 expression and activity examined. Co-immunoprecipitation served to evaluate TGM2-MMP7-interaction. TGM2 and MMP7 expression were correlated with invasion, migration, EMT marker expression (E-cadherin, N-cadherin, Slug, Snail), and ERK/MEK signaling. TGM2 overexpression enhanced MMP7 expression and activity, promoted cell invasion, migration and EMT, characterized by increased N-cadherin and Snail/Slug expression. TGM2 knockdown resulted in the opposite effects. Knocking down MMP7 was associated with reduced TGM2 protein expression, cell invasion and migration. Down-regulation of MMP7 diminished ERK/MEK signaling, whereas its up-regulation activated this pathway. The ERK-inhibitor GDC-0994 blocked phosphorylation of MEK/ERK and suppressed TGM2 and MMP7. TGM2 communicates with MMP7 in colon cancer cells forces cell migration and invasion by the MEK/ERK signaling pathway and triggers EMT. Inhibiting TGM2 could thus offer new therapeutic options to treat patients with colon cancer, particularly to prevent metastatic progression.
Collapse
Affiliation(s)
- Roman A Blaheta
- University Medical Center Mainz, Dept. of Urology and Pediatric Urology, 55131 Mainz, Germany.
| | - Jiaoyan Han
- Department for General, Visceral, Transplant and Thoracic Surgery, Goethe University, 60590 Frankfurt am Main, Germany
| | - Elsie Oppermann
- Department for General, Visceral, Transplant and Thoracic Surgery, Goethe University, 60590 Frankfurt am Main, Germany
| | - Wolf Otto Bechstein
- Department for General, Visceral, Transplant and Thoracic Surgery, Goethe University, 60590 Frankfurt am Main, Germany
| | - Katrin Burkhard
- Department for General, Visceral, Transplant and Thoracic Surgery, Goethe University, 60590 Frankfurt am Main, Germany; Current affiliation: Department of Legal Medicine, University of Saarland Medical School, 66421 Homburg, Germany
| | - Axel Haferkamp
- University Medical Center Mainz, Dept. of Urology and Pediatric Urology, 55131 Mainz, Germany
| | - Michael A Rieger
- Department of Medicine II, Hematology/Oncology, Goethe University, 60590 Frankfurt am Main, Germany; Cardio-Pulmonary-Institute, 60590 Frankfurt am Main, Germany; Frankfurt Cancer Institute, 60590 Frankfurt am Main, Germany; German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Patrizia Malkomes
- Department for General, Visceral, Transplant and Thoracic Surgery, Goethe University, 60590 Frankfurt am Main, Germany; Frankfurt Cancer Institute, 60590 Frankfurt am Main, Germany; German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| |
Collapse
|
5
|
Ancona P, Trentini A, Terrazzan A, Grassilli S, Navals P, Gates EWJ, Rosta V, Cervellati C, Bergamini CM, Pignatelli A, Keillor JW, Taccioli C, Bianchi N. Transcriptomics Studies Reveal Functions of Transglutaminase 2 in Breast Cancer Cells Using Membrane Permeable and Impermeable Inhibitors. J Mol Biol 2024; 436:168569. [PMID: 38604527 DOI: 10.1016/j.jmb.2024.168569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 04/02/2024] [Accepted: 04/05/2024] [Indexed: 04/13/2024]
Abstract
Transglutaminase 2 (TG2) performs many functions both under physiological and pathological conditions. In cancer, its expression is associated with aggressiveness, propensity to epithelial-mesenchymal transition, and metastasis. Since TG2 performs key functions both outside and inside the cell, using inhibitors with different membrane permeability we analyzed the changes in the transcriptome induced in two triple-negative cell lines (MDA-MB-436 and MDA-MB-231) with aggressive features. By characterizing pathways and gene networks, we were able to define the effects of TG2 inhibitors (AA9, membrane-permeable, and NCEG2, impermeable) in relation to the roles of the enzyme in the intra- and extracellular space within the context of breast cancer. The deregulated genes revealed p53 and integrin signaling to be the common pathways with some genes showing opposite changes in expression. In MDA-MB-436, AA9 induced apoptosis, modulated cadherin, Wnt, gastrin and cholecystokinin receptors (CCKR) mediated signaling, with RHOB and GNG2 playing significant roles, and affected the Warburg effect by decreasing glycolytic enzymes. In MDA-MB-231 cells, AA9 strongly impacted HIF-mediated hypoxia, including AKT and mTOR pathway. These effects suggest an anti-tumor activity by blocking intracellular TG2 functions. Conversely, the use of NCEG2 stimulated the expression of ATP synthase and proteins involved in DNA replication, indicating a potential promotion of cell proliferation through inhibition of extracellular TG2. To effectively utilize these molecules as an anti-tumor strategy, an appropriate delivery system should be evaluated to target specific functions and avoid adverse effects. Additionally, considering combinations with other pathway modulators is crucial.
