1
|
Zhou K, Zhang S, Shang J, Lan X. Exploring immune gene expression and potential regulatory mechanisms in anaplastic thyroid carcinoma using a combination of single-cell and bulk RNA sequencing data. Comput Biol Chem 2025; 115:108311. [PMID: 39674047 DOI: 10.1016/j.compbiolchem.2024.108311] [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: 04/22/2024] [Revised: 11/27/2024] [Accepted: 12/04/2024] [Indexed: 12/16/2024]
Abstract
Thyroid cancer includes papillary thyroid carcinoma (PTC) and anaplastic thyroid carcinoma (ATC). While PTC has an excellent prognosis, ATC has a dismal prognosis, necessitating the identification of novel targets in ATC to aid in ATC diagnosis and treatment. Therefore, we analyzed ATC single-cell RNA sequencing (scRNA-seq) and bulk RNA sequencing (bulk RNA-seq) data from the Gene Expression Omnibus (GEO), retrieved immune-related genes from the ImmPort database, and identified differentially expressed immune genes within single-cell subgroups. The AUCell package in R was used to calculate activity scores for single-cell subgroups and identify active cell populations. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis were performed on differentially expressed genes (DEGs) in active cell populations. Then, we integrated thyroid-cancer scRNA-seq and bulk RNA-seq data to identify overlapping DEGs. Relevant transcription factors (TFs) were retrieved from the TRRUST database. A protein-protein interaction (PPI) network for key TFs was created using the STRING database. Simultaneously, drugs associated with key TFs were obtained from DGIdb. ScRNA-seq cluster analysis showed that T/natural killer (NK) cells were more distributed in ATC and that thyrocytes cells were more distributed in PTC. We obtained 264 differential immune genes (DIGs) from the IMMPORT database and integrated scRNA-seq cluster analysis to identify the active cell T/NK cells and myeloid cells. Integrated bulk RNA-seq analysis obtained common immune genes (CIGs) such as TMSB4X, NFKB1, TNFRSF1B, and B2M. The nine CIG-related TFs (CEBPB, SPI1, NFKB1, RUNX1, NFE2L2, REL, CIITA, KLF6, and CEBPD) in myeloid cells and three TFs (NFKB1, FOXO1, and NR3C1) in T/NK cells were obtained from the TRRUST database. The key genes we identified represent potential targets for treating ATC.
Collapse
Affiliation(s)
- Kehui Zhou
- Postgraduate training base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang 310022, China; Department of Thyroid Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Shijia Zhang
- Postgraduate training base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang 310022, China; Department of Thyroid Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Jinbiao Shang
- Department of Thyroid Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China; Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou, Zhejiang 310022, China
| | - Xiabin Lan
- Postgraduate training base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang 310022, China; Department of Thyroid Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China; Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou, Zhejiang 310022, China.
| |
Collapse
|
2
|
Aftabi S, Barzegar Behrooz A, Cordani M, Rahiman N, Sadeghdoust M, Aligolighasemabadi F, Pistorius S, Alavizadeh SH, Taefehshokr N, Ghavami S. Therapeutic targeting of TGF-β in lung cancer. FEBS J 2025; 292:1520-1557. [PMID: 39083441 PMCID: PMC11970718 DOI: 10.1111/febs.17234] [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: 11/05/2023] [Revised: 05/22/2024] [Accepted: 07/19/2024] [Indexed: 08/02/2024]
Abstract
Transforming growth factor-β (TGF-β) plays a complex role in lung cancer pathophysiology, initially acting as a tumor suppressor by inhibiting early-stage tumor growth. However, its role evolves in the advanced stages of the disease, where it contributes to tumor progression not by directly promoting cell proliferation but by enhancing epithelial-mesenchymal transition (EMT) and creating a conducive tumor microenvironment. While EMT is typically associated with enhanced migratory and invasive capabilities rather than proliferation per se, TGF-β's influence on this process facilitates the complex dynamics of tumor metastasis. Additionally, TGF-β impacts the tumor microenvironment by interacting with immune cells, a process influenced by genetic and epigenetic changes within tumor cells. This interaction highlights its role in immune evasion and chemoresistance, further complicating lung cancer therapy. This review provides a critical overview of recent findings on TGF-β's involvement in lung cancer, its contribution to chemoresistance, and its modulation of the immune response. Despite the considerable challenges encountered in clinical trials and the development of new treatments targeting the TGF-β pathway, this review highlights the necessity for continued, in-depth investigation into the roles of TGF-β. A deeper comprehension of these roles may lead to novel, targeted therapies for lung cancer. Despite the intricate behavior of TGF-β signaling in tumors and previous challenges, further research could yield innovative treatment strategies.
Collapse
Affiliation(s)
- Sajjad Aftabi
- Department of Human Anatomy and Cell ScienceUniversity of Manitoba College of MedicineWinnipegCanada
- Paul Albrechtsen Research Institute, CancerCare ManitobaUniversity of ManitobaWinnipegCanada
- Department of Physics and AstronomyUniversity of ManitobaWinnipegCanada
| | - Amir Barzegar Behrooz
- Department of Human Anatomy and Cell ScienceUniversity of Manitoba College of MedicineWinnipegCanada
- Electrophysiology Research Center, Neuroscience InstituteTehran University of Medical SciencesIran
| | - Marco Cordani
- Department of Biochemistry and Molecular Biology, Faculty of BiologyComplutense UniversityMadridSpain
- Instituto de Investigaciones Sanitarias San Carlos (IdISSC)MadridSpain
| | - Niloufar Rahiman
- Nanotechnology Research Center, Pharmaceutical Technology InstituteMashhad University of Medical SciencesIran
- Department of Pharmaceutical Nanotechnology, School of PharmacyMashhad University of Medical SciencesIran
| | - Mohammadamin Sadeghdoust
- Division of BioMedical Sciences, Faculty of MedicineMemorial University of NewfoundlandSt. John'sCanada
| | - Farnaz Aligolighasemabadi
- Department of Human Anatomy and Cell ScienceUniversity of Manitoba College of MedicineWinnipegCanada
| | - Stephen Pistorius
- Department of Human Anatomy and Cell ScienceUniversity of Manitoba College of MedicineWinnipegCanada
- Paul Albrechtsen Research Institute, CancerCare ManitobaUniversity of ManitobaWinnipegCanada
- Department of Physics and AstronomyUniversity of ManitobaWinnipegCanada
| | - Seyedeh Hoda Alavizadeh
- Nanotechnology Research Center, Pharmaceutical Technology InstituteMashhad University of Medical SciencesIran
- Department of Pharmaceutical Nanotechnology, School of PharmacyMashhad University of Medical SciencesIran
| | - Nima Taefehshokr
- Apoptosis Research CentreChildren's Hospital of Eastern Ontario Research InstituteOttawaCanada
| | - Saeid Ghavami
- Department of Human Anatomy and Cell ScienceUniversity of Manitoba College of MedicineWinnipegCanada
- Paul Albrechtsen Research Institute, CancerCare ManitobaUniversity of ManitobaWinnipegCanada
- Faculty Academy of Silesia, Faculty of MedicineKatowicePoland
- Children Hospital Research Institute of ManitobaUniversity of ManitobaWinnipegCanada
| |
Collapse
|
3
|
Ungefroren H, von der Ohe J, Braun R, Gätje Y, Lapshyna O, Schrader J, Lehnert H, Marquardt JU, Konukiewitz B, Hass R. Characterization of Epithelial-Mesenchymal and Neuroendocrine Differentiation States in Pancreatic and Small Cell Ovarian Tumor Cells and Their Modulation by TGF-β1 and BMP-7. Cells 2024; 13:2010. [PMID: 39682758 PMCID: PMC11640004 DOI: 10.3390/cells13232010] [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: 07/14/2024] [Revised: 11/20/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has an extremely poor prognosis, due in part to early invasion and metastasis, which in turn involves epithelial-mesenchymal transition (EMT) of the cancer cells. Prompted by the discovery that two PDAC cell lines of the quasi-mesenchymal subtype (PANC-1, MIA PaCa-2) exhibit neuroendocrine differentiation (NED), we asked whether NED is associated with EMT. Using real-time PCR and immunoblotting, we initially verified endogenous expressions of various NED markers, i.e., chromogranin A (CHGA), synaptophysin (SYP), somatostatin receptor 2 (SSTR2), and SSTR5 in PANC-1 and MIA PaCa-2 cells. By means of immunohistochemistry, the expressions of CHGA, SYP, SSTR2, and the EMT markers cytokeratin 7 (CK7) and vimentin could be allocated to the neoplastic ductal epithelial cells of pancreatic ducts in surgically resected tissues from patients with PDAC. In HPDE6c7 normal pancreatic duct epithelial cells and in epithelial subtype BxPC-3 PDAC cells, the expression of CHGA, SYP, and neuron-specific enolase 2 (NSE) was either undetectable or much lower than in PANC-1 and MIA PaCa-2 cells. Parental cultures of PANC-1 cells exhibit EM plasticity (EMP) and harbor clonal subpopulations with both M- and E-phenotypes. Of note, M-type clones were found to display more pronounced NED than E-type clones. Inducing EMT in parental cultures of PANC-1 cells by treatment with transforming growth factor-β1 (TGF-β1) repressed epithelial genes and co-induced mesenchymal and NED genes, except for SSTR5. Surprisingly, treatment with bone morphogenetic protein (BMP)-7 differentially affected gene expressions in PANC-1, MIA PaCa-2, BxPC-3, and HPDE cells. It synergized with TGF-β1 in the induction of vimentin, SNAIL, SSTR2, and NSE but antagonized it in the regulation of CHGA and SSTR5. Phospho-immunoblotting in M- and E-type PANC-1 clones revealed that both TGF-β1 and, surprisingly, also BMP-7 activated SMAD2 and SMAD3 and that in M- but not E-type clones BMP-7 was able to dramatically enhance the activation of SMAD3. From these data, we conclude that in EMT of PDAC cells mesenchymal and NED markers are co-regulated, and that mesenchymal-epithelial transition (MET) is associated with a loss of both the mesenchymal and NED phenotypes. Analyzing NED in another tumor type, small cell carcinoma of the ovary hypercalcemic type (SCCOHT), revealed that two model cell lines of this disease (SCCOHT-1, BIN-67) do express CDH1, SNAI1, VIM, CHGA, SYP, ENO2, and SSTR2, but that in contrast to BMP-7, none of these genes was transcriptionally regulated by TGF-β1. Likewise, in BIN-67 cells, BMP-7 was able to reduce proliferation, while in SCCOHT-1 cells this occurred only upon combined treatment with TGF-β and BMP-7. We conclude that in PDAC-derived tumor cells, NED is closely linked to EMT and TGF-β signaling, which may have implications for the therapeutic use of TGF-β inhibitors in PDAC management.
Collapse
Affiliation(s)
- Hendrik Ungefroren
- First Department of Medicine, University Hospital Schleswig-Holstein, Campus Lübeck, 23538 Lübeck, Germany
- Institute of Pathology, University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Juliane von der Ohe
- Biochemistry and Tumor Biology Lab, Department of Obstetrics and Gynecology, Hannover Medical School, 30625 Hannover, Germany
| | - Rüdiger Braun
- Department of Surgery, University Hospital Schleswig-Holstein, Campus Lübeck, 23538 Lübeck, Germany
| | - Yola Gätje
- First Department of Medicine, University Hospital Schleswig-Holstein, Campus Lübeck, 23538 Lübeck, Germany
| | - Olha Lapshyna
- Department of Surgery, University Hospital Schleswig-Holstein, Campus Lübeck, 23538 Lübeck, Germany
| | - Jörg Schrader
- First Department of Medicine, University Hospital Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Hendrik Lehnert
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire (UHCW), Coventry CV2 2DX, UK
| | - Jens-Uwe Marquardt
- First Department of Medicine, University Hospital Schleswig-Holstein, Campus Lübeck, 23538 Lübeck, Germany
| | - Björn Konukiewitz
- Institute of Pathology, University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Ralf Hass
- Biochemistry and Tumor Biology Lab, Department of Obstetrics and Gynecology, Hannover Medical School, 30625 Hannover, Germany
| |
Collapse
|
4
|
Zhou L, Wang N, Feng W, Liu X, Wu Q, Chen J, Jiao X, Ning X, Qi Z, Xu Z, Jiang X, Zhao Q. Soluble TGF-β decoy receptor TGFBR3 exacerbates Alzheimer's disease pathology by modifying microglial function. Glia 2024; 72:2201-2216. [PMID: 39137117 DOI: 10.1002/glia.24606] [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: 01/09/2024] [Revised: 07/25/2024] [Accepted: 08/01/2024] [Indexed: 08/15/2024]
Abstract
Alzheimer's disease (AD) is a major cause of progressive dementia characterized by memory loss and progressive neurocognitive dysfunction. However, the molecular mechanisms are not fully understood. To elucidate the molecular mechanism contributing to AD, an integrated analytical workflow was deployed to identify pivotal regulatory target within the RNA-sequencing (RNA-seq) data of the temporal cortex from AD patients. Soluble transforming growth factor beta receptor 3 (sTGFBR3) was identified as a critical target in AD, which was abnormally elevated in AD patients and AD mouse models. We then demonstrated that sTGFBR3 deficiency restored spatial learning and memory deficits in amyloid precursor protein (APP)/PS1 and streptozotocin (STZ)-induced neuronal impairment mice after its expression was disrupted by a lentiviral (LV) vector expressing shRNA. Mechanistically, sTGFBR3 deficiency augments TGF-β signaling and suppressing the NF-κB pathway, thereby reduced the number of disease-associated microglia (DAMs), inhibited proinflammatory activity and increased the phagocytic activity of DAMs. Moreover, sTGFBR3 deficiency significantly mitigated acute neuroinflammation provoked by lipopolysaccharide (LPS) and alleviated neuronal dysfunction induced by STZ. Collectively, these results position sTGFBR3 as a promising candidate for therapeutic intervention in AD.
Collapse
Affiliation(s)
- Lijun Zhou
- Department of Clinical Pharmacy, Shenyang Pharmaceutical University, Shenyang, People's Republic of China
| | - Nan Wang
- Department of Clinical Pharmacy, Shenyang Pharmaceutical University, Shenyang, People's Republic of China
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, People's Republic of China
| | - Wenzheng Feng
- Department of Clinical Pharmacy, Shenyang Pharmaceutical University, Shenyang, People's Republic of China
| | - Xin Liu
- Department of Clinical Pharmacy, Shenyang Pharmaceutical University, Shenyang, People's Republic of China
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, People's Republic of China
| | - Qiong Wu
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, People's Republic of China
| | - Jiangxia Chen
- Department of Clinical Pharmacy, Shenyang Pharmaceutical University, Shenyang, People's Republic of China
| | - Xinming Jiao
- Department of Clinical Pharmacy, Shenyang Pharmaceutical University, Shenyang, People's Republic of China
| | - Xinyue Ning
- Department of Clinical Pharmacy, Shenyang Pharmaceutical University, Shenyang, People's Republic of China
| | - Zhentong Qi
- Department of Clinical Pharmacy, Shenyang Pharmaceutical University, Shenyang, People's Republic of China
| | - Zihua Xu
- Department of Clinical Pharmacy, Shenyang Pharmaceutical University, Shenyang, People's Republic of China
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, People's Republic of China
| | - Xiaowen Jiang
- College of Traditional Chinese Pharmacy, Shenyang Pharmaceutical University, Shenyang, People's Republic of China
| | - Qingchun Zhao
- Department of Clinical Pharmacy, Shenyang Pharmaceutical University, Shenyang, People's Republic of China
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, People's Republic of China
| |
Collapse
|
5
|
Santos da Silva T, da Silva-Júnior LN, Horvath-Pereira BDO, Valbão MCM, Garcia MHH, Lopes JB, Reis CHB, Barreto RDSN, Buchaim DV, Buchaim RL, Miglino MA. The Role of the Pancreatic Extracellular Matrix as a Tissue Engineering Support for the Bioartificial Pancreas. Biomimetics (Basel) 2024; 9:598. [PMID: 39451804 PMCID: PMC11505355 DOI: 10.3390/biomimetics9100598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/22/2024] [Accepted: 09/30/2024] [Indexed: 10/26/2024] Open
Abstract
Type 1 diabetes mellitus (T1DM) is a chronic condition primarily managed with insulin replacement, leading to significant treatment costs. Complications include vasculopathy, cardiovascular diseases, nephropathy, neuropathy, and reticulopathy. Pancreatic islet transplantation is an option but its success does not depend solely on adequate vascularization. The main limitations to clinical islet transplantation are the scarcity of human pancreas, the need for immunosuppression, and the inadequacy of the islet isolation process. Despite extensive research, T1DM remains a major global health issue. In 2015, diabetes affected approximately 415 million people, with projected expenditures of USD 1.7 trillion by 2030. Pancreas transplantation faces challenges due to limited organ availability and complex vascularization. T1DM is caused by the autoimmune destruction of insulin-producing pancreatic cells. Advances in biomaterials, particularly the extracellular matrix (ECM), show promise in tissue reconstruction and transplantation, offering structural and regulatory functions critical for cell migration, differentiation, and adhesion. Tissue engineering aims to create bioartificial pancreases integrating insulin-producing cells and suitable frameworks. This involves decellularization and recellularization techniques to develop biological scaffolds. The challenges include replicating the pancreas's intricate architecture and maintaining cell viability and functionality. Emerging technologies, such as 3D printing and advanced biomaterials, have shown potential in constructing bioartificial organs. ECM components, including collagens and glycoproteins, play essential roles in cell adhesion, migration, and differentiation. Clinical applications focus on developing functional scaffolds for transplantation, with ongoing research addressing immunological responses and long-term efficacy. Pancreatic bioengineering represents a promising avenue for T1DM treatment, requiring further research to ensure successful implementation.
