1
|
Gagnani R, Srivastava M, Suri M, Singh H, Shanker Navik U, Bali A. A focus on c-Jun-N-terminal kinase signaling in sepsis-associated multiple organ dysfunction: Mechanisms and therapeutic strategies. Int Immunopharmacol 2024; 143:113552. [PMID: 39536486 DOI: 10.1016/j.intimp.2024.113552] [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: 07/31/2024] [Revised: 09/19/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024]
Abstract
Sepsis is a life-threatening condition characterized by a widespread inflammatory response to infection, inevitably leading to multiple organ dysfunctions. Extensive research, both in vivo and in vitro, has revealed key factors contributing to sepsis, such as apoptosis, inflammation, cytokine release, oxidative stress, and systemic stress. The changes observed during sepsis-induced conditions are mainly attributed to altered signal transduction pathways, which play a critical role in cell proliferation, migration, and apoptosis. C-Jun N-terminal kinases, JNKs, and serine/threonine protein kinases in the mitogen-activated super family have gained considerable interest for their contribution to cellular events under sepsis conditions. JNK1 and JNK2 are present in various tissues like the lungs, liver, and intestine, while JNK3 is found in neurons. The JNK pathway plays a crucial role in the signal transduction of cytokines related to sepsis development, notably TNF-α and IL-1β. Activated JNK leads to apoptosis, causing tissue damage and organ dysfunction. Further, JNK activation is significant in several inflammatory conditions. Pharmacologically inhibiting JNK has been shown to prevent sepsis-associated damage across multiple organs, including the lungs, liver, intestines, heart, and kidneys. Multiple signaling pathways have been implicated in sepsis, including JNK/c-Myc, Mst1-JNK, MKK4-JNK, JNK-dependent autophagy, and Sirt1/FoxO3a. The review examines the role of JNK signaling in the development of sepsis-induced multiple-organ dysfunction through specific mechanisms. It also discusses different therapeutic approaches to target JNK. This review emphasizes the potential of JNKs as targets for the development of therapeutic agents for sepsis and the associated specific organ damage.
Collapse
Affiliation(s)
- Riya Gagnani
- Laboratory of Neuroendocrinology, Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, India.
| | - Mukul Srivastava
- Laboratory of Neuroendocrinology, Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, India
| | - Manisha Suri
- Laboratory of Neuroendocrinology, Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, India
| | - Harshita Singh
- Laboratory of Neuroendocrinology, Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, India
| | - Uma Shanker Navik
- Laboratory of Neuroendocrinology, Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, India
| | - Anjana Bali
- Laboratory of Neuroendocrinology, Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, India.
| |
Collapse
|
2
|
Yang B, Yang K, Xi R, Li S, Chen J, Wu Y. Inhibition of JNK signaling attenuates photoreceptor ferroptosis caused by all-trans-retinal. Free Radic Biol Med 2024; 227:179-189. [PMID: 39643129 DOI: 10.1016/j.freeradbiomed.2024.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 11/20/2024] [Accepted: 12/02/2024] [Indexed: 12/09/2024]
Abstract
The disruption of the visual cycle leads to the accumulation of all-trans-retinal (atRAL) in the retina, a hallmark of autosomal recessive Stargardt disease (STGD1) and dry age-related macular degeneration (AMD), both of which cause retinal degeneration. Although our previous studies have shown that atRAL induces ferroptosis and activates c-Jun N-terminal kinase (JNK) signaling in the retina, the relationship between JNK signaling and ferroptosis in atRAL-mediated photoreceptor damage remains unclear. Here, we reported that JNK activation by atRAL drove photoreceptor ferroptosis through ferritinophagy. In photoreceptor cells loaded with atRAL, activated JNK phosphorylated c-Jun, which facilitated its nuclear translocation and promoted the expression of the nuclear receptor coactivator 4 (NCOA4). Elevated NCOA4 induced ferritin degradation via lysosomal processing, a process known as ferritinophagy, thereby releasing a large amount of labile iron. Iron overload led to the generation of reactive oxygen species (ROS) and lipid peroxidation, ultimately culminating in ferroptosis. Treatment with the JNK inhibitor JNK-IN-8, as well as the knockout of Jnk1 and Jnk2 genes, significantly rescued atRAL-loaded photoreceptor cells from ferritinophagy-induced ferroptosis. Abca4-/-Rdh8-/- mice, which exhibit atRAL accumulation in the retina following light exposure, are commonly used to study the pathological processes of STGD1 and dry AMD. In these mice, light exposure activated the JNK/c-Jun/NCOA4 axis, resulting in ferritinophagy in the neural retina. Importantly, intraperitoneal administration of JNK-IN-8 significantly rescued retinal function and photoreceptors from ferritinophagy-induced ferroptosis and effectively mitigated retinal degeneration in light-exposed Abca4-/-Rdh8-/- mice. This study underscores the critical role of the JNK/c-Jun/NCOA4 axis in mediating atRAL-induced ferritinophagy, which drives ferroptosis and retinal atrophy, suggesting that targeting this pathway may offer a potential therapeutic approach for STGD1 and dry AMD.
Collapse
Affiliation(s)
- Bo Yang
- Department of Ophthalmology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361003, China; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Kunhuan Yang
- Department of Ophthalmology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361003, China; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Ruitong Xi
- Department of Ophthalmology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361003, China; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Shiying Li
- Department of Ophthalmology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361003, China
| | - Jingmeng Chen
- School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China; Shenzhen Research Institute of Xiamen University, Shenzhen, Guangdong, 518057, China
| | - Yalin Wu
- Department of Ophthalmology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361003, China; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China; Shenzhen Research Institute of Xiamen University, Shenzhen, Guangdong, 518057, China.
| |
Collapse
|
3
|
Taya M, Merenbakh-Lamin K, Zubkov A, Honig Z, Kurolap A, Mayer O, Shomron N, Wolf I, Rubinek T. Beyond endocrine resistance: estrogen receptor (ESR1) activating mutations mediate chemotherapy resistance through the JNK/c-Jun MDR1 pathway in breast cancer. Breast Cancer Res Treat 2024:10.1007/s10549-024-07507-3. [PMID: 39470848 DOI: 10.1007/s10549-024-07507-3] [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: 02/21/2024] [Accepted: 10/03/2024] [Indexed: 11/01/2024]
Abstract
PURPOSE All patients with metastatic breast cancer (MBC) expressing estrogen receptor-α (ESR1) will eventually develop resistance to endocrine therapies. In up to 40% of patients, this resistance is caused by activating mutations in the ligand-binding domain (LBD) of ESR1. Accumulating clinical evidence indicate adverse outcomes for these patients, beyond that expected by resistance to endocrine therapy. Here we aimed to study the role of ESR1 mutations in conferring chemoresistance in BC cells. METHODS MCF-7 cells harboring Y537S and D538G ESR1 mutations (mut-ER) were employed to study the response to chemotherapy drugs, paclitaxel and doxorubicin, using viability and apoptotic assay in vitro, and tumor growth in vivo. JNK/c-Jun/MDR1 pathway was studied using qRT-PCR, western-blot, gene-reporter and ChIP assays. MDR1 expression was analyzed in clinical samples using IHC. RESULTS Cell harboring ESR1 mutations displayed relative chemoresistance compared to WT-ER, evidenced by higher viability and reduced apoptosis as well as resistance to paclitaxel in vivo. To elucidate the underlying mechanism, MDR1 expression was examined and elevated levels were observed in mut-ER cells, and in clinical BC samples. MDR1 is regulated by the c-Jun pathway, and we showed high correlation between these two genes in BC using TCGA databases. Accordingly, we detected higher JNK/c-Jun expression and activity in ESR1-mutated cells, as well as increased occupancy of c-Jun in MDR1 promoter. Importantly, JNK inhibition decreased MDR1 expression and restored sensitivity to chemotherapy. CONCLUSIONS Taken together, these data indicate that ESR1 mutations confer chemoresistance through activation of the JNK/MDR1 axis. These finding suggest a novel treatment option for BC tumors expressing ESR1 mutations.