Collapse
Affiliation(s)
- Pietro Ancona
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy.
| | - Alessandro Trentini
- Department of Environmental Sciences and Prevention, University of Ferrara, Ferrara, Italy.
| | - Anna Terrazzan
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy.
| | - Silvia Grassilli
- Department of Environmental Sciences and Prevention, University of Ferrara, Ferrara, Italy.
| | - Pauline Navals
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada.
| | - Eric W J Gates
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada.
| | - Valentina Rosta
- Department of Environmental Sciences and Prevention, University of Ferrara, Ferrara, Italy.
| | - Carlo Cervellati
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy.
| | - Carlo M Bergamini
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy.
| | - Angela Pignatelli
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy.
| | - Jeffrey W Keillor
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada.
| | - Cristian Taccioli
- Department of Animal Medicine, Production and Health, University of Padua, Padua, Italy.
| | - Nicoletta Bianchi
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy.
| |
Collapse
|
6
|
Buccarelli M, Castellani G, Fiorentino V, Pizzimenti C, Beninati S, Ricci-Vitiani L, Scattoni ML, Mischiati C, Facchiano F, Tabolacci C. Biological Implications and Functional Significance of Transglutaminase Type 2 in Nervous System Tumors. Cells 2024; 13:667. [PMID: 38667282 PMCID: PMC11048792 DOI: 10.3390/cells13080667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 04/04/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
Transglutaminase type 2 (TG2) is the most ubiquitously expressed member of the transglutaminase family. TG2 catalyzes the transamidation reaction leading to several protein post-translational modifications and it is also implicated in signal transduction thanks to its GTP binding/hydrolyzing activity. In the nervous system, TG2 regulates multiple physiological processes, such as development, neuronal cell death and differentiation, and synaptic plasticity. Given its different enzymatic activities, aberrant expression or activity of TG2 can contribute to tumorigenesis, including in peripheral and central nervous system tumors. Indeed, TG2 dysregulation has been reported in meningiomas, medulloblastomas, neuroblastomas, glioblastomas, and other adult-type diffuse gliomas. The aim of this review is to provide an overview of the biological and functional relevance of TG2 in the pathogenesis of nervous system tumors, highlighting its involvement in survival, tumor inflammation, differentiation, and in the resistance to standard therapies.
Collapse
Affiliation(s)
- Mariachiara Buccarelli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.B.); (G.C.); (L.R.-V.); (F.F.)
| | - Giorgia Castellani
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.B.); (G.C.); (L.R.-V.); (F.F.)
| | - Vincenzo Fiorentino
- Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy;
| | - Cristina Pizzimenti
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, 98125 Messina, Italy;
| | - Simone Beninati
- Department of Biology, University of Rome “Tor Vergata”, 00133 Rome, Italy;
| | - Lucia Ricci-Vitiani
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.B.); (G.C.); (L.R.-V.); (F.F.)
| | - Maria Luisa Scattoni
- Research Coordination and Support Service, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy;
| | - Carlo Mischiati
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy;
| | - Francesco Facchiano
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.B.); (G.C.); (L.R.-V.); (F.F.)