Collapse
Affiliation(s)
- Thamires Santos da Silva
- Graduate Program in Anatomy of Domestic and Wild Animals, Faculty of Veterinary Medicine and Animal Science, University of São Paulo (FMVZ/USP), São Paulo 05508-270, Brazil; (T.S.d.S.); (L.N.d.S.-J.); (B.d.O.H.-P.); (R.d.S.N.B.); (D.V.B.); (R.L.B.)
| | - Leandro Norberto da Silva-Júnior
- Graduate Program in Anatomy of Domestic and Wild Animals, Faculty of Veterinary Medicine and Animal Science, University of São Paulo (FMVZ/USP), São Paulo 05508-270, Brazil; (T.S.d.S.); (L.N.d.S.-J.); (B.d.O.H.-P.); (R.d.S.N.B.); (D.V.B.); (R.L.B.)
- Postgraduate Department, University of Marília (UNIMAR), Marília 17525-902, Brazil; (M.C.M.V.); (M.H.H.G.); (J.B.L.)
| | - Bianca de Oliveira Horvath-Pereira
- Graduate Program in Anatomy of Domestic and Wild Animals, Faculty of Veterinary Medicine and Animal Science, University of São Paulo (FMVZ/USP), São Paulo 05508-270, Brazil; (T.S.d.S.); (L.N.d.S.-J.); (B.d.O.H.-P.); (R.d.S.N.B.); (D.V.B.); (R.L.B.)
| | - Maria Carolina Miglino Valbão
- Postgraduate Department, University of Marília (UNIMAR), Marília 17525-902, Brazil; (M.C.M.V.); (M.H.H.G.); (J.B.L.)
| | | | - Juliana Barbosa Lopes
- Postgraduate Department, University of Marília (UNIMAR), Marília 17525-902, Brazil; (M.C.M.V.); (M.H.H.G.); (J.B.L.)
| | - Carlos Henrique Bertoni Reis
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marilia (UNIMAR), Marília 17525-902, Brazil;
- UNIMAR Beneficent Hospital (HBU), Medical School, University of Marilia (UNIMAR), Marilia 17525-160, Brazil
| | - Rodrigo da Silva Nunes Barreto
- Graduate Program in Anatomy of Domestic and Wild Animals, Faculty of Veterinary Medicine and Animal Science, University of São Paulo (FMVZ/USP), São Paulo 05508-270, Brazil; (T.S.d.S.); (L.N.d.S.-J.); (B.d.O.H.-P.); (R.d.S.N.B.); (D.V.B.); (R.L.B.)
- Department of Animal Morphology and Physiology, Faculty of Agricultural and Veterinary Sciences, São Paulo State University, Jaboticabal 14884-900, Brazil
| | - Daniela Vieira Buchaim
- Graduate Program in Anatomy of Domestic and Wild Animals, Faculty of Veterinary Medicine and Animal Science, University of São Paulo (FMVZ/USP), São Paulo 05508-270, Brazil; (T.S.d.S.); (L.N.d.S.-J.); (B.d.O.H.-P.); (R.d.S.N.B.); (D.V.B.); (R.L.B.)
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marilia (UNIMAR), Marília 17525-902, Brazil;
- Medical School, University Center of Adamantina (UNIFAI), Adamantina 17800-000, Brazil
| | - Rogerio Leone Buchaim
- Graduate Program in Anatomy of Domestic and Wild Animals, Faculty of Veterinary Medicine and Animal Science, University of São Paulo (FMVZ/USP), São Paulo 05508-270, Brazil; (T.S.d.S.); (L.N.d.S.-J.); (B.d.O.H.-P.); (R.d.S.N.B.); (D.V.B.); (R.L.B.)
- Department of Biological Sciences, Bauru School of Dentistry (FOB/USP), University of Sao Paulo, Bauru 17012-901, Brazil
| | - Maria Angelica Miglino
- Postgraduate Department, University of Marília (UNIMAR), Marília 17525-902, Brazil; (M.C.M.V.); (M.H.H.G.); (J.B.L.)
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marilia (UNIMAR), Marília 17525-902, Brazil;
- Postgraduate Program in Animal Health, Production and Environment, University of Marilia (UNIMAR), Marilia 17525-902, Brazil
| |
Collapse
|
6
|
Zhang F, Duan Z, Chen Q, Wang X, Li H, Tao Z, Chen Z, Yu G, Yu H. Molecular characterization, expression and immune functional analysis of cystatin 10 in turbot. Mol Biol Rep 2024; 51:709. [PMID: 38824265 DOI: 10.1007/s11033-024-09634-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 05/10/2024] [Indexed: 06/03/2024]
Abstract
BACKGROUND Cystatin is a protease inhibitor that also regulates genes expression linked to inflammation and plays a role in defense and regulation. METHODS AND RESULTS Cystatin 10 (Smcys10) was cloned from Scophthalmus maximus and encodes a 145 amino acid polypeptide. The results of qRT-PCR showed that Smcys10 exhibited tissue-specific expression patterns, and its expression was significantly higher in the skin than in other tissues. The expression level of Smcys10 was significantly different in the skin, gill, head kidney, spleen and macrophages after Vibrio anguillarum infection, indicating that Smcys10 may play an important role in resistance to V. anguillarum infection. The recombinant Smcys10 protein showed binding and agglutinating activity in a Ca2+-dependent manner against bacteria. rSmcys10 treatment upregulated the expression of IL-10, TNF-α and TGF-β in macrophages of turbot and hindered the release of lactate dehydrogenase (LDH) from macrophages after V. anguillarum infection, which confirmed that rSmcys10 reduced the damage to macrophages by V. anguillarum. The NF-κB pathway was suppressed by Smcys10, as demonstrated by dual-luciferase analysis. CONCLUSIONS These results indicated that Smcys10 is involved in the host antibacterial immune response.
Collapse
Affiliation(s)
- Fan Zhang
- Key Laboratory of Marine Genetics and Breeding, Ministry of Education, Ocean University of China, Qingdao, 266003, Shandong, China
| | - Zhixiang Duan
- Key Laboratory of Marine Genetics and Breeding, Ministry of Education, Ocean University of China, Qingdao, 266003, Shandong, China
| | - Qiannan Chen
- Key Laboratory of Marine Genetics and Breeding, Ministry of Education, Ocean University of China, Qingdao, 266003, Shandong, China
| | - Xuangang Wang
- Key Laboratory of Marine Genetics and Breeding, Ministry of Education, Ocean University of China, Qingdao, 266003, Shandong, China
| | - Hengshun Li
- Key Laboratory of Marine Genetics and Breeding, Ministry of Education, Ocean University of China, Qingdao, 266003, Shandong, China
| | - Ze Tao
- Key Laboratory of Marine Genetics and Breeding, Ministry of Education, Ocean University of China, Qingdao, 266003, Shandong, China
| | - Zhentao Chen
- Key Laboratory of Marine Genetics and Breeding, Ministry of Education, Ocean University of China, Qingdao, 266003, Shandong, China
| | - Gan Yu
- Key Laboratory of Marine Genetics and Breeding, Ministry of Education, Ocean University of China, Qingdao, 266003, Shandong, China
| | - Haiyang Yu
- Key Laboratory of Marine Genetics and Breeding, Ministry of Education, Ocean University of China, Qingdao, 266003, Shandong, China.
| |
Collapse
|
7
|
Tolue Ghasaban F, Ghanei M, Mahmoudian RA, Taghehchian N, Abbaszadegan MR, Moghbeli M. MicroRNAs as the critical regulators of epithelial mesenchymal transition in pancreatic tumor cells. Heliyon 2024; 10:e30599. [PMID: 38726188 PMCID: PMC11079401 DOI: 10.1016/j.heliyon.2024.e30599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 04/26/2024] [Accepted: 04/30/2024] [Indexed: 05/12/2024] Open
Abstract
Pancreatic cancer (PC), as one of the main endocrine and digestive systems malignancies has the highest cancer related mortality in the world. Lack of the evident clinical symptoms and appropriate diagnostic markers in the early stages of tumor progression are the main reasons of the high mortality rate among PC patients. Therefore, it is necessary to investigate the molecular pathways involved in the PC progression, in order to introduce novel early diagnostic methods. Epithelial mesenchymal transition (EMT) is a critical cellular process associated with pancreatic tumor cells invasion and distant metastasis. MicroRNAs (miRNAs) are also important regulators of EMT process. In the present review, we discussed the role of miRNAs in regulation of EMT process during PC progression. It has been reported that the miRNAs mainly regulate the EMT process in pancreatic tumor cells through the regulation of EMT-specific transcription factors and several signaling pathways such as WNT, NOTCH, TGF-β, JAK/STAT, and PI3K/AKT. Considering the high stability of miRNAs in body fluids and their role in regulation of EMT process, they can be introduced as the non-invasive diagnostic markers in the early stages of malignant pancreatic tumors. This review paves the way to introduce a non-invasive EMT based panel marker for the early tumor detection among PC patients.
Collapse
Affiliation(s)
- Faezeh Tolue Ghasaban
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Ghanei
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reihaneh Alsadat Mahmoudian
- Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Cancer Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Negin Taghehchian
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Meysam Moghbeli
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
8
|
Moshe DL, Baghaie L, Leroy F, Skapinker E, Szewczuk MR. Metamorphic Effect of Angiogenic Switch in Tumor Development: Conundrum of Tumor Angiogenesis Toward Progression and Metastatic Potential. Biomedicines 2023; 11:2142. [PMID: 37626639 PMCID: PMC10452636 DOI: 10.3390/biomedicines11082142] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/27/2023] [Accepted: 07/28/2023] [Indexed: 08/27/2023] Open
Abstract
Our understanding of angiogenesis has significantly expanded over the past five decades. More recently, research has focused on this process at a more molecular level, looking at it through the signaling pathways that activate it and its non-direct downstream effects. This review discusses current findings in molecular angiogenesis, focusing on its impact on the immune system. Moreover, the impairment of this process in cancer progression and metastasis is highlighted, and current anti-angiogenic treatments and their effects on tumor growth are discussed.
Collapse
Affiliation(s)
- Daniel Leon Moshe
- Faculty of Health Sciences, Queen’s University, Kingston, ON K7L 3N9, Canada;
| | - Leili Baghaie
- Department of Biomedical & Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada;
| | - Fleur Leroy
- Faculté de médecine, Maïeutique et Sciences de la Santé, Université de Strasbourg, F-67000 Strasbourg, France;
| | - Elizabeth Skapinker
- Faculty of Arts and Science, Queen’s University, Kingston, ON K7L 3N9, Canada;
| | - Myron R. Szewczuk
- Faculty of Health Sciences, Queen’s University, Kingston, ON K7L 3N9, Canada;
| |
Collapse
|
9
|
Geerinckx B, Teuwen LA, Foo T, Vandamme T, Smith A, Peeters M, Price T. Novel therapeutic strategies in pancreatic cancer: moving beyond cytotoxic chemotherapy. Expert Rev Anticancer Ther 2023; 23:1237-1249. [PMID: 37842857 DOI: 10.1080/14737140.2023.2270161] [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: 04/20/2023] [Accepted: 10/09/2023] [Indexed: 10/17/2023]
Abstract
INTRODUCTION Prognosis of patients with metastatic pancreatic ductal adenocarcinoma (mPDAC) remains disappointing with a 5-year overall survival of only 3-5%. Compared to other cancers, the evolution in standard therapeutic options has been stagnant and polychemotherapy regimens (with well-known toxicity profile and resistance pattern) remain standard of care. Only for patients (5%-7%) with a breast cancer gene (BRCA) pathogenic germline variant, prognosis has improved by the use of olaparib (poly-ADP ribose polymerase (PARP) inhibitor). AREAS COVERED This review covers emerging treatment strategies in the management of mPDAC. One of the main topics is the rigid and immunological cold tumor microenvironment (TME) of PDAC and the search for agents that impact this TME and/or engage the immune system. In addition, the use of next-generation sequencing (NGS) has elicited for some patients new targeted therapies directed at alterations in the RTK/RAS/MAPK pathway and the deoxyribonucleic acid (DNA) damage repair pathway. Other evolving treatment strategies are also discussed. EXPERT OPINION The search for new, often combination, treatment strategies for mPDAC should be encouraged and implemented in early treatment lines given the significant decline of performance status of patients in later lines. NGS analysis should be used where available, although cost-effectiveness could be debatable.
Collapse
Affiliation(s)
- Barbara Geerinckx
- Department of Medical Oncology, The Queen Elizabeth Hospital, Woodville, Australia
- Department of Oncology and Multidisciplinary Oncological Center of Antwerp (MOCA), Antwerp University Hospital, Edegem, Belgium
| | - Laure-Anne Teuwen
- Department of Oncology and Multidisciplinary Oncological Center of Antwerp (MOCA), Antwerp University Hospital, Edegem, Belgium
| | - Tiffany Foo
- Department of Medical Oncology, The Queen Elizabeth Hospital, Woodville, Australia
| | - Timon Vandamme
- Department of Oncology and Multidisciplinary Oncological Center of Antwerp (MOCA), Antwerp University Hospital, Edegem, Belgium
| | - Annabel Smith
- Department of Medical Oncology, The Queen Elizabeth Hospital, Woodville, Australia
| | - Marc Peeters
- Department of Oncology and Multidisciplinary Oncological Center of Antwerp (MOCA), Antwerp University Hospital, Edegem, Belgium
| | - Timothy Price
- Department of Medical Oncology, The Queen Elizabeth Hospital, Woodville, Australia
- School of Medicine, University of Adelaide, Adelaide, Australia
| |
Collapse
|
10
|
Aouimeur I, Sagnial T, Coulomb L, Maurin C, Thomas J, Forestier P, Ninotta S, Perrache C, Forest F, Gain P, Thuret G, He Z. Investigating the Role of TGF-β Signaling Pathways in Human Corneal Endothelial Cell Primary Culture. Cells 2023; 12:1624. [PMID: 37371094 PMCID: PMC10297110 DOI: 10.3390/cells12121624] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 06/05/2023] [Accepted: 06/10/2023] [Indexed: 06/29/2023] Open
Abstract
Corneal endothelial diseases are the leading cause of corneal transplantation. The global shortage of donor corneas has resulted in the investigation of alternative methods, such as cell therapy and tissue-engineered endothelial keratoplasty (TEEK), using primary cultures of human corneal endothelial cells (hCECs). The main challenge is optimizing the hCEC culture process to increase the endothelial cell density (ECD) and overall yield while preventing endothelial-mesenchymal transition (EndMT). Fetal bovine serum (FBS) is necessary for hCEC expansion but contains TGF-βs, which have been shown to be detrimental to hCECs. Therefore, we investigated various TGF-β signaling pathways using inhibitors to improve hCEC culture. Initially, we confirmed that TGF-β1, 2, and 3 induced EndMT on confluent hCECs without FBS. Using this TGF-β-induced EndMT model, we validated NCAM as a reliable biomarker to assess EndMT. We then demonstrated that, in a culture medium containing 8% FBS for hCEC expansion, TGF-β1 and 3, but not 2, significantly reduced the ECD and caused EndMT. TGF-β receptor inhibition had an anti-EndMT effect. Inhibition of the ROCK pathway, notably that of the P38 MAPK pathway, increased the ECD, while inhibition of the ERK pathway decreased the ECD. In conclusion, the presence of TGF-β1 and 3 in 8% FBS leads to a reduction in ECD and induces EndMT. The use of SB431542 or LY2109761 may prevent EndMT, while Y27632 or Ripasudil, and SB203580 or SB202190, can increase the ECD.
