1
|
Li C, Syed MU, Nimbalkar A, Shen Y, Vieira MD, Fraser C, Inde Z, Qin X, Ouyang J, Kreuzer J, Clark SE, Kelley G, Hensley EM, Morris R, Lazaro R, Belmonte B, Oh A, Walcott M, Nabel CS, Caenepeel S, Saiki AY, Rex K, Lipford JR, Heist RS, Lin JJ, Haas W, Sarosiek K, Hughes PE, Hata AN. LKB1 regulates JNK-dependent stress signaling and apoptotic dependency of KRAS-mutant lung cancers. Nat Commun 2025; 16:4112. [PMID: 40316540 PMCID: PMC12048556 DOI: 10.1038/s41467-025-58753-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 04/01/2025] [Indexed: 05/04/2025] Open
Abstract
The efficacy of molecularly targeted therapies may be limited by co-occurring mutations within a tumor. Conversely, these alterations may confer collateral vulnerabilities that can be therapeutically leveraged. KRAS-mutant lung cancers are distinguished by recurrent loss of the tumor suppressor STK11/LKB1. Whether LKB1 modulates cellular responses to therapeutic stress seems unknown. Here we show that in LKB1-deficient KRAS-mutant lung cancer cells, inhibition of KRAS or its downstream effector MEK leads to hyperactivation of JNK due to loss of NUAK-mediated PP1B phosphatase activity. JNK-mediated inhibitory phosphorylation of BCL-XL rewires apoptotic dependencies, rendering LKB1-deficient cells vulnerable to MCL-1 inhibition. These results uncover an unknown role for LKB1 in regulating stress signaling and mitochondrial apoptosis independent of its tumor suppressor activity mediated by AMPK and SIK. Additionally, our study reveals a therapy-induced vulnerability in LKB1-deficient KRAS-mutant lung cancers that could be exploited as a genotype-informed strategy to improve the efficacy of KRAS-targeted therapies.
Collapse
Affiliation(s)
- Chendi Li
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | | | | | - Yi Shen
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | | | - Cameron Fraser
- John B. Little Center for Radiation Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Zintis Inde
- John B. Little Center for Radiation Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Xingping Qin
- John B. Little Center for Radiation Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Jian Ouyang
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
- Department of Biochemistry & Molecular Biology and Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Johannes Kreuzer
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Sarah E Clark
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Grace Kelley
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Emily M Hensley
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Robert Morris
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Raul Lazaro
- Amgen Research, Amgen Inc., Thousand Oaks, CA, USA
| | | | - Audris Oh
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Makeba Walcott
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Christopher S Nabel
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
- Koch Institute for Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Anne Y Saiki
- Amgen Research, Amgen Inc., Thousand Oaks, CA, USA
| | - Karen Rex
- Amgen Research, Amgen Inc., Thousand Oaks, CA, USA
| | | | - Rebecca S Heist
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Jessica J Lin
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Wilhelm Haas
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Kristopher Sarosiek
- John B. Little Center for Radiation Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Lab for Systems Pharmacology, Harvard Program in Therapeutics Science, Harvard Medical School, Boston, MA, USA
| | | | - Aaron N Hata
- Massachusetts General Hospital Cancer Center, Boston, MA, USA.
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
2
|
Khalaf MM, Malak MN, Alsahli TG, Althobaiti M, Hamzawy MA, Abdel-Fattah MM. Repurposing bosentan as an anticancer agent: EGFR/ERK/c-Jun modulation inhibits NSCLC tumor growth. Fundam Clin Pharmacol 2025; 39:e13052. [PMID: 39801131 DOI: 10.1111/fcp.13052] [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: 09/09/2024] [Revised: 11/29/2024] [Accepted: 12/18/2024] [Indexed: 05/02/2025]
Abstract
Drug repurposing of well-established drugs to be targeted against lung cancer has been a promising strategy. Bosentan is an endothelin 1 (ET-1) blocker widely used in pulmonary hypertension. The current experiment intends to inspect the anticancer and antiangiogenic mechanism of bosentan targeting epidermal growth factor receptor (EGFR) /extra-cellular Signal Regulated Kinase (ERK) /c-Jun/vascular endothelial growth factor (VEGF) carcinogenic pathway. BALB/c mice were randomized into four groups, the first received the vehicle, the second received 100 mg/kg oral bosentan alone, the third has non-small cell lung cancer (NSCLC) induced by two doses of 1.5 g/kg urethane i.p. and finally the fourth has NSCLC received bosentan. To determine the anti-proliferative impact of bosentan, cytokeratin 19 fragments (CYFRA 21-1) level was assessed, and Ki-67 positive cells were counted by immunohistochemical (IHC). Molecular expression of EGFR via IHC, relative expression of p-ERK1/2 and p-c-Jun via western blotting and caspase 3, Bcl-2 Associated X-protein (BAX)/B-cell lymphoma 2 (Bcl-2) ratio and VEGF via ELISA were quantified. Bosentan showed pronounced improvement in lung index and histopathological examinations. Bosentan exerted a noticeable arrest of lung cancer growth indicated by the attenuation of CYFRA 21-1 and Ki-67 positive cell counts besides the boost of BAX/Bcl-2 ratio and caspase 3. Bosentan induced a remarkable decline of EGFR, T-ERK1/2/p-ERK1/2, T-c-Jun/p-c-Jun, and VEGF. Bosentan induced cytotoxic and anti-angiogenic impact through regulation of EGFR/ERK/c-Jun/VEGF axis suggesting its potential therapeutic impact against lung cancer.
Collapse
Affiliation(s)
- Marwa M Khalaf
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, 62514, Egypt
| | - Marina N Malak
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, 62514, Egypt
| | - Tariq G Alsahli
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka, Aljouf, 72341, Saudi Arabia
| | - Musaad Althobaiti
- Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, Taif, Saudi Arabia
| | - Mohamed A Hamzawy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Fayoum University, Fayoum, 63514, Egypt
| | - Maha M Abdel-Fattah
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, 62514, Egypt
| |
Collapse
|
3
|
Loos B, Salas-Bastos A, Nordin A, Debbache J, Stierli S, Cheng PF, Rufli S, Wyss C, Levesque MP, Dummer R, Wong WWL, Pascolo S, Cantù C, Sommer L. TGFβ signaling sensitizes MEKi-resistant human melanoma to targeted therapy-induced apoptosis. Cell Death Dis 2024; 15:925. [PMID: 39709491 DOI: 10.1038/s41419-024-07305-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 11/29/2024] [Accepted: 12/11/2024] [Indexed: 12/23/2024]
Abstract
The TGFβ signaling pathway is known for its pleiotropic functions in a plethora of biological processes. In melanoma, TGFβ signaling promotes invasiveness and metastasis formation. However, its involvement in the response to therapy is controversial. While several studies have linked TGFβ signaling to elevated resistance to targeted therapy in melanoma, separate findings have indicated a favorable treatment response through TGFβ-mediated increase of cell death. We now found that the outcome of TGFβ signaling in the context of targeted therapy is dose dependent. Unlike low doses, high levels of TGFβ signal activation induce apoptosis upon simultaneous MAPK pathway inhibition, even in targeted therapy resistant melanoma cell lines. Using transcriptomic analyses, combined with genomic target identification of the critical TGFβ signaling effector SMAD4, we demonstrate that parallel activation of TGFβ signaling and MAPK pathway inhibition causes a complete switch of TGFβ target genes from promoting pro-invasive processes to fueling pro-apoptotic pathways. Investigations of underlying mechanisms identified a novel apoptosis-inducing gene signature. Functional validation of signature members highlighted a central role of the pro-apoptotic BCL2 family member BCL2L11 (BIM) in mediating apoptosis in this condition. Using a modified, synthetic version of the TGFB1 mRNA for intra-tumoral injections, we additionally showcase a potential therapeutic application of this treatment combination.
Collapse
Affiliation(s)
- Benjamin Loos
- University of Zürich, Institute of Anatomy, Winterthurerstrasse 190, 8057, Zürich, Switzerland
| | - Adrian Salas-Bastos
- University of Zürich, Institute of Anatomy, Winterthurerstrasse 190, 8057, Zürich, Switzerland
| | - Anna Nordin
- Wallenberg Centre for Molecular Medicine, Linköping University, 58185, Linköping, Sweden
- Department of Biomedical and Clinical Sciences, Division of Molecular Medicine and Virology; Faculty of Medicine and Health Sciences, Linköping University, 58185, Linköping, Sweden
| | - Julien Debbache
- University of Zürich, Institute of Anatomy, Winterthurerstrasse 190, 8057, Zürich, Switzerland
| | - Salome Stierli
- University of Zürich, Institute of Anatomy, Winterthurerstrasse 190, 8057, Zürich, Switzerland
| | - Phil F Cheng
- University of Zürich Hospital, University of Zürich, Department of Dermatology, Raemistrasse 100, 8091, Zürich, Switzerland
| | - Stefanie Rufli
- University of Zurich, Institute of Experimental Immunology, Winterthurerstrasse 190, 8057, Zürich, Switzerland
| | - Conrad Wyss
- University of Zürich Hospital, University of Zürich, Department of Dermatology, Raemistrasse 100, 8091, Zürich, Switzerland
| | - Mitchell P Levesque
- University of Zürich Hospital, University of Zürich, Department of Dermatology, Raemistrasse 100, 8091, Zürich, Switzerland
| | - Reinhard Dummer
- University of Zürich Hospital, University of Zürich, Department of Dermatology, Raemistrasse 100, 8091, Zürich, Switzerland
| | - Wendy Wei-Lynn Wong
- University of Zurich, Institute of Experimental Immunology, Winterthurerstrasse 190, 8057, Zürich, Switzerland
- Department of Molecular Life Sciences, University of Zürich, Zürich, Switzerland
| | - Steve Pascolo
- University of Zürich Hospital, University of Zürich, Department of Dermatology, Raemistrasse 100, 8091, Zürich, Switzerland
- Faculty of Medicine, University of Zürich, Zürich, Switzerland
| | - Claudio Cantù
- Wallenberg Centre for Molecular Medicine, Linköping University, 58185, Linköping, Sweden
- Department of Biomedical and Clinical Sciences, Division of Molecular Medicine and Virology; Faculty of Medicine and Health Sciences, Linköping University, 58185, Linköping, Sweden
| | - Lukas Sommer
- University of Zürich, Institute of Anatomy, Winterthurerstrasse 190, 8057, Zürich, Switzerland.
| |
Collapse
|
4
|
Chen JH, Wei CM, Lin QY, Wang Z, Zhang FM, Shi MN, Lan WJ, Sun CG, Lin WJ, Ma WZ. Notopterygium Incisum Extract Promotes Apoptosis by Preventing the Degradation of BIM in Colorectal Cancer. Curr Med Sci 2024; 44:833-840. [PMID: 38967889 DOI: 10.1007/s11596-024-2883-1] [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: 01/24/2024] [Accepted: 04/09/2024] [Indexed: 07/06/2024]
Abstract
OBJECTIVE Colorectal cancer (CRC), a prevalent malignancy worldwide, has prompted extensive research into anticancer drugs. Traditional Chinese medicinal materials offer promising avenues for cancer management due to their diverse pharmacological activities. This study investigated the effects of Notopterygium incisum, a traditional Chinese medicine named Qianghuo (QH), on CRC cells and the underlying mechanism. METHODS The sulforhodamine B assay and colony formation assay were employed to assess the effect of QH extract on the proliferation of CRC cell lines HCT116 and Caco-2. Propidium iodide (PI) staining was utilized to detect cell cycle progression, and PE Annexin V staining to detect apoptosis. Western blotting was conducted to examine the levels of apoptotic proteins, including B-cell lymphoma 2-interacting mediator of cell death (BIM), B-cell lymphoma 2 (Bcl-2), Bcl-2-associated X protein (BAX) and cleaved caspase-3, as well as BIM stability after treatment with the protein synthesis inhibitor cycloheximide. The expression of BAX was suppressed using lentivirus-mediated shRNA to validate the involvement of the BIM/BAX axis in QH-induced apoptosis. The in vivo effects of QH extract on tumor growth were observed using a xenograft model. Lastly, APCMin+ mice were used to study the effects of QH extract on primary intestinal tumors. RESULTS QH extract exhibited significant in vitro anti-CRC activities evidenced by the inhibition of cell proliferation, perturbation of cell cycle progression, and induction of apoptosis. Mechanistically, QH extract significantly increased the stability of BIM proteins, which undergo rapid degradation under unstressed conditions. Knockdown of BAX, the downstream effector of BIM, significantly rescued QH-induced apoptosis. Furthermore, the in vitro effect of QH extract was recapitulated in vivo. QH extract significantly inhibited the tumor growth of HCT116 xenografts in nude mice and decreased the number of intestinal polyps in the APCMin+ mice. CONCLUSION QH extract promotes the apoptosis of CRC cells by preventing the degradation of BIM.
Collapse
Affiliation(s)
- Jun-He Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, 999078, China
| | - Cheng-Ming Wei
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, 999078, China
| | - Qian-Yu Lin
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, 999078, China
| | - Zi Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, 999078, China
| | - Fu-Ming Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, 999078, China
| | - Mei-Na Shi
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, 999078, China
| | - Wen-Jian Lan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Chang-Gang Sun
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang, 261053, China
| | - Wan-Jun Lin
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, 999078, China
| | - Wen-Zhe Ma
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, 999078, China.
| |
Collapse
|
5
|
Kitai H, Choi PH, Yang YC, Boyer JA, Whaley A, Pancholi P, Thant C, Reiter J, Chen K, Markov V, Taniguchi H, Yamaguchi R, Ebi H, Evans J, Jiang J, Lee B, Wildes D, de Stanchina E, Smith JAM, Singh M, Rosen N. Combined inhibition of KRAS G12C and mTORC1 kinase is synergistic in non-small cell lung cancer. Nat Commun 2024; 15:6076. [PMID: 39025835 PMCID: PMC11258147 DOI: 10.1038/s41467-024-50063-z] [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: 12/21/2023] [Accepted: 06/28/2024] [Indexed: 07/20/2024] Open
Abstract
Current KRASG12C (OFF) inhibitors that target inactive GDP-bound KRASG12C cause responses in less than half of patients and these responses are not durable. A class of RASG12C (ON) inhibitors that targets active GTP-bound KRASG12C blocks ERK signaling more potently than the inactive-state inhibitors. Sensitivity to either class of agents is strongly correlated with inhibition of mTORC1 activity. We have previously shown that PI3K/mTOR and ERK-signaling pathways converge on key cellular processes and that inhibition of both pathways is required for inhibition of these processes and for significant antitumor activity. We find here that the combination of a KRASG12C inhibitor with a selective mTORC1 kinase inhibitor causes synergistic inhibition of Cyclin D1 expression and cap-dependent translation. Moreover, BIM upregulation by KRASG12C inhibition and inhibition of MCL-1 expression by the mTORC1 inhibitor are both required to induce significant cell death. In vivo, this combination causes deep, durable tumor regressions and is well tolerated. This study suggests that the ERK and PI3K/mTOR pathways each mitigate the effects of inhibition of the other and that combinatorial inhibition is a potential strategy for treating KRASG12C-dependent lung cancer.
