1
|
Yan Z, Ji L. Hck promotes IL-1β-induced extracellular matrix degradation, inflammation, and apoptosis in osteoarthritis via activation of the JAK-STAT3 signaling pathway. Adv Rheumatol 2024; 64:88. [PMID: 39696562 DOI: 10.1186/s42358-024-00427-2] [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: 08/28/2024] [Accepted: 11/28/2024] [Indexed: 12/20/2024] Open
Abstract
We investigated role of haematopoietic cell kinase (Hck) in osteoarthritis (OA) and to explore the underlying mechanisms driving its effects. An OA animal model was established and after OA induction, rats received intra-articular injections of lentivirus twice a week for four weeks. Rats were divided into four groups: control (healthy rats without OA), OA model (rats with induced OA), OA + Len-si-NC (OA rats treated with a non-targeting control lentivirus), and OA + Len-si-Hck (OA rats treated with lentivirus targeting Hck). Blood samples were collected, and serum cytokine levels were measured using ELISA. Afterward, the rats were sacrificed for histological analysis and TUNEL assay. In vitro, IL-1β-treated human chondrocytes were transfected with Hck, and the effects on cell viability, apoptosis, ECM degradation, and JAK-STAT3 signaling were assessed. Colivelin, a JAK-STAT3 agonist, was used to confirm the pathway's involvement. Results indicated increased Hck expression in the cartilage tissues of OA rats and in IL-1β-stimulated chondrocytes. Silencing Hck in vivo reduced IL-6 and TNF-α levels, apoptosis, and preserved cartilage structure. In vitro, Hck knockdown in IL-1β-treated chondrocytes resulted in enhanced cell viability, reduced apoptosis, and decreased ECM degradation. Notably, the expression of MMP3 and MMP13 was significantly lowered, while collagen II and aggrecan levels were restored. Additionally, Hck knockdown inhibited JAK-STAT3 activation, which was evident from reduced levels of phosphorylated JAK1 and STAT3. The addition of colivelin reversed these effects, confirming that Hck mediates its effects through the JAK-STAT3 pathway. Overall, our findings indicate that Hck is critical in OA progression by promoting inflammation, apoptosis, and ECM degradation through the JAK-STAT3 signaling pathway activation.
Collapse
Affiliation(s)
- Zhenzhong Yan
- Department of Orthopedics, Changzhou No. 7 People's Hospital, No. 288 Yanling East Road, Economic Development Zone, Changzhou, Jiangsu, 213100, China
- Department of Orthopedics, Changzhou Geriatric Hospital Affiliated to Soochow University, No. 288 Yanling East Road, Economic Development Zone, Changzhou, Jiangsu, 213100, China
| | - Lin Ji
- Department of Orthopedics, Changzhou No. 7 People's Hospital, No. 288 Yanling East Road, Economic Development Zone, Changzhou, Jiangsu, 213100, China.
- Department of Orthopedics, Changzhou Geriatric Hospital Affiliated to Soochow University, No. 288 Yanling East Road, Economic Development Zone, Changzhou, Jiangsu, 213100, China.
| |
Collapse
|
2
|
Wang Y, Yao J, Zhang Z, Wei L, Wang S. Generation of novel lipid metabolism-based signatures to predict prognosis and immunotherapy response for colorectal adenocarcinoma. Sci Rep 2024; 14:17158. [PMID: 39060344 PMCID: PMC11282063 DOI: 10.1038/s41598-024-67549-x] [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: 01/12/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
Lipid metabolism reprogramming involves in epithelial-mesenchymal transition (EMT), cancer stemness and immune checkpoints (ICs), which influence the metastasis of cancer. This study aimed to generate lipid metabolism-based signatures to predict prognosis, immunotherapy and chemotherapy response for colorectal adenocarcinoma (COAD). Transcriptome data and clinical information of COAD patients were collected from the cancer genome atlas (TCGA) database. The expression of EMT-, stem cell-, and IC-related genes were assessed between COAD and control samples. Modules and genes correlated EMT, ICs and stemness signatures were identified through weighted gene co-expression network analysis (WGCNA). Prognostic signatures were generated and then the distribution of risk genes was evaluated using single-cell RNA sequencing (scRNA-seq) data from GSE132465 dataset. COAD patients exhibited increased EMT score and stemness along with decreased ICs. Next, 12 hub genes (PIK3CG, ALOX5AP, PIK3R5, TNFAIP8L2, DPEP2, PIK3CD, PIK3R6, GGT5, ELOVL4, PTGIS, CYP7B1 and PRKD1) were found within green and yellow modules correlated with EMT, stemness and ICs. Lipid metabolism-based prognostic signatures were generated based on PIK3CG, GGT5 and PTGIS. Patients with high-risk group had poor prognosis, elevated ESTIMATEScore and StromalScore, 100% mutation rate and higher TIDE score. Samples in low-risk group had more immunogenicity on ICIs. Notably, PIK3CG was expressed in B cells, while GGT5 and PTGIS were expressed in stromal cells. This study generates lipid metabolism-based signatures correlated with EMT, stemness and ICs for predicting prognosis of COAD, and provides potential therapeutic targets for immunotherapy in COAD.