Collapse
Affiliation(s)
- Inès Aouimeur
- Laboratory of Biology, Engineering and Imaging for Ophthalmology (BiiO), EA2521, Faculty of Medicine, Jean Monnet University, 42270 Saint-Etienne, France
| | - Tomy Sagnial
- Laboratory of Biology, Engineering and Imaging for Ophthalmology (BiiO), EA2521, Faculty of Medicine, Jean Monnet University, 42270 Saint-Etienne, France
| | - Louise Coulomb
- Laboratory of Biology, Engineering and Imaging for Ophthalmology (BiiO), EA2521, Faculty of Medicine, Jean Monnet University, 42270 Saint-Etienne, France
| | - Corantin Maurin
- Laboratory of Biology, Engineering and Imaging for Ophthalmology (BiiO), EA2521, Faculty of Medicine, Jean Monnet University, 42270 Saint-Etienne, France
| | - Justin Thomas
- Laboratory of Biology, Engineering and Imaging for Ophthalmology (BiiO), EA2521, Faculty of Medicine, Jean Monnet University, 42270 Saint-Etienne, France
| | - Pierre Forestier
- Laboratory of Biology, Engineering and Imaging for Ophthalmology (BiiO), EA2521, Faculty of Medicine, Jean Monnet University, 42270 Saint-Etienne, France
| | - Sandrine Ninotta
- Laboratory of Biology, Engineering and Imaging for Ophthalmology (BiiO), EA2521, Faculty of Medicine, Jean Monnet University, 42270 Saint-Etienne, France
- Eye Bank, Etablissement Français du Sang (EFS) Auvergne-Rhône-Alpes, 42023 Saint-Etienne, France
| | - Chantal Perrache
- Laboratory of Biology, Engineering and Imaging for Ophthalmology (BiiO), EA2521, Faculty of Medicine, Jean Monnet University, 42270 Saint-Etienne, France
| | - Fabien Forest
- Laboratory of Biology, Engineering and Imaging for Ophthalmology (BiiO), EA2521, Faculty of Medicine, Jean Monnet University, 42270 Saint-Etienne, France
| | - Philippe Gain
- Laboratory of Biology, Engineering and Imaging for Ophthalmology (BiiO), EA2521, Faculty of Medicine, Jean Monnet University, 42270 Saint-Etienne, France
- Ophthalmology Department, University Hospital Center, 42055 Saint-Etienne, France
| | - Gilles Thuret
- Laboratory of Biology, Engineering and Imaging for Ophthalmology (BiiO), EA2521, Faculty of Medicine, Jean Monnet University, 42270 Saint-Etienne, France
- Ophthalmology Department, University Hospital Center, 42055 Saint-Etienne, France
| | - Zhiguo He
- Laboratory of Biology, Engineering and Imaging for Ophthalmology (BiiO), EA2521, Faculty of Medicine, Jean Monnet University, 42270 Saint-Etienne, France
| |
Collapse
|
11
|
Borlongan MC, Wang H. Profiling and targeting cancer stem cell signaling pathways for cancer therapeutics. Front Cell Dev Biol 2023; 11:1125174. [PMID: 37305676 PMCID: PMC10247984 DOI: 10.3389/fcell.2023.1125174] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 05/15/2023] [Indexed: 06/13/2023] Open
Abstract
Tumorigenic cancer stem cells (CSCs) represent a subpopulation of cells within the tumor that express genetic and phenotypic profiles and signaling pathways distinct from the other tumor cells. CSCs have eluded many conventional anti-oncogenic treatments, resulting in metastases and relapses of cancers. Effectively targeting CSCs' unique self-renewal and differentiation properties would be a breakthrough in cancer therapy. A better characterization of the CSCs' unique signaling mechanisms will improve our understanding of the pathology and treatment of cancer. In this paper, we will discuss CSC origin, followed by an in-depth review of CSC-associated signaling pathways. Particular emphasis is given on CSC signaling pathways' ligand-receptor engagement, upstream and downstream mechanisms, and associated genes, and molecules. Signaling pathways associated with regulation of CSC development stand as potential targets of CSC therapy, which include Wnt, TGFβ (transforming growth factor-β)/SMAD, Notch, JAK-STAT (Janus kinase-signal transducers and activators of transcription), Hedgehog (Hh), and vascular endothelial growth factor (VEGF). Lastly, we will also discuss milestone discoveries in CSC-based therapies, including pre-clinical and clinical studies featuring novel CSC signaling pathway cancer therapeutics. This review aims at generating innovative views on CSCs toward a better understanding of cancer pathology and treatment.
Collapse
Affiliation(s)
- Mia C. Borlongan
- Master Program of Pharmaceutical Science College of Graduate Studies, Elk Grove, CA, United States
| | - Hongbin Wang
- Master Program of Pharmaceutical Science College of Graduate Studies, Elk Grove, CA, United States
- Department of Pharmaceutical and Biomedical Sciences College of Pharmacy, Elk Grove, CA, United States
- Department of Basic Science College of Medicine, California Northstate University, Elk Grove, CA, United States
| |
Collapse
|
12
|
Mukhi D, Kolligundla LP, Maruvada S, Nishad R, Pasupulati AK. Growth hormone induces transforming growth factor-β1 in podocytes: Implications in podocytopathy and proteinuria. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119391. [PMID: 36400249 DOI: 10.1016/j.bbamcr.2022.119391] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 10/31/2022] [Accepted: 11/06/2022] [Indexed: 11/17/2022]
Abstract
Pituitary growth hormone (GH) is essential for growth, metabolism, and renal function. Overactive GH signaling is associated with impaired kidney function. Glomerular podocytes, a key kidney cell type, play an indispensable role in the renal filtration and express GH receptors (GHR), suggesting the direct action of GH on these cells. However, the precise mechanism and the downstream signaling events by which GH leads to diabetic nephropathy remain to be elucidated. Here we performed proteome analysis of the condition media from human podocytes and confirmed that GH-induces TGF-β1. Inhibition of GH/GHR stimulated-JAK2 signaling abrogates GH-induced TGF-β1 secretion. Mice administered with GH showed glomerular manifestations concomitant with proteinuria. Pharmacological inhibition of TGF-βR1 in mice prevented GH-induced TGF-β dependent SMAD signaling and proteinuria. Conditional deletion of GHR in podocytes protected mice from streptozotocin-induced diabetic nephropathy. GH and TGF-β1 signaling components expression was elevated in the kidneys of human diabetic nephropathy patients. Our study identifies that GH induces TGF-β1 in podocytes, contributing to diabetic nephropathy.
Collapse
Affiliation(s)
- Dhanunjay Mukhi
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | - Lakshmi P Kolligundla
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | - Saikrishna Maruvada
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | - Rajkishor Nishad
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | - Anil K Pasupulati
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, India.
| |
Collapse
|
13
|
Husanie H, Abu-Remaileh M, Maroun K, Abu-Tair L, Safadi H, Atlan K, Golan T, Aqeilan RI. Loss of tumor suppressor WWOX accelerates pancreatic cancer development through promotion of TGFβ/BMP2 signaling. Cell Death Dis 2022; 13:1074. [PMID: 36572673 PMCID: PMC9792466 DOI: 10.1038/s41419-022-05519-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 12/05/2022] [Accepted: 12/14/2022] [Indexed: 12/28/2022]
Abstract
Pancreatic cancer is one of the most lethal cancers, owing to its late diagnosis and resistance to chemotherapy. The tumor suppressor WW domain-containing oxidoreductase (WWOX), one of the most active fragile sites in the human genome (FRA16D), is commonly altered in pancreatic cancer. However, the direct contribution of WWOX loss to pancreatic cancer development and progression remains largely unknown. Here, we report that combined conditional deletion of Wwox and activation of KRasG12D in Ptf1a-CreER-expressing mice results in accelerated formation of precursor lesions and pancreatic carcinoma. At the molecular level, we found that WWOX physically interacts with SMAD3 and BMP2, which are known activators of the TGF-β signaling pathway. In the absence of WWOX, TGFβ/BMPs signaling was enhanced, leading to increased macrophage infiltration and enhanced cancer stemness. Finally, overexpression of WWOX in patient-derived xenografts led to diminished aggressiveness both in vitro and in vivo. Overall, our findings reveal an essential role of WWOX in pancreatic cancer development and progression and underscore its role as a bona fide tumor suppressor.
Collapse
Affiliation(s)
- Hussam Husanie
- grid.9619.70000 0004 1937 0538The Concern Foundation Laboratories, The Lautenberg Center for Immunology and Cancer Research, Department of Immunology and Cancer Research-IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Muhannad Abu-Remaileh
- grid.9619.70000 0004 1937 0538The Concern Foundation Laboratories, The Lautenberg Center for Immunology and Cancer Research, Department of Immunology and Cancer Research-IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Kian Maroun
- grid.9619.70000 0004 1937 0538The Concern Foundation Laboratories, The Lautenberg Center for Immunology and Cancer Research, Department of Immunology and Cancer Research-IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Lina Abu-Tair
- grid.9619.70000 0004 1937 0538The Concern Foundation Laboratories, The Lautenberg Center for Immunology and Cancer Research, Department of Immunology and Cancer Research-IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Hazem Safadi
- grid.9619.70000 0004 1937 0538The Concern Foundation Laboratories, The Lautenberg Center for Immunology and Cancer Research, Department of Immunology and Cancer Research-IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Karine Atlan
- grid.17788.310000 0001 2221 2926Department of Pathology, Hadassah Medical Center, Jerusalem, Israel
| | - Talia Golan
- grid.12136.370000 0004 1937 0546Oncology Institute, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel
| | - Rami I. Aqeilan
- grid.9619.70000 0004 1937 0538The Concern Foundation Laboratories, The Lautenberg Center for Immunology and Cancer Research, Department of Immunology and Cancer Research-IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
14
|
Ye Z, Kilic G, Dabelsteen S, Marinova IN, Thøfner JF, Song M, Rudjord-Levann AM, Bagdonaite I, Vakhrushev SY, Brakebusch CH, Olsen JV, Wandall HH. Characterization of TGF-β signaling in a human organotypic skin model reveals that loss of TGF-βRII induces invasive tissue growth. Sci Signal 2022; 15:eabo2206. [DOI: 10.1126/scisignal.abo2206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Transforming growth factor–β (TGF-β) signaling regulates various aspects of cell growth and differentiation and is often dysregulated in human cancers. We combined genetic engineering of a human organotypic three-dimensional (3D) skin model with global quantitative proteomics and phosphoproteomics to dissect the importance of essential components of the TGF-β signaling pathway, including the ligands TGF-β1, TGF-β2, and TGF-β3, the receptor TGF-βRII, and the intracellular effector SMAD4. Consistent with the antiproliferative effects of TGF-β signaling, the loss of TGF-β1 or SMAD4 promoted cell cycling and delayed epidermal differentiation. The loss of TGF-βRII, which abrogates both SMAD4-dependent and SMAD4-independent downstream signaling, more strongly affected cell proliferation and differentiation than did loss of SMAD4, and it induced invasive growth. TGF-βRII knockout reduced cell-matrix interactions, and the production of matrix proteins increased the production of cancer-associated cell-cell adhesion proteins and proinflammatory mediators and increased mitogen-activated protein kinase (MAPK) signaling. Inhibiting the activation of the ERK and p38 MAPK pathways blocked the development of the invasive phenotype upon the loss of TGF-βRII. This study provides a framework for exploring TGF-β signaling pathways in human epithelial tissue homeostasis and transformation using genetic engineering, 3D tissue models, and high-throughput quantitative proteomics and phosphoproteomics.
Collapse
Affiliation(s)
- Zilu Ye
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Gülcan Kilic
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Section of Oral Biology and Immunopathology, School of Dentistry, University of Copenhagen, Copenhagen, Denmark
| | - Sally Dabelsteen
- Section of Oral Biology and Immunopathology, School of Dentistry, University of Copenhagen, Copenhagen, Denmark
| | - Irina N. Marinova
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens F. B. Thøfner
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ming Song
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Asha M. Rudjord-Levann
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ieva Bagdonaite
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sergey Y. Vakhrushev
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Cord H. Brakebusch
- Biotech Research and Innovation Centre, Biomedical Institute, University of Copenhagen, Copenhagen, Denmark
| | - Jesper V. Olsen
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Hans H. Wandall
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
15
|
Mukherjee AG, Wanjari UR, Gopalakrishnan AV, Bradu P, Sukumar A, Patil M, Renu K, Dey A, Vellingiri B, George A, Ganesan R. Implications of cancer stem cells in diabetes and pancreatic cancer. Life Sci 2022; 312:121211. [PMID: 36414089 DOI: 10.1016/j.lfs.2022.121211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 11/15/2022] [Accepted: 11/16/2022] [Indexed: 11/21/2022]
Abstract
This review provides a detailed study of pancreatic cancer (PC) and the implication of different types of cancers concerning diabetes. The combination of anti-diabetic drugs with other anti-cancer drugs and phytochemicals can help prevent and treat this disease. PC cancer stem cells (CSCs) and how they migrate and develop into malignant tumors are discussed. A detailed explanation of the different mechanisms of diabetes development, which can enhance the pancreatic CSCs' proliferation by increasing the IGF factor levels, epigenetic modifications, DNA damage, and the influence of lifestyle factors like obesity, and inflammation, has been discussed. It also explains how cancer due to diabetes is associated with high mortality rates. One of the well-known diabetic drugs, metformin, can be combined with other anti-cancer drugs and prevent the development of PC and has been taken as one of the prime focus in this review. Overall, this paper provides insight into the relationship between diabetes and PC and the methods that can be employed to diagnose this disease at an earlier stage successfully.
Collapse
Affiliation(s)
- Anirban Goutam Mukherjee
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - Uddesh Ramesh Wanjari
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India.
| | - Pragya Bradu
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - Aarthi Sukumar
- Department of Integrative Biology, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - Megha Patil
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - Kaviyarasi Renu
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, 600077, Tamil Nadu, India
| | - Abhijit Dey
- Department of Life Sciences, Presidency University, Kolkata, West Bengal, 700073, India
| | - Balachandar Vellingiri
- Stem cell and Regenerative Medicine/Translational Research, Department of Zoology, School of Basic Sciences, Central University of Punjab (CUPB), Bathinda - 151401, Punjab, India
| | - Alex George
- Jubilee Centre for Medical Research, Jubilee Mission Medical College and Research Institute, Thrissur, 680005, Kerala, India
| | - Raja Ganesan
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, 24252, Republic of Korea
| |
Collapse
|
16
|
Devan AR, Pavithran K, Nair B, Murali M, Nath LR. Deciphering the role of transforming growth factor-beta 1 as a diagnostic-prognostic-therapeutic candidate against hepatocellular carcinoma. World J Gastroenterol 2022; 28:5250-5264. [PMID: 36185626 PMCID: PMC9521521 DOI: 10.3748/wjg.v28.i36.5250] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 04/30/2022] [Accepted: 08/16/2022] [Indexed: 02/06/2023] Open
Abstract
Transforming growth factor-beta (TGF-β) is a multifunctional cytokine that performs a dual role as a tumor suppressor and tumor promoter during cancer progression. Among different ligands of the TGF-β family, TGF-β1 modulates most of its biological outcomes. Despite the abundant expression of TGF-β1 in the liver, steatosis to hepatocellular carcinoma (HCC) progression triggers elevated TGF-β1 levels, contributing to poor prognosis and survival. Additionally, elevated TGF-β1 levels in the tumor microenvironment create an immunosuppressive stage via various mechanisms. TGF-β1 has a prime role as a diagnostic and prognostic biomarker in HCC. Moreover, TGF-β1 is widely studied as a therapeutic target either as monotherapy or combined with immune checkpoint inhibitors. This review provides clinical relevance and up-to-date information regarding the potential of TGF-β1 in diagnosis, prognosis, and therapy against HCC.
Collapse
Affiliation(s)
- Aswathy R Devan
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Kochi 682041, Kerala, India
| | - Keechilat Pavithran
- Department of Medical Oncology and Hematology, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi 682041, Kerala, India
| | - Bhagyalakshmi Nair
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Kochi 682041, Kerala, India
| | - Maneesha Murali
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Kochi 682041, Kerala, India
| | - Lekshmi R Nath
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Kochi 682041, Kerala, India
| |
Collapse
|
17
|
Raja Arul GL, Toruner MD, Gatenby RA, Carr RM. Ecoevolutionary biology of pancreatic ductal adenocarcinoma. Pancreatology 2022; 22:730-740. [PMID: 35821188 DOI: 10.1016/j.pan.2022.06.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 06/01/2022] [Indexed: 12/11/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC), the most common histological subtype of pancreatic cancer, is an aggressive disease predicted to be the 2nd cause of cancer mortality in the US by 2040. While first-line therapy has improved, 5-year overall survival has only increased from 5 to ∼10%, and surgical resection is only available for ∼20% of patients as most present with advanced disease, which is invariably lethal. PDAC has well-established highly recurrent mutations in four driver genes including KRAS, TP53, CDKN2A, and SMAD4. Unfortunately, these genetic drivers are not currently therapeutically actionable. Despite extensive sequencing efforts, few additional significantly recurrent and druggable drivers have been identified. In the absence of targetable mutations, chemotherapy remains the mainstay of treatment for most patients. Further, the role of the above driver mutations on PDAC initiation and early development is well-established. However, these mutations alone cannot account for PDAC heterogeneity nor discern early from advanced disease. Taken together, management of PDAC is an example highlighting the shortcomings of the current precision medicine paradigm. PDAC, like other malignancies, represents an ecoevolutionary process. Better understanding the disease through this lens can facilitate the development of novel therapeutic strategies to better control and cure PDAC. This review aims to integrate the current understanding of PDAC pathobiology into an ecoevolutionary framework.