Collapse
Affiliation(s)
- Hidenori Kitai
- Program in Molecular Pharmacology and Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Philip H Choi
- Program in Molecular Pharmacology and Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yu C Yang
- Department of Biology, Revolution Medicines Inc., Redwood City, CA, USA
| | - Jacob A Boyer
- Program in Molecular Pharmacology and Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Adele Whaley
- Program in Molecular Pharmacology and Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Priya Pancholi
- Program in Molecular Pharmacology and Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Claire Thant
- Program in Molecular Pharmacology and Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jason Reiter
- Program in Molecular Pharmacology and Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kevin Chen
- Antitumor Assessment Core, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Vladimir Markov
- Antitumor Assessment Core, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hirokazu Taniguchi
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Rui Yamaguchi
- Division of Cancer Systems Biology, Aichi Cancer Center Research Institute, Nagoya, Aichi, 464-8681, Japan
| | - Hiromichi Ebi
- Division of Molecular Therapeutics, Aichi Cancer Center Research Institute, Nagoya, Aichi, 464-8681, Japan
| | - James Evans
- Department of Biology, Revolution Medicines Inc., Redwood City, CA, USA
| | - Jingjing Jiang
- Department of Biology, Revolution Medicines Inc., Redwood City, CA, USA
| | - Bianca Lee
- Department of Biology, Revolution Medicines Inc., Redwood City, CA, USA
| | - David Wildes
- Department of Biology, Revolution Medicines Inc., Redwood City, CA, USA
| | - Elisa de Stanchina
- Antitumor Assessment Core, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Mallika Singh
- Department of Biology, Revolution Medicines Inc., Redwood City, CA, USA.
| | - Neal Rosen
- Program in Molecular Pharmacology and Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
6
|
Kesarwani M, Kincaid Z, Azhar M, Azam M. Enhanced MAPK signaling induced by CSF3R mutants confers dependence to DUSP1 for leukemic transformation. Blood Adv 2024; 8:2765-2776. [PMID: 38531054 PMCID: PMC11176961 DOI: 10.1182/bloodadvances.2023010830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 02/20/2024] [Accepted: 02/27/2024] [Indexed: 03/28/2024] Open
Abstract
ABSTRACT Elevated MAPK and the JAK-STAT signaling play pivotal roles in the pathogenesis of chronic neutrophilic leukemia and atypical chronic myeloid leukemia. Although inhibitors targeting these pathways effectively suppress the diseases, they fall short in providing enduring remission, largely attributed to the cytostatic nature of these drugs. Even combinations of these drugs are ineffective in achieving sustained remission. Enhanced MAPK signaling besides promoting proliferation and survival triggers a proapoptotic response. Consequently, malignancies reliant on elevated MAPK signaling use MAPK feedback regulators to intricately modulate the signaling output, prioritizing proliferation and survival while dampening the apoptotic stimuli. Herein, we demonstrate that enhanced MAPK signaling in granulocyte colony-stimulating factor 3 receptor (CSF3R)-driven leukemia upregulates the expression of dual specificity phosphatase 1 (DUSP1) to suppress the apoptotic stimuli crucial for leukemogenesis. Consequently, genetic deletion of Dusp1 in mice conferred synthetic lethality to CSF3R-induced leukemia. Mechanistically, DUSP1 depletion in leukemic context causes activation of JNK1/2 that results in induced expression of BIM and P53 while suppressing the expression of BCL2 that selectively triggers apoptotic response in leukemic cells. Pharmacological inhibition of DUSP1 by BCI (a DUSP1 inhibitor) alone lacked antileukemic activity due to ERK1/2 rebound caused by off-target inhibition of DUSP6. Consequently, a combination of BCI with a MEK inhibitor successfully cured CSF3R-induced leukemia in a preclinical mouse model. Our findings underscore the pivotal role of DUSP1 in leukemic transformation driven by enhanced MAPK signaling and advocate for the development of a selective DUSP1 inhibitor for curative treatment outcomes.
Collapse
Affiliation(s)
- Meenu Kesarwani
- Division of Pathology, Cincinnati Children’s Hospital, Cincinnati, OH
| | - Zachary Kincaid
- Division of Pathology, Cincinnati Children’s Hospital, Cincinnati, OH
| | - Mohammad Azhar
- Division of Pathology, Cincinnati Children’s Hospital, Cincinnati, OH
| | - Mohammad Azam
- Division of Pathology, Cincinnati Children’s Hospital, Cincinnati, OH
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital, Cincinnati, OH
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH
| |
Collapse
|
7
|
Jenkins LJ, Luk IY, Chionh F, Tan T, Needham K, Ayton J, Reehorst CM, Vukelic N, Sieber OM, Mouradov D, Gibbs P, Williams DS, Tebbutt NC, Desai J, Hollande F, Dhillon AS, Lee EF, Merino D, Fairlie WD, Mariadason JM. BCL-X L inhibitors enhance the apoptotic efficacy of BRAF inhibitors in BRAF V600E colorectal cancer. Cell Death Dis 2024; 15:183. [PMID: 38429301 PMCID: PMC10907349 DOI: 10.1038/s41419-024-06478-z] [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: 06/26/2023] [Revised: 01/09/2024] [Accepted: 01/17/2024] [Indexed: 03/03/2024]
Abstract
Metastatic BRAFV600E colorectal cancer (CRC) carries an extremely poor prognosis and is in urgent need of effective new treatments. While the BRAFV600E inhibitor encorafenib in combination with the EGFR inhibitor cetuximab (Enc+Cet) was recently approved for this indication, overall survival is only increased by 3.6 months and objective responses are observed in only 20% of patients. We have found that a limitation of Enc+Cet treatment is the failure to efficiently induce apoptosis in BRAFV600E CRCs, despite inducing expression of the pro-apoptotic protein BIM and repressing expression of the pro-survival protein MCL-1. Here, we show that BRAFV600E CRCs express high basal levels of the pro-survival proteins MCL-1 and BCL-XL, and that combining encorafenib with a BCL-XL inhibitor significantly enhances apoptosis in BRAFV600E CRC cell lines. This effect was partially dependent on the induction of BIM, as BIM deletion markedly attenuated BRAF plus BCL-XL inhibitor-induced apoptosis. As thrombocytopenia is an established on-target toxicity of BCL-XL inhibition, we also examined the effect of combining encorafenib with the BCL-XL -targeting PROTAC DT2216, and the novel BCL-2/BCL-XL inhibitor dendrimer conjugate AZD0466. Combining encorafenib with DT2216 significantly increased apoptosis induction in vitro, while combining encorafenib with AZD0466 was well tolerated in mice and further reduced growth of BRAFV600E CRC xenografts compared to either agent alone. Collectively, these findings demonstrate that combined BRAF and BCL-XL inhibition significantly enhances apoptosis in pre-clinical models of BRAFV600E CRC and is a combination regimen worthy of clinical investigation to improve outcomes for these patients.
Collapse
Affiliation(s)
- Laura J Jenkins
- Olivia Newton-John Cancer Research Institute, Melbourne, VIC, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, VIC, Australia
| | - Ian Y Luk
- Olivia Newton-John Cancer Research Institute, Melbourne, VIC, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, VIC, Australia
| | - Fiona Chionh
- Olivia Newton-John Cancer Research Institute, Melbourne, VIC, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, VIC, Australia
| | - Tao Tan
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Kristen Needham
- Olivia Newton-John Cancer Research Institute, Melbourne, VIC, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, VIC, Australia
| | - Jamieson Ayton
- Olivia Newton-John Cancer Research Institute, Melbourne, VIC, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, VIC, Australia
| | - Camilla M Reehorst
- Olivia Newton-John Cancer Research Institute, Melbourne, VIC, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, VIC, Australia
| | - Natalia Vukelic
- Olivia Newton-John Cancer Research Institute, Melbourne, VIC, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, VIC, Australia
| | - Oliver M Sieber
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
- Department of Surgery, The University of Melbourne, Melbourne, VIC, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, Australia
| | - Dmitri Mouradov
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Peter Gibbs
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | - David S Williams
- Olivia Newton-John Cancer Research Institute, Melbourne, VIC, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, VIC, Australia
- Department of Pathology, Austin Health, Melbourne, VIC, Australia
| | - Niall C Tebbutt
- Olivia Newton-John Cancer Research Institute, Melbourne, VIC, Australia
- Department of Surgery, The University of Melbourne, Melbourne, VIC, Australia
- Department of Medical Oncology, Austin Health, Melbourne, Australia
| | - Jayesh Desai
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Frédéric Hollande
- Department of Clinical Pathology, The University of Melbourne, Melbourne, VIC, Australia
- University of Melbourne Centre for Cancer Research, Victorian Comprehensive Cancer Centre, Melbourne, VIC, Australia
| | - Amardeep S Dhillon
- The institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Australia
| | - Erinna F Lee
- Olivia Newton-John Cancer Research Institute, Melbourne, VIC, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, VIC, Australia
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| | - Delphine Merino
- Olivia Newton-John Cancer Research Institute, Melbourne, VIC, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | - W Douglas Fairlie
- Olivia Newton-John Cancer Research Institute, Melbourne, VIC, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, VIC, Australia
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| | - John M Mariadason
- Olivia Newton-John Cancer Research Institute, Melbourne, VIC, Australia.
- School of Cancer Medicine, La Trobe University, Melbourne, VIC, Australia.
| |
Collapse
|
8
|
Abbas H, Derkaoui DK, Jeammet L, Adicéam E, Tiollier J, Sicard H, Braun T, Poyet JL. Apoptosis Inhibitor 5: A Multifaceted Regulator of Cell Fate. Biomolecules 2024; 14:136. [PMID: 38275765 PMCID: PMC10813780 DOI: 10.3390/biom14010136] [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: 01/17/2024] [Accepted: 01/19/2024] [Indexed: 01/27/2024] Open
Abstract
Apoptosis, or programmed cell death, is a fundamental process that maintains tissue homeostasis, eliminates damaged or infected cells, and plays a crucial role in various biological phenomena. The deregulation of apoptosis is involved in many human diseases, including cancer. One of the emerging players in the intricate regulatory network of apoptosis is apoptosis inhibitor 5 (API5), also called AAC-11 (anti-apoptosis clone 11) or FIF (fibroblast growth factor-2 interacting factor). While it may not have yet the same level of notoriety as some other cancer-associated proteins, API5 has garnered increasing attention in the cancer field in recent years, as elevated API5 levels are often associated with aggressive tumor behavior, resistance to therapy, and poor patient prognosis. This review aims to shed light on the multifaceted functions and regulatory mechanisms of API5 in cell fate decisions as well as its interest as therapeutic target in cancer.
Collapse
Affiliation(s)
- Hafsia Abbas
- Université Oran 1, Ahmed Ben Bella, Oran 31000, Algeria; (H.A.); (D.K.D.)
| | | | - Louise Jeammet
- Jalon Therapeutics, 75010 Paris, France; (L.J.); (J.T.); (H.S.)
| | - Emilie Adicéam
- Jalon Therapeutics, 75010 Paris, France; (L.J.); (J.T.); (H.S.)
| | - Jérôme Tiollier
- Jalon Therapeutics, 75010 Paris, France; (L.J.); (J.T.); (H.S.)
| | - Hélène Sicard
- Jalon Therapeutics, 75010 Paris, France; (L.J.); (J.T.); (H.S.)
| | - Thorsten Braun
- Laboratoire de Transfert des Leucémies, EA3518, Institut de Recherche Saint Louis, Hôpital Saint Louis, Université de Paris, 75010 Paris, France;
- AP-HP, Service d’Hématologie Clinique, Hôpital Avicenne, Université Paris XIII, 93000 Bobigny, France
- OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Hôpital Saint-Louis, 75010 Paris, France
| | - Jean-Luc Poyet
- INSERM UMRS976, Institut de Recherche Saint Louis, Hôpital Saint Louis, 75010 Paris, France
- Université Paris Cité, 75015 Paris, France
| |
Collapse
|
9
|
Carlin CR, Ngalula S. Loss of EGF receptor polarity enables homeostatic imbalance in epithelial-cell models. Mol Biol Cell 2023; 34:ar116. [PMID: 37647145 PMCID: PMC10846618 DOI: 10.1091/mbc.e23-04-0133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 07/26/2023] [Accepted: 08/22/2023] [Indexed: 09/01/2023] Open
Abstract
The polarized distribution of membrane proteins into apical and basolateral domains provides the basis for specialized functions of epithelial tissues. The EGF receptor (EGFR) plays important roles in embryonic development, adult-epithelial tissue homeostasis, and growth and survival of many carcinomas. Typically targeted to basolateral domains, there is also considerable evidence of EGFR sorting plasticity but very limited knowledge regarding domain-specific EGFR substrates. Here we have investigated effects of selective EGFR mistargeting because of inactive-basolateral sorting signals on epithelial-cell homeostatic responses to growth-induced stress in MDCK cell models. Aberrant EGFR localization was associated with multilayer formation, anchorage-independent growth, and upregulated expression of the intermediate filament-protein vimentin characteristically seen in cells undergoing epithelial-to-mesenchymal transition. EGFRs were selectively retained following their internalization from apical membranes, and a signaling pathway involving the signaling adaptor Gab1 protein and extracellular signal-regulated kinase ERK5 had an essential role integrating multiple responses to growth-induced stress. Our studies highlight the potential importance of cellular machinery specifying EGFR polarity in epithelial pathologies associated with homeostatic imbalance.