| | - Claudio Tabolacci
- Research Coordination and Support Service, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy;
| |
Collapse
|
7
|
Zaltron E, Vianello F, Ruzza A, Palazzo A, Brillo V, Celotti I, Scavezzon M, Rossin F, Leanza L, Severin F. The Role of Transglutaminase 2 in Cancer: An Update. Int J Mol Sci 2024; 25:2797. [PMID: 38474044 DOI: 10.3390/ijms25052797] [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: 01/22/2024] [Revised: 02/25/2024] [Accepted: 02/26/2024] [Indexed: 03/14/2024] Open
Abstract
Transglutaminase type 2 (TG2) is the most ubiquitously expressed and well characterized member of the transglutaminase family. It is a ubiquitous multifunctional enzyme implicated in the regulation of several cellular pathways that support the survival, death, and general homeostasis of eukaryotic cells. Due to its multiple localizations both inside and outside the cell, TG2 participates in the regulation of many crucial intracellular signaling cascades in a tissue- and cell-specific manner, making this enzyme an important player in disease development and progression. Moreover, TG2 is capable of modulating the tumor microenvironment, a process of dynamic tissue remodeling and biomechanical events, resulting in changes which influence tumor initiation, growth, and metastasis. Even if generally related to the Ca2+-dependent post-translational modification of proteins, a number of different biological functions have been ascribed to TG2, like those of a peptide isomerase, protein kinase, guanine nucleotide binder, and cytosolic-nuclear translocator. With respect to cancer, TG2's role is controversial and highly debated; it has been described both as an anti- and pro-apoptotic factor and is linked to all the processes of tumorigenesis. However, numerous pieces of evidence support a tissue-specific role of TG2 so that it can assume both oncogenic and tumor-suppressive roles.
Collapse
Affiliation(s)
| | | | - Alessia Ruzza
- Department of Biology, University of Padua, 35131 Padua, Italy
| | - Alberta Palazzo
- Department of Biology, University of Padua, 35131 Padua, Italy
| | | | - Ilaria Celotti
- Department of Biology, University of Padua, 35131 Padua, Italy
| | | | - Federica Rossin
- Department of Biology, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Luigi Leanza
- Department of Biology, University of Padua, 35131 Padua, Italy
| | - Filippo Severin
- Department of Biology, University of Padua, 35131 Padua, Italy
| |
Collapse
|
8
|
Liu J, Mouradian MM. Pathogenetic Contributions and Therapeutic Implications of Transglutaminase 2 in Neurodegenerative Diseases. Int J Mol Sci 2024; 25:2364. [PMID: 38397040 PMCID: PMC10888553 DOI: 10.3390/ijms25042364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 02/07/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
Neurodegenerative diseases encompass a heterogeneous group of disorders that afflict millions of people worldwide. Characteristic protein aggregates are histopathological hallmark features of these disorders, including Amyloid β (Aβ)-containing plaques and tau-containing neurofibrillary tangles in Alzheimer's disease, α-Synuclein (α-Syn)-containing Lewy bodies and Lewy neurites in Parkinson's disease and dementia with Lewy bodies, and mutant huntingtin (mHTT) in nuclear inclusions in Huntington's disease. These various aggregates are found in specific brain regions that are impacted by neurodegeneration and associated with clinical manifestations. Transglutaminase (TG2) (also known as tissue transglutaminase) is the most ubiquitously expressed member of the transglutaminase family with protein crosslinking activity. To date, Aβ, tau, α-Syn, and mHTT have been determined to be substrates of TG2, leading to their aggregation and implicating the involvement of TG2 in several pathophysiological events in neurodegenerative disorders. In this review, we summarize the biochemistry and physiologic functions of TG2 and describe recent advances in the pathogenetic role of TG2 in these diseases. We also review TG2 inhibitors tested in clinical trials and discuss recent TG2-targeting approaches, which offer new perspectives for the design of future highly potent and selective drugs with improved brain delivery as a disease-modifying treatment for neurodegenerative disorders.