Collapse
Affiliation(s)
- Marwa Taya
- Department of Oncology, Tel Aviv Sourasky Medical Center, 6 Weizmann St., 6423906, Tel Aviv, Israel.
- The Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel.
| | - Keren Merenbakh-Lamin
- Department of Oncology, Tel Aviv Sourasky Medical Center, 6 Weizmann St., 6423906, Tel Aviv, Israel
| | - Asia Zubkov
- Institute of Pathology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Zohar Honig
- Department of Oncology, Tel Aviv Sourasky Medical Center, 6 Weizmann St., 6423906, Tel Aviv, Israel
| | - Alina Kurolap
- The Genetics Institute and Genomics Center, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Ori Mayer
- The Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Noam Shomron
- The Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Ido Wolf
- Department of Oncology, Tel Aviv Sourasky Medical Center, 6 Weizmann St., 6423906, Tel Aviv, Israel.
- The Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel.
| | - Tami Rubinek
- Department of Oncology, Tel Aviv Sourasky Medical Center, 6 Weizmann St., 6423906, Tel Aviv, Israel.
- The Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
4
|
Sevrin T, Imoto H, Robertson S, Rauch N, Dyn'ko U, Koubova K, Wynne K, Kolch W, Rukhlenko OS, Kholodenko BN. Cell-specific models reveal conformation-specific RAF inhibitor combinations that synergistically inhibit ERK signaling in pancreatic cancer cells. Cell Rep 2024; 43:114710. [PMID: 39240715 PMCID: PMC11474227 DOI: 10.1016/j.celrep.2024.114710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 07/16/2024] [Accepted: 08/20/2024] [Indexed: 09/08/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) presents significant challenges for targeted clinical interventions due to prevalent KRAS mutations, rendering PDAC resistant to RAF and MEK inhibitors (RAFi and MEKi). In addition, responses to targeted therapies vary between patients. Here, we explored the differential sensitivities of PDAC cell lines to RAFi and MEKi and developed an isogenic pair comprising the most sensitive and resistant PDAC cells. To simulate patient- or tumor-specific variations, we constructed cell-line-specific mechanistic models based on protein expression profiling and differential properties of KRAS mutants. These models predicted synergy between two RAFi with different conformation specificity (type I½ and type II RAFi) in inhibiting phospho-ERK (ppERK) and reducing PDAC cell viability. This synergy was experimentally validated across all four studied PDAC cell lines. Our findings underscore the need for combination approaches to inhibit the ERK pathway in PDAC.
Collapse
Affiliation(s)
- Thomas Sevrin
- Systems Biology Ireland, University College Dublin, Dublin, Ireland
| | - Hiroaki Imoto
- Systems Biology Ireland, University College Dublin, Dublin, Ireland
| | - Sarah Robertson
- Systems Biology Ireland, University College Dublin, Dublin, Ireland
| | - Nora Rauch
- Systems Biology Ireland, University College Dublin, Dublin, Ireland
| | - Uscinnia Dyn'ko
- Systems Biology Ireland, University College Dublin, Dublin, Ireland
| | - Katerina Koubova
- Systems Biology Ireland, University College Dublin, Dublin, Ireland; Department of Histology and Embryology, Faculty of Medicine and Dentistry, Palacky University, 779 00 Olomouc, Czech Republic
| | - Kieran Wynne
- Systems Biology Ireland, University College Dublin, Dublin, Ireland
| | - Walter Kolch
- Systems Biology Ireland, University College Dublin, Dublin, Ireland; Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin, Ireland; School of Medicine and Medical Science, University College Dublin, Dublin, Ireland
| | | | - Boris N Kholodenko
- Systems Biology Ireland, University College Dublin, Dublin, Ireland; Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin, Ireland; School of Medicine and Medical Science, University College Dublin, Dublin, Ireland; Department of Pharmacology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
5
|
Berginski ME, Jenner MR, Joisa CU, Herrera Loeza G, Golitz BT, Lipner MB, Leary JR, Rashid N, Johnson GL, Yeh JJ, Gomez SM. Kinome state is predictive of cell viability in pancreatic cancer tumor and cancer-associated fibroblast cell lines. PeerJ 2024; 12:e17797. [PMID: 39221276 PMCID: PMC11365483 DOI: 10.7717/peerj.17797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 07/02/2024] [Indexed: 09/04/2024] Open
Abstract
Numerous aspects of cellular signaling are regulated by the kinome-the network of over 500 protein kinases that guides and modulates information transfer throughout the cell. The key role played by both individual kinases and assemblies of kinases organized into functional subnetworks leads to kinome dysregulation driving many diseases, particularly cancer. In the case of pancreatic ductal adenocarcinoma (PDAC), a variety of kinases and associated signaling pathways have been identified for their key role in the establishment of disease as well as its progression. However, the identification of additional relevant therapeutic targets has been slow and is further confounded by interactions between the tumor and the surrounding tumor microenvironment. In this work, we attempt to link the state of the human kinome, or kinotype, with cell viability in treated, patient-derived PDAC tumor and cancer-associated fibroblast cell lines. We applied classification models to independent kinome perturbation and kinase inhibitor cell screen data, and found that the inferred kinotype of a cell has a significant and predictive relationship with cell viability. We further find that models are able to identify a set of kinases whose behavior in response to perturbation drive the majority of viability responses in these cell lines, including the understudied kinases CSNK2A1/3, CAMKK2, and PIP4K2C. We next utilized these models to predict the response of new, clinical kinase inhibitors that were not present in the initial dataset for model devlopment and conducted a validation screen that confirmed the accuracy of the models. These results suggest that characterizing the perturbed state of the human protein kinome provides significant opportunity for better understanding of signaling behavior and downstream cell phenotypes, as well as providing insight into the broader design of potential therapeutic strategies for PDAC.
Collapse
Affiliation(s)
- Matthew E. Berginski
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, United States of America
| | - Madison R. Jenner
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, United States of America
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Chinmaya U. Joisa
- Joint Department of Biomedical Engineering at the University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, United States of America
| | - Gabriela Herrera Loeza
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Brian T. Golitz
- Eshelman Institute for Innovation, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Matthew B. Lipner
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, United States of America
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Jack R. Leary
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
- Department of Biostatistics, University of Florida, Gainsville, FL, United States of America
| | - Naim Rashid
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Gary L. Johnson
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, United States of America
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Jen Jen Yeh
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, United States of America
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
- Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Shawn M. Gomez
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, United States of America
- Joint Department of Biomedical Engineering at the University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, United States of America
| |
Collapse
|
6
|
Brown BA, Myers PJ, Adair SJ, Pitarresi JR, Sah-Teli SK, Campbell LA, Hart WS, Barbeau MC, Leong K, Seyler N, Kane W, Lee KE, Stelow E, Jones M, Simon MC, Koivunen P, Bauer TW, Stanger BZ, Lazzara MJ. A Histone Methylation-MAPK Signaling Axis Drives Durable Epithelial-Mesenchymal Transition in Hypoxic Pancreatic Cancer. Cancer Res 2024; 84:1764-1780. [PMID: 38471099 DOI: 10.1158/0008-5472.can-22-2945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 10/10/2023] [Accepted: 03/01/2024] [Indexed: 03/14/2024]
Abstract
The tumor microenvironment in pancreatic ductal adenocarcinoma (PDAC) plays a key role in tumor progression and response to therapy. The dense PDAC stroma causes hypovascularity, which leads to hypoxia. Here, we showed that hypoxia drives long-lasting epithelial-mesenchymal transition (EMT) in PDAC primarily through a positive-feedback histone methylation-MAPK signaling axis. Transformed cells preferentially underwent EMT in hypoxic tumor regions in multiple model systems. Hypoxia drove a cell autonomous EMT in PDAC cells, which, unlike EMT in response to growth factors, could last for weeks. Furthermore, hypoxia reduced histone demethylase KDM2A activity, suppressed PP2 family phosphatase expression, and activated MAPKs to post-translationally stabilize histone methyltransferase NSD2, leading to an H3K36me2-dependent EMT in which hypoxia-inducible factors played only a supporting role. Hypoxia-driven EMT could be antagonized in vivo by combinations of MAPK inhibitors. Collectively, these results suggest that hypoxia promotes durable EMT in PDAC by inducing a histone methylation-MAPK axis that can be effectively targeted with multidrug therapies, providing a potential strategy for overcoming chemoresistance. SIGNIFICANCE Integrated regulation of histone methylation and MAPK signaling by the low-oxygen environment of pancreatic cancer drives long-lasting EMT that promotes chemoresistance and shortens patient survival and that can be pharmacologically inhibited. See related commentary by Wirth and Schneider, p. 1739.