Collapse
Affiliation(s)
- Cathleen R. Carlin
- Department of Molecular Biology and Microbiology, Case Western Reserve University, Cleveland, OH 44106-4970
- Case Western Reserve University Comprehensive Cancer Center, School of Medicine, Case Western Reserve University, Cleveland, OH 44106-4970
| | - Syntyche Ngalula
- Department of Molecular Biology and Microbiology, Case Western Reserve University, Cleveland, OH 44106-4970
| |
Collapse
|
10
|
Martin-Vega A, Cobb MH. Navigating the ERK1/2 MAPK Cascade. Biomolecules 2023; 13:1555. [PMID: 37892237 PMCID: PMC10605237 DOI: 10.3390/biom13101555] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 10/16/2023] [Accepted: 10/18/2023] [Indexed: 10/29/2023] Open
Abstract
The RAS-ERK pathway is a fundamental signaling cascade crucial for many biological processes including proliferation, cell cycle control, growth, and survival; common across all cell types. Notably, ERK1/2 are implicated in specific processes in a context-dependent manner as in stem cells and pancreatic β-cells. Alterations in the different components of this cascade result in dysregulation of the effector kinases ERK1/2 which communicate with hundreds of substrates. Aberrant activation of the pathway contributes to a range of disorders, including cancer. This review provides an overview of the structure, activation, regulation, and mutational frequency of the different tiers of the cascade; with a particular focus on ERK1/2. We highlight the importance of scaffold proteins that contribute to kinase localization and coordinate interaction dynamics of the kinases with substrates, activators, and inhibitors. Additionally, we explore innovative therapeutic approaches emphasizing promising avenues in this field.
Collapse
Affiliation(s)
- Ana Martin-Vega
- Department of Pharmacology, UT Southwestern Medical Center, 6001 Forest Park Rd., Dallas, TX 75390, USA;
| | - Melanie H. Cobb
- Department of Pharmacology, UT Southwestern Medical Center, 6001 Forest Park Rd., Dallas, TX 75390, USA;
- Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, 6001 Forest Park Rd., Dallas, TX 75390, USA
| |
Collapse
|
11
|
Netterfield TS, Ostheimer GJ, Tentner AR, Joughin BA, Dakoyannis AM, Sharma CD, Sorger PK, Janes KA, Lauffenburger DA, Yaffe MB. Biphasic JNK-Erk signaling separates the induction and maintenance of cell senescence after DNA damage induced by topoisomerase II inhibition. Cell Syst 2023; 14:582-604.e10. [PMID: 37473730 PMCID: PMC10627503 DOI: 10.1016/j.cels.2023.06.005] [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: 06/14/2022] [Revised: 03/24/2023] [Accepted: 06/13/2023] [Indexed: 07/22/2023]
Abstract
Genotoxic stress in mammalian cells, including those caused by anti-cancer chemotherapy, can induce temporary cell-cycle arrest, DNA damage-induced senescence (DDIS), or apoptotic cell death. Despite obvious clinical importance, it is unclear how the signals emerging from DNA damage are integrated together with other cellular signaling pathways monitoring the cell's environment and/or internal state to control different cell fates. Using single-cell-based signaling measurements combined with tensor partial least square regression (t-PLSR)/principal component analysis (PCA) analysis, we show that JNK and Erk MAPK signaling regulates the initiation of cell senescence through the transcription factor AP-1 at early times after doxorubicin-induced DNA damage and the senescence-associated secretory phenotype (SASP) at late times after damage. These results identify temporally distinct roles for signaling pathways beyond the classic DNA damage response (DDR) that control the cell senescence decision and modulate the tumor microenvironment and reveal fundamental similarities between signaling pathways responsible for oncogene-induced senescence (OIS) and senescence caused by topoisomerase II inhibition. A record of this paper's transparent peer review process is included in the supplemental information.
Collapse
Affiliation(s)
- Tatiana S Netterfield
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Center for Precision Cancer Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Gerard J Ostheimer
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Andrea R Tentner
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Brian A Joughin
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Center for Precision Cancer Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Alexandra M Dakoyannis
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Center for Precision Cancer Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Charvi D Sharma
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Center for Precision Cancer Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Computer Science and Molecular Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Peter K Sorger
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Kevin A Janes
- Department of Biomedical Engineering and Department of Biochemistry & Molecular Genetics, University of Virginia, Charlottesville, VA 22908, USA
| | - Douglas A Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Michael B Yaffe
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Center for Precision Cancer Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Division of Acute Care Surgery, Trauma, and Surgical Critical Care, and Division of Surgical Oncology, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
12
|
Khan S, Budamagunta V, Zhou D. Targeting KRAS in pancreatic cancer: Emerging therapeutic strategies. Adv Cancer Res 2023; 159:145-184. [PMID: 37268395 DOI: 10.1016/bs.acr.2023.02.004] [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] [Indexed: 03/12/2023]
Abstract
KRAS, a predominant member of the RAS family, is the most frequently mutated oncogene in human pancreatic cancer (∼95% of cases). Mutations in KRAS lead to its constitutive activation and activation of its downstream signaling pathways such as RAF/MEK/ERK and PI3K/AKT/mTOR that promote cell proliferation and provide apoptosis evasion capabilities to cancer cells. KRAS had been considered 'undruggable' until the discovery of the first covalent inhibitor targeting the G12C mutation. While G12C mutations are frequently found in non-small cell lung cancer, these are relatively rare in pancreatic cancer. On the other hand, pancreatic cancer harbors other KRAS mutations such as G12D and G12V. The inhibitors targeting G12D mutation (such as MRTX1133) have been recently developed, whereas those targeting other mutations are still lacking. Unfortunately, KRAS inhibitor monotherapy-associated resistance hinders their therapeutic efficacy. Therefore, various combination strategies have been tested and some yielded promising results, such as combinations with receptor tyrosine kinase, SHP2, or SOS1 inhibitors. In addition, we recently demonstrated that the combination of sotorasib with DT2216 (a BCL-XL-selective degrader) synergistically inhibits G12C-mutated pancreatic cancer cell growth in vitro and in vivo. This is in part because KRAS-targeted therapies induce cell cycle arrest and cellular senescence, which contributes to therapeutic resistance, while their combination with DT2216 can more effectively induce apoptosis. Similar combination strategies may also work for G12D inhibitors in pancreatic cancer. This chapter will review KRAS biochemistry, signaling pathways, different mutations, emerging KRAS-targeted therapies, and combination strategies. Finally, we discuss challenges associated with KRAS targeting and future directions, emphasizing pancreatic cancer.
Collapse
Affiliation(s)
- Sajid Khan
- Department of Biochemistry & Structural Biology, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States; Mays Cancer Center, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States.
| | - Vivekananda Budamagunta
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, United States; Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, United States; Genetics and Genomics Graduate Program, Genetics Institute, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Daohong Zhou
- Department of Biochemistry & Structural Biology, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States; Mays Cancer Center, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States.
| |
Collapse
|
13
|
Liang X, Yadav SP, Batz ZA, Nellissery J, Swaroop A. Protein kinase CK2 modulates the activity of Maf-family bZIP transcription factor NRL in rod photoreceptors of mammalian retina. Hum Mol Genet 2023; 32:948-958. [PMID: 36226585 PMCID: PMC9991000 DOI: 10.1093/hmg/ddac256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 09/21/2022] [Accepted: 10/07/2022] [Indexed: 11/14/2022] Open
Abstract
Maf-family basic motif leucine zipper protein NRL specifies rod photoreceptor cell fate during retinal development and, in concert with homeodomain protein CRX and other regulatory factors, controls the expression of most rod-expressed genes including the visual pigment gene Rhodopsin (Rho). Transcriptional regulatory activity of NRL is modulated by post-translational modifications, especially phosphorylation, and mutations at specific phosphosites can lead to retinal degeneration. During our studies to elucidate NRL-mediated transcriptional regulation, we identified protein kinase CK2 in NRL-enriched complexes bound to Rho promoter-enhancer regions and in NRL-enriched high molecular mass fractions from the bovine retina. The presence of CK2 in NRL complexes was confirmed by co-immunoprecipitation from developing and adult mouse retinal extracts. In vitro kinase assay and bioinformatic analysis indicated phosphorylation of NRL at Ser117 residue by CK2. Co-transfection of Csnk2a1 cDNA encoding murine CK2 with human NRL and CRX reduced the bovine Rho promoter-driven luciferase expression in HEK293 cells and mutagenesis of NRL-Ser117 residue to Ala restored the reporter gene activity. In concordance, overexpression of CK2 in the mouse retina in vivo by electroporation resulted in reduction of Rho promoter-driven DsRed reporter expression as well as the transcript level of many phototransduction genes. Thus, our studies demonstrate that CK2 can phosphorylate Ser117 of NRL. Modulation of NRL activity by CK2 suggests intricate interdependence of transcriptional and signaling pathways in maintaining rod homeostasis.
Collapse
Affiliation(s)
- Xulong Liang
- Neurobiology-Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, 6 Center Drive, MSC0610, Bethesda, MD 20892, USA
| | - Sharda P Yadav
- Neurobiology-Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, 6 Center Drive, MSC0610, Bethesda, MD 20892, USA
| | - Zachary A Batz
- Neurobiology-Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, 6 Center Drive, MSC0610, Bethesda, MD 20892, USA
| | - Jacob Nellissery
- Neurobiology-Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, 6 Center Drive, MSC0610, Bethesda, MD 20892, USA
| | - Anand Swaroop
- Neurobiology-Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, 6 Center Drive, MSC0610, Bethesda, MD 20892, USA
| |
Collapse
|
14
|
Jenkins LJ, Luk IY, Fairlie WD, Lee EF, Palmieri M, Schoffer KL, Tan T, Ng I, Vukelic N, Tran S, Tse JW, Nightingale R, Alam Z, Chionh F, Iatropoulos G, Ernst M, Afshar-Sterle S, Desai J, Gibbs P, Sieber OM, Dhillon AS, Tebbutt NC, Mariadason JM. Genotype-Tailored ERK/MAPK Pathway and HDAC Inhibition Rewires the Apoptotic Rheostat to Trigger Colorectal Cancer Cell Death. Mol Cancer Ther 2023; 22:52-62. [PMID: 36343387 PMCID: PMC9808369 DOI: 10.1158/1535-7163.mct-22-0101] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 07/21/2022] [Accepted: 10/31/2022] [Indexed: 11/09/2022]
Abstract
The EGFR/RAS/MEK/ERK signaling pathway (ERK/MAPK) is hyperactivated in most colorectal cancers. A current limitation of inhibitors of this pathway is that they primarily induce cytostatic effects in colorectal cancer cells. Nevertheless, these drugs do induce expression of proapoptotic factors, suggesting they may prime colorectal cancer cells to undergo apoptosis. As histone deacetylase inhibitors (HDACis) induce expression of multiple proapoptotic proteins, we examined whether they could synergize with ERK/MAPK inhibitors to trigger colorectal cancer cell apoptosis. Combined MEK/ERK and HDAC inhibition synergistically induced apoptosis in colorectal cancer cell lines and patient-derived tumor organoids in vitro, and attenuated Apc-initiated adenoma formation in vivo. Mechanistically, combined MAPK/HDAC inhibition enhanced expression of the BH3-only proapoptotic proteins BIM and BMF, and their knockdown significantly attenuated MAPK/HDAC inhibitor-induced apoptosis. Importantly, we demonstrate that the paradigm of combined MAPK/HDAC inhibitor treatment to induce apoptosis can be tailored to specific MAPK genotypes in colorectal cancers, by combining an HDAC inhibitor with either an EGFR, KRASG12C or BRAFV600 inhibitor in KRAS/BRAFWT; KRASG12C, BRAFV600E colorectal cancer cell lines, respectively. These findings identify a series of ERK/MAPK genotype-tailored treatment strategies that can readily undergo clinical testing for the treatment of colorectal cancer.
Collapse
Affiliation(s)
- Laura J. Jenkins
- Olivia Newton-John Cancer Research Institute, Melbourne, Victoria, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, Victoria, Australia
| | - Ian Y. Luk
- Olivia Newton-John Cancer Research Institute, Melbourne, Victoria, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, Victoria, Australia
| | - W. Douglas Fairlie
- Olivia Newton-John Cancer Research Institute, Melbourne, Victoria, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, Victoria, Australia
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| | - Erinna F. Lee
- Olivia Newton-John Cancer Research Institute, Melbourne, Victoria, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, Victoria, Australia
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| | - Michelle Palmieri
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Kael L. Schoffer
- Olivia Newton-John Cancer Research Institute, Melbourne, Victoria, Australia
| | - Tao Tan
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Irvin Ng
- Olivia Newton-John Cancer Research Institute, Melbourne, Victoria, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, Victoria, Australia
| | - Natalia Vukelic
- Olivia Newton-John Cancer Research Institute, Melbourne, Victoria, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, Victoria, Australia
| | - Sharon Tran
- Olivia Newton-John Cancer Research Institute, Melbourne, Victoria, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, Victoria, Australia
| | - Janson W.T. Tse
- Olivia Newton-John Cancer Research Institute, Melbourne, Victoria, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, Victoria, Australia
| | - Rebecca Nightingale
- Olivia Newton-John Cancer Research Institute, Melbourne, Victoria, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, Victoria, Australia
| | - Zakia Alam
- Olivia Newton-John Cancer Research Institute, Melbourne, Victoria, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, Victoria, Australia
| | - Fiona Chionh
- Olivia Newton-John Cancer Research Institute, Melbourne, Victoria, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, Victoria, Australia
| | - George Iatropoulos
- Olivia Newton-John Cancer Research Institute, Melbourne, Victoria, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, Victoria, Australia
| | - Matthias Ernst
- Olivia Newton-John Cancer Research Institute, Melbourne, Victoria, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, Victoria, Australia
| | - Shoukat Afshar-Sterle
- Olivia Newton-John Cancer Research Institute, Melbourne, Victoria, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, Victoria, Australia
| | - Jayesh Desai
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Peter Gibbs
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Oliver M. Sieber
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, Victoria, Australia
- Department of Surgery, The University of Melbourne, Melbourne, Victoria, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Victoria, Australia
| | - Amardeep S. Dhillon
- Olivia Newton-John Cancer Research Institute, Melbourne, Victoria, Australia
- The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Australia
| | - Niall C. Tebbutt
- Olivia Newton-John Cancer Research Institute, Melbourne, Victoria, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, Victoria, Australia
- Department of Surgery, The University of Melbourne, Melbourne, Victoria, Australia
| | - John M. Mariadason
- Olivia Newton-John Cancer Research Institute, Melbourne, Victoria, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, Victoria, Australia
- Corresponding Author: John M. Mariadason, Olivia Newton-John Cancer Wellness and Research Centre, 145-163 Studley Road, Melbourne, Victoria, 3084, Australia. Phone: 613-9496-3068; E-mail:
| |
Collapse
|
15
|
Harabuchi S, Khan O, Bassiri H, Yoshida T, Okada Y, Takizawa M, Ikeda O, Katada A, Kambayashi T. Manipulation of diacylglycerol and ERK-mediated signaling differentially controls CD8 + T cell responses during chronic viral infection. Front Immunol 2022; 13:1032113. [PMID: 36846018 PMCID: PMC9951774 DOI: 10.3389/fimmu.2022.1032113] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/07/2022] [Indexed: 11/25/2022] Open
Abstract
Introduction Activation of T cell receptor (TCR) signaling is critical for clonal expansion of CD8+ T cells. However, the effects of augmenting TCR signaling during chronic antigen exposure is less understood. Here, we investigated the role of diacylglycerol (DAG)-mediated signaling downstream of the TCR during chronic lymphocytic choriomeningitis virus clone 13 (LCMV CL13) infection by blocking DAG kinase zeta (DGKζ), a negative regulator of DAG. Methods We examined the activation, survival, expansion, and phenotype of virus-specific T cell in the acute and chronic phases of LCMV CL13-infected in mice after DGKζ blockade or selective activation of ERK. Results Upon LCMV CL13 infection, DGKζ deficiency promoted early short-lived effector cell (SLEC) differentiation of LCMV-specific CD8+ T cells, but this was followed by abrupt cell death. Short-term inhibition of DGKζ with ASP1570, a DGKζ-selective pharmacological inhibitor, augmented CD8+ T cell activation without causing cell death, which reduced virus titers both in the acute and chronic phases of LCMV CL13 infection. Unexpectedly, the selective enhancement of ERK, one key signaling pathway downstream of DAG, lowered viral titers and promoted expansion, survival, and a memory phenotype of LCMV-specific CD8+ T cells in the acute phase with fewer exhausted T cells in the chronic phase. The difference seen between DGKζ deficiency and selective ERK enhancement could be potentially explained by the activation of the AKT/mTOR pathway by DGKζ deficiency, since the mTOR inhibitor rapamycin rescued the abrupt cell death seen in virus-specific DGKζ KO CD8+ T cells. Discussion Thus, while ERK is downstream of DAG signaling, the two pathways lead to distinct outcomes in the context of chronic CD8+ T cell activation, whereby DAG promotes SLEC differentiation and ERK promotes a memory phenotype.