Collapse
Affiliation(s)
| | - M. Maral Mouradian
- RWJMS Institute for Neurological Therapeutics and Department of Neurology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA;
| |
Collapse
|
9
|
Kim GR, Kang JH, Kim HJ, Im E, Bae J, Kwon WS, Rha SY, Chung HC, Cho EY, Kim SY, Kim YC. Discovery of novel 1H-benzo[d]imidazole-4,7-dione based transglutaminase 2 inhibitors as p53 stabilizing anticancer agents in renal cell carcinoma. Bioorg Chem 2024; 143:107061. [PMID: 38154386 DOI: 10.1016/j.bioorg.2023.107061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/21/2023] [Accepted: 12/23/2023] [Indexed: 12/30/2023]
Abstract
Overexpression of transglutaminase 2 (TGase 2; TG2) has been implicated in the progression of renal cell carcinoma (RCC) through the inactivation of p53 by forming a protein complex. Because most p53 in RCC has no mutations, apoptosis can be increased by inhibiting the binding between TG2 and p53 to increase the stability of p53. In the present study, a novel TG2 inhibitor was discovered by investigating the structure of 1H-benzo[d]imidazole-4,7-dione as a simpler chemotype based on the amino-1,4-benzoquinone moiety of streptonigrin, a previously reported inhibitor. Through structure-activity relationship (SAR) studies, compound 8j (MD102) was discovered as a potent TG2 inhibitor with an IC50 value of 0.35 µM, p53 stabilization effect and anticancer effects in the ACHN and Caki-1 RCC cell lines with sulforhodamine B (SRB) GI50 values of 2.15 µM and 1.98 µM, respectively. The binding property of compound 8j (MD102) with TG2 was confirmed to be reversible in a competitive enzyme assay, and the binding interaction was expected to be formed at the β-sandwich domain, a p53 binding site, in the SPR binding assay with mutant proteins. The mode of binding of compound 8j (MD102) to the β-sandwich domain of TG2 was analyzed by molecular docking using the crystal structure of the active conformation of human TG2. Compound 8j (MD102) induced a decrease in the downstream signaling of p-AKT and p-mTOR through the stabilization of p53 by TG2 inhibition, resulting in tumor cell apoptosis. In a xenograft animal model using ACHN cancer cells, oral administration and intraperitoneal injection of compound 8j (MD102) showed an inhibitory effect on tumor growth, confirming increased levels of p53 and decreased levels of Ki-67 in tumor tissues through immunohistochemical (IHC) tissue staining. These results indicated that the inhibition of TG2 by compound 8j (MD102) could enhance p53 stabilization, thereby ultimately showing anticancer effects in RCC. Compound 8j (MD102), a novel TG2 inhibitor, can be further applied for the development of an anticancer candidate drug targeting RCC.
Collapse
Affiliation(s)
- Ga-Ram Kim
- School of Life Sciences, Gwangju Institute of Science and Technology, 123 Cheomdangwagi-ro, Buk-gu, Gwangju 61005, Republic of Korea
| | - Joon Hee Kang
- Graduate School of Cancer Science and Policy, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang 10408, Republic of Korea
| | - Hyeon Joo Kim
- MDbiopharm Corp., 114 Beobwon-ro, Songpa-gu, Seoul 05854, Republic of Korea
| | - Eunji Im
- MDbiopharm Corp., 114 Beobwon-ro, Songpa-gu, Seoul 05854, Republic of Korea
| | - Jinsu Bae
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, 123 Cheomdangwagi-ro, Buk-gu, Gwangju 61005, Republic of Korea
| | - Woo Sun Kwon
- Songdang Institute for Cancer Research, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Sun Young Rha
- Songdang Institute for Cancer Research, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea; Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea; Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Hyun Cheol Chung
- MDbiopharm Corp., 114 Beobwon-ro, Songpa-gu, Seoul 05854, Republic of Korea; Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Eun Yi Cho
- MDbiopharm Corp., 114 Beobwon-ro, Songpa-gu, Seoul 05854, Republic of Korea.