Collapse
Affiliation(s)
- Brooke A Brown
- Department of Chemical Engineering, University of Virginia, Charlottesville, Virginia
| | - Paul J Myers
- Department of Chemical Engineering, University of Virginia, Charlottesville, Virginia
| | - Sara J Adair
- Department of Surgery, University of Virginia, Charlottesville, Virginia
| | - Jason R Pitarresi
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Shiv K Sah-Teli
- Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Logan A Campbell
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia
| | - William S Hart
- Department of Chemical Engineering, University of Virginia, Charlottesville, Virginia
| | | | - Kelsey Leong
- Engineering Science, University of Virginia, Charlottesville, Virginia
| | - Nicholas Seyler
- Department of Chemical Engineering, University of Virginia, Charlottesville, Virginia
| | - William Kane
- Department of Surgery, University of Virginia, Charlottesville, Virginia
| | - Kyoung Eun Lee
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan
| | - Edward Stelow
- Department of Pathology, University of Virginia, Charlottesville, Virginia
| | - Marieke Jones
- Claude Moore Health Sciences Library, University of Virginia, Charlottesville, Virginia
| | - M Celeste Simon
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Peppi Koivunen
- Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Todd W Bauer
- Department of Surgery, University of Virginia, Charlottesville, Virginia
| | - Ben Z Stanger
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Matthew J Lazzara
- Department of Chemical Engineering, University of Virginia, Charlottesville, Virginia
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
7
|
Kearney JF, Trembath HE, Chan PS, Morrison AB, Xu Y, Luan CF, McCabe IC, Zarmer SA, Kim HJ, Peng XL, Yeh JJ. Myofibroblastic cancer-associated fibroblast subtype heterogeneity in pancreatic cancer. J Surg Oncol 2024; 129:860-868. [PMID: 38233984 PMCID: PMC11307498 DOI: 10.1002/jso.27582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 12/01/2023] [Accepted: 12/29/2023] [Indexed: 01/19/2024]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) has a fibrotic stroma that has both tumor-promoting and tumor-restraining properties. Different types of cancer-associated fibroblasts (CAFs) have been described. Here, we investigated whether CAFs within the same subtype exhibit heterogeneous functions. METHODS We evaluated the gene and protein expression differences in two myofibroblastic CAF (myCAF) lines using single-cell and bulk RNA-sequencing. We utilized proliferation and migration assays to determine the effect of different CAF lines on a tumor cell line. RESULTS We found that myCAF lines express an activated stroma subtype gene signature, which is associated with a shorter survival in patients. Although both myCAF lines expressed α-smooth muscle actin (α-SMA), platelet-derived growth factor-α (PDGFR-α), fibroblast-activated protein (FAP), and vimentin, we observed heterogeneity between the two lines. Similarly, despite being consistent with myCAF gene expression overall, heterogeneity within specific genes was observed. We found that these differences extended to the functional level where the two myCAF lines had different effects on the same tumor cell line. The myCAF216 line, which had slightly increased inflammatory CAF-like gene expression and higher protein expression of α-SMA, PDGFR-α, and FAP was found to restrain migration of tumor cells. CONCLUSIONS We found that two myCAF lines with globally similar expression characteristics had different effects on the same tumor cell line, one promoting and the other restraining migration. Our study highlights that there may be unappreciated heterogeneity within CAF subtypes. Further investigation and attention to specific genes or proteins that may drive this heterogeneity will be important.
Collapse
Affiliation(s)
- Joseph F. Kearney
- The University of North Carolina at Chapel Hill Department of Surgery, Chapel Hill, North Carolina, USA
| | - Hannah E. Trembath
- The University of North Carolina at Chapel Hill Department of Surgery, Chapel Hill, North Carolina, USA
| | - Priscilla S. Chan
- The University of North Carolina at Chapel Hill Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina, USA
| | - Ashley B. Morrison
- The University of North Carolina at Chapel Hill Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina, USA
| | - Yi Xu
- The University of North Carolina at Chapel Hill Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina, USA
| | - Chang Fei Luan
- The University of North Carolina at Chapel Hill Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina, USA
| | - Ian C. McCabe
- The University of North Carolina at Chapel Hill Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina, USA
| | - Sandra A. Zarmer
- The University of North Carolina at Chapel Hill Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina, USA
| | - Hong Jin Kim
- The University of North Carolina at Chapel Hill Department of Surgery, Chapel Hill, North Carolina, USA
| | - Xianlu L. Peng
- The University of North Carolina at Chapel Hill Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina, USA
| | - Jen Jen Yeh
- The University of North Carolina at Chapel Hill Department of Surgery, Chapel Hill, North Carolina, USA
| |
Collapse
|
8
|
Joisa CU, Chen KA, Berginski ME, Golitz BT, Jenner MR, Herrera Loeza G, Yeh JJ, Gomez SM. Integrated single-dose kinome profiling data is predictive of cancer cell line sensitivity to kinase inhibitors. PeerJ 2023; 11:e16342. [PMID: 38025707 PMCID: PMC10657565 DOI: 10.7717/peerj.16342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 10/03/2023] [Indexed: 12/01/2023] Open
Abstract
Protein kinase activity forms the backbone of cellular information transfer, acting both individually and as part of a broader network, the kinome. Their central role in signaling leads to kinome dysfunction being a common driver of disease, and in particular cancer, where numerous kinases have been identified as having a causal or modulating role in tumor development and progression. As a result, the development of therapies targeting kinases has rapidly grown, with over 70 kinase inhibitors approved for use in the clinic and over double this number currently in clinical trials. Understanding the relationship between kinase inhibitor treatment and their effects on downstream cellular phenotype is thus of clear importance for understanding treatment mechanisms and streamlining compound screening in therapy development. In this work, we combine two large-scale kinome profiling data sets and use them to link inhibitor-kinome interactions with cell line treatment responses (AUC/IC50). We then built computational models on this data set that achieve a high degree of prediction accuracy (R2 of 0.7 and RMSE of 0.9) and were able to identify a set of well-characterized and understudied kinases that significantly affect cell responses. We further validated these models experimentally by testing predicted effects in breast cancer cell lines and extended the model scope by performing additional validation in patient-derived pancreatic cancer cell lines. Overall, these results demonstrate that broad quantification of kinome inhibition state is highly predictive of downstream cellular phenotypes.
Collapse
Affiliation(s)
- Chinmaya U. Joisa
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC, United States of America
| | - Kevin A. Chen
- Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Matthew E. Berginski
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Brian T. Golitz
- Eshelman Institute for Innovation, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Madison R. Jenner
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Gabriela Herrera Loeza
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Jen Jen Yeh
- Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Shawn M. Gomez
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC, United States of America
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| |
Collapse
|
9
|
Tam SY, Islam Khan MZ, Chen JY, Yip JHY, Yan HY, Tam TY, Law HKW. Proteomic Profiling of Chemotherapy Responses in FOLFOX-Resistant Colorectal Cancer Cells. Int J Mol Sci 2023; 24:9899. [PMID: 37373047 DOI: 10.3390/ijms24129899] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/04/2023] [Accepted: 06/05/2023] [Indexed: 06/29/2023] Open
Abstract
Chemoresistance mechanisms of colorectal cancer remain largely elusive. We aim to compare the difference of chemotherapy responses between FOLFOX-resistant and wild-type colorectal cancer cells by proteomic profiling to suggest novel treatment targets. FOLFOX-resistant colorectal cancer cells DLD1-R and HCT116-R were developed by chronic exposure to progressive FOLFOX doses. Proteomic profiling of FOLFOX-resistant and wild-type cells under FOLFOX exposure were conducted by mass-spectrometry-based protein-analysis technology. Verification of selected KEGG pathways was conducted by Western blot. DLD1-R had significantly higher FOLFOX-chemoresistance (10.81 times) than its wild-type counterpart. A total of 309 and 90 differentially expressed proteins were identified in DLD1-R and HCT116-R, respectively. In terms of gene ontology molecular function, RNA binding and cadherin binding ranked first for DLD1 and HCT116 groups, respectively. For gene set enrichment analysis, ribosome pathway and DNA replication were significantly up-regulated and down-regulated in DLD1-R, respectively. The most significantly up-regulated pathway in HCT116-R was regulation of the actin cytoskeleton. Up-regulations in the ribosome pathway (DLD1-R) and actin cytoskeleton (HCT116-R) were verified by Western blot. There were several significantly altered signaling pathways in FOLFOX-resistant colorectal cancer cells under FOLFOX with notable up-regulations in the ribosomal process and actin cytoskeleton.