Collapse
Affiliation(s)
- Shohei Harabuchi
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
- Department of Otolaryngology-Head and Neck surgery, Asahikawa Medical University, Asahikawa, Japan
| | - Omar Khan
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Hamid Bassiri
- Division of Infectious Diseases, Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Taku Yoshida
- Immuno-Oncology, Astellas Pharma Inc., Tsukuba, Japan
| | - Yohei Okada
- Immuno-Oncology, Astellas Pharma Inc., Tsukuba, Japan
| | - Masaomi Takizawa
- Research Program Management-Applied Research Management, Astellas Pharma Inc., Tokyo, Japan
| | - Osamu Ikeda
- Immuno-Oncology, Astellas Pharma Inc., Tsukuba, Japan
| | - Akihiro Katada
- Department of Otolaryngology-Head and Neck surgery, Asahikawa Medical University, Asahikawa, Japan
| | - Taku Kambayashi
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
16
|
Valencia-Expósito A, Gómez-Lamarca MJ, Widmann TJ, Martín-Bermudo MD. Integrins Cooperate With the EGFR/Ras Pathway to Preserve Epithelia Survival and Architecture in Development and Oncogenesis. Front Cell Dev Biol 2022; 10:892691. [PMID: 35769262 PMCID: PMC9234701 DOI: 10.3389/fcell.2022.892691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 05/09/2022] [Indexed: 11/13/2022] Open
Abstract
Adhesion to the extracellular matrix (ECM) is required for normal epithelial cell survival. Disruption of this interaction leads to a specific type of apoptosis known as anoikis. Yet, there are physiological and pathological situations in which cells not connected to the ECM are protected from anoikis, such as during cell migration or metastasis. The main receptors transmitting signals from the ECM are members of the integrin family. However, although integrin-mediated cell-ECM anchorage has been long recognized as crucial for epithelial cell survival, the in vivo significance of this interaction remains to be weighed. In this work, we have used the Drosophila wing imaginal disc epithelium to analyze the importance of integrins as survival factors during epithelia morphogenesis. We show that reducing integrin expression in the wing disc induces caspase-dependent cell death and basal extrusion of the dead cells. In this case, anoikis is mediated by the activation of the JNK pathway, which in turn triggers expression of the proapoptotic protein Hid. In addition, our results strongly suggest that, during wing disc morphogenesis, the EGFR pathway protects cells undergoing cell shape changes upon ECM detachment from anoikis. Furthermore, we show that oncogenic activation of the EGFR/Ras pathway in integrin mutant cells rescues them from apoptosis while promoting their extrusion from the epithelium. Altogether, our results support the idea that integrins promote cell survival during normal tissue morphogenesis and prevent the extrusion of transformed cells.
Collapse
Affiliation(s)
| | - M. Jesús Gómez-Lamarca
- Centro Andaluz de Biología del Desarrollo CSIC-Universidad Pablo de Olavide, Sevilla, Spain
- Departamento de Biología Celular, Universidad de Sevilla, Sevilla, Spain
| | | | - María D. Martín-Bermudo
- Centro Andaluz de Biología del Desarrollo CSIC-Universidad Pablo de Olavide, Sevilla, Spain
- *Correspondence: María D. Martín-Bermudo,
| |
Collapse
|
17
|
Seiler K, Humbert M, Minder P, Mashimo I, Schläfli AM, Krauer D, Federzoni EA, Vu B, Moresco JJ, Yates JR, Sadowski MC, Radpour R, Kaufmann T, Sarry JE, Dengjel J, Tschan MP, Torbett BE. Hexokinase 3 enhances myeloid cell survival via non-glycolytic functions. Cell Death Dis 2022; 13:448. [PMID: 35538058 PMCID: PMC9091226 DOI: 10.1038/s41419-022-04891-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 04/10/2022] [Accepted: 04/28/2022] [Indexed: 12/14/2022]
Abstract
The family of hexokinases (HKs) catalyzes the first step of glycolysis, the ATP-dependent phosphorylation of glucose to glucose-6-phosphate. While HK1 and HK2 are ubiquitously expressed, the less well-studied HK3 is primarily expressed in hematopoietic cells and tissues and is highly upregulated during terminal differentiation of some acute myeloid leukemia (AML) cell line models. Here we show that expression of HK3 is predominantly originating from myeloid cells and that the upregulation of this glycolytic enzyme is not restricted to differentiation of leukemic cells but also occurs during ex vivo myeloid differentiation of healthy CD34+ hematopoietic stem and progenitor cells. Within the hematopoietic system, we show that HK3 is predominantly expressed in cells of myeloid origin. CRISPR/Cas9 mediated gene disruption revealed that loss of HK3 has no effect on glycolytic activity in AML cell lines while knocking out HK2 significantly reduced basal glycolysis and glycolytic capacity. Instead, loss of HK3 but not HK2 led to increased sensitivity to ATRA-induced cell death in AML cell lines. We found that HK3 knockout (HK3-null) AML cells showed an accumulation of reactive oxygen species (ROS) as well as DNA damage during ATRA-induced differentiation. RNA sequencing analysis confirmed pathway enrichment for programmed cell death, oxidative stress, and DNA damage response in HK3-null AML cells. These signatures were confirmed in ATAC sequencing, showing that loss of HK3 leads to changes in chromatin configuration and increases the accessibility of genes involved in apoptosis and stress response. Through isoform-specific pulldowns, we furthermore identified a direct interaction between HK3 and the proapoptotic BCL-2 family member BIM, which has previously been shown to shorten myeloid life span. Our findings provide evidence that HK3 is dispensable for glycolytic activity in AML cells while promoting cell survival, possibly through direct interaction with the BH3-only protein BIM during ATRA-induced neutrophil differentiation.
Collapse
Affiliation(s)
- Kristina Seiler
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
- Graduate School of Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA, USA
| | - Magali Humbert
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - Petra Minder
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA, USA
| | - Iris Mashimo
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA, USA
| | - Anna M Schläfli
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - Deborah Krauer
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - Elena A Federzoni
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA, USA
| | - Bich Vu
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - James J Moresco
- Department of Molecular Medicine, Scripps Research, La Jolla, CA, USA
| | - John R Yates
- Department of Molecular Medicine, Scripps Research, La Jolla, CA, USA
| | - Martin C Sadowski
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - Ramin Radpour
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Tumor Immunology, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Thomas Kaufmann
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Jean-Emmanuel Sarry
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, CNRS, Toulouse, France
- Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Joern Dengjel
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Mario P Tschan
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland.
- Graduate School of Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland.
| | - Bruce E Torbett
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA, USA.
- Department of Pediatrics, School of Medicine, University of Washington, Seattle, WA, USA.
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA, USA.
- Institute for Stem Cell and Regenerative Medicine, Seattle, WA, USA.
| |
Collapse
|
18
|
van der Zwet JCG, Buijs-Gladdines JGCAM, Cordo' V, Debets DO, Smits WK, Chen Z, Dylus J, Zaman GJR, Altelaar M, Oshima K, Bornhauser B, Bourquin JP, Cools J, Ferrando AA, Vormoor J, Pieters R, Vormoor B, Meijerink JPP. MAPK-ERK is a central pathway in T-cell acute lymphoblastic leukemia that drives steroid resistance. Leukemia 2021; 35:3394-3405. [PMID: 34007050 DOI: 10.1038/s41375-021-01291-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 04/17/2021] [Accepted: 05/07/2021] [Indexed: 02/04/2023]
Abstract
(Patho-)physiological activation of the IL7-receptor (IL7R) signaling contributes to steroid resistance in pediatric T-cell acute lymphoblastic leukemia (T-ALL). Here, we show that activating IL7R pathway mutations and physiological IL7R signaling activate MAPK-ERK signaling, which provokes steroid resistance by phosphorylation of BIM. By mass spectrometry, we demonstrate that phosphorylated BIM is impaired in binding to BCL2, BCLXL and MCL1, shifting the apoptotic balance toward survival. Treatment with MEK inhibitors abolishes this inactivating phosphorylation of BIM and restores its interaction with anti-apoptotic BCL2-protein family members. Importantly, the MEK inhibitor selumetinib synergizes with steroids in both IL7-dependent and IL7-independent steroid resistant pediatric T-ALL PDX samples. Despite the anti-MAPK-ERK activity of ruxolitinib in IL7-induced signaling and JAK1 mutant cells, ruxolitinib only synergizes with steroid treatment in IL7-dependent steroid resistant PDX samples but not in IL7-independent steroid resistant PDX samples. Our study highlights the central role for MAPK-ERK signaling in steroid resistance in T-ALL patients, and demonstrates the broader application of MEK inhibitors over ruxolitinib to resensitize steroid-resistant T-ALL cells. These findings strongly support the enrollment of T-ALL patients in the current phase I/II SeluDex trial (NCT03705507) and contributes to the optimization and stratification of newly designed T-ALL treatment regimens.
Collapse
Affiliation(s)
| | | | - Valentina Cordo'
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Donna O Debets
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center of Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| | - Willem K Smits
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Zhongli Chen
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Jelle Dylus
- Netherlands Translational Research Center B.V., Oss, the Netherlands
| | - Guido J R Zaman
- Netherlands Translational Research Center B.V., Oss, the Netherlands
| | - Maarten Altelaar
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center of Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| | - Koichi Oshima
- Institute of Cancer Genetics, Columbia University Medical Center, New York, NY, USA
| | - Beat Bornhauser
- Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Jean-Pierre Bourquin
- Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Jan Cools
- KU Leuven Center for Human Genetics & VIB Center for Cancer Biology, Leuven, Belgium
| | - Adolfo A Ferrando
- Institute of Cancer Genetics, Columbia University Medical Center, New York, NY, USA
| | - Josef Vormoor
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Newcastle University, Newcastle upon Tyne, UK
| | - Rob Pieters
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Britta Vormoor
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | | |
Collapse
|
19
|
Xie X, Zhu L, Jie Z, Li Y, Gu M, Zhou X, Wang H, Chang JH, Ko CJ, Cheng X, Sun SC. TRAF2 regulates T cell immunity by maintaining a Tpl2-ERK survival signaling axis in effector and memory CD8 T cells. Cell Mol Immunol 2021; 18:2262-2274. [PMID: 33203937 PMCID: PMC8429472 DOI: 10.1038/s41423-020-00583-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 10/21/2020] [Indexed: 11/09/2022] Open
Abstract
Generation and maintenance of antigen-specific effector and memory T cells are central events in immune responses against infections. We show that TNF receptor-associated factor 2 (TRAF2) maintains a survival signaling axis in effector and memory CD8 T cells required for immune responses against infections. This signaling axis involves activation of Tpl2 and its downstream kinase ERK by NF-κB-inducing kinase (NIK) and degradation of the proapoptotic factor Bim. NIK mediates Tpl2 activation by stimulating the phosphorylation and degradation of the Tpl2 inhibitor p105. Interestingly, while NIK is required for Tpl2-ERK signaling under normal conditions, uncontrolled NIK activation due to loss of its negative regulator, TRAF2, causes constitutive degradation of p105 and Tpl2, leading to severe defects in ERK activation and effector/memory CD8 T cell survival. Thus, TRAF2 controls a previously unappreciated signaling axis mediating effector/memory CD8 T cell survival and protective immunity.