Collapse
Affiliation(s)
| | - Merih D Toruner
- Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Robert A Gatenby
- Department of Integrated Mathematical Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Ryan M Carr
- Department of Oncology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
18
|
Balsano R, Kruize Z, Lunardi M, Comandatore A, Barone M, Cavazzoni A, Re Cecconi AD, Morelli L, Wilmink H, Tiseo M, Garajovà I, van Zuylen L, Giovannetti E, Piccirillo R. Transforming Growth Factor-Beta Signaling in Cancer-Induced Cachexia: From Molecular Pathways to the Clinics. Cells 2022; 11:2671. [PMID: 36078078 PMCID: PMC9454487 DOI: 10.3390/cells11172671] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 08/23/2022] [Accepted: 08/25/2022] [Indexed: 02/06/2023] Open
Abstract
Cachexia is a metabolic syndrome consisting of massive loss of muscle mass and function that has a severe impact on the quality of life and survival of cancer patients. Up to 20% of lung cancer patients and up to 80% of pancreatic cancer patients are diagnosed with cachexia, leading to death in 20% of them. The main drivers of cachexia are cytokines such as interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-α), macrophage inhibitory cytokine 1 (MIC-1/GDF15) and transforming growth factor-beta (TGF-β). Besides its double-edged role as a tumor suppressor and activator, TGF-β causes muscle loss through myostatin-based signaling, involved in the reduction in protein synthesis and enhanced protein degradation. Additionally, TGF-β induces inhibin and activin, causing weight loss and muscle depletion, while MIC-1/GDF15, a member of the TGF-β superfamily, leads to anorexia and so, indirectly, to muscle wasting, acting on the hypothalamus center. Against this background, the blockade of TGF-β is tested as a potential mechanism to revert cachexia, and antibodies against TGF-β reduced weight and muscle loss in murine models of pancreatic cancer. This article reviews the role of the TGF-β pathway and to a minor extent of other molecules including microRNA in cancer onset and progression with a special focus on their involvement in cachexia, to enlighten whether TGF-β and such other players could be potential targets for therapy.
Collapse
Affiliation(s)
- Rita Balsano
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- Medical Oncology Unit, University Hospital of Parma, 43100 Parma, Italy
| | - Zita Kruize
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Martina Lunardi
- Department of Neurosciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy
| | - Annalisa Comandatore
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56124 Pisa, Italy
| | - Mara Barone
- Department of Neurosciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy
| | - Andrea Cavazzoni
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Andrea David Re Cecconi
- Department of Neurosciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy
| | - Luca Morelli
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56124 Pisa, Italy
| | - Hanneke Wilmink
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Marcello Tiseo
- Medical Oncology Unit, University Hospital of Parma, 43100 Parma, Italy
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Ingrid Garajovà
- Medical Oncology Unit, University Hospital of Parma, 43100 Parma, Italy
| | - Lia van Zuylen
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- Fondazione Pisana per La Scienza, 56124 Pisa, Italy
| | - Rosanna Piccirillo
- Department of Neurosciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy
| |
Collapse
|
19
|
Zhang Y, Qin S, Chao J, Luo Y, Sun Y, Duan J. The In-Vitro Antitumor Effects of AST-3424 Monotherapy and Combination Therapy With Oxaliplatin or 5-Fluorouracil in Primary Liver Cancer. Front Oncol 2022; 12:885139. [PMID: 35936728 PMCID: PMC9354847 DOI: 10.3389/fonc.2022.885139] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 06/06/2022] [Indexed: 01/08/2023] Open
Abstract
Background Primary liver cancer (PLC) is a common and highly lethal malignancy in the world. Approximately 85% of PLC is hepatocellular carcinoma (HCC), and this study mainly focuses on HCC. The onset of liver cancer is insidious and often complicated with basic liver disease. Meanwhile, its clinical symptoms are atypical, and the degree of malignancy is high. What is worse is that its treatment is difficult, and the prognosis is poor. All these factors make its mortality close to its incidence. AST-3424 is a prodrug of a potent nitrogen mustard, which targets the tumor by its specific and selective mode of activation and results in the concentration of the drug in the tumor and plays a higher intensity of antitumor effect with reduced side effects. The purpose of this study was to explore the in-vitro antitumor activity and mechanism of AST-3424 monotherapy and combination therapy with oxaliplatin (OXA) or 5-fluorouracil (5-Fu). Moreover, it can provide an experimental basis for further studies. Methods Tumor growth of HCC cells was examined by using the Cell Counting Kit-8 (CCK-8), flow cytometry, and clone formation assays. Tumor migration of HCC cells was examined by using the Transwell assay. The in-vitro antitumor activity of AST-3424 monotherapy and combination therapy with OXA and 5-Fu was quantified by growth and metastasis inhibition rate. The underlying molecular mechanism was investigated by using Western blotting. Results The inhibiting effects of AST-3424 were significant in both HepG2 cells and PLC/PRF/5 cells. Moreover, HepG2 cells showed higher sensitivity to AST-3424. With increasing AST-3424 concentration, AKR1C3 protein expression level was downregulated significantly. The inhibition of AST-3424 was significantly higher than OXA, 5-Fu, Sor (sorafenib), and Apa (apatinib) in both HCC cells. AST-3424 monotherapy and combination therapy with OXA or 5-Fu all strongly inhibited the proliferation of HCC cells, blocked HCC cells in the S phase, promoted apoptosis induction, and suppressed the migration of HCC cells. Among them, the antitumor effect of AST-3424 in combination with OXA was obviously enhanced. Western blotting analysis demonstrated the regulation of P21, Bax, Caspase3, PARP, MMP-2, MMP-9, and p-Smad proteins in the presence of AST-3424 monotherapy and combination therapy with OXA or 5-Fu, indicating that its antitumor mechanisms may be associated with the regulation of the TGF-β signaling cascade. Conclusion The in-vitro studies revealed that AST-3424 in combination with both OXA and 5-Fu showed an increased antitumor effect, and the combination with OXA resulted in a synergistic effect. Together with the in-vitro results, additional in-vitro and in-vivo studies are warranted to further certify its antitumor effects and explore more potential antitumor mechanisms.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Graduate School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shukui Qin
- Department of Medical Oncology Center, BaYi Affiliated Hospital, Nanjing, China
- *Correspondence: Shukui Qin,
| | - Jiaojiao Chao
- Department of Graduate School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yan Luo
- Department of Biochemistry and Cancer Institute of the Second Affiliated Hospital (Key Laboratory of Cancer Prevention and Intervention of China National Ministry of Education (MOE)), Zhejiang University School of Medicine, Hangzhou, China
| | - Yandi Sun
- Department of Biochemistry and Cancer Institute of the Second Affiliated Hospital (Key Laboratory of Cancer Prevention and Intervention of China National Ministry of Education (MOE)), Zhejiang University School of Medicine, Hangzhou, China
| | - Jianxin Duan
- Ascentawits Pharmaceuticals, Ltd., Shenzhen, China
| |
Collapse
|
20
|
Liao J, Liu Q, Chen J, Lu Z, Mo H, Jia J. A risk score model based on TGF-β pathway-related genes predicts survival, tumor microenvironment and immunotherapy for liver hepatocellular carcinoma. Proteome Sci 2022; 20:11. [PMID: 35733217 PMCID: PMC9215003 DOI: 10.1186/s12953-022-00192-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 05/06/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Transforming growth factor-beta (TGF-β) signal is an important pathway involved in all stages of liver hepatocellular carcinoma (LIHC) initiation and progression. Therefore, targeting TGF- β pathway may be a potential therapeutic strategy for LIHC. Prediction of patients' tumor cells response requires effective biomarkers. METHODS From 54 TGF-β-related genes, this research determined the genes showing the greatest relation to LIHC prognosis, and developed a risk score model with 8 TGF-β-related genes. The model divided LIHC patients from different datasets and platforms into low- and high-risk groups. Multivariate Cox regression analysis confirmed that the model was an independent prognostic factor for LIHC. The differences in genetic mutation, immune cell infiltration, biological pathway, response to immunotherapy or chemotherapy, and tumor microenvironment in LIHC samples showing different risks were analyzed. RESULTS Compared with low-risk group, in the training set and test set, high-risk group showed shorter survival, lower stromal score and higher M0 macrophages scores, regulatory T cells (Tregs), helper follicular T cells. Moreover, high-risk samples showed higher sensitivity to cisplatin, imatinib, sorafenib and salubrinal and pyrimethamine. High-risk group demonstrated a significantly higher Tumor Immune Dysfunction and Exclusion (TIDE) score, but would significantly benefit less from taking immunotherapy and was less likely to respond to immune checkpoint inhibitors. CONCLUSIONS In general, this work provided a risk scoring model based on 8 TGF-β pathway-related genes, which might be a new potential tool for predicting LIHC.
Collapse
Affiliation(s)
- Jingsheng Liao
- Department of Medical Oncology, Affiliated Dongguan Hospital, Southern Medical University, 78 Wandao Road, Dongguan City, 523000, Guangdong Province, China.,Department of Medical Oncology, Dongguan Institute of Clinical Cancer Research, 78 Wandao Road, Dongguan City, 523000, Guangdong Province, China
| | - Qi Liu
- Department of Medical Oncology, Affiliated Dongguan Hospital, Southern Medical University, 78 Wandao Road, Dongguan City, 523000, Guangdong Province, China.,Department of Medical Oncology, Dongguan Institute of Clinical Cancer Research, 78 Wandao Road, Dongguan City, 523000, Guangdong Province, China
| | - Jingtang Chen
- Department of Medical Oncology, Affiliated Dongguan Hospital, Southern Medical University, 78 Wandao Road, Dongguan City, 523000, Guangdong Province, China.,Department of Medical Oncology, Dongguan Institute of Clinical Cancer Research, 78 Wandao Road, Dongguan City, 523000, Guangdong Province, China
| | - Zhibin Lu
- Department of Medical Oncology, Affiliated Dongguan Hospital, Southern Medical University, 78 Wandao Road, Dongguan City, 523000, Guangdong Province, China.,Department of Medical Oncology, Dongguan Institute of Clinical Cancer Research, 78 Wandao Road, Dongguan City, 523000, Guangdong Province, China
| | - Huiting Mo
- Department of Medical Oncology, Affiliated Dongguan Hospital, Southern Medical University, 78 Wandao Road, Dongguan City, 523000, Guangdong Province, China.,Department of Medical Oncology, Dongguan Institute of Clinical Cancer Research, 78 Wandao Road, Dongguan City, 523000, Guangdong Province, China
| | - Jun Jia
- Department of Medical Oncology, Affiliated Dongguan Hospital, Southern Medical University, 78 Wandao Road, Dongguan City, 523000, Guangdong Province, China. .,Department of Medical Oncology, Dongguan Institute of Clinical Cancer Research, 78 Wandao Road, Dongguan City, 523000, Guangdong Province, China.
| |
Collapse
|
21
|
Tosti E, Almeida AS, Tran TTT, Barbachan E Silva M, Broin PÓ, Dubin R, Chen K, Beck AP, Mclellan AS, Vilar E, Golden A, O'Toole PW, Edelmann W. Loss of MMR and TGFBR2 Increases the Susceptibility to Microbiota-Dependent Inflammation-Associated Colon Cancer. Cell Mol Gastroenterol Hepatol 2022; 14:693-717. [PMID: 35688320 PMCID: PMC9421583 DOI: 10.1016/j.jcmgh.2022.05.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 04/29/2022] [Accepted: 05/18/2022] [Indexed: 12/10/2022]
Abstract
BACKGROUND AND AIMS Mutations in DNA mismatch repair (MMR) genes are causative in Lynch syndrome and a significant proportion of sporadic colorectal cancers (CRCs). MMR-deficient (dMMR) CRCs display increased mutation rates, with mutations frequently accumulating at short repetitive DNA sequences throughout the genome (microsatellite instability). The TGFBR2 gene is one of the most frequently mutated genes in dMMR CRCs. Therefore, we generated an animal model to study how the loss of both TGFBR2 signaling impacts dMMR-driven intestinal tumorigenesis in vivo and explore the impact of the gut microbiota. METHODS We generated VCMsh2/Tgfbr2 mice in which Msh2loxP and Tgfbr2loxP alleles are inactivated by Villin-Cre recombinase in the intestinal epithelium. VCMsh2/Tgfbr2 mice were analyzed for their rate of intestinal cancer development and for the mutational spectra and gene expression profiles of tumors. In addition, we assessed the impact of chemically induced chronic inflammation and gut microbiota composition on colorectal tumorigenesis. RESULTS VCMsh2/Tgfbr2 mice developed small intestinal adenocarcinomas and CRCs with histopathological features highly similar to CRCs in Lynch syndrome patients. The CRCs in VCMsh2/Tgfbr2 mice were associated with the presence of colitis and displayed genetic and histological features that resembled inflammation-associated CRCs in human patients. The development of CRCs in VCMsh2/Tgfbr2 mice was strongly modulated by the gut microbiota composition, which in turn was impacted by the TGFBR2 status of the tumors. CONCLUSIONS Our results demonstrate a synergistic interaction between MMR and TGFBR2 inactivation in inflammation-associated colon tumorigenesis and highlight the crucial impact of the gut microbiota on modulating the incidence of inflammation-associated CRCs.
Collapse
Affiliation(s)
- Elena Tosti
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York.
| | - Ana S Almeida
- APC Microbiome Ireland and School of Microbiology, University College Cork, Cork, Ireland
| | - Tam T T Tran
- University of Science and Technology of Hanoi, Vietnam Academy of Science and Technology, Hanoi, Vietnam
| | - Mariel Barbachan E Silva
- School of Mathematics, Statistics and Applied Mathematics, National University of Ireland Galway, Galway, Ireland
| | - Pilib Ó Broin
- School of Mathematics, Statistics and Applied Mathematics, National University of Ireland Galway, Galway, Ireland
| | - Robert Dubin
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York
| | - Ken Chen
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York
| | - Amanda P Beck
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York
| | - Andrew S Mclellan
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, New York
| | - Eduardo Vilar
- Department of Clinical Cancer Prevention, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Aaron Golden
- School of Mathematics, Statistics and Applied Mathematics, National University of Ireland Galway, Galway, Ireland
| | - Paul W O'Toole
- APC Microbiome Ireland and School of Microbiology, University College Cork, Cork, Ireland
| | - Winfried Edelmann
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York.
| |
Collapse
|
22
|
Nathan J, Shameera R, Palanivel G. Studying molecular signaling in major angiogenic diseases. Mol Cell Biochem 2022; 477:2433-2450. [PMID: 35581517 DOI: 10.1007/s11010-022-04452-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 04/24/2022] [Indexed: 10/18/2022]
Abstract
The growth of blood vessels from already existing vasculature is angiogenesis and it is one of the fundamental processes in fetal development, tissue damage or repair, and the reproductive cycle. In a healthy person, angiogenesis is regulated by the balance between pro- and anti-angiogenic factors. However, when the balance is disturbed, it results in various diseases or disorders. The angiogenesis pathway is a sequential cascade and differs based on the stimuli. Therefore, targeting one of the factors involved in the process can help us find a therapeutic strategy to treat irregular angiogenesis. In the past three decades of cancer research, angiogenesis has been at its peak, where an anti-angiogenic agent inhibiting vascular endothelial growth factor acts as a promising substance to treat cancer. In addition, cancer can be assessed based on the expression of angiogenic factors and its response to therapies. Angiogenesis is important for all tissues, which might be normal or pathologically changed and occur through ages. In clinical therapeutics, target therapy focusing on discovery of novel anti-angiogenic agents like bevacizumab, cetuximab, sunitinib, imatinib, lenvatinib, thalidomide, everolimus etc., to block or inhibit the angiogenesis pathway is well explored in recent times. In this review, we will discuss about the molecular signaling pathways involved in major angiogenic diseases in detail.
Collapse
Affiliation(s)
- Jhansi Nathan
- Zebrafish Developmental Biology Laboratory, AUKBC Research Centre, Anna University, Chennai, Tamil Nadu, 600044, India.
| | - Rabiathul Shameera
- Zebrafish Developmental Biology Laboratory, AUKBC Research Centre, Anna University, Chennai, Tamil Nadu, 600044, India
| | - Gajalakshmi Palanivel
- Zebrafish Developmental Biology Laboratory, AUKBC Research Centre, Anna University, Chennai, Tamil Nadu, 600044, India
| |
Collapse
|
23
|
Role of tumour-derived exosomes in metastasis. Biomed Pharmacother 2022; 147:112657. [DOI: 10.1016/j.biopha.2022.112657] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 01/15/2022] [Accepted: 01/18/2022] [Indexed: 12/15/2022] Open
|
24
|
Dharmalingam P, Venkatakrishnan K, Tan B. Nanoplatform to Investigate Tumor-Initiating Cancer Stem Cells: Breaking the Diagnostic Barrier. ACS APPLIED MATERIALS & INTERFACES 2022; 14:6370-6386. [PMID: 35090345 DOI: 10.1021/acsami.1c21998] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Drug-resistant capacity in a small population of tumor-initiating cancer stem cells (tiCSCs) can be due to aberrant epigenetic changes. However, currently available conventional detection methods are inappropriate and cannot be applied to investigate the scarce population (tiCSCs). In addition, selective inhibitor drugs are shown to reverse epigenetic changes; however, each cancer type is discrete. Hence, it is essential to probe the resultant changes in tiCSCs even after therapy. Therefore, we have developed a multimode nanoplatform to investigate tiCSCs, detect epigenetic changes, and subsequently explore their transformation signals following drug therapy. We performed this by developing a surface-enhanced Raman scattering (SERS)-active nanoplatform integrated with n-dopant using an ultrafast laser ionization technique. The dopant functionalization enhances Raman scattering ability and permits label-free analysis of biomarkers in tiCSCs with the resolution down to the cellular level. Here, we investigated epigenetic biomarkers of tiCSCs in pancreatic and lung cancers. An extended study using inhibitor drugs demonstrates an unexpected increase of tiCSCs from lung cancer; this difference can be attributed to transformation changes in lung tiCSC. Thus, our work brings new insight into the differentiation abilities of CSCs upon epigenetic reversal, emphasizing unique perceptions in cancer treatment.