Collapse
Affiliation(s)
- Shing-Yau Tam
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Md Zahirul Islam Khan
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Ju-Yu Chen
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Jerica Hiu-Yui Yip
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Hong-Yiu Yan
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Tsz-Yan Tam
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Helen Ka-Wai Law
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| |
Collapse
|
10
|
Gao P, Duan W, Shi H, Wang Q. Silencing circPalm2 inhibits sepsis-induced acute lung injury by sponging miR-376b-3p and targeting MAP3K1. Toxicol Res 2023; 39:275-294. [PMID: 37008689 PMCID: PMC10050541 DOI: 10.1007/s43188-022-00169-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 12/26/2022] [Accepted: 12/28/2022] [Indexed: 01/18/2023] Open
Abstract
The apoptosis and inflammation of pulmonary epithelial cells are important pathogenic factors of sepsis-induced acute lung injury (ALI). Upregulation of circPalm2 (circ_0001212) expression levels has been previously detected in the lung tissue of ALI rats. Herein, the biological significance and detailed mechanism of circPalm2 in ALI pathogenesis were investigated. In vivo models of sepsis-induced ALI were established by treating C57BL/6 mice with cecal ligation and puncture (CLP) surgery. Murine pulmonary epithelial cells (MLE-12 cells) were stimulated with lipopolysaccharide (LPS) to establish in vitro septic ALI models. MLE-12 cell viability and apoptosis were evaluated by CCK-8 assay and flow cytometry analysis, respectively. The pathological alterations of the lung tissue were analysed based on hematoxylin-eosin (H&E) staining. Cell apoptosis in the lung tissue samples was examined by TUNEL staining assay. LPS administration suppressed the viability and accelerated the inflammation and apoptotic behaviours of MLE-12 cells. CircPalm2 displayed high expression in LPS-stimulated MLE-12 cells and possessed circular characteristics. The silencing of circPalm2 impeded apoptosis and inflammation in LPS-stimulated MLE-12 cells. Mechanistically, circPalm2 bound with miR-376b-3p, which targeted MAP3K1. In rescue assays, MAP3K1 enhancement reversed the repressive effects of circPalm2 depletion on LPS-triggered inflammatory injury and MLE-12 cell apoptosis. Furthermore, the lung tissue collected from CLP model mice displayed low miR-376b-3p expression and high levels of circPalm2 and MAP3K1. CircPalm2 positively regulated MAP3K1 expression by downregulating miR-376b-3p in murine lung tissues. Importantly, circPalm2 knockdown attenuated CLP-induced inflammation, apoptosis, and pathological alterations in lung tissues collected from mice. Silenced circPalm2 inhibits LPS-induced pulmonary epithelial cell dysfunction and mitigates abnormalities in lung tissues collected from CLP-stimulated mice via the miR-376b-3p/MAP3K1 axis in septic ALI. Supplementary Information The online version contains supplementary material available at 10.1007/s43188-022-00169-7.
Collapse
Affiliation(s)
- Pengfei Gao
- Shanghai East Clinical Medical College, Nanjing Medical University, No. 150, Jimo Road, Pudong New Area, Shanghai, 200120 China
- Department of Anesthesiology, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Huaian, Jiangsu 223300 China
| | - Wenying Duan
- Shanghai East Clinical Medical College, Nanjing Medical University, No. 150, Jimo Road, Pudong New Area, Shanghai, 200120 China
| | - Huiyan Shi
- Jinzhou Medical University, Jinzhou, Liaoning 121001 China
| | - Qingxiu Wang
- Shanghai East Clinical Medical College, Nanjing Medical University, No. 150, Jimo Road, Pudong New Area, Shanghai, 200120 China
- Shanghai East Hopital, Tongji University School of Medicine, Shanghai, 200120 China
| |
Collapse
|
11
|
Kumar S, Singh SK, Srivastava P, Suresh S, Rana B, Rana A. Interplay between MAP kinases and tumor microenvironment: Opportunity for immunotherapy in pancreatic cancer. Adv Cancer Res 2023. [PMID: 37268394 DOI: 10.1016/bs.acr.2023.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC), commonly called pancreatic cancer, is aggressive cancer usually detected at a late stage, limiting treatment options with modest clinical responses. It is projected that by 2030, PDAC will be the second most common cause of cancer-related mortality in the United States. Drug resistance in PDAC is common and significantly affects patients' overall survival (OS). Oncogenic KRAS mutations are nearly uniform in PDAC, affecting over 90% of patients. However, effective drugs directed to target prevalent KRAS mutants in pancreatic cancer are not in clinical practice. Accordingly, efforts are continued on identifying alternative druggable target(s) or approaches to improve patient outcomes with PDAC. In most PDAC cases, the KRAS mutations turn-on the RAF-MEK-MAPK pathways, leading to pancreatic tumorigenesis. The MAPK signaling cascade (MAP4K→MAP3K→MAP2K→MAPK) plays a central role in the pancreatic cancer tumor microenvironment (TME) and chemotherapy resistance. The immunosuppressive pancreatic cancer TME is another unfavorable factor affecting the therapeutic efficacy of chemotherapy and immunotherapy. The immune checkpoint proteins (ICPs), including CTLA-4, PD-1, PD-L1, and PD-L2, are critical players in T cell dysfunction and pancreatic tumor cell growth. Here, we review the activation of MAPKs, a molecular trait of KRAS mutations and their impact on pancreatic cancer TME, chemoresistance, and expression of ICPs that could influence the clinical outcomes in PDAC patients. Therefore, understanding the interplay between MAPK pathways and TME could help to design rational therapy combining immunotherapy and MAPK inhibitors for pancreatic cancer treatment.
Collapse
|
12
|
Non-kinase targeting of oncogenic c-Jun N-terminal kinase (JNK) signaling: the future of clinically viable cancer treatments. Biochem Soc Trans 2022; 50:1823-1836. [PMID: 36454622 PMCID: PMC9788565 DOI: 10.1042/bst20220808] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/28/2022] [Accepted: 11/15/2022] [Indexed: 01/09/2023]
Abstract
c-Jun N-terminal Kinases (JNKs) have been identified as key disease drivers in a number of pathophysiological settings and central oncogenic signaling nodes in various cancers. Their roles in driving primary tumor growth, positively regulating cancer stem cell populations, promoting invasion and facilitating metastatic outgrowth have led JNKs to be considered attractive targets for anti-cancer therapies. However, the homeostatic, apoptotic and tumor-suppressive activities of JNK proteins limit the use of direct JNK inhibitors in a clinical setting. In this review, we will provide an overview of the different JNK targeting strategies developed to date, which include various ATP-competitive, non-kinase and substrate-competitive inhibitors. We aim to summarize their distinct mechanisms of action, review some of the insights they have provided regarding JNK-targeting in cancer, and outline the limitations as well as challenges of all strategies that target JNKs directly. Furthermore, we will highlight alternate drug targets within JNK signaling complexes, including recently identified scaffold proteins, and discuss how these findings may open up novel therapeutic options for targeting discrete oncogenic JNK signaling complexes in specific cancer settings.