Collapse
Affiliation(s)
- Yi Wang
- Department of Oncology and Hematology, Suzhou Kowloon Hospital, Shanghai Jiao Tong University School of Medicine, Suzhou, 215127, China
| | - Jun Yao
- Department of General Surgery, The Fourth Affiliated Hospital of Soochow University, Suzhou, 215127, China
| | - Zhe Zhang
- Department of General Surgery, The Fourth Affiliated Hospital of Soochow University, Suzhou, 215127, China
| | - Luxin Wei
- Department of General Surgery, The Fourth Affiliated Hospital of Soochow University, Suzhou, 215127, China
| | - Sheng Wang
- Department of General Surgery, The Fourth Affiliated Hospital of Soochow University, Suzhou, 215127, China.
| |
Collapse
|
3
|
Carvalho MFL, de Almeida BO, Bueno MLP, Vicari HP, Lima K, Rego EM, Roversi FM, Machado-Neto JA. Comprehensive analysis of the HCK gene in myeloid neoplasms: Insights into biological functions, prognosis, and response to antineoplastic agents. Hematol Transfus Cell Ther 2024; 46:273-282. [PMID: 38326180 PMCID: PMC11221266 DOI: 10.1016/j.htct.2023.11.007] [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: 08/19/2023] [Revised: 11/21/2023] [Accepted: 11/29/2023] [Indexed: 02/09/2024] Open
Abstract
Myeloid neoplasms result from molecular alterations in hematopoietic stem cells, with acute myeloid leukemia (AML) being one of the most aggressive and with a poor prognosis. Hematopoietic cell kinase (HCK) is a proto-oncogene that encodes a protein-tyrosine kinase of the Scr family, and it is highly expressed in AML. The present study investigated HCK expression in normal hematopoietic cells across myeloid differentiation stages and myeloid neoplasm patients. Within the AML cohort, we explored the impact of HCK expression on clinical outcomes and its correlation with clinical, genetic, and laboratory characteristics. Furthermore, we evaluated the association between HCK expression and the response to antineoplastic agents using ex vivo assay data from AML patients. HCK expression is higher in differentiated subpopulations of myeloid cells. High HCK expression was observed in patients with chronic myelomonocytic leukemia, chronic myeloid leukemia, and AML. In patients with AML, high levels of HCK negatively impacted overall and disease-free survival. High HCK expression was also associated with worse molecular risk groups and white blood cell count; however, it was not an independent prognostic factor. In functional genomic analyses, high HCK expression was associated with several biological and molecular processes relevant to leukemogenesis. HCK expression was also associated with sensitivity and resistance to several drugs currently used in the clinic. In conclusion, our analysis confirmed the differential expression of HCK in myeloid neoplasms and its potential association with unfavorable molecular risks in AML. We also provide new insights into HCK biological functions, prognosis, and response to antineoplastic agents.
Collapse
Affiliation(s)
| | | | - Maura Lima Pereira Bueno
- Hematology and Transfusion Medicine Center, University of Campinas, Hemocentro-UNICAMP, Campinas, São Paulo, Brazil
| | - Hugo Passos Vicari
- Institute of Biomedical Sciences, University of São Paulo (USP), SP, Brazil
| | - Keli Lima
- Institute of Biomedical Sciences, University of São Paulo (USP), SP, Brazil; Laboratory of Medical Investigation in Pathogenesis and Targeted Therapy in Onco-Immuno-Hematology (LIM-31), Department of Internal Medicine, Hematology Division, Faculdade de Medicina, University of São Paulo, São Paulo, Brazil
| | - Eduardo Magalhães Rego
- Laboratory of Medical Investigation in Pathogenesis and Targeted Therapy in Onco-Immuno-Hematology (LIM-31), Department of Internal Medicine, Hematology Division, Faculdade de Medicina, University of São Paulo, São Paulo, Brazil
| | - Fernanda Marconi Roversi
- Hematology and Transfusion Medicine Center, University of Campinas, Hemocentro-UNICAMP, Campinas, São Paulo, Brazil; Department of Surgery Division Emory University, Atlanta, GA, USA
| | | |
Collapse
|
4
|
Shang Z, Fan Y, Xi S, Zhang S, Shen W, Tao L, Xu C, Tan J, Fan M, Ma H, Lai Y, Sun D, Cheng H. Arenobufagin enhances T-cell anti-tumor immunity in colorectal cancer by modulating HSP90β accessibility. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155497. [PMID: 38640855 DOI: 10.1016/j.phymed.2024.155497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 02/01/2024] [Accepted: 02/26/2024] [Indexed: 04/21/2024]
Abstract
BACKGROUND Colorectal cancer (CRC) is a significant public health issue, ranking as one of the predominant cancer types globally in terms of incidence. Intriguingly, Arenobufagin (Are), a compound extracted from toad venom, has demonstrated the potential to inhibit tumor growth effectively. PURPOSE This study aimed to explore Are's molecular targets and unravel its antitumor mechanism in CRC. Specifically, we were interested in its impact on immune checkpoint modulation and correlations with HSP90β-STAT3-PD-L1 axis activity. METHODS We investigated the in vivo antitumor effects of Are by constructing a colorectalcancer subcutaneous xenograft mouse model. Subsequently, we employed single-cell multi-omics technology to study the potential mechanism by which Are inhibits CRC. Utilizing target-responsive accessibility profiling (TRAP) technology, we identified heatshock protein 90β (HSP90β) as the direct target of Are, and confirmed this through a microscale thermophoresis experiment (MST). Further downstream mechanisms were explored through techniques such as co-immunoprecipitation, Western blotting, qPCR, and immunofluorescence. Concurrently, we arrived at the same research conclusion at the organoid level by co-cultivating with immune cells. RESULTS We observed that Are inhibits PD-Ll expression in CRC tumor xenografts at low concentrations. Moreover, TRAP revealed that HSP90β's accessibility significantly decreased upon Are binding. We demonstrated a decrease in the activity of the HSP90β-STAT3-PD-Ll axis following low-concentration Are treatment in vivo. The PDO analysis showed improved enrichment of lymphocytes, particularly T cells, on the PDOs following Are treatment. CONCLUSION Contrary to previous research focusing on the direct cytotoxicity of Are towards tumor cells, our findings indicate that it can also inhibit tumor growth at lower concentrations through the modulation of immune checkpoints. This study unveils a novel anti-tumor mechanism of Are and stimulates contemplation on the dose-response relationship of natural products, which is beneficial for the clinical translational application of Are.