Collapse
Affiliation(s)
- Xiaoping Xie
- Department of Immunology, The University of Texas MD Anderson Cancer Center, 7455 Fannin Street, Box 902, Houston, TX, 77030, USA
| | - Lele Zhu
- Department of Immunology, The University of Texas MD Anderson Cancer Center, 7455 Fannin Street, Box 902, Houston, TX, 77030, USA
| | - Zuliang Jie
- Department of Immunology, The University of Texas MD Anderson Cancer Center, 7455 Fannin Street, Box 902, Houston, TX, 77030, USA
| | - Yanchuan Li
- Department of Immunology, The University of Texas MD Anderson Cancer Center, 7455 Fannin Street, Box 902, Houston, TX, 77030, USA
| | - Meidi Gu
- Department of Immunology, The University of Texas MD Anderson Cancer Center, 7455 Fannin Street, Box 902, Houston, TX, 77030, USA
| | - Xiaofei Zhou
- Department of Immunology, The University of Texas MD Anderson Cancer Center, 7455 Fannin Street, Box 902, Houston, TX, 77030, USA
| | - Hui Wang
- Department of Immunology, The University of Texas MD Anderson Cancer Center, 7455 Fannin Street, Box 902, Houston, TX, 77030, USA
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China
| | - Jae-Hoon Chang
- Department of Immunology, The University of Texas MD Anderson Cancer Center, 7455 Fannin Street, Box 902, Houston, TX, 77030, USA
- College of Pharmacy, Yeungnam University, Gyeongsan, 712-749, Republic of Korea
| | - Chun-Jung Ko
- Department of Immunology, The University of Texas MD Anderson Cancer Center, 7455 Fannin Street, Box 902, Houston, TX, 77030, USA
| | - Xuhong Cheng
- Department of Immunology, The University of Texas MD Anderson Cancer Center, 7455 Fannin Street, Box 902, Houston, TX, 77030, USA
| | - Shao-Cong Sun
- Department of Immunology, The University of Texas MD Anderson Cancer Center, 7455 Fannin Street, Box 902, Houston, TX, 77030, USA.
- MD Anderson Cancer Center UT Health Graduate School of Biomedical Sciences, Houston, TX, 77030, USA.
| |
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
|
Serrano-Del Valle A, Reina-Ortiz C, Benedi A, Anel A, Naval J, Marzo I. Future prospects for mitosis-targeted antitumor therapies. Biochem Pharmacol 2021; 190:114655. [PMID: 34129859 DOI: 10.1016/j.bcp.2021.114655] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/09/2021] [Accepted: 06/10/2021] [Indexed: 11/17/2022]
Abstract
Dysregulation of cell cycle progression is a hallmark of cancer cells. In recent years, efforts have been devoted to the development of new therapies that target proteins involved in cell cycle regulation and mitosis. Novel targeted antimitotic drugs include inhibitors of aurora kinase family, polo-like kinase 1, Mps1, Eg5, CENP-5 and the APC/cyclosome complex. While certain new inhibitors reached the clinical trial stage, most were discontinued due to negative results. However, these therapies should not be readily dismissed. Based on recent advances concerning their mechanisms of action, new strategies could be devised to increase their efficacy and promote further clinical trials. Here we discuss three main lines of action to empower these therapeutic approaches: increasing cell death signals during mitotic arrest, targeting senescent cells and facilitating antitumor immune response through immunogenic cell death (ICD).
Collapse
Affiliation(s)
| | - Chantal Reina-Ortiz
- Dept. Biochemistry, Molecular and Cell Biology, University of Zaragoza and IIS Aragón, Spain
| | - Andrea Benedi
- Dept. Biochemistry, Molecular and Cell Biology, University of Zaragoza and IIS Aragón, Spain
| | - Alberto Anel
- Dept. Biochemistry, Molecular and Cell Biology, University of Zaragoza and IIS Aragón, Spain
| | - Javier Naval
- Dept. Biochemistry, Molecular and Cell Biology, University of Zaragoza and IIS Aragón, Spain
| | - Isabel Marzo
- Dept. Biochemistry, Molecular and Cell Biology, University of Zaragoza and IIS Aragón, Spain.
| |
Collapse
|
22
|
Mlakar V, Morel E, Mlakar SJ, Ansari M, Gumy-Pause F. A review of the biological and clinical implications of RAS-MAPK pathway alterations in neuroblastoma. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:189. [PMID: 34103089 PMCID: PMC8188681 DOI: 10.1186/s13046-021-01967-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 04/27/2021] [Indexed: 02/07/2023]
Abstract
Neuroblastoma is the most common extra-cranial solid tumor in children, representing approximately 8% of all malignant childhood tumors and 15% of pediatric cancer-related deaths. Recent sequencing and transcriptomics studies have demonstrated the RAS-MAPK pathway’s contribution to the development and progression of neuroblastoma. This review compiles up-to-date evidence of this pathway’s involvement in neuroblastoma. We discuss the RAS-MAPK pathway’s general functioning, the clinical implications of its deregulation in neuroblastoma, and current promising therapeutics targeting proteins involved in signaling.
Collapse
Affiliation(s)
- Vid Mlakar
- CANSEARCH Research Platform for Pediatric Oncology and Hematology, Department of Pediatrics, Gynecology and Obstetrics, Faculty of Medicine, University of Geneva, Avenue de la Roseraie 64, 1205, Geneva, Switzerland
| | - Edouard Morel
- CANSEARCH Research Platform for Pediatric Oncology and Hematology, Department of Pediatrics, Gynecology and Obstetrics, Faculty of Medicine, University of Geneva, Avenue de la Roseraie 64, 1205, Geneva, Switzerland
| | - Simona Jurkovic Mlakar
- CANSEARCH Research Platform for Pediatric Oncology and Hematology, Department of Pediatrics, Gynecology and Obstetrics, Faculty of Medicine, University of Geneva, Avenue de la Roseraie 64, 1205, Geneva, Switzerland
| | - Marc Ansari
- CANSEARCH Research Platform for Pediatric Oncology and Hematology, Department of Pediatrics, Gynecology and Obstetrics, Faculty of Medicine, University of Geneva, Avenue de la Roseraie 64, 1205, Geneva, Switzerland.,Division of Pediatric Oncology and Hematology, Department of Women, Child and Adolescent, University Hospital of Geneva, Rue Willy-Donzé 6, 1205, Geneva, Switzerland
| | - Fabienne Gumy-Pause
- CANSEARCH Research Platform for Pediatric Oncology and Hematology, Department of Pediatrics, Gynecology and Obstetrics, Faculty of Medicine, University of Geneva, Avenue de la Roseraie 64, 1205, Geneva, Switzerland. .,Division of Pediatric Oncology and Hematology, Department of Women, Child and Adolescent, University Hospital of Geneva, Rue Willy-Donzé 6, 1205, Geneva, Switzerland.
| |
Collapse
|
23
|
Activation of the Integrated Stress Response and ER Stress Protect from Fluorizoline-Induced Apoptosis in HEK293T and U2OS Cell Lines. Int J Mol Sci 2021; 22:ijms22116117. [PMID: 34204139 PMCID: PMC8201103 DOI: 10.3390/ijms22116117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/29/2021] [Accepted: 06/03/2021] [Indexed: 02/06/2023] Open
Abstract
The prohibitin (PHB)-binding compound fluorizoline as well as PHB-downregulation activate the integrated stress response (ISR) in HEK293T and U2OS human cell lines. This activation is denoted by phosphorylation of eIF2α and increases in ATF4, ATF3, and CHOP protein levels. The blockage of the activation of the ISR by overexpression of GRP78, as well as an increase in IRE1 activity, indicate the presence of ER stress after fluorizoline treatment. The inhibition of the ER stress response in HEK293T and U2OS led to increased sensitivity to fluorizoline-induced apoptosis, indicating a pro-survival role of this pathway after fluorizoline treatment in these cell lines. Fluorizoline induced an increase in calcium concentration in the cytosol and the mitochondria. Finally, two different calcium chelators reduced fluorizoline-induced apoptosis in U2OS cells. Thus, we have found that fluorizoline causes increased ER stress and activation of the integrated stress response, which in HEK293T and U2OS cells are protective against fluorizoline-induced apoptosis.
Collapse
|
24
|
Ullah R, Yin Q, Snell AH, Wan L. RAF-MEK-ERK pathway in cancer evolution and treatment. Semin Cancer Biol 2021; 85:123-154. [PMID: 33992782 DOI: 10.1016/j.semcancer.2021.05.010] [Citation(s) in RCA: 235] [Impact Index Per Article: 58.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/03/2021] [Accepted: 05/06/2021] [Indexed: 12/13/2022]
Abstract
The RAF-MEK-ERK signaling cascade is a well-characterized MAPK pathway involved in cell proliferation and survival. The three-layered MAPK signaling cascade is initiated upon RTK and RAS activation. Three RAF isoforms ARAF, BRAF and CRAF, and their downstream MEK1/2 and ERK1/2 kinases constitute a coherently orchestrated signaling module that directs a range of physiological functions. Genetic alterations in this pathway are among the most prevalent in human cancers, which consist of numerous hot-spot mutations such as BRAFV600E. Oncogenic mutations in this pathway often override otherwise tightly regulated checkpoints to open the door for uncontrolled cell growth and neoplasia. The crosstalk between the RAF-MEK-ERK axis and other signaling pathways further extends the proliferative potential of this pathway in human cancers. In this review, we summarize the molecular architecture and physiological functions of the RAF-MEK-ERK pathway with emphasis on its dysregulations in human cancers, as well as the efforts made to target the RAF-MEK-ERK module using small molecule inhibitors.
Collapse
Affiliation(s)
- Rahim Ullah
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Qing Yin
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Aidan H Snell
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Lixin Wan
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA; Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA.
| |
Collapse
|
25
|
Nagamura Y, Miyazaki M, Nagano Y, Yuki M, Fukami K, Yanagihara K, Sasaki K, Sakai R, Yamaguchi H. PLEKHA5 regulates the survival and peritoneal dissemination of diffuse-type gastric carcinoma cells with Met gene amplification. Oncogenesis 2021; 10:25. [PMID: 33677467 PMCID: PMC7936979 DOI: 10.1038/s41389-021-00314-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 02/10/2021] [Accepted: 02/11/2021] [Indexed: 02/07/2023] Open
Abstract
Met gene amplification has been found in a subset of malignant carcinomas, including diffuse-type gastric carcinoma (DGC), which has a poor prognosis owing to rapid infiltrative invasion and frequent peritoneal dissemination. Met is considered a promising therapeutic target for DGC. However, DGC cells with Met gene amplification eventually acquire resistance to Met inhibitors. Therefore, identification of alternate targets that mediate Met signaling and confer malignant phenotypes is critical. In this study, we conducted a phosphoproteomic analysis of DGC cells possessing Met gene amplification and identified Pleckstrin Homology Domain Containing A5 (PLEKHA5) as a protein that is tyrosine-phosphorylated downstream of Met. Knockdown of PLEKHA5 selectively suppressed the growth of DGC cells with Met gene amplification by inducing apoptosis, even though they had acquired resistance to Met inhibitors. Moreover, PLEKHA5 silencing abrogated the malignant phenotypes of Met-addicted DGC cells, including peritoneal dissemination in vivo. Mechanistically, PLEKHA5 knockdown dysregulates glycolytic metabolism, leading to activation of the JNK pathway that promotes apoptosis. These results indicate that PLEKHA5 is a novel downstream effector of amplified Met and is required for the malignant progression of Met-addicted DGC.
Collapse
Affiliation(s)
- Yuko Nagamura
- Department of Cancer Cell Research, Sasaki Institute, Sasaki Foundation, Tokyo, Japan
| | - Makoto Miyazaki
- Department of Cancer Cell Research, Sasaki Institute, Sasaki Foundation, Tokyo, Japan
| | - Yoshiko Nagano
- Department of Cancer Cell Research, Sasaki Institute, Sasaki Foundation, Tokyo, Japan
| | - Masako Yuki
- Department of Cancer Cell Research, Sasaki Institute, Sasaki Foundation, Tokyo, Japan.,Laboratory of Genome and Biosignal, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Kiyoko Fukami
- Laboratory of Genome and Biosignal, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Kazuyoshi Yanagihara
- Division of Biomarker Discovery, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Chiba, Japan
| | - Kazuki Sasaki
- Department of Peptidomics, Sasaki Institute, Sasaki Foundation, Tokyo, Japan
| | - Ryuichi Sakai
- Department of Biochemistry, Kitasato University School of Medicine, Kanagawa, Japan
| | - Hideki Yamaguchi
- Department of Cancer Cell Research, Sasaki Institute, Sasaki Foundation, Tokyo, Japan.
| |
Collapse
|
26
|
ERK signalling: a master regulator of cell behaviour, life and fate. Nat Rev Mol Cell Biol 2020; 21:607-632. [PMID: 32576977 DOI: 10.1038/s41580-020-0255-7] [Citation(s) in RCA: 643] [Impact Index Per Article: 128.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/07/2020] [Indexed: 12/13/2022]
Abstract
The proteins extracellular signal-regulated kinase 1 (ERK1) and ERK2 are the downstream components of a phosphorelay pathway that conveys growth and mitogenic signals largely channelled by the small RAS GTPases. By phosphorylating widely diverse substrates, ERK proteins govern a variety of evolutionarily conserved cellular processes in metazoans, the dysregulation of which contributes to the cause of distinct human diseases. The mechanisms underlying the regulation of ERK1 and ERK2, their mode of action and their impact on the development and homeostasis of various organisms have been the focus of much attention for nearly three decades. In this Review, we discuss the current understanding of this important class of kinases. We begin with a brief overview of the structure, regulation, substrate recognition and subcellular localization of ERK1 and ERK2. We then systematically discuss how ERK signalling regulates six fundamental cellular processes in response to extracellular cues. These processes are cell proliferation, cell survival, cell growth, cell metabolism, cell migration and cell differentiation.
Collapse
|
27
|
Li KP, Ladle BH, Kurtulus S, Sholl A, Shanmuganad S, Hildeman DA. T-cell receptor signal strength and epigenetic control of Bim predict memory CD8 + T-cell fate. Cell Death Differ 2020; 27:1214-1224. [PMID: 31558776 PMCID: PMC7206134 DOI: 10.1038/s41418-019-0410-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 05/31/2019] [Accepted: 08/21/2019] [Indexed: 12/24/2022] Open
Abstract
Most effector CD8+ T cells die, while some persist and become either "effector" (TEM) or "central" (TCM) memory T cells. Paradoxically, effector CD8+ T cells with greater memory potential have higher levels of the pro-apoptotic molecule Bim. Here, we report, using a novel Bim-mCherry knock-in mouse, that cells with high levels of Bim preferentially develop into TCM cells. Bim levels remained stable and were regulated by DNA methylation at the Bim promoter. Notably, high levels of Bcl-2 were required for Bimhi cells to survive. Using Nur77-GFP mice as an indicator of TCR signal strength, Nur77 levels correlated with Bim expression and Nur77hi cells also selectively developed into TCM cells. Altogether, these data show that Bim levels and TCR signal strength are predictive of TEM- vs. TCM-cell fate. Further, given the many other biologic functions of Bim, these mice will have broad utility beyond CD8+ T-cell fate.