Collapse
|
13
|
Shi J, Yang X, Kang Q, Lu J, Denzinger M, Kornmann M, Traub B. JNK inhibitor IX restrains pancreatic cancer through p53 and p21. Front Oncol 2022; 12:1006131. [PMID: 36568248 PMCID: PMC9768178 DOI: 10.3389/fonc.2022.1006131] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022] Open
Abstract
Novel treatment options for pancreatic cancer are desperately needed. De-regulated kinases can be regularly detected in pancreatic cancer. Multiple pathway inhibitors were developed to exploit these features, among them selective inhibitors of the c-Jun N-terminal kinase isoforms 1 and 2 (JNK1 and 2). We evaluated the effectiveness of four different JNK inhibitors on pancreatic cancer cell lines. Cell mobility and migration were evaluated in scratch assay and Boyden chamber assay. Mechanism of cell death was analyzed via apoptosis assays in FACS and immunoblotting as well as cell cycle analysis via FACS, and qPCR. JNK2 knockout cells were generated using siRNA transfection. Among the inhibitors, JNK inhibitor IX (JNK-in-IX), designed as specific inhibitor against JNK2 was proven highly effective in inhibiting cell growth, mobility and migration. We were able to show that JNK-in-IX caused DNA damage resulting in G2 arrest mediated through p53 and p21. Interestingly, JNK-in-IX acted independently of its primary target JNK2. In summary, JNK-in-IX was shown highly effective in pancreatic cancer. This study underlines the need for modeling systems in testing therapeutic options as JNK2 was previously not indicated as a potential target.
Collapse
Affiliation(s)
- Jingwei Shi
- Department of Cardiothoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China,Department of General and Visceral Surgery, Ulm University Hospital, Ulm, Germany
| | - Xing Yang
- Department of General and Visceral Surgery, Ulm University Hospital, Ulm, Germany
| | - Qi Kang
- Department of General and Visceral Surgery, Ulm University Hospital, Ulm, Germany
| | - Jian Lu
- Department of General and Visceral Surgery, Ulm University Hospital, Ulm, Germany
| | - Maximilian Denzinger
- Department of General and Visceral Surgery, Ulm University Hospital, Ulm, Germany
| | - Marko Kornmann
- Department of General and Visceral Surgery, Ulm University Hospital, Ulm, Germany
| | - Benno Traub
- Department of General and Visceral Surgery, Ulm University Hospital, Ulm, Germany,*Correspondence: Benno Traub,
| |
Collapse
|
14
|
Al-Tamimi M, Khan AQ, Anver R, Ahmad F, M Mateo J, Raza SS, Alam M, Buddenkotte J, Steinhoff M, Uddin S. Pristimerin mediated anticancer effects and sensitization of human skin cancer cells through modulation of MAPK signaling pathways. Biomed Pharmacother 2022; 156:113950. [PMID: 36411635 DOI: 10.1016/j.biopha.2022.113950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 10/22/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022] Open
|
15
|
Chen H, Jiang Y, Liu R, Deng J, Chen Q, Chen L, Liang G, Chen X, Xu Z. Curcumin Derivative C66 Suppresses Pancreatic Cancer Progression through the Inhibition of JNK-Mediated Inflammation. Molecules 2022; 27:3076. [PMID: 35630552 PMCID: PMC9147000 DOI: 10.3390/molecules27103076] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/05/2022] [Accepted: 05/09/2022] [Indexed: 11/16/2022] Open
Abstract
Pancreatic adenocarcinoma is by far the deadliest type of cancer. Inflammation is one of the important risk factors in tumor development. However, it is not yet clear whether deterioration in pancreatic cancer patients is related to inflammation, as well as the underlying mechanism. In addition, JNK is abnormally activated in pancreatic cancer cells and the JNK inhibitor C66 reduces the inflammatory microenvironment in the tumor. Therefore, the aim of this study was to evaluate the role of C66 in the proliferation and migration of pancreatic cancer. Our results showed that various inflammatory cytokines, such as IL-1β, IL-6, IL-8, and IL-15, were more expressed in pancreatic cancer than in the matching normal tissue. Furthermore, C66, a curcumin analogue with good anti-inflammatory activity, inhibited the proliferation and migration of pancreatic cancer cells in a dose-dependent manner, and effectively inhibited the expression of the above inflammatory factors. Our previous research demonstrated that C66 prevents the inflammatory response by targeting JNK. Therefore, in this study, JNK activity in pancreatic cancer cells was investigated, revealing that JNK was highly activated, and the treatment with C66 inhibited the phosphorylation of JNK. Next, shJNK was used to knockdown JNK expression in pancreatic cancer cells to further confirm the role of JNK in the proliferation and migration of this tumor, as well as in the inflammatory tumor microenvironment (TME). The results demonstrated that JNK knockdown could significantly inhibit the proliferation and migration of pancreatic cancer. Moreover, the low JNK expression in pancreatic cancer cells significantly inhibited the expression of various inflammatory factors. These results indicated that C66 inhibited the progression of pancreatic cancer through the inhibition of JNK-mediated inflammation.
Collapse
Affiliation(s)
- Hongjin Chen
- Translational Medicine Research Center, Guizhou Medical University, Guiyang 550000, China
| | - Yuchen Jiang
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, China
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital Hangzhou Medical College, Hangzhou 310000, China
| | - Rongdiao Liu
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325000, China
| | - Jie Deng
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325000, China
| | - Qinbo Chen
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325000, China
| | - Lingfeng Chen
- School of Pharmacy, Hangzhou Medical College, Hangzhou 310000, China
| | - Guang Liang
- School of Pharmacy, Hangzhou Medical College, Hangzhou 310000, China
| | - Xiong Chen
- Department of Endocrinology, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325000, China
| | - Zheng Xu
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325000, China
| |
Collapse
|
16
|
Exploration of the System-Level Mechanisms of the Herbal Drug FDY003 for Pancreatic Cancer Treatment: A Network Pharmacological Investigation. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:7160209. [PMID: 35591866 PMCID: PMC9113891 DOI: 10.1155/2022/7160209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 04/12/2022] [Indexed: 11/18/2022]
Abstract
Pancreatic cancer (PC) is the most lethal cancer with the lowest survival rate globally. Although the prescription of herbal drugs against PC is gaining increasing attention, their polypharmacological therapeutic mechanisms are yet to be fully understood. Based on network pharmacology, we explored the anti-PC properties and system-level mechanisms of the herbal drug FDY003. FDY003 decreased the viability of human PC cells and strengthened their chemosensitivity. Network pharmacological analysis of FDY003 indicated the presence of 16 active phytochemical components and 123 PC-related pharmacological targets. Functional enrichment analysis revealed that the PC-related targets of FDY003 participate in the regulation of cell growth and proliferation, cell cycle process, cell survival, and cell death. In addition, FDY003 was shown to target diverse key pathways associated with PC pathophysiology, namely, the PIK3-Akt, MAPK, FoxO, focal adhesion, TNF, p53, HIF-1, and Ras pathways. Our network pharmacological findings advance the mechanistic understanding of the anti-PC properties of FDY003 from a system perspective.