Collapse
Affiliation(s)
- Zhihao Shang
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, 210046, China; The First School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, 210046, China
| | - Yiping Fan
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, 210046, China; The First School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, 210046, China; Jiaxing Hospital of Traditional Chinese Medicine, Jiaxing 314000, China
| | - Songyang Xi
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, 210046, China; The First School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, 210046, China; Zhenjiang Hospital of Chinese Traditional and Western Medicine, Zhenjiang, 212000, China
| | - Shang Zhang
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, 210046, China; The First School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, 210046, China
| | - Weixing Shen
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, 210046, China; The First School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, 210046, China
| | - Lihuiping Tao
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, 210046, China; The First School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, 210046, China
| | - Changliang Xu
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, 210046, China; The First School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, 210046, China
| | - Jiani Tan
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, 210046, China; The First School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, 210046, China
| | - Minmin Fan
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, 210046, China; The First School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, 210046, China
| | - Hongyue Ma
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210046, China
| | - Yueyang Lai
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, 210046, China; The First School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, 210046, China.
| | - Dongdong Sun
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, 210046, China; School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210046, China.
| | - Haibo Cheng
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, 210046, China; The First School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, 210046, China.
| |
Collapse
|
5
|
Thilakasiri P, O'Keefe RN, To SQ, Chisanga D, Eissmann MF, Carli ALE, Duscio B, Baloyan D, Dmello RS, Williams D, Mariadason J, Poh AR, Pal B, Kile BT, Vissers JH, Harvey KF, Buchert M, Shi W, Ernst M, Chand AL. Mechanisms of cellular crosstalk in the gastric tumor microenvironment are mediated by YAP1 and STAT3. Life Sci Alliance 2024; 7:e202302411. [PMID: 37957015 PMCID: PMC10643184 DOI: 10.26508/lsa.202302411] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 10/29/2023] [Accepted: 10/31/2023] [Indexed: 11/16/2023] Open
Abstract
Deregulation of the Hippo pathway is a driver for cancer progression and treatment resistance. In the context of gastric cancer, YAP1 is a biomarker for poor patient prognosis. Although genomic tumor profiling provides information of Hippo pathway activation, the present study demonstrates that inhibition of Yap1 activity has anti-tumor effects in gastric tumors driven by oncogenic mutations and inflammatory cytokines. We show that Yap1 is a key regulator of cell metabolism, proliferation, and immune responses in normal and neoplastic gastric epithelium. We propose that the Hippo pathway is targetable across gastric cancer subtypes and its therapeutic benefits are likely to be mediated by both cancer cell-intrinsic and -extrinsic mechanisms.