Collapse
Affiliation(s)
- Kun-Po Li
- Immunology Graduate Program, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA
| | - Brian H Ladle
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA
- Johns Hopkins Hospital, 1800 Orleans Street, The Charlotte R. Bloomberg Children's Center Building, 11th Floor, Baltimore, MD, 21287, USA
| | - Sema Kurtulus
- Immunology Graduate Program, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA
| | - Allyson Sholl
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA
| | - Sharmila Shanmuganad
- Immunology Graduate Program, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA
| | - David A Hildeman
- Immunology Graduate Program, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA.
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA.
| |
Collapse
|
28
|
Barez SR, Atar AM, Aghaei M. Mechanism of inositol-requiring enzyme 1-alpha inhibition in endoplasmic reticulum stress and apoptosis in ovarian cancer cells. J Cell Commun Signal 2020; 14:403-415. [PMID: 32200504 DOI: 10.1007/s12079-020-00562-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Accepted: 03/09/2020] [Indexed: 02/06/2023] Open
Abstract
IRE1α endonuclease is a key regulator of endoplasmic reticulum (ER) stress that controls cell survival/apoptosis in cancers. Inhibition of IRE1α endonuclease leads to decreased splice XBP1 which decreases cell proliferation and increases cell death in cancer cells. Therefore, this study investigated the effects and mechanism of STF-083010 (an IRE1α inhibitor) on the cell growth/apoptosis of ovarian malignant cells via the XBP1-CHOP-Bim pathway following the induction of ER stress (ERS). ERS in OVCAR3 and SKOV3 cells was measured using Thioflavin T staining. The expression of ER stress response genes was evaluated by QRT-PCR. The levels of XBP1(s), PERK, phospho-PERK, p-PP2A, ATF4, BIP/GRP78, CHOP, and Bim proteins were evaluated using western blotting. Cell viability and apoptosis in STF-083010 and Tunicamycin (Tm) co-treated cells were assessed using BrdU, MTT, Annexin V-FITC/PI staining, and caspases-12 and -3 activity assays. The results showed increased XBP1, CHOP, and ATF-4 mRNA expression levels as well as high protein aggregation in STF-083010 and Tm co-treated cells. The IRE1α inhibitor down-regulated sXBP1 and BIP proteins, while XBP-1, p-PERK, ATF-4, CHOP, and Bim proteins were up-regulated. STF-083010 reduced cell proliferation and induced apoptosis through the activation of caspases-12 and -3 and Bax/Bcl-2 protein expression. In summary, the present data revealed the effects of STF-083010 in ER stress and apoptosis as well as signaling via XBP1/CHOP/Bim mediators. Thus, STF-083010 is proposed as a new target for the control of ERS in ovarian cancer cells.
Collapse
Affiliation(s)
- Shekufe Rezghi Barez
- Department of Clinical Biochemistry, School of Pharmacy & Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ahmad Movahedian Atar
- Department of Clinical Biochemistry, School of Pharmacy & Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mahmoud Aghaei
- Department of Clinical Biochemistry, School of Pharmacy & Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
29
|
Mei AHC, Tung K, Han J, Perumal D, Laganà A, Keats J, Auclair D, Chari A, Jagannath S, Parekh S, Cho HJ. MAGE-A inhibit apoptosis and promote proliferation in multiple myeloma through regulation of BIM and p21 Cip1. Oncotarget 2020; 11:727-739. [PMID: 32133047 PMCID: PMC7041939 DOI: 10.18632/oncotarget.27488] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 01/29/2020] [Indexed: 12/22/2022] Open
Abstract
The type I Melanoma Antigen Gene (MAGE) A3 is a functional target associated with survival and proliferation in multiple myeloma (MM). To investigate the mechanisms of these oncogenic functions, we performed gene expression profiling (GEP) of p53 wild-type human myeloma cell lines (HMCL) after MAGE-A knockdown, which identified a set of 201 differentially expressed genes (DEG) associated with apoptosis, DNA repair, and cell cycle regulation. MAGE knockdown increased protein levels of pro-apoptotic BIM and of the endogenous cyclin-dependent kinase (CDK) inhibitor p21Cip1. Depletion of MAGE-A in HMCL increased sensitivity to the alkylating agent melphalan but not to proteasome inhibition. High MAGEA3 was associated with the MYC and Cell Cycling clusters defined by a network model of GEP data from the CoMMpass database of newly diagnosed, untreated MM patients. Comparative analysis of CoMMpass subjects based on high or low MAGEA3 expression revealed a set of 6748 DEG that also had significant functional associations with cell cycle and DNA replication pathways, similar to that observed in HMCL. High MAGEA3 expression correlated with shorter overall survival after melphalan chemotherapy and autologous stem cell transplantation (ASCT). These results demonstrate that MAGE-A3 regulates Bim and p21Cip1 transcription and protein expression, inhibits apoptosis, and promotes proliferation.
Collapse
Affiliation(s)
- Anna Huo-Chang Mei
- Tisch Cancer Institute, Icahn School of Medicine at Mt. Sinai, New York, NY, USA
| | - Kaity Tung
- Tisch Cancer Institute, Icahn School of Medicine at Mt. Sinai, New York, NY, USA
| | - Jessie Han
- Tisch Cancer Institute, Icahn School of Medicine at Mt. Sinai, New York, NY, USA
| | - Deepak Perumal
- Tisch Cancer Institute, Icahn School of Medicine at Mt. Sinai, New York, NY, USA
| | - Alessandro Laganà
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mt. Sinai, New York, NY, USA
- Institute for Next Generation Healthcare, Icahn School of Medicine at Mt. Sinai, New York, NY, USA
| | - Jonathan Keats
- Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Daniel Auclair
- The Multiple Myeloma Research Foundation, Norwalk, CT, USA
| | - Ajai Chari
- Tisch Cancer Institute, Icahn School of Medicine at Mt. Sinai, New York, NY, USA
| | - Sundar Jagannath
- Tisch Cancer Institute, Icahn School of Medicine at Mt. Sinai, New York, NY, USA
| | - Samir Parekh
- Tisch Cancer Institute, Icahn School of Medicine at Mt. Sinai, New York, NY, USA
| | - Hearn Jay Cho
- Tisch Cancer Institute, Icahn School of Medicine at Mt. Sinai, New York, NY, USA
- The Multiple Myeloma Research Foundation, Norwalk, CT, USA
| |
Collapse
|
30
|
Dynamics and Regulations of BimEL Ser65 and Thr112 Phosphorylation in Porcine Granulosa Cells during Follicular Atresia. Cells 2020; 9:cells9020402. [PMID: 32050589 PMCID: PMC7072439 DOI: 10.3390/cells9020402] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 01/31/2020] [Accepted: 02/05/2020] [Indexed: 01/07/2023] Open
Abstract
BimEL protein is involved in follicular atresia by regulating granulosa cell apoptosis, but the dynamic changes of BimEL phosphorylation during follicular atresia are poorly understood. The aim of this study was to explore the changes of key BimEL phosphorylation sites and their upstream regulatory pathways. First, the levels of BimEL-Ser65 and BimEL-Thr112 phosphorylation (p-BimEL-S65, p-BimEL-T112) in granulosa cells (GC) from healthy (H), slightly-atretic (SA), and atretic (A) follicles and in cultured GC after different treatments were detected by Western blotting. Next, the effects of the corresponding site mutations of BIM on apoptosis of GC were investigated. Finally, the pathways of two phosphorylation sites were investigated by kinase inhibitors. The results revealed that p-BimEL-S65 levels were higher in GC from H than SA and A, whereas p-BimEL-T112 was reversed. The prosurvival factors like FSH and IGF-1 upregulated the level of p-BimEL-S65, while the proapoptotic factor, heat stress, increased the level of p-BimEL-T112 in cultured GC. Compared with the overexpression of wild BimEL, the apoptotic rate of the GC overexpressed BimEL-S65A (replace Ser65 with Ala) mutant was significantly higher, but the apoptotic rate of the cells overexpressing BimEL-T112A did not differ. In addition, inhibition of the ERK1/2 or JNK pathway by specific inhibitors reduced the levels of p-BimEL-S65 and p-BimEL-T112. In conclusion, the levels of p-BimEL-S65 and p-BimEL-T112 were reversed during follicular atresia. Prosurvival factors promote p-BimEL-S65 levels via ERK1/2 to inhibit GC apoptosis, whereas proapoptotic factor upregulates the level of p-BimEL-T112 via JNK to induce GC apoptosis.
Collapse
|
31
|
Han Y, Wang S, Wang Y, Zeng S. IGF-1 Inhibits Apoptosis of Porcine Primary Granulosa Cell by Targeting Degradation of Bim EL. Int J Mol Sci 2019; 20:ijms20215356. [PMID: 31661816 PMCID: PMC6861984 DOI: 10.3390/ijms20215356] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 09/01/2019] [Accepted: 09/04/2019] [Indexed: 01/02/2023] Open
Abstract
Insulin-like growth factor-1 (IGF-1) is an intra-ovarian growth factor that plays important endocrine or paracrine roles during ovarian development. IGF-1 affects ovarian function and female fertility through reducing apoptosis of granulosa cells, yet the underlying mechanism remains poorly characterized. Here, we aimed to address these knowledge gaps using porcine primary granulosa cells and examining the anti-apoptotic mechanisms of IGF-1. IGF-1 prevented the granulosa cell from apoptosis, as shown by TUNEL and Annexin V/PI detection, and gained the anti-apoptotic index, the ratio of Bcl-2/Bax. This process was partly mediated by reducing the pro-apoptotic BimEL (Bcl-2 Interacting Mediator of Cell Death-Extra Long) protein level. Western blotting showed that IGF-1 promoted BimEL phosphorylation through activating p-ERK1/2, and that the proteasome system was responsible for degradation of phosphorylated BimEL. Meanwhile, IGF-1 enhanced the Beclin1 level and the rate of LC3 II/LC3 I, indicating that autophagy was induced by IGF-1. By blocking the proteolysis processes of both proteasome and autophagy flux with MG132 and chloroquine, respectively, the BimEL did not reduce and the phosphorylated BimEL protein accumulated, thereby indicating that both proteasome and autophagy pathways were involved in the degradation of BimEL stimulated by IGF-1. In conclusion, IGF-1 inhibited porcine primary granulosa cell apoptosis via degradation of pro-apoptotic BimEL. This study is critical for us to further understand the mechanisms of follicular survival and atresia regulated by IGF-1. Moreover, it provides a direction for the treatment of infertility caused by ovarian dysplasia, such as polycystic ovary syndrome and the improvement of assisted reproductive technology.
Collapse
Affiliation(s)
- Ying Han
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding, and Reproduction of the Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Shumin Wang
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding, and Reproduction of the Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Yingzheng Wang
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding, and Reproduction of the Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Shenming Zeng
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding, and Reproduction of the Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
32
|
Trastuzumab in combination with paclitaxel enhances antitumor activity by promoting apoptosis in human epidermal growth factor receptor 2-positive trastuzumab-resistant gastric cancer xenograft models. Anticancer Drugs 2019; 31:241-250. [PMID: 31633500 DOI: 10.1097/cad.0000000000000853] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Trastuzumab, a humanized anti-human epidermal growth factor receptor 2 antibody drug, is the first-line therapy for human epidermal growth factor receptor 2-positive breast and gastric cancer. For breast cancer, the benefit of continuous treatment with trastuzumab after it becomes refractory to first-line therapy has been demonstrated. However, it is unclear whether trastuzumab can show similar efficacy as a second-line treatment for gastric cancer. Here, we report that trastuzumab in combination with paclitaxel exhibits increased antitumor efficacy even for trastuzumab-resistant xenografted tumors. We derived the trastuzumab-resistant models from previously established human epidermal growth factor receptor 2-positive gastric cancer patient-derived cells. Human epidermal growth factor receptor 2 expression, PIK3CA mutation, and phosphatase and tensin homolog expression in these resistant models was equivalent to those in the trastuzumab-sensitive parental model, whereas cyclin-dependent kinase inhibitors, such as p16, p15, and p21, were downregulated. Trastuzumab in combination with paclitaxel enhanced antitumor activity in both the sensitive and resistant models. In the trastuzumab-sensitive model, the combination of trastuzumab and paclitaxel resulted in suppression of the AKT-p27-retinoblastoma protein pathway and induction of apoptosis. Although this combination did not suppress retinoblastoma protein phosphorylation in the trastuzumab-resistant model, it did markedly decrease epidermal growth factor receptor and human epidermal growth factor receptor 2 phosphorylation and further enhance paclitaxel-mediated apoptosis. These results suggested that trastuzumab in combination with paclitaxel can still exert more potent antitumor efficacy than each agent alone in trastuzumab-resistant models, providing evidence that trastuzumab remains beneficial in the treatment of trastuzumab-resistant tumors.
Collapse
|
33
|
健愉 冯, 玉山 朱, 陈 权, 凌 林, Jianyu F, Yushan Z, Quan C, Jialing L. [Physiological Function and Structural Basis of Bcl-2 Family Proteins]. ZHONGGUO XI BAO SHENG WU XUE XUE BAO 2019; 41:1477-1489. [PMID: 34249113 PMCID: PMC8265309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Apoptosis is an important biological process that plays a key role in the regulation of cell fate and homeostasis. The B-cell lymphoma-2 (Bcl-2) family proteins are important regulators of the apoptotic pathway, and their dysfunction is associated with a variety of diseases, including cancer, neurodegenerative and autoimmune diseases. In the past decade, a large number of research work on the physiological functions and atomic structures of Bcl-2 family proteins have been reported, which has deepened our understanding of the molecular mechanism and pathological significance of Bcl-2 family proteins. Recently, new drugs targeting different Bcl-2 proteins have been developed and used in clinics or tested in clinical trials. However, the complexity and diversity in functions and structures of Bcl-2 family have left many unsolved problems. This article summarizes current knowledge of the structure and function of Bcl-2 family proteins and discusses the pharmacological significance of Bcl-2 proteins as effective therapeutic targets.