Collapse
|
17
|
Tong Z, Gaudy A, Tatosian D, Ramirez-Valle F, Liu H, Chen J, Hoffmann M, Surapaneni S. Assessment of drug-drug interactions of CC-90001, a potent and selective inhibitor of c-Jun N-terminal kinase. Xenobiotica 2022; 51:1416-1426. [PMID: 35000550 DOI: 10.1080/00498254.2022.2027553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
CC-90001 is predominantly metabolised via glucuronidation, while oxidative metabolism is a minor pathway in human hepatocytes and liver microsomes. In vitro, CC-90001 glucuronidation was catalysed by UGT1A9, UGT1A4, and UGT1A1, while oxidative metabolism was primarily mediated by CYP3A4/5 with minor contributions from CYP1A2, CYP2C9, CYP2B6, and CYP2D6.CC-90001 in vitro inhibits CYP2A6, CYP2B6, CYP2C8, CYP2C9, CYP2C19, and CYP3A4 ≤ 55% at 100 μM, and the inhibition was negligible at ≤30 μM. CC-90001 is not a time-dependent CYP inhibitor.In human hepatocytes CC-90001 is an inducer of CYP2B6 and CYP3A, with mRNA levels increased 34.4% to 52.8% relative to positive controls.In vitro CC-90001 is a substrate of P-gp, and an inhibitor of P-gp, BCRP, OAT3, OATP1B1, OATP1B3, OCT2, MATE1, and MATE2k with IC50 values of 30.3, 25.8, 17.7, 0.417, 19.9, 0.605, 4.17, and 20 μM, respectively.A clinical study demonstrated that CC-90001 has no or little impact on the exposure of warfarin (CYP2C9), omeprazole (CYP2C19), midazolam (CYP3A) or metformin (OCT2, MATE1/2k). CC-90001 co-administration increases the AUCt and Cmax 176% and 339% for rosuvastatin (BCRP/OATP1B1/3), 116% and 171% for digoxin (P-gp), and 266% and 321% for nintedanib (CYP3A & P-gp), respectively.In conclusion, CC-90001 in unlikely to be a victim or perpetrator of clinically relevant interactions involving CYPs or UGTs. Weak to moderate interactions are expected in clinic with substrates of P-gp and OATP1B1 due to CC-90001 inhibition of these transporters.
Collapse
Affiliation(s)
- Zeen Tong
- Nonclinical Development, Bristol Myers Squibb, Summit, NJ, USA
| | - Allison Gaudy
- Clinical Pharmacology, Bristol Myers Squibb, Summit, NJ, USA
| | - Daniel Tatosian
- Clinical Pharmacology, Bristol Myers Squibb, Summit, NJ, USA
| | | | - Hong Liu
- Nonclinical Development, Bristol Myers Squibb, Summit, NJ, USA
| | - Jian Chen
- Nonclinical Development, Bristol Myers Squibb, Summit, NJ, USA
| | | | | |
Collapse
|
18
|
Tu M, Klein L, Espinet E, Georgomanolis T, Wegwitz F, Li X, Urbach L, Danieli-Mackay A, Küffer S, Bojarczuk K, Mizi A, Günesdogan U, Chapuy B, Gu Z, Neesse A, Kishore U, Ströbel P, Hessmann E, Hahn SA, Trumpp A, Papantonis A, Ellenrieder V, Singh SK. TNF-α-producing macrophages determine subtype identity and prognosis via AP1 enhancer reprogramming in pancreatic cancer. NATURE CANCER 2021; 2:1185-1203. [PMID: 35122059 DOI: 10.1038/s43018-021-00258-w] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Accepted: 08/19/2021] [Indexed: 12/11/2022]
Abstract
Large-scale genomic profiling of pancreatic cancer (PDAC) has revealed two distinct subtypes: 'classical' and 'basal-like'. Their variable coexistence within the stromal immune microenvironment is linked to differential prognosis; however, the extent to which these neoplastic subtypes shape the stromal immune landscape and impact clinical outcome remains unclear. By combining preclinical models, patient-derived xenografts, as well as FACS-sorted PDAC patient biopsies, we show that the basal-like neoplastic state is sustained via BRD4-mediated cJUN/AP1 expression, which induces CCL2 to recruit tumor necrosis factor (TNF)-α-secreting macrophages. TNF-α+ macrophages force classical neoplastic cells into an aggressive phenotypic state via lineage reprogramming. Integration of ATAC-, ChIP- and RNA-seq data revealed distinct JUNB/AP1 (classical) and cJUN/AP1 (basal-like)-driven regulation of PDAC subtype identity. Pharmacological inhibition of BRD4 led to suppression of the BRD4-cJUN-CCL2-TNF-α axis, restoration of classical subtype identity and a favorable prognosis. Hence, patient-tailored therapy for a cJUNhigh/TNF-αhigh subtype is paramount in overcoming highly inflamed and aggressive PDAC states.
Collapse
Affiliation(s)
- Mengyu Tu
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Göttingen, Göttingen, Germany
| | - Lukas Klein
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Göttingen, Göttingen, Germany
| | - Elisa Espinet
- Division of Stem Cells and Cancer, DKFZ, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbh), Heidelberg, Germany
| | | | - Florian Wegwitz
- Department of Gynecology and Obstetrics, University Medical Center Göttingen, Göttingen, Germany
| | - Xiaojuan Li
- Department of Developmental Biology, Göttingen Center for Molecular Biosciences, Göttingen, Germany
| | - Laura Urbach
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Göttingen, Göttingen, Germany
| | - Adi Danieli-Mackay
- Institute of Pathology, University Medical Center Göttingen, Göttingen, Germany
| | - Stefan Küffer
- Institute of Pathology, University Medical Center Göttingen, Göttingen, Germany
| | - Kamil Bojarczuk
- Department of Hematology and Medical Oncology, University Medical Center Göttingen, Göttingen, Germany
| | - Athanasia Mizi
- Institute of Pathology, University Medical Center Göttingen, Göttingen, Germany
| | - Ufuk Günesdogan
- Department of Developmental Biology, Göttingen Center for Molecular Biosciences, Göttingen, Germany
| | - Björn Chapuy
- Department of Hematology and Medical Oncology, University Medical Center Göttingen, Göttingen, Germany
| | - Zuguang Gu
- Bioinformatics and Omics Data Analytics, DKFZ, Heidelberg, Germany
- Division of Cancer Epigenomics, DKFZ, Heidelberg, Germany
| | - Albrecht Neesse
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Göttingen, Göttingen, Germany
| | - Uday Kishore
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, UK
| | - Philipp Ströbel
- Institute of Pathology, University Medical Center Göttingen, Göttingen, Germany
| | - Elisabeth Hessmann
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Göttingen, Göttingen, Germany
| | - Stephan A Hahn
- Faculty of Medicine, Department of Molecular GI Oncology, Ruhr University Bochum, Bochum, Germany
| | - Andreas Trumpp
- Division of Stem Cells and Cancer, DKFZ, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbh), Heidelberg, Germany
| | - Argyris Papantonis
- Institute of Pathology, University Medical Center Göttingen, Göttingen, Germany
| | - Volker Ellenrieder
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Göttingen, Göttingen, Germany
| | - Shiv K Singh
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Göttingen, Göttingen, Germany.
| |
Collapse
|
19
|
Traub B, Roth A, Kornmann M, Knippschild U, Bischof J. Stress-activated kinases as therapeutic targets in pancreatic cancer. World J Gastroenterol 2021; 27:4963-4984. [PMID: 34497429 PMCID: PMC8384741 DOI: 10.3748/wjg.v27.i30.4963] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 05/17/2021] [Accepted: 07/20/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is a dismal disease with high incidence and poor survival rates. With the aim to improve overall survival of pancreatic cancer patients, new therapeutic approaches are urgently needed. Protein kinases are key regulatory players in basically all stages of development, maintaining physiologic functions but also being involved in pathogenic processes. c-Jun N-terminal kinases (JNK) and p38 kinases, representatives of the mitogen-activated protein kinases, as well as the casein kinase 1 (CK1) family of protein kinases are important mediators of adequate response to cellular stress following inflammatory and metabolic stressors, DNA damage, and others. In their physiologic roles, they are responsible for the regulation of cell cycle progression, cell proliferation and differentiation, and apoptosis. Dysregulation of the underlying pathways consequently has been identified in various cancer types, including pancreatic cancer. Pharmacological targeting of those pathways has been the field of interest for several years. While success in earlier studies was limited due to lacking specificity and off-target effects, more recent improvements in small molecule inhibitor design against stress-activated protein kinases and their use in combination therapies have shown promising in vitro results. Consequently, targeting of JNK, p38, and CK1 protein kinase family members may actually be of particular interest in the field of precision medicine in patients with highly deregulated kinase pathways related to these kinases. However, further studies are warranted, especially involving in vivo investigation and clinical trials, in order to advance inhibition of stress-activated kinases to the field of translational medicine.