Collapse
Affiliation(s)
- Priya Dharmalingam
- Ultrashort Laser Nanomanufacturing Research Facility, Department of Mechanical and Industrial Engineering, Ryerson University, 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
- Institute for Biomedical Engineering, Science and Technology (I-BEST), Partnership between Ryerson University and St. Michael's Hospital, Toronto, Ontario M5B 1W8, Canada
- Nanocharacterization Laboratory, Faculty of Engineering and Architectural Science, Ryerson University, 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
- Nano-Bio Interface Facility, Department of Mechanical and Industrial Engineering, Ryerson University, 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
| | - Krishnan Venkatakrishnan
- Ultrashort Laser Nanomanufacturing Research Facility, Department of Mechanical and Industrial Engineering, Ryerson University, 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
- Keenan Research Center, St. Michael's Hospital, 209 Victoria Street, Toronto, Ontario M5B 1T8, Canada
- Nano-Bio Interface Facility, Department of Mechanical and Industrial Engineering, Ryerson University, 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
| | - Bo Tan
- Nanocharacterization Laboratory, Faculty of Engineering and Architectural Science, Ryerson University, 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
- Keenan Research Center, St. Michael's Hospital, 209 Victoria Street, Toronto, Ontario M5B 1T8, Canada
| |
Collapse
|
25
|
Wu L, Zhao X, Ma H, Zhang L, Li X. Discoidin Domain Receptor 1, a Potential Biomarker and Therapeutic Target in Hepatocellular Carcinoma. Int J Gen Med 2022; 15:2037-2044. [PMID: 35237068 PMCID: PMC8882470 DOI: 10.2147/ijgm.s348110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 02/10/2022] [Indexed: 11/24/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is still one of the most lethal human cancers in the world due to its high degree of malignancy, easy invasion and metastasis, poor therapeutic effect and poor prognosis. Nowadays, there is no very effective diagnosis and treatment method. It is crucial to elucidate the underlying pathogenesis and mechanisms of HCC for developing new and effective diagnostic/prognostic biomarkers and therapies. Discoidin domain receptors (DDRs) belong to the family of transmembrane receptor tyrosine kinases (RTKs) and are recognized as playing central regulatory roles in a variety of high incidence human diseases, including tumors. DDRs have two members, DDR1 and DDR2. The role of DDR1 in several tumors has been extensively studied, and many researchers have identified it as a powerful candidate target for the development of functional and effective tumor treatment inhibitors. However, its role and mechanism in HCC are ill defined. In this article, we review the advanced insights into the progression of DDR1 in HCC, particularly the ligands and mechanisms in invasion and metastasis, which may open new avenues for the therapeutic utility of HCC.
Collapse
Affiliation(s)
- Linghong Wu
- Department of Gastroenterology, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan, People’s Republic of China
| | - Xinhua Zhao
- Department of Gastroenterology, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan, People’s Republic of China
| | - Huan Ma
- Department of Gastroenterology, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan, People’s Republic of China
| | - Lili Zhang
- Department of Gastroenterology, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan, People’s Republic of China
| | - Xiaoan Li
- Department of Gastroenterology, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan, People’s Republic of China
- Correspondence: Xiaoan Li, Department of Gastroenterology, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 12 Changjia Lane, Jingzhong Street, Fucheng District, Mianyang, 621000, Sichuan, People’s Republic of China, Tel +86 816 224 3593 Email
| |
Collapse
|
26
|
Bhave S, Ho HK. Exploring the Gamut of Receptor Tyrosine Kinases for Their Promise in the Management of Non-Alcoholic Fatty Liver Disease. Biomedicines 2021; 9:1776. [PMID: 34944593 PMCID: PMC8698495 DOI: 10.3390/biomedicines9121776] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/18/2021] [Accepted: 11/19/2021] [Indexed: 11/17/2022] Open
Abstract
Recently, non-alcoholic fatty liver disease (NAFLD) has emerged as a predominant health concern affecting approximately a quarter of the world's population. NAFLD is a spectrum of liver ailments arising from nascent lipid accumulation and leading to inflammation, fibrosis or even carcinogenesis. Despite its prevalence and severity, no targeted pharmacological intervention is approved to date. Thus, it is imperative to identify suitable drug targets critical to the development and progression of NAFLD. In this quest, a ray of hope is nestled within a group of proteins, receptor tyrosine kinases (RTKs), as targets to contain or even reverse NAFLD. RTKs control numerous vital biological processes and their selective expression and activity in specific diseases have rendered them useful as drug targets. In this review, we discuss the recent advancements in characterizing the role of RTKs in NAFLD progression and qualify their suitability as pharmacological targets. Available data suggests inhibition of Epidermal Growth Factor Receptor, AXL, Fibroblast Growth Factor Receptor 4 and Vascular Endothelial Growth Factor Receptor, and activation of cellular mesenchymal-epithelial transition factor and Fibroblast Growth Factor Receptor 1 could pave the way for novel NAFLD therapeutics. Thus, it is important to characterize these RTKs for target validation and proof-of-concept through clinical trials.
Collapse
Affiliation(s)
| | - Han Kiat Ho
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore 117559, Singapore;
| |
Collapse
|
27
|
Maneshi P, Mason J, Dongre M, Öhlund D. Targeting Tumor-Stromal Interactions in Pancreatic Cancer: Impact of Collagens and Mechanical Traits. Front Cell Dev Biol 2021; 9:787485. [PMID: 34901028 PMCID: PMC8656238 DOI: 10.3389/fcell.2021.787485] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/01/2021] [Indexed: 01/18/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has one of the worst outcomes among cancers with a 5-years survival rate of below 10%. This is a result of late diagnosis and the lack of effective treatments. The tumor is characterized by a highly fibrotic stroma containing distinct cellular components, embedded within an extracellular matrix (ECM). This ECM-abundant tumor microenvironment (TME) in PDAC plays a pivotal role in tumor progression and resistance to treatment. Cancer-associated fibroblasts (CAFs), being a dominant cell type of the stroma, are in fact functionally heterogeneous populations of cells within the TME. Certain subtypes of CAFs are the main producer of the ECM components of the stroma, with the most abundant one being the collagen family of proteins. Collagens are large macromolecules that upon deposition into the ECM form supramolecular fibrillar structures which provide a mechanical framework to the TME. They not only bring structure to the tissue by being the main structural proteins but also contain binding domains that interact with surface receptors on the cancer cells. These interactions can induce various responses in the cancer cells and activate signaling pathways leading to epithelial-to-mesenchymal transition (EMT) and ultimately metastasis. In addition, collagens are one of the main contributors to building up mechanical forces in the tumor. These forces influence the signaling pathways that are involved in cell motility and tumor progression and affect tumor microstructure and tissue stiffness by exerting solid stress and interstitial fluid pressure on the cells. Taken together, the TME is subjected to various types of mechanical forces and interactions that affect tumor progression, metastasis, and drug response. In this review article, we aim to summarize and contextualize the recent knowledge of components of the PDAC stroma, especially the role of different collagens and mechanical traits on tumor progression. We furthermore discuss different experimental models available for studying tumor-stromal interactions and finally discuss potential therapeutic targets within the stroma.
Collapse
Affiliation(s)
- Parniyan Maneshi
- Department of Radiation Sciences, Umeå University, Umeå, Sweden
- Wallenberg Centre for Molecular Medicine, Umeå University, Umeå, Sweden
| | - James Mason
- Department of Radiation Sciences, Umeå University, Umeå, Sweden
- Wallenberg Centre for Molecular Medicine, Umeå University, Umeå, Sweden
| | - Mitesh Dongre
- Department of Radiation Sciences, Umeå University, Umeå, Sweden
- Wallenberg Centre for Molecular Medicine, Umeå University, Umeå, Sweden
| | - Daniel Öhlund
- Department of Radiation Sciences, Umeå University, Umeå, Sweden
- Wallenberg Centre for Molecular Medicine, Umeå University, Umeå, Sweden
| |
Collapse
|
28
|
Flores-Vergara R, Olmedo I, Aránguiz P, Riquelme JA, Vivar R, Pedrozo Z. Communication Between Cardiomyocytes and Fibroblasts During Cardiac Ischemia/Reperfusion and Remodeling: Roles of TGF-β, CTGF, the Renin Angiotensin Axis, and Non-coding RNA Molecules. Front Physiol 2021; 12:716721. [PMID: 34539441 PMCID: PMC8446518 DOI: 10.3389/fphys.2021.716721] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 07/26/2021] [Indexed: 11/20/2022] Open
Abstract
Communication between cells is a foundational concept for understanding the physiology and pathology of biological systems. Paracrine/autocrine signaling, direct cell-to-cell interplay, and extracellular matrix interactions are three types of cell communication that regulate responses to different stimuli. In the heart, cardiomyocytes, fibroblasts, and endothelial cells interact to form the cardiac tissue. Under pathological conditions, such as myocardial infarction, humoral factors released by these cells may induce tissue damage or protection, depending on the type and concentration of molecules secreted. Cardiac remodeling is also mediated by the factors secreted by cardiomyocytes and fibroblasts that are involved in the extensive reciprocal interactions between these cells. Identifying the molecules and cellular signal pathways implicated in these processes will be crucial for creating effective tissue-preserving treatments during or after reperfusion. Numerous therapies to protect cardiac tissue from reperfusion-induced injury have been explored, and ample pre-clinical research has attempted to identify drugs or techniques to mitigate cardiac damage. However, despite great success in animal models, it has not been possible to completely translate these cardioprotective effects to human applications. This review provides a current summary of the principal molecules, pathways, and mechanisms underlying cardiomyocyte and cardiac fibroblast crosstalk during ischemia/reperfusion injury. We also discuss pre-clinical molecules proposed as treatments for myocardial infarction and provide a clinical perspective on these potential therapeutic agents.
Collapse
Affiliation(s)
- Raúl Flores-Vergara
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, Santiago de Chile, Chile.,Programa de Fisiología y Biofísica, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago de Chile, Chile
| | - Ivonne Olmedo
- Programa de Fisiopatología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago de Chile, Chile.,Red para el Estudio de Enfermedades Cardiopulmonares de alta letalidad (REECPAL), Universidad de Chile, Santiago de Chile, Chile
| | - Pablo Aránguiz
- Escuela de Química y Farmacia, Facultad de Medicina, Universidad Andrés Bello, Viña del Mar, Chile
| | - Jaime Andrés Riquelme
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, Santiago de Chile, Chile.,Departamento de Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago de Chile, Chile
| | - Raúl Vivar
- Programa de Farmacología Molecular y Clínica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago de Chile, Chile
| | - Zully Pedrozo
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, Santiago de Chile, Chile.,Programa de Fisiología y Biofísica, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago de Chile, Chile.,Red para el Estudio de Enfermedades Cardiopulmonares de alta letalidad (REECPAL), Universidad de Chile, Santiago de Chile, Chile
| |
Collapse
|
29
|
Grasso M, Caruso G, Godos J, Bonaccorso A, Carbone C, Castellano S, Currenti W, Grosso G, Musumeci T, Caraci F. Improving Cognition with Nutraceuticals Targeting TGF-β1 Signaling. Antioxidants (Basel) 2021; 10:1075. [PMID: 34356309 PMCID: PMC8301008 DOI: 10.3390/antiox10071075] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 06/29/2021] [Accepted: 06/30/2021] [Indexed: 12/11/2022] Open
Abstract
Rescue of cognitive function represents an unmet need in the treatment of neurodegenerative disorders such as Alzheimer's disease (AD). Nutraceuticals deliver a concentrated form of a presumed bioactive(s) agent(s) that can improve cognitive function alone or in combination with current approved drugs for the treatment of cognitive disorders. Nutraceuticals include different natural compounds such as flavonoids and their subclasses (flavan-3-ols, catechins, anthocyanins, and flavonols), omega-3, and carnosine that can improve synaptic plasticity and rescue cognitive deficits through multiple molecular mechanisms. A deficit of transforming growth factor-β1 (TGF-β1) pathway is an early event in the pathophysiology of cognitive impairment in different neuropsychiatric disorders, from depression to AD. In the present review, we provide evidence that different nutraceuticals, such as Hypericum perforatum (hypericin and hyperforin), flavonoids such as hesperidin, omega-3, and carnosine, can target TGF-β1 signaling and increase TGF-β1 production in the central nervous system as well as cognitive function. The bioavailability of these nutraceuticals, in particular carnosine, can be significantly improved with novel formulations (nanoparticulate systems, nanoliposomes) that increase the efficacy and stability of this peptide. Overall, these studies suggest that the synergism between nutraceuticals targeting the TGF-β1 pathway and current approved drugs might represent a novel pharmacological approach for reverting cognitive deficits in AD patients.
Collapse
Affiliation(s)
- Margherita Grasso
- Department of Drug and Health Sciences, University of Catania, 95125 Catania, Italy; (M.G.); (G.C.); (A.B.); (C.C.); (T.M.); (F.C.)
- Oasi Research Institute—IRCCS, 94018 Troina, Italy
| | - Giuseppe Caruso
- Department of Drug and Health Sciences, University of Catania, 95125 Catania, Italy; (M.G.); (G.C.); (A.B.); (C.C.); (T.M.); (F.C.)
| | - Justyna Godos
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (J.G.); (W.C.)
| | - Angela Bonaccorso
- Department of Drug and Health Sciences, University of Catania, 95125 Catania, Italy; (M.G.); (G.C.); (A.B.); (C.C.); (T.M.); (F.C.)
| | - Claudia Carbone
- Department of Drug and Health Sciences, University of Catania, 95125 Catania, Italy; (M.G.); (G.C.); (A.B.); (C.C.); (T.M.); (F.C.)
| | - Sabrina Castellano
- Department of Educational Sciences, University of Catania, 95124 Catania, Italy;
| | - Walter Currenti
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (J.G.); (W.C.)
| | - Giuseppe Grosso
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (J.G.); (W.C.)
| | - Teresa Musumeci
- Department of Drug and Health Sciences, University of Catania, 95125 Catania, Italy; (M.G.); (G.C.); (A.B.); (C.C.); (T.M.); (F.C.)
| | - Filippo Caraci
- Department of Drug and Health Sciences, University of Catania, 95125 Catania, Italy; (M.G.); (G.C.); (A.B.); (C.C.); (T.M.); (F.C.)
- Oasi Research Institute—IRCCS, 94018 Troina, Italy
| |
Collapse
|
30
|
Apu MNH, Aktar MN, Rahman MM, Mostaid MS. Association of TGFB1 gene polymorphisms with cervical cancer in Bangladeshi women: A case-control study. Tumour Biol 2021; 43:27-35. [PMID: 33935123 DOI: 10.3233/tub-200061] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
OBJECTIVES Genetic susceptibility to cervical cancer in relation to transforming growth factor beta 1 (TGFB1) gene polymorphisms has not been investigated extensively among the women in Bangladesh. So, the aim of this study was to find out the correlation of the polymorphisms of TGFB1 C509T (rs1800469) and T869C (rs1800470) with the risk of cervical cancer among the Bangladeshi women. STUDY DESIGN 134 cervical cancer patients and 102 age-sex matched healthy controls were included from two institutions in Bangladesh. Polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) method was used for genotyping two TGFB1 single nucleotide polymorphisms C509T (rs1800469) and T869C (rs1800470) in patients and controls. RESULTS No significant correlation was found between polymorphisms C509T (rs1800469) and T869C (rs1800470) of TGFB1 gene with cervical cancer in Bangladeshi women. In case of the cervical cancer patients who had first degree relatives with cancer were prone to carry the polymorphic version of the TGFB1 gene polymorphism at C509T (OR = 5.597, 95% CI = 1.224-25.597, p < 0.05) but may not result in the increase of developing cervical cancer. CONCLUSION In summary, two polymorphisms C509T and T869C of TGFB1 gene may not be associated with cervical cancer risk in Bangladeshi women.