Collapse
Affiliation(s)
- Pathum Thilakasiri
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Heidelberg, Australia
| | - Ryan N O'Keefe
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Heidelberg, Australia
| | - Sarah Q To
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Heidelberg, Australia
| | - David Chisanga
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Heidelberg, Australia
| | - Moritz F Eissmann
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Heidelberg, Australia
| | - Annalisa LE Carli
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Heidelberg, Australia
| | - Belinda Duscio
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Heidelberg, Australia
| | - David Baloyan
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Heidelberg, Australia
| | - Rhynelle S Dmello
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Heidelberg, Australia
| | - David Williams
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Heidelberg, Australia
- Department of Pathology, Austin Health, Heidelberg, Australia
| | - John Mariadason
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Heidelberg, Australia
| | - Ashleigh R Poh
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Heidelberg, Australia
| | - Bhupinder Pal
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Heidelberg, Australia
| | - Benjamin T Kile
- Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia
| | | | - Kieran F Harvey
- Peter MacCallum Cancer Centre, Melbourne, Australia
- Department of Anatomy and Developmental Biology, and Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Michael Buchert
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Heidelberg, Australia
| | - Wei Shi
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Heidelberg, Australia
| | - Matthias Ernst
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Heidelberg, Australia
| | - Ashwini L Chand
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Heidelberg, Australia
| |
Collapse
|
6
|
Choi YJ, Choi M, Park J, Park M, Kim MJ, Lee JS, Oh SJ, Lee YJ, Shim WS, Kim JW, Kim MJ, Kim YC, Kang KW. Therapeutic strategy using novel RET/YES1 dual-target inhibitor in lung cancer. Biomed Pharmacother 2024; 171:116124. [PMID: 38198957 DOI: 10.1016/j.biopha.2024.116124] [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: 08/04/2023] [Revised: 12/20/2023] [Accepted: 01/02/2024] [Indexed: 01/12/2024] Open
Abstract
Lung cancer represents a significant global health concern and stands as the leading cause of cancer-related mortality worldwide. The identification of specific genomic alterations such as EGFR and KRAS in lung cancer has paved the way for the development of targeted therapies. While targeted therapies for lung cancer exhibiting EGFR, MET and ALK mutations have been well-established, the options for RET mutations remain limited. Importantly, RET mutations have been found to be mutually exclusive from other genomic mutations and to be related with high incidences of brain metastasis. Given these facts, it is imperative to explore the development of RET-targeting therapies and to elucidate the mechanisms underlying metastasis in RET-expressing lung cancer cells. In this study, we investigated PLM-101, a novel dual-target inhibitor of RET/YES1, which exhibits notable anti-cancer activities against CCDC6-RET-positive cancer cells and anti-metastatic effects against YES1-positive cancer cells. Our findings shed light on the significance of the YES1-Cortactin-actin remodeling pathway in the metastasis of lung cancer cells, establishing YES1 as a promising target for suppression of metastasis. This paper unveils a novel inhibitor that effectively targets both RET and YES1, thereby demonstrating its potential to impede the growth and metastasis of RET rearrangement lung cancer.
Collapse
Affiliation(s)
- Yong June Choi
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Munkyung Choi
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Jaewoo Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Miso Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea; Department of Pharmacy, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Myung Jun Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Jae-Sun Lee
- R&D Center, PeLeMed, Co. Ltd., Seoul 06100, Republic of Korea
| | - Su-Jin Oh
- R&D Center, PeLeMed, Co. Ltd., Seoul 06100, Republic of Korea
| | - Young Joo Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Wan Seob Shim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Ji Won Kim
- Jeju Research Institute of Pharmaceutical Sciences, College of Pharmacy, Jeju National University, Jeju 63243, Republic of Korea
| | - Myung Jin Kim
- R&D Center, PeLeMed, Co. Ltd., Seoul 06100, Republic of Korea
| | - Yong-Chul Kim
- R&D Center, PeLeMed, Co. Ltd., Seoul 06100, Republic of Korea; School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Keon Wook Kang
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
7
|
O'Keefe RN, Carli ALE, Baloyan D, Chisanga D, Shi W, Afshar-Sterle S, Eissmann MF, Poh AR, Pal B, Seillet C, Locksley RM, Ernst M, Buchert M. A tuft cell - ILC2 signaling circuit provides therapeutic targets to inhibit gastric metaplasia and tumor development. Nat Commun 2023; 14:6872. [PMID: 37898600 PMCID: PMC10613282 DOI: 10.1038/s41467-023-42215-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 10/04/2023] [Indexed: 10/30/2023] Open
Abstract
Although gastric cancer is a leading cause of cancer-related deaths, systemic treatment strategies remain scarce. Here, we report the pro-tumorigenic properties of the crosstalk between intestinal tuft cells and type 2 innate lymphoid cells (ILC2) that is evolutionarily optimized for epithelial remodeling in response to helminth infection. We demonstrate that tuft cell-derived interleukin 25 (IL25) drives ILC2 activation, inducing the release of IL13 and promoting epithelial tuft cell hyperplasia. While the resulting tuft cell - ILC2 feed-forward circuit promotes gastric metaplasia and tumor formation, genetic depletion of tuft cells or ILC2s, or therapeutic targeting of IL13 or IL25 alleviates these pathologies in mice. In gastric cancer patients, tuft cell and ILC2 gene signatures predict worsening survival in intestinal-type gastric cancer where ~40% of the corresponding cancers show enriched co-existence of tuft cells and ILC2s. Our findings suggest a role for ILC2 and tuft cells, along with their associated cytokine IL13 and IL25 as gatekeepers and enablers of metaplastic transformation and gastric tumorigenesis, thereby providing an opportunity to therapeutically inhibit early-stage gastric cancer through repurposing antibody-mediated therapies.
Collapse
Affiliation(s)
- Ryan N O'Keefe
- Olivia Newton-John Cancer Research Institute, Heidelberg, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, Australia
| | - Annalisa L E Carli
- Olivia Newton-John Cancer Research Institute, Heidelberg, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, Australia
| | - David Baloyan
- Olivia Newton-John Cancer Research Institute, Heidelberg, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, Australia
| | - David Chisanga
- Olivia Newton-John Cancer Research Institute, Heidelberg, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, Australia
| | - Wei Shi
- Olivia Newton-John Cancer Research Institute, Heidelberg, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, Australia
| | - Shoukat Afshar-Sterle
- Olivia Newton-John Cancer Research Institute, Heidelberg, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, Australia
| | - Moritz F Eissmann
- Olivia Newton-John Cancer Research Institute, Heidelberg, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, Australia
| | - Ashleigh R Poh
- Olivia Newton-John Cancer Research Institute, Heidelberg, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, Australia
| | - Bhupinder Pal
- Olivia Newton-John Cancer Research Institute, Heidelberg, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, Australia
| | - Cyril Seillet
- Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Richard M Locksley
- Department of Medicine, University of California San Francisco, San Francisco, USA
- Howard Hughes Medical Institute, University of California San Francisco, San Francisco, USA
| | - Matthias Ernst
- Olivia Newton-John Cancer Research Institute, Heidelberg, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, Australia
| | - Michael Buchert
- Olivia Newton-John Cancer Research Institute, Heidelberg, Australia.