| | - Soo-Youl Kim
- Cancer Molecular Biology Branch, Division of Cancer Biology, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang 10408, Republic of Korea.
| | - Yong-Chul Kim
- School of Life Sciences, Gwangju Institute of Science and Technology, 123 Cheomdangwagi-ro, Buk-gu, Gwangju 61005, Republic of Korea.
| |
Collapse
|
10
|
Selcuk K, Leitner A, Braun L, Le Blanc F, Pacak P, Pot S, Vogel V. Transglutaminase 2 has higher affinity for relaxed than for stretched fibronectin fibers. Matrix Biol 2024; 125:113-132. [PMID: 38135164 DOI: 10.1016/j.matbio.2023.12.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 11/20/2023] [Accepted: 12/18/2023] [Indexed: 12/24/2023]
Abstract
Transglutaminase 2 (TG2) plays a vital role in stabilizing extracellular matrix (ECM) proteins through enzymatic crosslinking during tissue growth, repair, and inflammation. TG2 also binds non-covalently to fibronectin (FN), an essential component of the ECM, facilitating cell adhesion, migration, proliferation, and survival. However, the interaction between TG2 and fibrillar FN remains poorly understood, as most studies have focused on soluble or surface-adsorbed FN or FN fragments, which differ in their conformations from insoluble FN fibers. Using a well-established in vitro FN fiber stretch assay, we discovered that the binding of a crosslinking enzyme to ECM fibers is mechano-regulated. TG2 binding to FN is tuned by the mechanical tension of FN fibers, whereby TG2 predominantly co-localizes to low-tension FN fibers, while fiber stretching reduces their affinity for TG2. This mechano-regulated binding relies on the proximity between the N-terminal β-sandwich and C-terminal β-barrels of TG2. Crosslinking mass spectrometry (XL-MS) revealed a novel TG2-FN synergy site within TG2's C-terminal β-barrels that interacts with FN regions located outside of the canonical gelatin binding domain, specifically FNI2 and FNIII14-15. Combining XL-MS distance restraints with molecular docking revealed the mechano-regulated binding mechanism between TG2 and modules FNI7-9 by which mechanical forces regulate TG2-FN interactions. This highlights a previously unrecognized role of TG2 as a tension sensor for FN fibers. This novel interaction mechanism has significant implications in physiology and mechanobiology, including how forces regulate cell adhesion, spreading, migration, phenotype modulation, depending on the tensional state of ECM fibers. Data are available via ProteomeXchange with identifier PXD043976.
Collapse
Affiliation(s)
- Kateryna Selcuk
- Department of Health Sciences and Technology, Institute of Translational Medicine, Laboratory of Applied Mechanobiology, ETH Zurich, Gloriastrasse 37-39 GLC G11, CH-8092 Zurich, Switzerland
| | - Alexander Leitner
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Otto-Stern-Weg 3, CH-8093 Zurich, Switzerland
| | - Lukas Braun
- Department of Health Sciences and Technology, Institute of Translational Medicine, Laboratory of Applied Mechanobiology, ETH Zurich, Gloriastrasse 37-39 GLC G11, CH-8092 Zurich, Switzerland
| | - Fanny Le Blanc
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Otto-Stern-Weg 3, CH-8093 Zurich, Switzerland
| | - Paulina Pacak
- Department of Health Sciences and Technology, Institute of Translational Medicine, Laboratory of Applied Mechanobiology, ETH Zurich, Gloriastrasse 37-39 GLC G11, CH-8092 Zurich, Switzerland
| | - Simon Pot
- Department of Health Sciences and Technology, Institute of Translational Medicine, Laboratory of Applied Mechanobiology, ETH Zurich, Gloriastrasse 37-39 GLC G11, CH-8092 Zurich, Switzerland
| | - Viola Vogel
- Department of Health Sciences and Technology, Institute of Translational Medicine, Laboratory of Applied Mechanobiology, ETH Zurich, Gloriastrasse 37-39 GLC G11, CH-8092 Zurich, Switzerland.