Collapse
Affiliation(s)
- Mohd Nazmul Hasan Apu
- Department of Clinical Pharmacy and Pharmacology, Faculty of Pharmacy, University of Dhaka, Dhaka, Bangladesh
| | - Most Nasrin Aktar
- Department of Pharmaceutical Sciences, North South University, Dhaka, Bangladesh
| | - Md Morshadur Rahman
- Department of Statistics, Faculty of Sciences, University of Dhaka, Dhaka, Bangladesh
| | - Md Shaki Mostaid
- Department of Clinical Pharmacy and Pharmacology, Faculty of Pharmacy, University of Dhaka, Dhaka, Bangladesh.,Department of Pharmaceutical Sciences, North South University, Dhaka, Bangladesh
| |
Collapse
|
31
|
Novák Š, Kolář M, Szabó A, Vernerová Z, Lacina L, Strnad H, Šáchová J, Hradilová M, Havránek J, Španko M, Čoma M, Urban L, Kaňuchová M, Melegová N, Gürlich R, Dvořák J, Smetana K, Gál P, Szabo P. Desmoplastic Crosstalk in Pancreatic Ductal Adenocarcinoma Is Reflected by Different Responses of Panc-1, MIAPaCa-2, PaTu-8902, and CAPAN-2 Cell Lines to Cancer-associated/Normal Fibroblasts. Cancer Genomics Proteomics 2021; 18:221-243. [PMID: 33893076 DOI: 10.21873/cgp.20254] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/25/2021] [Accepted: 03/04/2021] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND/AIM Pancreatic ductal adenocarcinoma (PDAC) still represents one of the most aggressive cancers. Understanding of the epithelial-mesenchymal crosstalk as a crucial part of the tumor microenvironment should pave the way for therapies to improve patient survival rates. Well-established cell lines present a useful and reproducible model to study PDAC biology. However, the tumor-stromal interactions between cancer cells and cancer-associated fibroblasts (CAFs) are still poorly understood. MATERIALS AND METHODS We studied interactions between four PDAC cell lines (Panc-1, CAPAN-2, MIAPaCa-2, and PaTu-8902) and conditioned media derived from primary cultures of normal fibroblasts/PDAC-derived CAFs (PANFs). RESULTS When the tested PDAC cell lines were stimulated by PANF-derived conditioned media, the most aggressive behavior was acquired by the Panc-1 cell line (increased number and size of colonies, remaining expression of vimentin and keratin 8 as well as increase of epithelial-to-mesenchymal polarization markers), whereas PaTu-8902 cells were rather inhibited. Of note, administration of the conditioned media to MIAPaCa-2 cells resulted in an inverse effect on the size and number of colonies, whereas CAPAN-2 cells were rather stimulated. To explain the heterogeneous pattern of the observed PDAC crosstalk at the in vitro level, we further compared the phenotype of primary cultures of cells derived from ascitic fluid with that of the tested PDAC cell lines, analyzed tumor samples of PDAC patients, and performed gene expression profiling of PANFs. Immuno-cyto/histo-chemical analysis found specific phenotype differences within the group of examined patients and tested PDAC cell lines, whereas the genomic approach in PANFs found the key molecules (IL6, IL8, MFGE8 and periostin) that may contribute to the cancer aggressive behavior. CONCLUSION The desmoplastic patient-specific regulation of cancer cells by CAFs (also demonstrated by the heterogeneous response of PDAC cell lines to fibroblasts) precludes simple targeting and development of an effective treatment strategy and rather requires establishment of an individualized tumor-specific treatment protocol.
Collapse
Affiliation(s)
- Štepán Novák
- Institute of Anatomy, First Faculty of Medicine, Charles University, Prague, Czech Republic.,Department of Otorhinolaryngology, Head and Neck Surgery, First Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Michal Kolář
- Laboratory of Genomics and Bioinformatics, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Arpád Szabó
- Department of Pathology, Third Faculty of Medicine, Charles University and University Hospital Královske Vinohrady, Prague, Czech Republic
| | - Zdena Vernerová
- Department of Pathology, Third Faculty of Medicine, Charles University and University Hospital Královske Vinohrady, Prague, Czech Republic
| | - Lukáš Lacina
- Institute of Anatomy, First Faculty of Medicine, Charles University, Prague, Czech Republic.,BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czech Republic.,Department of Dermatology and Venereology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Hynek Strnad
- Laboratory of Genomics and Bioinformatics, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Jana Šáchová
- Laboratory of Genomics and Bioinformatics, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Miluše Hradilová
- Laboratory of Genomics and Bioinformatics, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Jan Havránek
- Laboratory of Genomics and Bioinformatics, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic.,Department of Informatics and Chemistry, Faculty of Chemical Technology, University of Chemistry and Technology, Prague, Czech Republic
| | - Michal Španko
- Institute of Anatomy, First Faculty of Medicine, Charles University, Prague, Czech Republic.,Department of Stomatology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Matúš Čoma
- Department of Pharmacology, Pavol Jozef Šafárik University, Košice, Slovak Republic.,Department of Biomedical Research, East-Slovak Institute of Cardiovascular Diseases, Košice, Slovak Republic
| | - Lukáš Urban
- Department of Biomedical Research, East-Slovak Institute of Cardiovascular Diseases, Košice, Slovak Republic.,Laboratory of Cell Interactions, Center of Clinical and Preclinical Research MediPark, Pavol Jozef Šafárik University, Košice, Slovak Republic
| | - Miriam Kaňuchová
- Laboratory of Cell Interactions, Center of Clinical and Preclinical Research MediPark, Pavol Jozef Šafárik University, Košice, Slovak Republic
| | - Nikola Melegová
- Laboratory of Cell Interactions, Center of Clinical and Preclinical Research MediPark, Pavol Jozef Šafárik University, Košice, Slovak Republic
| | - Robert Gürlich
- Department of Surgery, Third Faculty of Medicine, Charles University and University Hospital Královske Vinohrady, Prague, Czech Republic
| | - Josef Dvořák
- Department of Oncology, First Faculty of Medicine, Charles University and Thomayer Hospital, Prague, Czech Republic
| | - Karel Smetana
- Institute of Anatomy, First Faculty of Medicine, Charles University, Prague, Czech Republic.,BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czech Republic
| | - Peter Gál
- Department of Biomedical Research, East-Slovak Institute of Cardiovascular Diseases, Košice, Slovak Republic; .,Laboratory of Cell Interactions, Center of Clinical and Preclinical Research MediPark, Pavol Jozef Šafárik University, Košice, Slovak Republic.,Prague Burn Centre, Third Faculty of Medicine, Charles University and University Hospital Královske Vinohrady, Prague, Czech Republic
| | - Pavol Szabo
- Institute of Anatomy, First Faculty of Medicine, Charles University, Prague, Czech Republic; .,BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czech Republic.,Department of Biomedical Research, East-Slovak Institute of Cardiovascular Diseases, Košice, Slovak Republic
| |
Collapse
|
32
|
Liot S, Balas J, Aubert A, Prigent L, Mercier-Gouy P, Verrier B, Bertolino P, Hennino A, Valcourt U, Lambert E. Stroma Involvement in Pancreatic Ductal Adenocarcinoma: An Overview Focusing on Extracellular Matrix Proteins. Front Immunol 2021; 12:612271. [PMID: 33889150 PMCID: PMC8056076 DOI: 10.3389/fimmu.2021.612271] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 02/23/2021] [Indexed: 12/11/2022] Open
Abstract
Pancreatic cancer is the seventh leading cause of cancer-related deaths worldwide and is predicted to become second in 2030 in industrialized countries if no therapeutic progress is made. Among the different types of pancreatic cancers, Pancreatic Ductal Adenocarcinoma (PDAC) is by far the most represented one with an occurrence of more than 90%. This specific cancer is a devastating malignancy with an extremely poor prognosis, as shown by the 5-years survival rate of 2–9%, ranking firmly last amongst all cancer sites in terms of prognostic outcomes for patients. Pancreatic tumors progress with few specific symptoms and are thus at an advanced stage at diagnosis in most patients. This malignancy is characterized by an extremely dense stroma deposition around lesions, accompanied by tissue hypovascularization and a profound immune suppression. Altogether, these combined features make access to cancer cells almost impossible for conventional chemotherapeutics and new immunotherapeutic agents, thus contributing to the fatal outcomes of the disease. Initially ignored, the Tumor MicroEnvironment (TME) is now the subject of intensive research related to PDAC treatment and could contain new therapeutic targets. In this review, we will summarize the current state of knowledge in the field by focusing on TME composition to understand how this specific compartment could influence tumor progression and resistance to therapies. Attention will be paid to Tenascin-C, a matrix glycoprotein commonly upregulated during cancer that participates to PDAC progression and thus contributes to poor prognosis.
Collapse
Affiliation(s)
- Sophie Liot
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| | - Jonathan Balas
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| | - Alexandre Aubert
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| | - Laura Prigent
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| | - Perrine Mercier-Gouy
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| | - Bernard Verrier
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| | - Philippe Bertolino
- Cancer Research Center of Lyon, UMR INSERM 1052, CNRS 5286, Lyon, France
| | - Ana Hennino
- Cancer Research Center of Lyon, UMR INSERM 1052, CNRS 5286, Lyon, France
| | - Ulrich Valcourt
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| | - Elise Lambert
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| |
Collapse
|
33
|
You X, Wang Y, Meng J, Han S, Liu L, Sun Y, Zhang J, Sun S, Li X, Sun W, Dong Y, Zhang Y. Exosomal miR‑663b exposed to TGF‑β1 promotes cervical cancer metastasis and epithelial‑mesenchymal transition by targeting MGAT3. Oncol Rep 2021; 45:12. [PMID: 33649791 PMCID: PMC7877003 DOI: 10.3892/or.2021.7963] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 01/18/2021] [Indexed: 12/24/2022] Open
Abstract
Transforming growth factor (TGF)‑β1 is a key cytokine affecting the pathogenesis and progression of cervical cancer. Tumor‑derived exosomes contain microRNAs (miRNAs/miRs) that interact with cancer and stromal cells, thereby contributing to tissue remodeling in the tumor microenvironment (TME). The present study was designed to clarify how TGF‑β1 affects tumor biological functions through exosomes released by cervical cancer cells. Deep RNA sequencing found that TGF‑β1 stimulated cervical cancer cells to secrete more miR‑663b‑containing exosomes, which could be transferred into new target cells to promote metastasis. Further studies have shown that miR‑663b directly targets the 3'-untranslated regions (3'‑UTR) of mannoside acetylglucosaminyltransferase 3 (MGAT3) and is involved in the epithelial‑mesenchymal transition (EMT) process. Remarkably, the overexpression of MGAT3 suppressed cervical cancer cell metastasis promoted by exosomal miR‑663b, causing increased expression of epithelial differentiation marker E‑cadherin and decreased expression of mesenchymal markers N‑cadherin and β‑catenin. Throughout our study, online bioinformation tools and dual luciferase reporter assay were applied to identify MGAT3 as a novel direct target of miR‑663b. Exosome PKH67‑labeling experiment verified that exosomal miR‑663b could be endocytosed by cervical cancer cells and subsequently influence its migration and invasion functions which were measured by wound healing and Transwell assays. The expression of miR‑663b and MGAT3 and the regulation of the EMT pathway caused by MGAT3 were detected by quantitative real‑time transcription‑polymerase chain reaction (qPCR) and western blot analysis. These results, thus, provide evidence that cancer cell‑derived exosomal miR‑663b is endocytosed by cervical cancer cells adjacent or distant after TGF‑β1 exposure and inhibits the expression of MGAT3, thereby accelerating the EMT process and ultimately promoting local and distant metastasis.
Collapse
Affiliation(s)
- Xuewu You
- Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Ying Wang
- Department of Obstetrics and Gynecology, Yidu Central Hospital of Weifang, Weifang, Shandong 262500, P.R. China
| | - Jinyu Meng
- Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Sai Han
- Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Lu Liu
- Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Yu Sun
- Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Junhua Zhang
- Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Shuqin Sun
- Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Xinyue Li
- Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Wenxiong Sun
- Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Yajie Dong
- Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Youzhong Zhang
- Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
34
|
Wang J, Xiang H, Lu Y, Wu T. Role and clinical significance of TGF‑β1 and TGF‑βR1 in malignant tumors (Review). Int J Mol Med 2021; 47:55. [PMID: 33604683 PMCID: PMC7895515 DOI: 10.3892/ijmm.2021.4888] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 01/27/2021] [Indexed: 12/24/2022] Open
Abstract
The appearance and growth of malignant tumors is a complicated process that is regulated by a number of genes. In recent years, studies have revealed that the transforming growth factor-β (TGF-β) signaling pathway serves an important role in cell cycle regulation, growth and development, differentiation, extracellular matrix synthesis and immune response. Notably, two members of the TGF-β signaling pathway, TGF-β1 and TGF-β receptor 1 (TGF-βR1), are highly expressed in a variety of tumors, such as breast cancer, colon cancer, gastric cancer and hepatocellular carcinoma. Moreover, an increasing number of studies have demonstrated that TGF-β1 and TGF-βR1 promote proliferation, migration and epithelial-mesenchymal transition of tumor cells by activating other signaling pathways, signaling molecules or microRNAs (miRs), such as the NF-κB signaling pathway and miR-133b. In addition, some inhibitors targeting TGF-β1 and TGF-βR1 have exhibited positive effects in in vitro experiments. The present review discusses the association between TGF-β1 or TGF-βR1 and tumors, and the development of some inhibitors, hoping to provide more approaches to help identify novel tumor markers to restrain and cure tumors.
Collapse
Affiliation(s)
- Junmin Wang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Hongjiao Xiang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Yifei Lu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Tao Wu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| |
Collapse
|
35
|
Dysregulation of Transcription Factor Activity During Formation of Cancer-Associated Fibroblasts. Int J Mol Sci 2020; 21:ijms21228749. [PMID: 33228208 PMCID: PMC7699520 DOI: 10.3390/ijms21228749] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/08/2020] [Accepted: 11/17/2020] [Indexed: 01/22/2023] Open
Abstract
The reciprocal interactions between cancer cells and the quiescent fibroblasts leading to the activation of cancer-associated fibroblasts (CAFs) serve an important role in cancer progression. Here, we investigated the activation of transcription factors (TFs) in prostate fibroblasts (WPMY cell line) co-cultured with normal prostate or tumorous cells (RWPE1 and RWPE2 cell lines, respectively). After indirect co-cultures, we performed mRNA-seq and predicted TF activity using mRNA expression profiles with the Systems EPigenomics Inference of Regulatory Activity (SEPIRA) package and the GTEx and mRNA-seq data of 483 cultured fibroblasts. The initial differential expression analysis between time points and experimental conditions showed that co-culture with normal epithelial cells mainly promotes an inflammatory response in fibroblasts, whereas with the cancerous epithelial, it stimulates transformation by changing the expression of the genes associated with microfilaments. TF activity analysis revealed only one positively regulated TF in the RWPE1 co-culture alone, while we observed dysregulation of 45 TFs (7 decreased activity and 38 increased activity) uniquely in co-culture with RWPE2. Pathway analysis showed that these 45 dysregulated TFs in fibroblasts co-cultured with RWPE2 cells may be associated with the RUNX1 and PTEN pathways. Moreover, we showed that observed dysregulation could be associated with FER1L4 expression. We conclude that phenotypic changes in fibroblast responses to co-culturing with cancer epithelium result from orchestrated dysregulation of signaling pathways that favor their transformation and motility rather than proinflammatory status. This dysregulation can be observed both at the TF and transcriptome levels.
Collapse
|
36
|
Xie J, Zhu H, Chang HM, Klausen C, Dong M, Leung PCK. GDF8 Promotes the Cell Invasiveness in Human Trophoblasts by Upregulating the Expression of Follistatin-Like 3 Through the ALK5-SMAD2/3 Signaling Pathway. Front Cell Dev Biol 2020; 8:573781. [PMID: 33195207 PMCID: PMC7655915 DOI: 10.3389/fcell.2020.573781] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 09/28/2020] [Indexed: 11/30/2022] Open
Abstract
Growth differentiation factor 8 (GDF8) and its antagonist follistatin-like 3 (FSTL3) are expressed in the placenta during early pregnancy. These two factors may have a role to play in the regulation of normal placentation. However, whether GDF8 can regulate the expression of FSTL3 in human trophoblasts remains to be elucidated. In this study, we aimed to investigate the effects of GDF8 on the expression of FSTL3 and the underlying molecular mechanisms using human trophoblasts as a study model. Our results showed that GDF8 significantly upregulates the expression and production of FSTL3, which further promotes cell invasiveness in immortalized extravillous cytotrophoblast cells and primary extravillous cytotrophoblast cells obtained from human first-trimester placentae. Additionally, using an siRNA-mediated knockdown approach, we found that this regulatory effect is most likely mediated by the ALK5-Sma- and Mad-related protein (SMAD)2/3-induced signaling pathway. These findings deepen our understanding of the functional roles of GDF8 and FSTL3 in the regulation of cell invasiveness of trophoblasts.