- School of Cancer Medicine, La Trobe University, Bundoora, Australia.
| |
Collapse
|
8
|
Chen M, Menon MC, Wang W, Fu J, Yi Z, Sun Z, Liu J, Li Z, Mou L, Banu K, Lee SW, Dai Y, Anandakrishnan N, Azeloglu EU, Lee K, Zhang W, Das B, He JC, Wei C. HCK induces macrophage activation to promote renal inflammation and fibrosis via suppression of autophagy. Nat Commun 2023; 14:4297. [PMID: 37463911 PMCID: PMC10354075 DOI: 10.1038/s41467-023-40086-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 07/12/2023] [Indexed: 07/20/2023] Open
Abstract
Renal inflammation and fibrosis are the common pathways leading to progressive chronic kidney disease (CKD). We previously identified hematopoietic cell kinase (HCK) as upregulated in human chronic allograft injury promoting kidney fibrosis; however, the cellular source and molecular mechanisms are unclear. Here, using immunostaining and single cell sequencing data, we show that HCK expression is highly enriched in pro-inflammatory macrophages in diseased kidneys. HCK-knockout (KO) or HCK-inhibitor decreases macrophage M1-like pro-inflammatory polarization, proliferation, and migration in RAW264.7 cells and bone marrow-derived macrophages (BMDM). We identify an interaction between HCK and ATG2A and CBL, two autophagy-related proteins, inhibiting autophagy flux in macrophages. In vivo, both global or myeloid cell specific HCK-KO attenuates renal inflammation and fibrosis with reduces macrophage numbers, pro-inflammatory polarization and migration into unilateral ureteral obstruction (UUO) kidneys and unilateral ischemia reperfusion injury (IRI) models. Finally, we developed a selective boron containing HCK inhibitor which can reduce macrophage pro-inflammatory activity, proliferation, and migration in vitro, and attenuate kidney fibrosis in the UUO mice. The current study elucidates mechanisms downstream of HCK regulating macrophage activation and polarization via autophagy in CKD and identifies that selective HCK inhibitors could be potentially developed as a new therapy for renal fibrosis.
Collapse
Affiliation(s)
- Man Chen
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Critical Care Medicine, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, China
- Department of Critical Care Medicine, Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Madhav C Menon
- Division of Nephrology, Yale School of Medicine, New Haven, CT, USA
| | - Wenlin Wang
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jia Fu
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Zhengzi Yi
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Zeguo Sun
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jessica Liu
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Zhengzhe Li
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lingyun Mou
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Khadija Banu
- Division of Nephrology, Yale School of Medicine, New Haven, CT, USA
| | - Sui-Wan Lee
- Center for Comparative Medicine and Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ying Dai
- Center for Comparative Medicine and Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nanditha Anandakrishnan
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Evren U Azeloglu
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kyung Lee
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Weijia Zhang
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bhaskar Das
- Arnold and Marie Schwartz College of Pharmacy and Health Sciences, Long Island University, Brooklyn, NY, USA.
| | - John Cijiang He
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Renal Section, James J. Peters VAMC, Bronx, NY, USA.
| | - Chengguo Wei
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
9
|
Dunker W, Zaver SA, Pineda JMB, Howard CJ, Bradley RK, Woodward JJ. The proto-oncogene SRC phosphorylates cGAS to inhibit an antitumor immune response. JCI Insight 2023; 8:e167270. [PMID: 37166992 PMCID: PMC10371251 DOI: 10.1172/jci.insight.167270] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 05/09/2023] [Indexed: 05/12/2023] Open
Abstract
Cyclic GMP-AMP synthase (cGAS) is a DNA sensor and responsible for inducing an antitumor immune response. Recent studies reveal that cGAS is frequently inhibited in cancer, and therapeutic targets to promote antitumor cGAS function remain elusive. SRC is a proto-oncogene tyrosine kinase and is expressed at elevated levels in numerous cancers. Here, we demonstrate that SRC expression in primary and metastatic bladder cancer negatively correlates with innate immune gene expression and immune cell infiltration. We determine that SRC restricts cGAS signaling in human cell lines through SRC small molecule inhibitors, depletion, and overexpression. cGAS and SRC interact in cells and in vitro, while SRC directly inhibits cGAS enzymatic activity and DNA binding in a kinase-dependent manner. SRC phosphorylates cGAS, and inhibition of cGAS Y248 phosphorylation partially reduces SRC inhibition. Collectively, our study demonstrates that cGAS antitumor signaling is hindered by the proto-oncogene SRC and describes how cancer-associated proteins can regulate the innate immune system.