| |
Collapse
|
11
|
Ganesh RA, Venkataraman K, Sirdeshmukh R. GPR56 signaling pathway network and its dynamics in the mesenchymal transition of glioblastoma. J Cell Commun Signal 2023:10.1007/s12079-023-00792-5. [PMID: 37980704 DOI: 10.1007/s12079-023-00792-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 11/02/2023] [Indexed: 11/21/2023] Open
Abstract
G protein-coupled receptor 56 (GPR56/ADGRG1) is a multifunctional adhesion GPCR involved in diverse biological processes ranging from development to cancer. In our earlier study, we reported that GPR56 is expressed heterogeneously in glioblastoma (GBM) and is involved in the mesenchymal transition, making it a promising therapeutic target (Ganesh et al., 2022). Despite its important role in cancer, its mechanism of action or signaling is not completely understood. Thus, based on transcriptomic, proteomic, and phosphoproteomic differential expression data of GPR56 knockdown U373-GBM cells included in our above study along with detailed literature mining of the molecular events plausibly associated with GPR56 activity, we have constructed a signaling pathway map of GPR56 as may be applicable in mesenchymal transition in GBM. The map incorporates more than 100 molecular entities including kinases, receptors, ion channels, and others associated with Wnt, integrin, calcium signaling, growth factors, and inflammation signaling pathways. We also considered intracellular and extracellular factors that may influence the activity of the pathway entities. Here we present a curated signaling map of GPR56 in the context of GBM and discuss the relevance and plausible cross-connectivity across different axes attributable to GPR56 function. GPR56 signaling and mesenchymal transition.
Collapse
Affiliation(s)
- Raksha A Ganesh
- Mazumdar Shaw Center for Translational Research, Narayana Health, Mazumdar Shaw Medical Foundation, Bangalore, 560099, India
- Center for Bio-Separation Technology, Vellore Institute of Technology, Vellore, 632104, India
| | - Krishnan Venkataraman
- Center for Bio-Separation Technology, Vellore Institute of Technology, Vellore, 632104, India
| | - Ravi Sirdeshmukh
- Mazumdar Shaw Center for Translational Research, Narayana Health, Mazumdar Shaw Medical Foundation, Bangalore, 560099, India.
- Institute of Bioinformatics, International Tech Park, Bangalore, 560066, India.
- Manipal Academy of Higher Education, Manipal, 576104, India.
| |
Collapse
|
12
|
Gates EWJ, Calvert ND, Cundy NJ, Brugnoli F, Navals P, Kirby A, Bianchi N, Adhikary G, Shuhendler AJ, Eckert RL, Keillor JW. Cell-Impermeable Inhibitors Confirm That Intracellular Human Transglutaminase 2 Is Responsible for the Transglutaminase-Associated Cancer Phenotype. Int J Mol Sci 2023; 24:12546. [PMID: 37628729 PMCID: PMC10454375 DOI: 10.3390/ijms241612546] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/24/2023] [Accepted: 07/31/2023] [Indexed: 08/27/2023] Open
Abstract
Transglutaminase 2 (TG2) is a multifunctional enzyme primarily responsible for crosslinking proteins. Ubiquitously expressed in humans, TG2 can act either as a transamidase by crosslinking two substrates through formation of an Nε(ɣ-glutaminyl)lysine bond or as an intracellular G-protein. These discrete roles are tightly regulated by both allosteric and environmental stimuli and are associated with dramatic changes in the conformation of the enzyme. The pleiotropic nature of TG2 and multi-faceted activities have resulted in TG2 being implicated in numerous disease pathologies including celiac disease, fibrosis, and cancer. Targeted TG2 therapies have not been selective for subcellular localization, such that currently no tools exist to selectively target extracellular over intracellular TG2. Herein, we have designed novel TG2-selective inhibitors that are not only highly potent and irreversible, but also cell impermeable, targeting only extracellular TG2. We have also further derivatized the scaffold to develop probes that are intrinsically fluorescent or bear an alkyne handle, which target both intra- and extracellular TG2, in order to facilitate cellular labelling and pull-down assays. The fluorescent probes were internalized and imaged in cellulo, and provide the first implicit experimental evidence that by comparison with their cell-impermeable analogues, it is specifically intracellular TG2, and presumably its G-protein activity, that contributes to transglutaminase-associated cancer progression.