Collapse
Affiliation(s)
| | | | | | - 林家 凌
- 俄克拉荷马大学健康科学中心生物化学与分子生物学系, 俄克拉何马城 73126-0901
| | - Feng Jianyu
- College of Life Sciences, Nankai University, Tianjin 300074, China
| | - Zhu Yushan
- College of Life Sciences, Nankai University, Tianjin 300074, China
| | - Chen Quan
- College of Life Sciences, Nankai University, Tianjin 300074, China
| | - Lin Jialing
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma 73126, USA
| |
Collapse
|
34
|
Zhang M, Wang X, Guo F, Jia Q, Liu N, Chen Y, Yan Y, Huang M, Tang H, Deng Y, Huang S, Zhou Z, Zhang L, Zhang L. Cdc42 Deficiency Leads To Epidermal Barrier Dysfunction by Regulating Intercellular Junctions and Keratinization of Epidermal Cells during Mouse Skin Development. Am J Cancer Res 2019; 9:5065-5084. [PMID: 31410202 PMCID: PMC6691388 DOI: 10.7150/thno.34014] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 06/08/2019] [Indexed: 01/19/2023] Open
Abstract
Rationale: Cdc42 is a Rho GTPase that regulates diverse cellular functions. Here, we used genetic techniques to investigate the role of Cdc42 in epidermal development and epidermal barrier formation. Methods: Keratinocyte-restricted Cdc42 knockout mice were generated with the Cre-LoxP system under the keratin 14 (K14) promoter. The skin and other tissues were collected from mutant and wild-type mice, and their cellular, molecular, morphological, and physiological features were analyzed. Results: Loss of Cdc42 in the epidermis in vivo resulted in neonatal lethality and impairment of epidermal barrier formation. Cdc42 deficiency led to the loss of epidermal stem cells. The absence of Cdc42 led to increased thickening of the epidermis, which was associated with increased proliferation and reduced apoptosis of keratinocytes. In addition, Cdc42 deficiency damaged tight junctions, adherens junctions and desmosomes. RNA sequencing results showed that the most significantly altered genes were enriched by the terms of “keratinization” and “cornified envelope” (CE). Among the differentially expressed genes in the CE term, several members of the small proline-rich protein (SPRR) family were upregulated. Further study revealed that there may be a Cdc42-SPRR pathway, which may correlate with epidermal barrier function. Conclusions: Our study indicates that Cdc42 is essential for epidermal development and epidermal barrier formation. Defects in Cdc42-SPRR signaling may be associated with skin barrier dysfunction and a variety of skin diseases.
Collapse
|
35
|
Aurora kinase A drives the evolution of resistance to third-generation EGFR inhibitors in lung cancer. Nat Med 2018; 25:111-118. [PMID: 30478424 PMCID: PMC6324945 DOI: 10.1038/s41591-018-0264-7] [Citation(s) in RCA: 203] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 10/04/2018] [Indexed: 12/30/2022]
Abstract
Although targeted therapies often elicit profound initial patient responses, these effects are transient due to residual disease leading to acquired resistance. How tumors transition between drug responsiveness, tolerance and resistance, especially in the absence of preexisting subclones, remains unclear. In epidermal growth factor receptor (EGFR)-mutant lung adenocarcinoma cells, we demonstrate that residual disease and acquired resistance in response to EGFR inhibitors requires Aurora kinase A (AURKA) activity. Nongenetic resistance through the activation of AURKA by its coactivator TPX2 emerges in response to chronic EGFR inhibition where it mitigates drug-induced apoptosis. Aurora kinase inhibitors suppress this adaptive survival program, increasing the magnitude and duration of EGFR inhibitor response in preclinical models. Treatment-induced activation of AURKA is associated with resistance to EGFR inhibitors in vitro, in vivo and in most individuals with EGFR-mutant lung adenocarcinoma. These findings delineate a molecular path whereby drug resistance emerges from drug-tolerant cells and unveils a synthetic lethal strategy for enhancing responses to EGFR inhibitors by suppressing AURKA-driven residual disease and acquired resistance.
Collapse
|
36
|
González R, Molina-Ruiz FJ, Bárcena JA, Padilla CA, Muntané J. Regulation of Cell Survival, Apoptosis, and Epithelial-to-Mesenchymal Transition by Nitric Oxide-Dependent Post-Translational Modifications. Antioxid Redox Signal 2018; 29:1312-1332. [PMID: 28795583 DOI: 10.1089/ars.2017.7072] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
SIGNIFICANCE Nitric oxide (NO) is a physiopathological messenger generating different reactive nitrogen species (RNS) according to hypoxic, acidic and redox conditions. Recent Advances: RNS and reactive oxygen species (ROS) promote relevant post-translational modifications, such as nitrosation, nitration, and oxidation, in critical components of cell proliferation and death, epithelial-to-mesenchymal transition, and metastasis. CRITICAL ISSUES The pro- or antitumoral properties of NO are dependent on local concentration, redox state, cellular status, duration of exposure, and compartmentalization of NO generation. The increased expression of NO synthase has been associated with cancer progression. However, the experimental strategies leading to high intratumoral NO generation have been shown to exert antitumoral properties. The effect of NO and ROS on cell signaling is critically altered by factors modulating tumor progression such as oxygen content, metabolism, and inflammatory response. The review describes the alteration of key components involved in cell survival and death, metabolism, and metastasis induced by RNS- and ROS-related post-translational modifications. FUTURE DIRECTIONS The identification of the molecular targets affected by nitrosation, nitration, and oxidation, as well as their interactions with other post-translational modifications, will improve the understanding on the complex signaling and cell fate decision in cancer. The therapeutic NO-based strategies have to address the complex crosstalk among NO and ROS with regard to critical components affecting tumor cell survival, metabolism, and metastasis in the progression of cancer, as well as close interaction with ionizing radiation and chemotherapy.
Collapse
Affiliation(s)
- Raúl González
- 1 Institute of Biomedicine of Seville (IBiS), IBiS/"Virgen del Rocío" University Hospital/CSIC/University of Seville , Seville, Spain
| | - Francisco J Molina-Ruiz
- 1 Institute of Biomedicine of Seville (IBiS), IBiS/"Virgen del Rocío" University Hospital/CSIC/University of Seville , Seville, Spain
| | - J Antonio Bárcena
- 2 Department of Biochemistry and Molecular Biology, Maimonides Biomedical Research Institute of Córdoba (IMIBIC), University of Córdoba , Córdoba, Spain
| | - C Alicia Padilla
- 2 Department of Biochemistry and Molecular Biology, Maimonides Biomedical Research Institute of Córdoba (IMIBIC), University of Córdoba , Córdoba, Spain
| | - Jordi Muntané
- 3 Department of General Surgery, "Virgen del Rocío" University Hospital/IBiS/CSIC/University of Seville , Seville, Spain .,4 Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd) , Madrid, Spain
| |
Collapse
|
37
|
Wang Y, Zeng S. Melatonin Promotes Ubiquitination of Phosphorylated Pro-Apoptotic Protein Bcl-2-Interacting Mediator of Cell Death-Extra Long (Bim EL) in Porcine Granulosa Cells. Int J Mol Sci 2018; 19:ijms19113431. [PMID: 30388852 PMCID: PMC6274928 DOI: 10.3390/ijms19113431] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 10/27/2018] [Accepted: 10/27/2018] [Indexed: 12/31/2022] Open
Abstract
Melatonin (N-acetyl-5-methoxytryptamine) is found in ovarian follicular fluid, and its concentration is closely related to follicular health status. Nevertheless, the molecular mechanisms underlying melatonin function in follicles are uncertain. In this study, melatonin concentration was measured in porcine follicular fluid at different stages of health. The melatonin concentration decreased as the follicles underwent atresia, suggesting that melatonin may participate in the maintenance of follicular health. The molecular pathway through which melatonin may regulate follicular development was further investigated. The pro-apoptotic protein BimEL (Bcl-2-interacting mediator of cell death-Extra Long), a key protein controlling granulosa cell apoptosis during follicular atresia, was selected as the target molecule. BimEL was downregulated when porcine granulosa cells were cultured in medium containing 10−9 M melatonin and isolated cumulus oocyte complexes (COCs) or follicle stimulating hormone (FSH). Interestingly, ERK-mediated phosphorylation was a prerequisite for the melatonin-induced decline in BimEL, and melatonin only promoted the ubiquitination of phosphorylated BimEL, and did not affect the activities of the lysosome or the proteasome. Moreover, the melatonin-induced downregulation of BimEL was independent of its receptor and its antioxidant properties. In conclusion, melatonin may maintain follicular health by inducing BimEL ubiquitination to inhibit the apoptosis of granulosa cells.
Collapse
Affiliation(s)
- Yingzheng Wang
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China.
| | - Shenming Zeng
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China.
| |
Collapse
|
38
|
Oudenaarden CRL, van de Ven RAH, Derksen PWB. Re-inforcing the cell death army in the fight against breast cancer. J Cell Sci 2018; 131:131/16/jcs212563. [DOI: 10.1242/jcs.212563] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
ABSTRACT
Metastatic breast cancer is responsible for most breast cancer-related deaths. Disseminated cancer cells have developed an intrinsic ability to resist anchorage-dependent apoptosis (anoikis). Anoikis is caused by the absence of cellular adhesion, a process that underpins lumen formation and maintenance during mammary gland development and homeostasis. In healthy cells, anoikis is mostly governed by B-cell lymphoma-2 (BCL2) protein family members. Metastatic cancer cells, however, have often developed autocrine BCL2-dependent resistance mechanisms to counteract anoikis. In this Review, we discuss how a pro-apoptotic subgroup of the BCL2 protein family, known as the BH3-only proteins, controls apoptosis and anoikis during mammary gland homeostasis and to what extent their inhibition confers tumor suppressive functions in metastatic breast cancer. Specifically, the role of the two pro-apoptotic BH3-only proteins BCL2-modifying factor (BMF) and BCL2-interacting mediator of cell death (BIM) will be discussed here. We assess current developments in treatment that focus on mimicking the function of the BH3-only proteins to induce apoptosis, and consider their applicability to restore normal apoptotic responses in anchorage-independent disseminating tumor cells.
Collapse
Affiliation(s)
- Clara R. L. Oudenaarden
- UMC Utrecht, Department of Pathology, Heidelberglaan 100, 3584CX Utrecht, The Netherlands
- Lund University, Department of Experimental Oncology, Scheelevägen 2, 22363 Lund, Sweden
| | - Robert A. H. van de Ven
- UMC Utrecht, Department of Pathology, Heidelberglaan 100, 3584CX Utrecht, The Netherlands
- Harvard Medical School, Department of Cell Biology, 250 Longwood Avenue, Boston, MA 02115, USA
| | - Patrick W. B. Derksen
- UMC Utrecht, Department of Pathology, Heidelberglaan 100, 3584CX Utrecht, The Netherlands
| |
Collapse
|
39
|
Singh PK, Weber A, Häcker G. The established and the predicted roles of dynein light chain in the regulation of mitochondrial apoptosis. Cell Cycle 2018; 17:1037-1047. [PMID: 30019621 DOI: 10.1080/15384101.2018.1464851] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Abstract
The mitochondrial pathway of apoptosis is regulated by the interplay between the members of Bcl-2 family. Within this family, BH3-only proteins are the sensors of apoptotic stimuli and can trigger apoptosis either by inhibiting the anti-apoptotic Bcl-2-family proteins or by directly activating the effectors Bax and Bak. An expanding body of research suggests that a number of non-Bcl-2 proteins can also interact with Bcl-2 proteins and contribute to the decision of cell fate. Dynein light chain (LC8, DYNLL or DLC), a hub protein and a dimerizing engine has been proposed to regulate the pro-apoptotic activity of two BH3-only proteins, Bim and Bmf. Our recent work has provided insight into the mechanisms through which DLC1 (DYNLL1) modulates Bim activity. Here we discuss the present day understanding of Bim-DLC interaction and endeavor to evaluate this interaction in the light of information from studies of DLC with other binding partners.
Collapse
Affiliation(s)
- Prafull Kumar Singh
- a Institute of Medical Microbiology and Hygiene, Faculty of Medicine , Medical Center-University of Freiburg , Freiburg , Germany
| | - Arnim Weber
- a Institute of Medical Microbiology and Hygiene, Faculty of Medicine , Medical Center-University of Freiburg , Freiburg , Germany
| | - Georg Häcker
- a Institute of Medical Microbiology and Hygiene, Faculty of Medicine , Medical Center-University of Freiburg , Freiburg , Germany.,b BIOSS Centre for Biological Signalling Studies , University of Freiburg , Freiburg , Germany
| |
Collapse
|
40
|
Girnius N, Davis RJ. JNK Promotes Epithelial Cell Anoikis by Transcriptional and Post-translational Regulation of BH3-Only Proteins. Cell Rep 2018; 21:1910-1921. [PMID: 29141222 DOI: 10.1016/j.celrep.2017.10.067] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 09/27/2017] [Accepted: 10/18/2017] [Indexed: 11/18/2022] Open
Abstract
Developmental morphogenesis, tissue injury, and oncogenic transformation can cause the detachment of epithelial cells. These cells are eliminated by a specialized form of apoptosis (anoikis). While the processes that contribute to this form of cell death have been studied, the underlying mechanisms remain unclear. Here, we tested the role of the cJUN NH2-terminal kinase (JNK) signaling pathway using murine models with compound JNK deficiency in mammary and kidney epithelial cells. These studies demonstrated that JNK is required for efficient anoikis in vitro and in vivo. Moreover, JNK-promoted anoikis required pro-apoptotic members of the BCL2 family of proteins. We show that JNK acts through a BAK/BAX-dependent apoptotic pathway by increasing BIM expression and phosphorylating BMF, leading to death of detached epithelial cells.
Collapse
Affiliation(s)
- Nomeda Girnius
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Roger J Davis
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA; Howard Hughes Medical Institute, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
41
|
Conage-Pough JE, Boise LH. Phosphorylation alters Bim-mediated Mcl-1 stabilization and priming. FEBS J 2018; 285:2626-2640. [PMID: 29775995 DOI: 10.1111/febs.14505] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 04/15/2018] [Accepted: 05/14/2018] [Indexed: 12/18/2022]
Abstract
Mcl-1 is a highly labile protein, subject to extensive post-translational regulation. This distinguishes Mcl-1 from other antiapoptotic proteins and necessitates further study to better understand how interactions with proapoptotic Bcl-2 proteins affect its regulation. One such protein, Bim, is known to stabilize Mcl-1, and Bim phosphorylation has been associated with increased Mcl-1 binding. Consequently, we investigated the potential impact of Bim phosphorylation on Mcl-1 stability. We found that Bim stabilizes and primes Mcl-1 in RPCI-WM1 cells and is constitutively phosphorylated. Additionally, introduction of several phospho-mimetic and unphosphosphorylateable Bim mutations resulted in altered Mcl-1 stability and distinct Bim binding to antiapoptotic proteins. These findings suggest Bim phosphorylation not only regulates Mcl-1 stability but also is a potential mechanism for enforcing Mcl-1 dependence.