Collapse
Affiliation(s)
| | - Shivam A. Zaver
- Department of Microbiology and
- Medical Scientist Training Program, University of Washington, Seattle, Washington, USA
| | - Jose Mario Bello Pineda
- Medical Scientist Training Program, University of Washington, Seattle, Washington, USA
- Public Health Sciences and Basic Sciences Divisions, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
| | | | - Robert K. Bradley
- Public Health Sciences and Basic Sciences Divisions, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
| | | |
Collapse
|
10
|
Xia S, Li JD, Yan SB, Huang ZG, Liu ZS, Jing SW, Li DZ, Song C, Chen Y, Wang LT, Zhou YH, Huang R, Shi N, Lan SY, Chen G, Fan XH. Clinicopathological value of hematopoietic cell kinase overexpression in laryngeal squamous cell carcinoma tissues. Pathol Res Pract 2023; 247:154534. [PMID: 37201466 DOI: 10.1016/j.prp.2023.154534] [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: 11/21/2022] [Revised: 03/26/2023] [Accepted: 05/11/2023] [Indexed: 05/20/2023]
Abstract
Laryngeal squamous cell carcinoma (LSCC) is the most lethal cancer in head and neck tumors. Although hematopoietic cell kinase (HCK) has been proven to be an oncogene in several solid tumors, its roles in LSCC remain obscure. This is the first study to evaluate the clinical value of HCK in LSCC, with the aim of exploring its expression status and potential molecular mechanisms underlying LSCC. LSCC tissue-derived gene chips and RNA-seq data were collected for a quantitive integration of HCK mRNA expression level. To confirm the protein expression level of HCK, a total of 82 LSCC tissue specimens and 56 non-tumor laryngeal epithelial controls were collected for in-house tissue microarrays and immunohistochemical staining. Kaplan-Meier curves were generated to determine the ability of HCK in predicting overall survival, progress-free survival, and disease-free survival of LSCC patients. LSCC overexpressed genes and HCK co-expressed genes were intersected to preliminarily explore the enriched signaling pathways of HCK. It was noticed that HCK mRNA was markedly overexpressed in 323 LSCC tissues compared with 196 non-LSCC controls (standardized mean difference = 0.81, p < 0.0001). Upregulated HCK mRNA displayed a moderate discriminatory ability between LSCC tissues and non-tumor laryngeal epithelial controls (area under the curve = 0.78, sensitivity = 0.76, specificity = 0.68). The higher expression level of HCK mRNA could predict worse overall survival and disease-free survival for LSCC patients (p = 0.041 and p = 0.013). Lastly, upregulated co-expression genes of HCK were significantly enriched in leukocyte cell-cell adhesion, secretory granule membrane, and extracellular matrix structural constituent. Immune-related pathways were the predominantly activated signals, such as cytokine-cytokine receptor interaction, Th17 cell differentiation, and Toll-like receptor signaling pathway. In conclusion, HCK was upregulated in LSCC tissues and could be utilized as a risk predictor. HCK may promote the development of LSCC by disturbing immune signaling pathways.
Collapse
Affiliation(s)
- Shuang Xia
- Department of Human Anatomy, Guangxi Medical University, 22 Shuangyong Road, Nanning, Guangxi, Zhuang Autonomous Region 530021, PR China
| | - Jian-Di Li
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, Guangxi, Zhuang Autonomous Region 530021, PR China
| | - Shi-Bai Yan
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, Guangxi, Zhuang Autonomous Region 530021, PR China
| | - Zhi-Guang Huang
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, Guangxi, Zhuang Autonomous Region 530021, PR China
| | - Zhi-Su Liu
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, Guangxi, Zhuang Autonomous Region 530021, PR China
| | - Shu-Wen Jing
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, Guangxi, Zhuang Autonomous Region 530021, PR China
| | - Da-Zhi Li
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, Guangxi, Zhuang Autonomous Region 530021, PR China
| | - Chang Song
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, Guangxi, Zhuang Autonomous Region 530021, PR China
| | - Yi Chen
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, Guangxi, Zhuang Autonomous Region 530021, PR China
| | - Li-Ting Wang
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, Guangxi, Zhuang Autonomous Region 530021, PR China
| | - Yu-Hong Zhou
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, Guangxi, Zhuang Autonomous Region 530021, PR China
| | - Rong Huang
- Department of Human Anatomy, Guangxi Medical University, 22 Shuangyong Road, Nanning, Guangxi, Zhuang Autonomous Region 530021, PR China
| | - Nan Shi
- Department of Otorhinolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, Guangxi, Zhuang Autonomous Region 530021, PR China
| | - Song-Yao Lan
- Department of Human Anatomy, Guangxi Medical University, 22 Shuangyong Road, Nanning, Guangxi, Zhuang Autonomous Region 530021, PR China
| | - Gang Chen
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, Guangxi, Zhuang Autonomous Region 530021, PR China
| | - Xiao-Hui Fan
- Department of Immunology and Microbiology, Guangxi Medical University, 22 Shuangyong Road, Nanning, Guangxi, Zhuang Autonomous Region 530021, PR China.