Collapse
Affiliation(s)
- Eric W. J. Gates
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada; (E.W.J.G.); (N.D.C.); (N.J.C.); (P.N.); (A.K.); (A.J.S.)
| | - Nicholas D. Calvert
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada; (E.W.J.G.); (N.D.C.); (N.J.C.); (P.N.); (A.K.); (A.J.S.)
| | - Nicholas J. Cundy
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada; (E.W.J.G.); (N.D.C.); (N.J.C.); (P.N.); (A.K.); (A.J.S.)
| | - Federica Brugnoli
- Department of Translational Medicine, University of Ferrara, 44021 Ferrara, Italy; (F.B.); (N.B.)
| | - Pauline Navals
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada; (E.W.J.G.); (N.D.C.); (N.J.C.); (P.N.); (A.K.); (A.J.S.)
| | - Alexia Kirby
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada; (E.W.J.G.); (N.D.C.); (N.J.C.); (P.N.); (A.K.); (A.J.S.)
| | - Nicoletta Bianchi
- Department of Translational Medicine, University of Ferrara, 44021 Ferrara, Italy; (F.B.); (N.B.)
| | - Gautam Adhikary
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (G.A.); (R.L.E.)
| | - Adam J. Shuhendler
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada; (E.W.J.G.); (N.D.C.); (N.J.C.); (P.N.); (A.K.); (A.J.S.)
| | - Richard L. Eckert
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (G.A.); (R.L.E.)
| | - Jeffrey W. Keillor
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada; (E.W.J.G.); (N.D.C.); (N.J.C.); (P.N.); (A.K.); (A.J.S.)
| |
Collapse
|
13
|
Motta C, Pellegrini A, Camaione S, Geoghegan J, Speziale P, Barbieri G, Pietrocola G. von Willebrand factor-binding protein (vWbp)-activated factor XIII and transglutaminase 2 (TG2) promote cross-linking between FnBPA from Staphylococcus aureus and fibrinogen. Sci Rep 2023; 13:11683. [PMID: 37468579 DOI: 10.1038/s41598-023-38972-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 07/18/2023] [Indexed: 07/21/2023] Open
Abstract
The secreted von Willebrand factor-binding protein (vWbp) from Staphylococcus aureus interacts with the coagulation factors prothrombin and fibrinogen (Fbg), leading to the non-proteolytic transglutaminase activation of Factor XIII (FXIII). In this study we found that vWbp-activated FXIII catalyses the incorporation of amino-donor dansylcadaverine into region A of fibronectin-binding protein A (FnBPA). Incubation of Fbg with recombinant region A of S. aureus Fbg-binding proteins FnBPA, FnBPB, ClfA or ClfB in presence of vWbp-activated FXIII resulted in the formation of high molecular heteropolymers with FnBPA only, suggesting a specificity of the cross-linking reaction between fibrin(ogen) and the staphylococcal surface. As previously observed, cross-linking sites were mapped to the α-chain and the N1 subdomain of fibrin(ogen) and region A of FnBPA, respectively. Comparable results were obtained when tissue tranglutaminase-2 (TG2) was tested for cross-linking of FnBPA and Fbg. Of note, FnBPA-mediated covalent cross-linking promoted by vWbp-activated FXIII was also observed when bacteria were allowed to attach to fibrin(ogen). Together these findings suggest a novel pathogenetic mechanism by which the transglutaminase action of FXIII and/or TG2 contributes to entrapment and persistence of S. aureus in blood and host tissues.
Collapse
Affiliation(s)
- Chiara Motta
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | | | - Stefano Camaione
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Joan Geoghegan
- Institute of Microbiology and Infection, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Pietro Speziale
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Giulia Barbieri
- Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia, Italy
| | | |
Collapse
|