Collapse
Affiliation(s)
- Benno Traub
- Department of General and Visceral Surgery, Ulm University Hospital, Ulm 89081, Germany
| | - Aileen Roth
- Department of General and Visceral Surgery, Ulm University Hospital, Ulm 89081, Germany
| | - Marko Kornmann
- Department of General and Visceral Surgery, Ulm University Hospital, Ulm 89081, Germany
| | - Uwe Knippschild
- Department of General and Visceral Surgery, Ulm University Hospital, Ulm 89081, Germany
| | - Joachim Bischof
- Department of General and Visceral Surgery, Ulm University Hospital, Ulm 89081, Germany
| |
Collapse
|
20
|
Sun L, Patai ÁV, Hogenson TL, Fernandez-Zapico ME, Qin B, Sinicrope FA. Irreversible JNK blockade overcomes PD-L1-mediated resistance to chemotherapy in colorectal cancer. Oncogene 2021; 40:5105-5115. [PMID: 34193942 DOI: 10.1038/s41388-021-01910-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 06/11/2021] [Accepted: 06/15/2021] [Indexed: 11/09/2022]
Abstract
Colorectal cancer (CRC) cells have low or absent tumor cell PD-L1 expression that we previously demonstrated can confer chemotherapy resistance. Here, we demonstrate that PD-L1 depletion enhances JNK activity resulting in increased BimThr116 phosphorylation and its sequestration by MCL-1 and BCL-2. Activated JNK signaling in PD-L1-depeted cells was due to reduced mRNA stability of the CYLD deubiquitinase. PD-L1 was found to compete with the ribonuclease EXOSC10 for binding to CYLD mRNA. Thus, loss of PD-L1 promoted binding and degradation of CYLD mRNA by EXOSC10 which enhanced JNK activity. An irreversible JNK inhibitor (JNK-IN-8) reduced BimThr116 phosphorylation and unsequestered Bim from MCL-1 and BCL-2 to promote apoptosis. In cells lacking PD-L1, treatment with JNK-IN-8, an MCL-1 antagonist (AZD5991), or their combination promoted apoptosis and reduced long-term clonogenic survival by anticancer drugs. Similar effects of the JNK inhibitor on cell viability were observed in CRC organoids with suppression of PD-L1. These data indicate that JNK inhibition may represent a promising strategy to overcome drug resistance in CRC cells with low or absent PD-L1 expression.
Collapse
Affiliation(s)
- Lei Sun
- Gastrointestinal Research Unit, Mayo Clinic, Rochester, MN, USA.,Department of Gastrointestinal Surgery, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Árpád V Patai
- Gastrointestinal Research Unit, Mayo Clinic, Rochester, MN, USA
| | - Tara L Hogenson
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN, USA
| | - Martin E Fernandez-Zapico
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN, USA
| | - Bo Qin
- Gastrointestinal Research Unit, Mayo Clinic, Rochester, MN, USA.
| | - Frank A Sinicrope
- Gastrointestinal Research Unit, Mayo Clinic, Rochester, MN, USA. .,Departments of Medicine and Oncology, Mayo Clinic, Rochester, MN, USA. .,Mayo Comprehensive Cancer Center, Rochester, MN, USA.
| |
Collapse
|
21
|
Mallya K, Gautam SK, Aithal A, Batra SK, Jain M. Modeling pancreatic cancer in mice for experimental therapeutics. Biochim Biophys Acta Rev Cancer 2021; 1876:188554. [PMID: 33945847 DOI: 10.1016/j.bbcan.2021.188554] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/19/2021] [Accepted: 04/23/2021] [Indexed: 02/06/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive malignancy that is characterized by early metastasis, low resectability, high recurrence, and therapy resistance. The experimental mouse models have played a central role in understanding the pathobiology of PDAC and in the preclinical evaluation of various therapeutic modalities. Different mouse models with targetable pathological hallmarks have been developed and employed to address the unique challenges associated with PDAC progression, metastasis, and stromal heterogeneity. Over the years, mouse models have evolved from simple cell line-based heterotopic and orthotopic xenografts in immunocompromised mice to more complex and realistic genetically engineered mouse models (GEMMs) involving multi-gene manipulations. The GEMMs, mostly driven by KRAS mutation(s), have been widely accepted for therapeutic optimization due to their high penetrance and ability to recapitulate the histological, molecular, and pathological hallmarks of human PDAC, including comparable precursor lesions, extensive metastasis, desmoplasia, perineural invasion, and immunosuppressive tumor microenvironment. Advanced GEMMs modified to express fluorescent proteins have allowed cell lineage tracing to provide novel insights and a new understanding about the origin and contribution of various cell types in PDAC pathobiology. The syngeneic mouse models, GEMMs, and target-specific transgenic mice have been extensively used to evaluate immunotherapies and study therapy-induced immune modulation in PDAC yielding meaningful results to guide various clinical trials. The emerging mouse models for parabiosis, hepatic metastasis, cachexia, and image-guided implantation, are increasingly appreciated for their high translational significance. In this article, we describe the contribution of various experimental mouse models to the current understanding of PDAC pathobiology and their utility in evaluating and optimizing therapeutic modalities for this lethal malignancy.
Collapse
Affiliation(s)
- Kavita Mallya
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Shailendra K Gautam
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA.
| | - Abhijit Aithal
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA; Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Maneesh Jain
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA; Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
22
|
Low HB, Wong ZL, Wu B, Kong LR, Png CW, Cho YL, Li CW, Xiao F, Xin X, Yang H, Loo JM, Lee FYX, Tan IBH, DasGupta R, Shen HM, Schwarz H, Gascoigne NRJ, Goh BC, Xu X, Zhang Y. DUSP16 promotes cancer chemoresistance through regulation of mitochondria-mediated cell death. Nat Commun 2021; 12:2284. [PMID: 33863904 PMCID: PMC8052345 DOI: 10.1038/s41467-021-22638-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 03/18/2021] [Indexed: 02/02/2023] Open
Abstract
Drug resistance is a major obstacle to the treatment of most human tumors. In this study, we find that dual-specificity phosphatase 16 (DUSP16) regulates resistance to chemotherapy in nasopharyngeal carcinoma, colorectal cancer, gastric and breast cancer. Cancer cells expressing higher DUSP16 are intrinsically more resistant to chemotherapy-induced cell death than cells with lower DUSP16 expression. Overexpression of DUSP16 in cancer cells leads to increased resistance to cell death upon chemotherapy treatment. In contrast, knockdown of DUSP16 in cancer cells increases their sensitivity to treatment. Mechanistically, DUSP16 inhibits JNK and p38 activation, thereby reducing BAX accumulation in mitochondria to reduce apoptosis. Analysis of patient survival in head & neck cancer and breast cancer patient cohorts supports DUSP16 as a marker for sensitivity to chemotherapy and therapeutic outcome. This study therefore identifies DUSP16 as a prognostic marker for the efficacy of chemotherapy, and as a therapeutic target for overcoming chemoresistance in cancer.