| |
Collapse
|
11
|
Luo S, Du S, Tao M, Cao J, Cheng P. Insights on hematopoietic cell kinase: An oncogenic player in human cancer. Biomed Pharmacother 2023; 160:114339. [PMID: 36736283 DOI: 10.1016/j.biopha.2023.114339] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/18/2023] [Accepted: 01/27/2023] [Indexed: 02/04/2023] Open
Abstract
Hematopoietic cell kinase (Hck) is a member of the Src family and is expressed in hematopoietic cells. By regulating multiple signaling pathways, HCK can interact with multiple receptors to regulate signaling events involved in cell adhesion, proliferation, migration, invasion, apoptosis, and angiogenesis. However, aberrant expression of Hck in various hematopoietic cells and solid tumors plays a crucial role in tumor-related properties, including cell proliferation and epithelial-mesenchymal transition. In addition, Hck signaling regulates the function of immune cells such as macrophages, contributing to an immunosuppressive tumor microenvironment. The clinical success of various kinase inhibitors targeting the Src kinase family has validated the efficacy of targeting Src, and therapies with highly selective Hck kinase inhibitors are in clinical trials. This article reviews Hck inhibition as an emerging cancer treatment strategy, focusing on the expressions and functions of Hck in tumors and its impact on the tumor microenvironment. It also explores preclinical and clinical pharmacological strategies for Hck targeting to shed light on Hck-targeted tumor therapy.
Collapse
Affiliation(s)
- Shuyan Luo
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Shaonan Du
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Mei Tao
- Department of Thyroid and Neck Tumor, Tianjin Medical University Cancer Institute and Hospital, 300060 Tianjin, China
| | - Jingyuan Cao
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Peng Cheng
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China.
| |
Collapse
|
12
|
The link between rheumatic disorders and inborn errors of immunity. EBioMedicine 2023; 90:104501. [PMID: 36870198 PMCID: PMC9996386 DOI: 10.1016/j.ebiom.2023.104501] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 12/11/2022] [Accepted: 02/10/2023] [Indexed: 03/06/2023] Open
Abstract
Inborn errors of immunity (IEIs) are immunological disorders characterized by variable susceptibility to infections, immune dysregulation and/or malignancies, as a consequence of damaging germline variants in single genes. Though initially identified among patients with unusual, severe or recurrent infections, non-infectious manifestations and especially immune dysregulation in the form of autoimmunity or autoinflammation can be the first or dominant phenotypic aspect of IEIs. An increasing number of IEIs causing autoimmunity or autoinflammation, including rheumatic disease have been reported over the last decade. Despite their rarity, identification of those disorders provided insight into the pathomechanisms of immune dysregulation, which may be relevant for understanding the pathogenesis of systemic rheumatic disorders. In this review, we present novel IEIs primarily causing autoimmunity or autoinflammation along with their pathogenic mechanisms. In addition, we explore the likely pathophysiological and clinical relevance of IEIs in systemic rheumatic disorders.
Collapse
|
13
|
Yang F, Zhang D, Jiang H, Ye J, Zhang L, Bagley SJ, Winkler J, Gong Y, Fan Y. Small-molecule toosendanin reverses macrophage-mediated immunosuppression to overcome glioblastoma resistance to immunotherapy. Sci Transl Med 2023; 15:eabq3558. [PMID: 36791206 PMCID: PMC10394757 DOI: 10.1126/scitranslmed.abq3558] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 01/17/2023] [Indexed: 02/17/2023]
Abstract
T cell-based immunotherapy holds promise for treating solid tumors, but its therapeutic efficacy is limited by intratumoral immune suppression. This immune suppressive tumor microenvironment is largely driven by tumor-associated myeloid cells, including macrophages. Here, we report that toosendanin (TSN), a small-molecule compound, reprograms macrophages to enforce antitumor immunity in glioblastoma (GBM) in mouse models. Our functional screen of genetically probed macrophages with a chemical library identifies that TSN reverses macrophage-mediated tumor immunosuppression, leading to enhanced T cell infiltration, activation, and reduced exhaustion. Chemoproteomic and structural analyses revealed that TSN interacts with Hck and Lyn to abrogate suppressive macrophage immunity. In addition, a combination of immune checkpoint blockade and TSN therapy induced regression of syngeneic GBM tumors in mice. Furthermore, TSN treatment sensitized GBM to Egfrviii chimeric antigen receptor (CAR) T cell therapy. These findings suggest that TSN may serve as a therapeutic compound that blocks tumor immunosuppression and circumvents tumor resistance to T cell-based immunotherapy in GBM and other solid tumors that warrants further investigation.
Collapse
Affiliation(s)
- Fan Yang
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Duo Zhang
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Haowen Jiang
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA
| | - Jiangbin Ye
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA
| | - Lin Zhang
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Stephen J. Bagley
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jeffery Winkler
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yanqing Gong
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yi Fan
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
14
|
Morrow RJ, Allam AH, Konecnik J, Baloyan D, Dijkstra C, Eissmann MF, Jacob SP, O’Brien M, Poh AR, Ernst M. Tumor Growth Remains Refractory to Myc Ablation in Host Macrophages. Cells 2022; 11:cells11244104. [PMID: 36552868 PMCID: PMC9777527 DOI: 10.3390/cells11244104] [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: 11/17/2022] [Revised: 12/06/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Aberrant expression of the oncoprotein c-Myc (Myc) is frequently observed in solid tumors and is associated with reduced overall survival. In addition to well-recognized cancer cell-intrinsic roles of Myc, studies have also suggested tumor-promoting roles for Myc in cells of the tumor microenvironment, including macrophages and other myeloid cells. Here, we benchmark Myc inactivation in tumor cells against the contribution of its expression in myeloid cells of murine hosts that harbor endogenous or allograft tumors. Surprisingly, we observe that LysMCre-mediated Myc ablation in host macrophages does not attenuate tumor growth regardless of immunogenicity, the cellular origin of the tumor, the site it develops, or the stage along the tumor progression cascade. Likewise, we find no evidence for Myc ablation to revert or antagonize the polarization of alternatively activated immunosuppressive macrophages. Thus, we surmise that systemic targeting of Myc activity may confer therapeutic benefits primarily through limiting Myc activity in tumor cells rather than reinvigorating the anti-tumor activity of macrophages.