Collapse
Affiliation(s)
- Jiamin Xie
- Department of Obstetrics and Gynaecology, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Hua Zhu
- Department of Obstetrics and Gynaecology, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Hsun-Ming Chang
- Department of Obstetrics and Gynaecology, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Christian Klausen
- Department of Obstetrics and Gynaecology, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Minyue Dong
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Women's Reproductive Health of Zhejiang Province, Hangzhou, China.,Key Laboratory of Reproductive Genetics, Ministry of Education, Hangzhou, China
| | - Peter C K Leung
- Department of Obstetrics and Gynaecology, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
37
|
Ismail MMF, El-Zahabi HSA, Ibrahim RS, Mehany ABM. Design and synthesis of novel tranilast analogs: Docking, antiproliferative evaluation and in-silico screening of TGFβR1 inhibitors. Bioorg Chem 2020; 105:104368. [PMID: 33091671 DOI: 10.1016/j.bioorg.2020.104368] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 09/22/2020] [Accepted: 10/08/2020] [Indexed: 11/17/2022]
Abstract
The discovery of the antiproliferative potential of tranilast prompted additional studies directed at understanding the mechanisms of tranilast action. Its inhibitory effect on cell proliferation depends principally on the capacity of tranilast to interfere with transforming growth factor beta (TGFβR1) signaling. This work summarizes design, synthesis and biological evaluation of sixteen novel tranilast analogs on different tumors such as PC-3, HepG-2 and MCF-7 cell lines. The in vitro cytotoxicity was evaluated using MTT assay showed that, twelve compounds out of sixteen showed higher cytotoxic activities (IC50's 1.1-6.29 µM), than that of the reference standard, 5-FU (IC50 7.53 µM). The promising cytotoxic hits (4b, 7a, b and 14c-e), proved to be selective to cancer cells when their cytotoxicity's are examined on human normal cell line (WI-38). Then they are investigated for their possible mode of action as TGFβR1 inhibitors; remarkable inhibition of TGFβR1 by these hits was observed at the range of IC50 0.087-3.276 μM. The cell cycle analysis of the most potent TGFβR1 inhibitor, 4b revealed cell cycle arrest at G2/M phase on prostate cancer cells. Additionally, it is clearly indicated apoptosis induction at Pre-G1 phase, this is substantiated by significant increase in the expression on the tumor suppressor gene, p53 and up regulation the level of apoptosis mediator, caspase-3. In addition, in silico study was performed for validating the physicochemical and ADME properties which revealed that, all compounds are orally bioavailable with no side effects complying with Lipinski rule. The proposed mode of action can be further explored on the light of molecular modeling simulation of the most potent compounds, 4b and 14e which were docked into the active sites of TGFβR1 to predict their affinities toward the receptor.
Collapse
Affiliation(s)
- Magda M F Ismail
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy (Girls), Al-Azhar University, 11754 Nasr City, Cairo, Egypt.
| | - Heba S A El-Zahabi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy (Girls), Al-Azhar University, 11754 Nasr City, Cairo, Egypt
| | - Rabab S Ibrahim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy (Girls), Al-Azhar University, 11754 Nasr City, Cairo, Egypt
| | - Ahmed B M Mehany
- Department of Zoology, Faculty of Science (Boys), Al-Azhar University, 11754 Nasr City, Cairo, Egypt
| |
Collapse
|
38
|
Zhang B, Shang L, Zhang Y, Li T, Fang Y. The effect of bone marrow mesenchymal stem cells on highly metastatic MHCC97-H hepatocellular carcinoma cells following OPN and TGFβ1 gene silencing. Exp Ther Med 2020; 20:3633-3642. [PMID: 32855715 PMCID: PMC7444374 DOI: 10.3892/etm.2020.9106] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 06/10/2020] [Indexed: 12/12/2022] Open
Abstract
The metastatic behavior of hepatocellular carcinoma (HCC) is one of the key factors that leads to poor prognosis. The aim of the current study was to determine the changes in metastasis and the proliferation potential of bone marrow mesenchymal stem cells (BMSCs) in high metastatic potential hepatocellular carcinoma (MHCC97-H) following gene silencing. The osteopontin (OPN) and transforming growth factor-β (TGFβ1 ) genes, which are associated with metastasis and tumor proliferation, were silenced in MHCC97-H cells. Transwell assays were used to evaluate the migration of MHCC97-H cells in vitro. Additionally, a murine model of MHCC97-H lung metastasis was established. Following OPN and TGFβ1 silencing, the migration of MHCC97-H cells was significantly reduced following BMSC intervention (P<0.01). Furthermore, there were few MHCC97-H cells in the lung tissues of the OPN- and TGFβ1 -silenced animals, and their integrated optical density (IOD) value was significantly lower compared with controls (P<0.05). Immunofluorescence of lung metastasis in the MHCC97-H model revealed that there was no significant difference in the IOD value of integrin αvβ3 expression in the OPN- and TGFβ1 -silenced groups compared with controls (P>0.05). The metastasis and proliferation potential of MHCC97-H following BMSC intervention were significantly reduced in vitro and in vivo, especially in the TGFβ1-silenced group. The decrease in the metastatic potential in gene-silenced MHCC97-H cells was not associated with integrin αvβ3 expression. Therefore, OPN and TGFβ1 may be potential targets for HCC treatment, and TGFβ1 may have a higher therapeutic potential for BMSC intervention.
Collapse
Affiliation(s)
- Beibei Zhang
- Department of Radiology, the Fourth Medical Center of Chinese PLA General Hospital, Beijing 100048, P.R. China
| | - Liutong Shang
- Department of Radiology, the Fourth Medical Center of Chinese PLA General Hospital, Beijing 100048, P.R. China
| | - Yi Zhang
- Department of Radiology, the Fourth Medical Center of Chinese PLA General Hospital, Beijing 100048, P.R. China
| | - Tianran Li
- Department of Radiology, the Fourth Medical Center of Chinese PLA General Hospital, Beijing 100048, P.R. China
| | - Yuan Fang
- Department of Radiology, the Fourth Medical Center of Chinese PLA General Hospital, Beijing 100048, P.R. China
| |
Collapse
|
39
|
Hegde S. Pancreatic Cancer Immuno-oncology in the Era of Precision Medicine. Indian J Surg Oncol 2020; 12:118-127. [PMID: 33994737 DOI: 10.1007/s13193-020-01192-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 08/19/2020] [Indexed: 10/23/2022] Open
Abstract
Pancreatic malignancies carry a dismal prognosis globally, with pancreatic adenocarcinomas (PDAC) being particularly aggressive and stubborn. Unfortunately, several therapeutic strategies that show promise in other cancers have failed to make sizeable impact on pancreatic tumor outcomes. Responses to immunotherapies are especially rare in pancreatic cancer, and patients are in need of innovative approaches that can result in more durable responses. Current research in preclinical models and humans has suggested this resistance is due to a uniquely inflammatory and dysfunctional tumor microenvironment; these findings lay the groundwork for targeting these barriers and improving outcomes. Clinical analyses have also revealed unprecedented heterogeneity in tumor and stromal biology of PDAC, underscoring the need for more personalized approaches and combinatorial therapies. This review will highlight the current state of translational research focusing on PDAC immunity, summarize ongoing clinical efforts to tackle PDAC vulnerabilities, and underscore some unresolved challenges in implementing therapies more broadly. A better understanding of immune contexture and tumor heterogeneity in this disease will greatly accelerate drug discovery and implementation of precision medicine for PDAC.
Collapse
Affiliation(s)
- Samarth Hegde
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY USA
| |
Collapse
|
40
|
Gerner MC, Ziegler LS, Schmidt RLJ, Krenn M, Zimprich F, Uyanik‐Ünal K, Konstantopoulou V, Derdak S, Del Favero G, Schwarzinger I, Boztug K, Schmetterer KG. The TGF-b/SOX4 axis and ROS-driven autophagy co-mediate CD39 expression in regulatory T-cells. FASEB J 2020; 34:8367-8384. [PMID: 32319705 PMCID: PMC7317981 DOI: 10.1096/fj.201902664] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 04/10/2020] [Indexed: 12/16/2022]
Abstract
The ectonucleotidase CD39 on human regulatory T-cells (Treg) is an important immune regulator which is dysregulated in autoimmune diseases and cancer immunosuppression. We here define that CD39 expression on Treg is independent of the Treg-specific transcription factors FOXP3 and HELIOS and promoted by canonical TGF-b- and mTOR-signaling. Furthermore, the TGF-b mediated upregulation of CD39 is counteracted by reactive oxygen species (ROS)-driven autophagy. In line, CD39+ peripheral blood Treg constitute a distinct lineage with low autophagic flux and absent ROS production. Patients with rare genetic defects in autophagy show supraphysiological levels of CD39+ Treg, validating our observations in vivo. These biological processes rely on a distinct transcriptional program with CD39+ Treg expressing low levels of two genes with putative involvement in autophagy, NEFL and PLAC8. Furthermore, the TGF-b downstream transcription factor SOX4 is selectively upregulated in CD39+ Treg. Overexpression of SOX4 in Treg strongly increases CD39 expression, while Crispr/Cas9-mediated knockout of SOX4 in Treg has the opposing effect. Thus, we identify a crucial role of SOX4 in immune regulation and provide new insights involving the interplay of tolerogenic cues and autophagy in Treg.
Collapse
Affiliation(s)
- Marlene C. Gerner
- Department of Laboratory MedicineMedical University of ViennaViennaAustria
| | - Liesa S. Ziegler
- Department of Laboratory MedicineMedical University of ViennaViennaAustria
| | - Ralf L. J. Schmidt
- Department of Laboratory MedicineMedical University of ViennaViennaAustria
| | - Martin Krenn
- Department of NeurologyMedical University of ViennaViennaAustria
| | - Fritz Zimprich
- Department of NeurologyMedical University of ViennaViennaAustria
| | | | | | - Sophia Derdak
- Core Facility GenomicsMedical University of ViennaViennaAustria
| | - Giorgia Del Favero
- Department of Food Chemistry and ToxicologyFaculty of ChemistryUniversity of ViennaViennaAustria
| | - Ilse Schwarzinger
- Department of Laboratory MedicineMedical University of ViennaViennaAustria
| | - Kaan Boztug
- Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
| | | |
Collapse
|
41
|
Fang WY, Kuo YZ, Chang JY, Hsiao JR, Kao HY, Tsai ST, Wu LW. The Tumor Suppressor TGFBR3 Blocks Lymph Node Metastasis in Head and Neck Cancer. Cancers (Basel) 2020; 12:cancers12061375. [PMID: 32471132 PMCID: PMC7352722 DOI: 10.3390/cancers12061375] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 05/23/2020] [Accepted: 05/25/2020] [Indexed: 12/11/2022] Open
Abstract
The TGF-β type III receptor (TGFBR3) is an essential constituent of the TGF-β signaling. In this study, we observed a down-regulation of TGFBR3 in oral cancer, a subtype of head and neck cancer (HNC), and patients with low TGFBR3 had poor clinical outcomes. Ectopic expression of TGFBR3 decreased migration and invasion of oral cancer cells and lymph node metastasis of tumors, whereas depletion of TGFBR3 had the opposite effect. In SMAD4-positive OC-2 oral cancer cells, TGFBR3-mediated suppression requires both of its cytoplasmic interacting partners ARRB2 and GIPC1. We demonstrated that TGFBR3 induces the abundance of secreted angiogenin (ANG), a known pro-angiogenic factor, and ANG is essential and sufficient to mediate TGFBR3-dependent inhibition of migration and invasion of oral cancer cells. Notably, in SMAD4-deficient CAL-27 oral cancer cells, only GIPC1 is essential for TGFBR3-induced suppressive activity. Accordingly, HNC patients with low expressions of both TGFBR3 and GIPC1 had the poorest overall survival. In summary, we conclude that TGFBR3 is as a tumor suppressor via SMAD4-dependent and -independent manner in both tumor and stromal cells during oral carcinogenesis. Our study should facilitate the possibility of using TGFBR3-mediated tumor suppression for HNC treatment.
Collapse
Affiliation(s)
- Wei-Yu Fang
- Institutes of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan;
| | - Yi-Zih Kuo
- Department of Otolaryngology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan; (Y.-Z.K.); (J.-R.H.)
| | - Jang-Yang Chang
- National Institute of Cancer Research, National Health Research Institutes, Tainan 70456, Taiwan;
- Division of Hematology/Oncology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Jenn-Ren Hsiao
- Department of Otolaryngology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan; (Y.-Z.K.); (J.-R.H.)
| | - Hung-Ying Kao
- Department of Biochemistry, School of Medicine, Case Western Reserve University, Cleveland, OH 43210, USA;
| | - Sen-Tien Tsai
- Department of Otolaryngology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan; (Y.-Z.K.); (J.-R.H.)
- Correspondence: (S.-T.T.); (L.-W.W.); Tel.: +886-6-2353535 (ext. 5315) (S.-T.T.); +886-6-2353535 (ext. 3618) (L.-W.W.); Fax: +886-6-2095845 (L.-W.W.)
| | - Li-Wha Wu
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
- Department of Laboratory Science and Technology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Correspondence: (S.-T.T.); (L.-W.W.); Tel.: +886-6-2353535 (ext. 5315) (S.-T.T.); +886-6-2353535 (ext. 3618) (L.-W.W.); Fax: +886-6-2095845 (L.-W.W.)
| |
Collapse
|
42
|
Xu Z, Pang TCY, Liu AC, Pothula SP, Mekapogu AR, Perera CJ, Murakami T, Goldstein D, Pirola RC, Wilson JS, Apte MV. Targeting the HGF/c-MET pathway in advanced pancreatic cancer: a key element of treatment that limits primary tumour growth and eliminates metastasis. Br J Cancer 2020; 122:1486-1495. [PMID: 32203220 PMCID: PMC7217847 DOI: 10.1038/s41416-020-0782-1] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 02/04/2020] [Accepted: 02/18/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Stromal-tumour interactions facilitate pancreatic cancer (PC) progression. The hepatocyte growth factor (HGF)/c-MET pathway is upregulated in PC and mediates the interaction between cancer cells and stromal pancreatic stellate cells (PSCs). This study assessed the effect of HGF/c-MET inhibition plus gemcitabine (G) on the progression of advanced PC. METHODS Orthotopic PC was produced by implantation of luciferase-tagged human cancer cells + human PSCs into mouse pancreas. Tumours were allowed to develop without treatment for 4 weeks. Mice were then treated for 6 weeks with one of the following: IgG, G, HGF inhibitor (Hi), c-MET inhibitor (Ci), Hi + Ci, Hi + G, Ci + G, or Hi + Ci + G. RESULTS Bioluminescence imaging showed similar tumour sizes in all mice at the initiation of treatments. Triple therapy (Hi + Ci + G): (1) completely eliminated metastasis; (2) significantly reduced tumour size as assessed by bioluminescence and at necropsy; (3) significantly reduced proliferating cancer cell density and stem cell marker DCLK1 expression in tumours. In vitro 3D culture studies supported our in vivo findings. CONCLUSION Even at an advanced disease stage, a two-pronged approach, targeting (a) HGF/c-MET with relevant inhibitors and (b) cancer cells with chemotherapy, completely eliminated metastasis and significantly decreased tumour growth, suggesting that this is a promising treatment approach for PC.
Collapse
Affiliation(s)
- Zhihong Xu
- Pancreatic Research Group, South Western Sydney Clinical School, Faculty of Medicine, The University of New South Wales, Sydney, NSW, Australia
- Ingham Institute for Applied Medical Research, Sydney, NSW, Australia
| | - Tony C Y Pang
- Pancreatic Research Group, South Western Sydney Clinical School, Faculty of Medicine, The University of New South Wales, Sydney, NSW, Australia
- Ingham Institute for Applied Medical Research, Sydney, NSW, Australia
| | - Adele C Liu
- Pancreatic Research Group, South Western Sydney Clinical School, Faculty of Medicine, The University of New South Wales, Sydney, NSW, Australia
- Ingham Institute for Applied Medical Research, Sydney, NSW, Australia
| | - Srinivasa P Pothula
- Pancreatic Research Group, South Western Sydney Clinical School, Faculty of Medicine, The University of New South Wales, Sydney, NSW, Australia
- Ingham Institute for Applied Medical Research, Sydney, NSW, Australia
| | - Alpha Raj Mekapogu
- Pancreatic Research Group, South Western Sydney Clinical School, Faculty of Medicine, The University of New South Wales, Sydney, NSW, Australia
- Ingham Institute for Applied Medical Research, Sydney, NSW, Australia
| | - Chamini J Perera
- Pancreatic Research Group, South Western Sydney Clinical School, Faculty of Medicine, The University of New South Wales, Sydney, NSW, Australia
- Ingham Institute for Applied Medical Research, Sydney, NSW, Australia
| | | | - David Goldstein
- Pancreatic Research Group, South Western Sydney Clinical School, Faculty of Medicine, The University of New South Wales, Sydney, NSW, Australia
| | - Romano C Pirola
- Pancreatic Research Group, South Western Sydney Clinical School, Faculty of Medicine, The University of New South Wales, Sydney, NSW, Australia
| | - Jeremy S Wilson
- Pancreatic Research Group, South Western Sydney Clinical School, Faculty of Medicine, The University of New South Wales, Sydney, NSW, Australia
- Ingham Institute for Applied Medical Research, Sydney, NSW, Australia
| | - Minoti V Apte
- Pancreatic Research Group, South Western Sydney Clinical School, Faculty of Medicine, The University of New South Wales, Sydney, NSW, Australia.