Collapse
Affiliation(s)
- Heng Boon Low
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Immunology Programme, the Life Science Institute, National University of Singapore, Singapore, Singapore
| | - Zhen Lim Wong
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Immunology Programme, the Life Science Institute, National University of Singapore, Singapore, Singapore
| | - Bangyuan Wu
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Immunology Programme, the Life Science Institute, National University of Singapore, Singapore, Singapore
- College of Life Science, China West Normal University, Nanchong, Sichuan, China
| | - Li Ren Kong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Chin Wen Png
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Immunology Programme, the Life Science Institute, National University of Singapore, Singapore, Singapore
| | - Yik-Lam Cho
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Chun-Wei Li
- Department of Otorhinolaryngology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Fengchun Xiao
- Department of Pathology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Xuan Xin
- Department of Mathematics, National University of Singapore, Singapore, Singapore
| | - Henry Yang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Jia Min Loo
- Genome Institute of Singapore, Agency of Science Technology and Research (A*Star), Singapore, Singapore
| | - Fiona Yi Xin Lee
- Division of Medical Oncology, National Cancer Center, Singapore, Singapore
| | - Iain Bee Huat Tan
- Division of Medical Oncology, National Cancer Center, Singapore, Singapore
| | - Ramanuj DasGupta
- Genome Institute of Singapore, Agency of Science Technology and Research (A*Star), Singapore, Singapore
| | - Han-Ming Shen
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Faculty of Health Sciences, University of Macau, Macau, China
| | - Herbert Schwarz
- Immunology Programme, the Life Science Institute, National University of Singapore, Singapore, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Nicholas R J Gascoigne
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Immunology Programme, the Life Science Institute, National University of Singapore, Singapore, Singapore
| | - Boon Cher Goh
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Xiaohong Xu
- Department of Breast Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China.
| | - Yongliang Zhang
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Immunology Programme, the Life Science Institute, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
23
|
JNK signaling as a target for anticancer therapy. Pharmacol Rep 2021; 73:405-434. [PMID: 33710509 DOI: 10.1007/s43440-021-00238-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 01/30/2021] [Accepted: 02/15/2021] [Indexed: 12/15/2022]
Abstract
The JNKs are members of mitogen-activated protein kinases (MAPK) which regulate many physiological processes including inflammatory responses, macrophages, cell proliferation, differentiation, survival, and death. It is increasingly clear that the continuous activation of JNKs has a role in cancer development and progression. Therefore, JNKs represent attractive oncogenic targets for cancer therapy using small molecule kinase inhibitors. Studies showed that the two major JNK proteins JNK1 and JNK2 have opposite functions in different types of cancers, which need more specification in the design of JNK inhibitors. Some of ATP- competitive and ATP non-competitive inhibitors have been developed and widely used in vitro, but this type of inhibitors lack selectivity and inhibits phosphorylation of all JNK substrates and may lead to cellular toxicity. In this review, we summarized and discussed the strategies of JNK binding inhibitors and the role of JNK signaling in the pathogenesis of different solid and hematological malignancies.
Collapse
|
24
|
Musicant AM, Parag-Sharma K, Gong W, Sengupta M, Chatterjee A, Henry EC, Tsai YH, Hayward MC, Sheth S, Betancourt R, Hackman TG, Padilla RJ, Parker JS, Giudice J, Flaveny CA, Hayes DN, Amelio AL. CRTC1/MAML2 directs a PGC-1α-IGF-1 circuit that confers vulnerability to PPARγ inhibition. Cell Rep 2021; 34:108768. [PMID: 33626346 PMCID: PMC7955229 DOI: 10.1016/j.celrep.2021.108768] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 11/22/2020] [Accepted: 01/27/2021] [Indexed: 01/03/2023] Open
Abstract
Mucoepidermoid carcinoma (MEC) is a life-threatening salivary gland cancer that is driven primarily by a transcriptional coactivator fusion composed of cyclic AMP-regulated transcriptional coactivator 1 (CRTC1) and mastermind-like 2 (MAML2). The mechanisms by which the chimeric CRTC1/MAML2 (C1/M2) oncoprotein rewires gene expression programs that promote tumorigenesis remain poorly understood. Here, we show that C1/M2 induces transcriptional activation of the non-canonical peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α) splice variant PGC-1α4, which regulates peroxisome proliferator-activated receptor gamma (PPARγ)-mediated insulin-like growth factor 1 (IGF-1) expression. This mitogenic transcriptional circuitry is consistent across cell lines and primary tumors. C1/M2-positive tumors exhibit IGF-1 pathway activation, and small-molecule drug screens reveal that tumor cells harboring the fusion gene are selectively sensitive to IGF-1 receptor (IGF-1R) inhibition. Furthermore, this dependence on autocrine regulation of IGF-1 transcription renders MEC cells susceptible to PPARγ inhibition with inverse agonists. These results yield insights into the aberrant coregulatory functions of C1/M2 and identify a specific vulnerability that can be exploited for precision therapy.
Collapse
Affiliation(s)
- Adele M Musicant
- Graduate Curriculum in Genetics and Molecular Biology, Biological and Biomedical Sciences Program, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kshitij Parag-Sharma
- Graduate Curriculum in Cell Biology and Physiology, Biological and Biomedical Sciences Program, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Weida Gong
- Bioinformatics Core, Lineberger Comprehensive Cancer Center, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Monideepa Sengupta
- Graduate Curriculum in Pharmacological and Physiological Sciences, School of Medicine, Saint Louis University, Saint Louis, MO, USA
| | - Arindam Chatterjee
- Department of Pharmacology and Physiology, School of Medicine, Saint Louis University, Saint Louis, MO, USA
| | - Erin C Henry
- Division of Oral and Craniofacial Health Sciences, UNC Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Yi-Hsuan Tsai
- Bioinformatics Core, Lineberger Comprehensive Cancer Center, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Michele C Hayward
- Lineberger Comprehensive Cancer Center, Cancer Genetics Program, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Siddharth Sheth
- Division of Hematology/Oncology, Department of Medicine, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Renee Betancourt
- Department of Pathology and Laboratory Medicine, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Trevor G Hackman
- Department of Otolaryngology/Head and Neck Surgery, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ricardo J Padilla
- Division of Diagnostic Sciences, UNC Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Joel S Parker
- Lineberger Comprehensive Cancer Center, Cancer Genetics Program, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Genetics, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jimena Giudice
- Department of Cell Biology and Physiology, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; McAllister Heart Institute, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Colin A Flaveny
- Department of Pharmacology and Physiology, School of Medicine, Saint Louis University, Saint Louis, MO, USA
| | - David N Hayes
- Lineberger Comprehensive Cancer Center, Cancer Genetics Program, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Medical Oncology, University of Tennessee Health Sciences West Cancer Center, Memphis, TN, USA
| | - Antonio L Amelio
- Division of Oral and Craniofacial Health Sciences, UNC Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Cell Biology and Physiology, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Biomedical Research Imaging Center, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Lineberger Comprehensive Cancer Center, Cancer Cell Biology Program, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
25
|
Schröder SK, Asimakopoulou A, Tillmann S, Koschmieder S, Weiskirchen R. TNF-α controls Lipocalin-2 expression in PC-3 prostate cancer cells. Cytokine 2020; 135:155214. [PMID: 32712458 DOI: 10.1016/j.cyto.2020.155214] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/30/2020] [Accepted: 07/16/2020] [Indexed: 12/21/2022]
Abstract
Prostate cancer (PCa) is one of the most common and deadly cancers in men worldwide. The surrounding tumor microenvironment (TME) is important in tumor progression, as cytokines and soluble mediators including tumor necrosis factor (TNF-α) or lipocalin-2 (LCN2) can influence tumor growth and formation of metastasis. The exact mechanisms on how these pleiotropic factors affect PCa are still unknown. In this study, we showed for the first time that LCN2 mRNA and protein expression are strongly inducible by TNF-α in the highly metastatic human PCa cell line PC-3. In addition, we observed higher levels of secreted LCN2 in cell culture medium of TNF-α-treated PC-3 cells. We found that different signaling pathways such as p38, NF-κB or JNK were activated shortly after TNF-α treatment. Moreover, the mRNA levels of IL-1β and IL-8 were also significantly increased after 24 h stimulation. Mechanistically, the NF-κB pathway and the JNK signaling axis are directly responsible for LCN2 upregulation. This was shown by the fact that pretreatment with the JNK inhibitors SP600125 or JNK-IN-8 strongly downregulated phosphorylation of c-Jun protein and markedly reduced TNF-α-mediated LCN2 upregulation in PC-3 cells. Likewise, the NF-κB inhibitor QNZ was able to repress TNF-α-induced LCN2 expression in PC-3 cells. Taking into consideration that LCN2 has been described as a tumor promoting factor in PCa, our results indicate that JNK regulates LCN2 expression and unmasks the JNK signaling axis as a possible therapeutic target for patients with PCa.
Collapse
Affiliation(s)
- Sarah K Schröder
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, Aachen, Germany
| | - Anastasia Asimakopoulou
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, Aachen, Germany
| | - Stefan Tillmann
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Steffen Koschmieder
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, Aachen, Germany.
| |
Collapse
|