Collapse
|
15
|
Rouzbahani E, Majidpoor J, Najafi S, Mortezaee K. Cancer stem cells in immunoregulation and bypassing anti-checkpoint therapy. Biomed Pharmacother 2022; 156:113906. [DOI: 10.1016/j.biopha.2022.113906] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/16/2022] [Accepted: 10/19/2022] [Indexed: 11/26/2022] Open
|
16
|
Daly RJ, Scott AM, Klein O, Ernst M. Enhancing therapeutic anti-cancer responses by combining immune checkpoint and tyrosine kinase inhibition. Mol Cancer 2022; 21:189. [PMID: 36175961 PMCID: PMC9523960 DOI: 10.1186/s12943-022-01656-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 09/19/2022] [Indexed: 11/10/2022] Open
Abstract
Over the past decade, immune checkpoint inhibitor (ICI) therapy has been established as the standard of care for many types of cancer, but the strategies employed have continued to evolve. Recently, much clinical focus has been on combining targeted therapies with ICI for the purpose of manipulating the immune setpoint. The latter concept describes the equilibrium between factors that promote and those that suppress anti-cancer immunity. Besides tumor mutational load and other cancer cell-intrinsic determinants, the immune setpoint is also governed by the cells of the tumor microenvironment and how they are coerced by cancer cells to support the survival and growth of the tumor. These regulatory mechanisms provide therapeutic opportunities to intervene and reduce immune suppression via application of small molecule inhibitors and antibody-based therapies against (receptor) tyrosine kinases and thereby improve the response to ICIs. This article reviews how tyrosine kinase signaling in the tumor microenvironment can promote immune suppression and highlights how therapeutic strategies directed against specific tyrosine kinases can be used to lower the immune setpoint and elicit more effective anti-tumor immunity.
Collapse
Affiliation(s)
- Roger J Daly
- Cancer Program, Monash Biomedicine Discovery Institute, Monash University, 23 Innovation Walk, Clayton, VIC, 3800, Australia.
- Department of Biochemistry & Molecular Biology, Monash University, 23 Innovation Walk, Clayton, VIC, 3800, Australia.
| | - Andrew M Scott
- Department of Biochemistry & Molecular Biology, Monash University, 23 Innovation Walk, Clayton, VIC, 3800, Australia
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, 145 Studley Rd, Melbourne-Heidelberg, VIC, 3084, Australia
- Department of Molecular Imaging & Therapy, Austin Health, and Faculty of Medicine, University of Melbourne, 145 Studley Rd, Melbourne-Heidelberg, VIC, 3084, Australia
| | - Oliver Klein
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, 145 Studley Rd, Melbourne-Heidelberg, VIC, 3084, Australia
| | - Matthias Ernst
- Department of Biochemistry & Molecular Biology, Monash University, 23 Innovation Walk, Clayton, VIC, 3800, Australia.
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, 145 Studley Rd, Melbourne-Heidelberg, VIC, 3084, Australia.
| |
Collapse
|
17
|
Wang D, Ye Q, Gu H, Chen Z. The role of lipid metabolism in tumor immune microenvironment and potential therapeutic strategies. Front Oncol 2022; 12:984560. [PMID: 36172157 PMCID: PMC9510836 DOI: 10.3389/fonc.2022.984560] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 08/01/2022] [Indexed: 11/20/2022] Open
Abstract
Aberrant lipid metabolism is nonnegligible for tumor cells to adapt to the tumor microenvironment (TME). It plays a significant role in the amount and function of immune cells, including tumor-associated macrophages, T cells, dendritic cells and marrow-derived suppressor cells. It is well-known that the immune response in TME is suppressed and lipid metabolism is closely involved in this process. Immunotherapy, containing anti-PD1/PDL1 therapy and adoptive T cell therapy, is a crucial clinical cancer therapeutic strategy nowadays, but they display a low-sensibility in certain cancers. In this review, we mainly discussed the importance of lipid metabolism in the formation of immunosuppressive TME, and explored the effectiveness and sensitivity of immunotherapy treatment by regulating the lipid metabolism.
Collapse
Affiliation(s)
- Danting Wang
- Department of Breast Surgery (Surgical Oncology), Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qizhen Ye
- Department of Breast Surgery (Surgical Oncology), Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Haochen Gu
- Department of Breast Surgery (Surgical Oncology), Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhigang Chen
- Department of Breast Surgery (Surgical Oncology), Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University, Hangzhou, China
- Cancer Centre, Zhejiang University, Hangzhou, China
- Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|