- Ingham Institute for Applied Medical Research, Sydney, NSW, Australia.
| |
Collapse
|
43
|
Larson C, Oronsky B, Carter CA, Oronsky A, Knox SJ, Sher D, Reid TR. TGF-beta: a master immune regulator. Expert Opin Ther Targets 2020; 24:427-438. [PMID: 32228232 DOI: 10.1080/14728222.2020.1744568] [Citation(s) in RCA: 144] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: Transforming Growth Factor-Beta (TGF-β) is a master regulator of numerous cellular functions including cellular immunity. In cancer, TGF-β can function as a tumor promoter via several mechanisms including immunosuppression. Since the immune checkpoint pathways are co-opted in cancer to induce T cell tolerance, this review posits that TGF-β is a master checkpoint in cancer, whose negative regulatory influence overrides and controls that of other immune checkpoints.Areas Covered: This review examines therapeutic agents that target TGF-β and its signaling pathways for the treatment of cancer which may be classifiable as checkpoint inhibitors in the broadest sense. This concept is supported by the observations that 1) only a subset of patients benefit from current checkpoint inhibitor therapies, 2) the presence of TGF-β in the tumor microenvironment is associated with excluded or cold tumors, and resistance to checkpoint inhibitors, and 3) existing biomarkers such as PD-1, PD-L1, microsatellite instability and tumor mutational burden are inadequate to reliably and adequately identify immuno-responsive patients. By contrast, TGF-β overexpression is a widespread and profoundly negative molecular hallmark in multiple tumor types.Expert Opinion: TGF-β status may serve as a biomarker to predict responsiveness and as a therapeutic target to increase the activity of immunotherapies.
Collapse
Affiliation(s)
| | | | | | - Arnold Oronsky
- EpicentRx, San Diego, CA, USA.,InterWest Partners, Menlo Park, CA, USA
| | - Susan J Knox
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - David Sher
- Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Tony R Reid
- Department of Medical Oncology, UC San Diego School of Medicine, La Jolla, CA, USA
| |
Collapse
|
44
|
Conedera FM, Quintela Pousa AM, Presby DM, Mercader N, Enzmann V, Tschopp M. Diverse Signaling by TGFβ Isoforms in Response to Focal Injury is Associated with Either Retinal Regeneration or Reactive Gliosis. Cell Mol Neurobiol 2020; 41:43-62. [PMID: 32219603 PMCID: PMC7811507 DOI: 10.1007/s10571-020-00830-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 03/11/2020] [Indexed: 12/13/2022]
Abstract
Müller cells may have stem cell-like capability as they regenerate photoreceptor loss upon injury in some vertebrates, but not in mammals. Indeed, mammalian Müller cells undergo major cellular and molecular changes summarized as reactive gliosis. Transforming growth factor beta (TGFβ) isoforms are multifunctional cytokines that play a central role, both in wound healing and in tissue repair. Here, we studied the role of TGFβ isoforms and their signaling pathways in response to injury induction during tissue regeneration in zebrafish and scar formation in mouse. Our transcriptome analysis showed a different activation of canonical and non-canonical signaling pathways and how they shaped the injury response. In particular, TGFβ3 promotes retinal regeneration via Smad-dependent canonical pathway upon regulation of junb gene family and mycb in zebrafish Müller cells. However, in mice, TGFβ1 and TGFβ2 evoke the p38MAPK signaling pathway. The activation of this non-canonical pathway leads to retinal gliosis. Thus, the regenerative versus reparative effect of the TGFβ pathway observed may rely on the activation of different signaling cascades. This provides one explanation of the different injury response in zebrafish and mouse retina.
Collapse
Affiliation(s)
- Federica Maria Conedera
- Department of Ophthalmology, University Hospital of Bern, University of Bern, Bern, Switzerland.,Department of BioMedical Research, University of Bern, Bern, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Ana Maria Quintela Pousa
- Department of Ophthalmology, University Hospital of Bern, University of Bern, Bern, Switzerland.,Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - David Mikal Presby
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Nadia Mercader
- Institute of Anatomy, University of Bern, Bern, Switzerland
| | - Volker Enzmann
- Department of Ophthalmology, University Hospital of Bern, University of Bern, Bern, Switzerland. .,Department of BioMedical Research, University of Bern, Bern, Switzerland.
| | - Markus Tschopp
- Department of Ophthalmology, University Hospital of Bern, University of Bern, Bern, Switzerland. .,Department of Ophthalmology, Cantonal Hospital Aarau, Aarau, Switzerland.
| |
Collapse
|
45
|
Weidle UH, Birzele F, Nopora A. Pancreatic Ductal Adenocarcinoma: MicroRNAs Affecting Tumor Growth and Metastasis in Preclinical In Vivo Models. Cancer Genomics Proteomics 2020; 16:451-464. [PMID: 31659100 DOI: 10.21873/cgp.20149] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 07/30/2019] [Accepted: 08/01/2019] [Indexed: 02/08/2023] Open
Abstract
Patients with pancreatic ductal adenocarcinoma have a dismall prognosis because at the time of diagnosis, in the vast majority of patients the tumor has already disseminated to distant organs and the therapeutic benefit of approved agents such as gemcitabine is limited. Therefore, the identification and preclinical and clinical validation of therapeutic agents covering new targets is of paramount importance. In this review we have summarized microRNAs and corresponding targets which affect growth and metastasis of pancreatic tumors in preclinical mouse in vivo models. We identified four up-regulated and 16 down-regulated miRs in PDAC in comparison to corresponding normal tissues. Three sub-categories of miRs have emerged: miRs affecting tumor growth and miRs with an impact on both, tumor growth and metastasis or metastasis only. Finally, we discuss technical and therapeutic aspects of miR-related therapeutic agents for the treatment of pancreatic ductal adenocarcinoma.
Collapse
Affiliation(s)
- Ulrich H Weidle
- Roche Innovation Center Munich, Roche Diagnostics GmbH, Penzberg, Germany
| | - Fabian Birzele
- Roche Innovation Center Basel, F. Hofman La Roche, Basel, Switzerland
| | - Adam Nopora
- Roche Innovation Center Munich, Roche Diagnostics GmbH, Penzberg, Germany
| |
Collapse
|
46
|
Wang M, Ye Q, Mao D, Li H. Research Progress in Liver-Regenerating Microenvironment and DNA Methylation in Hepatocellular Carcinoma: The Role of Traditional Chinese Medicine. Med Sci Monit 2020; 26:e920310. [PMID: 32144233 PMCID: PMC7077739 DOI: 10.12659/msm.920310] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 12/10/2019] [Indexed: 12/11/2022] Open
Abstract
The development, progression, recurrence, and metastasis of hepatocellular carcinoma (HCC) are closely associated with an abnormal liver-regenerating microenvironment (LRM). Therefore, preventing and reversing an abnormal LRM is a potential therapeutic strategy against HCC. Studies are increasingly focusing on the impact of regeneration, fibrosis, angiogenesis, inflammation, immunomodulation, and hepatic stem cells on HCC development and progression. As a key epigenetic mechanism, DNA methylation is extensively involved in regulating physiological and pathological pathways. In this review, we summarize recent findings on the role of DNA methylation in the fibrotic, angiogenic, inflammatory/immune, and stem cell microenvironments of HCC, and discuss new advances in Traditional Chinese Medicine (TCM) on influencing the abnormal LRM, so as to gain new insights into alleviating the abnormal LRM via regulating DNA methylation by TCM.
Collapse
Affiliation(s)
- Minggang Wang
- Hubei University of Traditional Chinese Medicine, Wuhan, Hubei, P.R. China
| | - Qianling Ye
- Guangxi University of Traditional Chinese Medicine, Nanning, Guangxi, P.R. China
| | - Dewen Mao
- The First Affiliated Hospital of Guangxi University of Traditional Chinese Medicine, Nanning, Guangxi, P.R. China
| | - Hanmin Li
- Hubei University of Traditional Chinese Medicine, Wuhan, Hubei, P.R. China
| |
Collapse
|
47
|
Mardhian DF, Vrynas A, Storm G, Bansal R, Prakash J. FGF2 engineered SPIONs attenuate tumor stroma and potentiate the effect of chemotherapy in 3D heterospheroidal model of pancreatic tumor. Nanotheranostics 2020; 4:26-39. [PMID: 31911892 PMCID: PMC6940204 DOI: 10.7150/ntno.38092] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 11/22/2019] [Indexed: 12/12/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC), characterized with abundant tumor stroma, is a highly malignant tumor with poor prognosis. The tumor stroma largely consists of cancer-associated fibroblasts (CAFs) and extracellular matrix (ECM), and is known to promote tumor growth and progression as well as acts as a barrier to chemotherapy. Inhibition of tumor stroma is highly crucial to induce the effect of chemotherapy. In this study, we delivered fibroblast growth factor 2 (FGF2) to human pancreatic stellate cells (hPSCs), the precursors of CAFs, using superparamagnetic iron oxide nanoparticles (SPIONs). FGF2 was covalently conjugated to functionalized PEGylated dextran-coated SPIONs. FGF2-SPIONs significantly reduced TGF-β induced hPSCs differentiation (α-SMA and collagen-1 expression) by inhibiting pSmad2/3 signaling and inducing ERK1/2 activity, as shown with western blot analysis. Then, we established a stroma-rich self-assembling 3D heterospheroid model by co-culturing PANC-1 and hPSCs in 3D environment. We found that FGF2-SPIONs treatment alone inhibited the tumor stroma-induced spheroid growth. In addition, they also potentiated the effect of gemcitabine, as shown by measuring the spheroid size and ATP content. These effects were attributed to the reduced expression of the hPSC activation and differentiation marker, α-SMA. Furthermore, to demonstrate an application of SPIONs, we applied an external magnetic field to spheroids while incubated with FGF2-SPIONs. This resulted in an enhanced effect of gemcitabine in our 3D model. In conclusion, this study presents a novel approach to target FGF2 to tumor stroma using SPIONs and thereby enhancing the effect of gemcitabine as demonstrated in the complex 3D tumor spheroid model.
Collapse
Affiliation(s)
- Deby Fajar Mardhian
- Targeted Therapeutics, Department of Biomaterials Science and Technology, Technical Medical Centre, Faculty of Science and technology, University of Twente, Enschede, The Netherlands
| | - Aggelos Vrynas
- Targeted Therapeutics, Department of Biomaterials Science and Technology, Technical Medical Centre, Faculty of Science and technology, University of Twente, Enschede, The Netherlands
| | - Gert Storm
- Targeted Therapeutics, Department of Biomaterials Science and Technology, Technical Medical Centre, Faculty of Science and technology, University of Twente, Enschede, The Netherlands
| | - Ruchi Bansal
- Targeted Therapeutics, Department of Biomaterials Science and Technology, Technical Medical Centre, Faculty of Science and technology, University of Twente, Enschede, The Netherlands
| | - Jai Prakash
- Targeted Therapeutics, Department of Biomaterials Science and Technology, Technical Medical Centre, Faculty of Science and technology, University of Twente, Enschede, The Netherlands
| |
Collapse
|
48
|
The role of contextual signal TGF-β1 inducer of epithelial mesenchymal transition in metastatic lung adenocarcinoma patients with brain metastases: an update on its pathological significance and therapeutic potential. Contemp Oncol (Pozn) 2019; 23:187-194. [PMID: 31992949 PMCID: PMC6978756 DOI: 10.5114/wo.2019.91543] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 11/04/2019] [Indexed: 02/07/2023] Open
Abstract
Lung adenocarcinoma (LA) is the most common cause of cancer-related death worldwide. Despite the advances over last decade in new targeted therapies, cancer genetics, diagnostics, staging, and surgical techniques as well as new chemotherapy and radiotherapy protocols, the death rate from LA remains high. The tumour microenvironment is composed of several cytokines, one of which is transforming growth factor β1 (TGF-β1), which modulates and mediates the expression of epithelial-mesenchymal transition (EMT), correlated with invasive growth in LAs, and exhibits its pleiotropic effects through binding to transmembrane receptors TβR-1 (also termed activin receptor-like kinases – ALKs) and TβR-2. Accordingly, there is an urgent need to elucidate the molecular mechanisms associated with the tumoural spreading process and therapeutic resistance of this serious pathology. In this review, we briefly discuss the current role of contextual signal TGF-β1 inducer of epithelial mesenchymal transition in metastatic lung adenocarcinoma patients with brain metastases, and give an overview of our current mechanistic understanding of the TGF-β1-related pathways in brain metastases progression, TGF-β1 pathway inhibitors that could be used for clinical treatment, and examination of models used to study these processes. Finally, we summarise the current progress in the therapeutic approaches targeting TGF-β1.
Collapse
|
49
|
Kang JH, Jung MY, Choudhury M, Leof EB. Transforming growth factor beta induces fibroblasts to express and release the immunomodulatory protein PD-L1 into extracellular vesicles. FASEB J 2019; 34:2213-2226. [PMID: 31907984 DOI: 10.1096/fj.201902354r] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 11/15/2019] [Accepted: 11/20/2019] [Indexed: 01/14/2023]
Abstract
Transforming growth factor-beta (TGFβ) is an enigmatic protein with various roles in healthy tissue homeostasis/development as well as the development or progression of cancer, wound healing, fibrotic disorders, and immune modulation, to name a few. As TGFβ is causal to various fibroproliferative disorders featuring localized or systemic tissue/organ fibrosis as well as the activated stroma observed in various malignancies, characterizing the pathways and players mediating its action is fundamental. In the current study, we found that TGFβ induces the expression of the immunoinhibitory molecule Programed death-ligand 1 (PD-L1) in human and murine fibroblasts in a Smad2/3- and YAP/TAZ-dependent manner. Furthermore, PD-L1 knockdown decreased the TGFβ-dependent induction of extracellular matrix proteins, including collagen Iα1 (colIα1) and alpha-smooth muscle actin (α-SMA), and cell migration/wound healing. In addition to an endogenous role for PD-L1 in profibrotic TGFβ signaling, TGFβ stimulated-human lung fibroblast-derived PD-L1 into extracellular vesicles (EVs) capable of inhibiting T cell proliferation in response to T cell receptor stimulation and mediating fibroblast cell migration. These findings provide new insights and potential targets for a variety of fibrotic and malignant diseases.
Collapse
Affiliation(s)
- Jeong-Han Kang
- Thoracic Diseases Research Unit, Division of Pulmonary and Critical Care Medicine, Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Mi-Yeon Jung
- Thoracic Diseases Research Unit, Division of Pulmonary and Critical Care Medicine, Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Malay Choudhury
- Thoracic Diseases Research Unit, Division of Pulmonary and Critical Care Medicine, Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Edward B Leof
- Thoracic Diseases Research Unit, Division of Pulmonary and Critical Care Medicine, Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN, USA
| |
Collapse
|
50
|
Li Y, Zhao Z, Liu H, Fetse JP, Jain A, Lin CY, Cheng K. Development of a Tumor-Responsive Nanopolyplex Targeting Pancreatic Cancer Cells and Stroma. ACS APPLIED MATERIALS & INTERFACES 2019; 11:45390-45403. [PMID: 31769963 PMCID: PMC7372733 DOI: 10.1021/acsami.9b15116] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Desmoplasia plays a pivotal role in promoting pancreatic cancer progression and is associated with poor clinical outcome. Targeting the desmoplastic tumor microenvironment in combination with chemotherapy is therefore a promising strategy for pancreatic cancer therapy. Here, we report a novel biodegradable copolymer to codeliver LY2109761 (a TGF-β receptor I/II inhibitor) and CPI-613 (a novel chemotherapy agent) to desmoplastic stroma and tumor cells, respectively, in the tumor microenvironment. Hydrophobic CPI-613 is conjugated to the hydrophilic copolymer via a newly designed MMP-2-responsive linker to form a trigger-responsive nanopolyplex. LY2109761 is hydrophobic and encapsulated into the hydrophobic core of the nanopolyplex. The resulting nanopolyplex is modified with a plectin-1-targeting peptide to enhance the accumulation of the nanopolyplex in pancreatic tumors. The nanopolyplex aims to normalize the stroma by blocking the interaction between tumor cells and pancreatic stellate cells to inhibit the activation of pancreatic stellate cells and subsequently reduce the dense extracellular matrix. Normalized stroma increases the penetration of the nanopolyplex into the tumor. The nanopolyplex shows enhanced accumulation in xenograft pancreatic tumors in a biodistribution study. Moreover, the targeted nanopolyplex markedly inhibits tumor growth in an orthotopic pancreatic cancer mouse model by dual-targeting tumor cells and stroma. Overall, the multifunctional nanopolyplex is a promising platform for pancreatic cancer therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Kun Cheng
- Corresponding author: Kun Cheng, Ph.D., University of Missouri Curators’ Distinguished Professor, Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, Phone: (816) 235-2425, Fax: (816) 235-5779,
| |
Collapse
|