Collapse
Affiliation(s)
- Jason E Conage-Pough
- Cancer Biology Graduate Program, Winship Cancer Institute, Emory University, Atlanta, GA, USA.,Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Lawrence H Boise
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| |
Collapse
|
42
|
Fahl SP, Daamen AR, Crittenden RB, Bender TP. c-Myb Coordinates Survival and the Expression of Genes That Are Critical for the Pre-BCR Checkpoint. THE JOURNAL OF IMMUNOLOGY 2018; 200:3450-3463. [PMID: 29654210 DOI: 10.4049/jimmunol.1302303] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Accepted: 03/13/2018] [Indexed: 11/19/2022]
Abstract
The c-Myb transcription factor is required for adult hematopoiesis, yet little is known about c-Myb function during lineage-specific differentiation due to the embryonic lethality of Myb-null mutations. We previously used tissue-specific inactivation of the murine Myb locus to demonstrate that c-Myb is required for differentiation to the pro-B cell stage, survival during the pro-B cell stage, and the pro-B to pre-B cell transition during B lymphopoiesis. However, few downstream mediators of c-Myb-regulated function have been identified. We demonstrate that c-Myb regulates the intrinsic survival of CD19+ pro-B cells in the absence of IL-7 by repressing expression of the proapoptotic proteins Bmf and Bim and that levels of Bmf and Bim mRNA are further repressed by IL-7 signaling in pro-B cells. c-Myb regulates two crucial components of the IL-7 signaling pathway: the IL-7Rα-chain and the negative regulator SOCS3 in CD19+ pro-B cells. Bypassing IL-7R signaling through constitutive activation of Stat5b largely rescues survival of c-Myb-deficient pro-B cells, whereas constitutively active Akt is much less effective. However, rescue of pro-B cell survival is not sufficient to rescue proliferation of pro-B cells or the pro-B to small pre-B cell transition, and we further demonstrate that c-Myb-deficient large pre-B cells are hypoproliferative. Analysis of genes crucial for the pre-BCR checkpoint demonstrates that, in addition to IL-7Rα, the genes encoding λ5, cyclin D3, and CXCR4 are downregulated in the absence of c-Myb, and λ5 is a direct c-Myb target. Thus, c-Myb coordinates survival with the expression of genes that are required during the pre-BCR checkpoint.
Collapse
Affiliation(s)
- Shawn P Fahl
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia Health System, Charlottesville, VA 22908; and
| | - Andrea R Daamen
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia Health System, Charlottesville, VA 22908; and
| | - Rowena B Crittenden
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia Health System, Charlottesville, VA 22908; and
| | - Timothy P Bender
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia Health System, Charlottesville, VA 22908; and .,Beirne B. Carter Center for Immunology Research, University of Virginia Health System, Charlottesville, VA 22908
| |
Collapse
|
43
|
Seo SU, Woo SM, Min KJ, Kwon TK. Z-FL-COCHO, a cathepsin S inhibitor, enhances oxaliplatin-induced apoptosis through upregulation of Bim expression. Biochem Biophys Res Commun 2018. [DOI: 10.1016/j.bbrc.2018.03.068] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
44
|
Stevenson AJ, Ager EI, Proctor MA, Škalamera D, Heaton A, Brown D, Gabrielli BG. Mechanism of action of the third generation benzopyrans and evaluation of their broad anti-cancer activity in vitro and in vivo. Sci Rep 2018; 8:5144. [PMID: 29572477 PMCID: PMC5865165 DOI: 10.1038/s41598-018-22882-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 01/19/2018] [Indexed: 11/10/2022] Open
Abstract
Successive rounds of chemical modification in three generations of benzopyran molecules have shown to select for different mechanisms of actions and progressive increases in anti-cancer activity. In this study, we investigated the mechanism of action of the third-generation benzopyran compounds, TRX-E-002-1 and TRX-E-009-1. High-content screening of a panel of 240 cancer cell lines treated with TRX-E-009-1 demonstrated it has broad anti-cancer potential. Within this screen, melanoma cell lines showed a range of sensitivities and subsequently a second independent panel of 21 melanoma 3D spheroid lines were assessed for their responses to both TRX-E-002-1 and TRX-E-009-1 compounds. Time-lapse microscopy illustrated both of these compounds caused mitotic delays in treated cells, resulting in either mitotic slippage or apoptosis. This finding along with immunostaining, in vitro polymerization assays, and animal experiments in both athymic and immunocompetent mice, demonstrates that these third-generation benzopyran compounds are potent tubulin polymerization inhibitors in vitro and in vivo, and this is the molecular basis of their anti-cancer activity in melanoma. These findings indicate these BP compounds may offer a novel anti-microtubule strategy for cancer intervention and provides the basis for further investigation into biomarkers of clinical sensitivity.
Collapse
Affiliation(s)
- Alexander J Stevenson
- Mater Research Institute, The University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
| | | | - Martina A Proctor
- Mater Research Institute, The University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
| | - Dubravka Škalamera
- Mater Research Institute, The University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
| | - Andrew Heaton
- Novogen Ltd., Hornsby, New South Wales, Australia.,School of Medical Sciences, University of New South Wales Australia, Sydney, New South Wales, Australia
| | - David Brown
- Novogen Ltd., Hornsby, New South Wales, Australia.,School of Medical Sciences, University of New South Wales Australia, Sydney, New South Wales, Australia
| | - Brian G Gabrielli
- Mater Research Institute, The University of Queensland, Translational Research Institute, Brisbane, QLD, Australia.
| |
Collapse
|
45
|
Micro-Economics of Apoptosis in Cancer: ncRNAs Modulation of BCL-2 Family Members. Int J Mol Sci 2018; 19:ijms19040958. [PMID: 29570632 PMCID: PMC5979352 DOI: 10.3390/ijms19040958] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 03/20/2018] [Accepted: 03/21/2018] [Indexed: 12/31/2022] Open
Abstract
In the last few years, non-coding RNAs (ncRNAs) have been a hot topic in cancer research. Many ncRNAs were found to regulate the apoptotic process and to play a role in tumor cell resistance to treatment. The apoptotic program is on the frontline as self-defense from cancer onset, and evasion of apoptosis has been classified as one of the hallmarks of cancer responsible for therapy failure. The B-cell lymphoma 2 (BCL-2) family members are key players in the regulation of apoptosis and mediate the activation of the mitochondrial death machinery in response to radiation, chemotherapeutic agents and many targeted therapeutics. The balance between the pro-survival and the pro-apoptotic BCL-2 proteins is strictly controlled by ncRNAs. Here, we highlight the most common mechanisms exerted by microRNAs, long non-coding RNAs and circular RNAs on the main mediators of the intrinsic apoptotic cascade with particular focus on their significance in cancer biology.
Collapse
|
46
|
Girnius N, Edwards YJK, Davis RJ. The cJUN NH 2-terminal kinase (JNK) pathway contributes to mouse mammary gland remodeling during involution. Cell Death Differ 2018; 25:1702-1715. [PMID: 29511338 PMCID: PMC6143629 DOI: 10.1038/s41418-018-0081-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 01/23/2018] [Accepted: 01/29/2018] [Indexed: 11/18/2022] Open
Abstract
Involution returns the lactating mammary gland to a quiescent state after weaning. The mechanism of involution involves collapse of the mammary epithelial cell compartment. To test whether the cJUN NH2-terminal kinase (JNK) signal transduction pathway contributes to involution, we established mice with JNK deficiency in the mammary epithelium. We found that JNK is required for efficient involution. JNK deficiency did not alter the STAT3/5 or SMAD2/3 signaling pathways that have been previously implicated in this process. Nevertheless, JNK promotes the expression of genes that drive involution, including matrix metalloproteases, cathepsins, and BH3-only proteins. These data demonstrate that JNK has a key role in mammary gland involution post lactation.
Collapse
Affiliation(s)
- Nomeda Girnius
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Yvonne J K Edwards
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Roger J Davis
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, 01605, USA. .,Howard Hughes Medical Institute, University of Massachusetts Medical School, Worcester, MA, 01605, USA.
| |
Collapse
|
47
|
Rasmussen ML, Ortolano NA, Romero-Morales AI, Gama V. Wnt Signaling and Its Impact on Mitochondrial and Cell Cycle Dynamics in Pluripotent Stem Cells. Genes (Basel) 2018; 9:genes9020109. [PMID: 29463061 PMCID: PMC5852605 DOI: 10.3390/genes9020109] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 02/13/2018] [Accepted: 02/14/2018] [Indexed: 12/17/2022] Open
Abstract
The core transcriptional network regulating stem cell self-renewal and pluripotency remains an intense area of research. Increasing evidence indicates that modified regulation of basic cellular processes such as mitochondrial dynamics, apoptosis, and cell cycle are also essential for pluripotent stem cell identity and fate decisions. Here, we review evidence for Wnt regulation of pluripotency and self-renewal, and its connections to emerging features of pluripotent stem cells, including (1) increased mitochondrial fragmentation, (2) increased sensitivity to cell death, and (3) shortened cell cycle. We provide a general overview of the stem cell–specific mechanisms involved in the maintenance of these uncharacterized hallmarks of pluripotency and highlight potential links to the Wnt signaling pathway. Given the physiological importance of stem cells and their enormous potential for regenerative medicine, understanding fundamental mechanisms mediating the crosstalk between Wnt, organelle-dynamics, apoptosis, and cell cycle will be crucial to gain insight into the regulation of stemness.
Collapse
Affiliation(s)
- Megan L Rasmussen
- Department of Cell and Developmental Biology; Vanderbilt University, Nashville, TN37232, United States.
| | - Natalya A Ortolano
- Department of Cell and Developmental Biology; Vanderbilt University, Nashville, TN37232, United States.
| | | | - Vivian Gama
- Department of Cell and Developmental Biology; Vanderbilt University, Nashville, TN37232, United States.
- Vanderbilt Center for Stem Cell Biology; Vanderbilt University, Nashville, TN37232, United States.
- Vanderbilt Ingram Cancer Center; Vanderbilt University, Nashville, TN37232, United States.
| |
Collapse
|
48
|
Shee K, Yang W, Hinds JW, Hampsch RA, Varn FS, Traphagen NA, Patel K, Cheng C, Jenkins NP, Kettenbach AN, Demidenko E, Owens P, Faber AC, Golub TR, Straussman R, Miller TW. Therapeutically targeting tumor microenvironment-mediated drug resistance in estrogen receptor-positive breast cancer. J Exp Med 2018; 215:895-910. [PMID: 29436393 PMCID: PMC5839765 DOI: 10.1084/jem.20171818] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 12/29/2017] [Accepted: 01/01/2018] [Indexed: 12/22/2022] Open
Abstract
Tumor microenvironment (TME) cytokine screening revealed FGF2 as a clinically relevant mechanism of resistance to anti-estrogens, mTORC1 inhibition, and PI3K inhibition in ER+ breast cancer. Shee et al. highlight an underdeveloped aspect of precision oncology: treating patients according to their TME constitution. Drug resistance to approved systemic therapies in estrogen receptor–positive (ER+) breast cancer remains common. We hypothesized that factors present in the human tumor microenvironment (TME) drive drug resistance. Screening of a library of recombinant secreted microenvironmental proteins revealed fibroblast growth factor 2 (FGF2) as a potent mediator of resistance to anti-estrogens, mTORC1 inhibition, and phosphatidylinositol 3-kinase inhibition in ER+ breast cancer. Phosphoproteomic analyses identified ERK1/2 as a major output of FGF2 signaling via FGF receptors (FGFRs), with consequent up-regulation of Cyclin D1 and down-regulation of Bim as mediators of drug resistance. FGF2-driven drug resistance in anti-estrogen–sensitive and –resistant models, including patient-derived xenografts, was reverted by neutralizing FGF2 or FGFRs. A transcriptomic signature of FGF2 signaling in primary tumors predicted shorter recurrence-free survival independently of age, grade, stage, and FGFR amplification status. These findings delineate FGF2 signaling as a ligand-based drug resistance mechanism and highlights an underdeveloped aspect of precision oncology: characterizing and treating patients according to their TME constitution.
Collapse
Affiliation(s)
- Kevin Shee
- Department of Molecular and Systems Biology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH
| | - Wei Yang
- Department of Molecular and Systems Biology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH
| | - John W Hinds
- Department of Molecular and Systems Biology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH
| | - Riley A Hampsch
- Department of Molecular and Systems Biology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH
| | - Frederick S Varn
- Department of Molecular and Systems Biology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH.,Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Lebanon, NH
| | - Nicole A Traphagen
- Department of Molecular and Systems Biology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH
| | - Kishan Patel
- Department of Molecular and Systems Biology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH
| | - Chao Cheng
- Department of Molecular and Systems Biology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH.,Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Lebanon, NH
| | - Nicole P Jenkins
- Department of Biochemistry and Cell Biology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH
| | - Arminja N Kettenbach
- Department of Biochemistry and Cell Biology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH
| | - Eugene Demidenko
- Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Lebanon, NH
| | - Philip Owens
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN.,Research Medicine, Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN
| | - Anthony C Faber
- VCU Philips Institute for Oral Health Research, School of Dentistry and Massey Cancer Center, Virginia Commonwealth University, Richmond, VA
| | - Todd R Golub
- Broad Institute of MIT and Harvard, Cambridge, MA
| | - Ravid Straussman
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Todd W Miller
- Department of Molecular and Systems Biology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH .,Comprehensive Breast Program, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH
| |
Collapse
|
49
|
Aira LE, Villa E, Colosetti P, Gamas P, Signetti L, Obba S, Proics E, Gautier F, Bailly-Maitre B, Jacquel A, Robert G, Luciano F, Juin PP, Ricci JE, Auberger P, Marchetti S. The oncogenic tyrosine kinase Lyn impairs the pro-apoptotic function of Bim. Oncogene 2018; 37:2122-2136. [PMID: 29391601 DOI: 10.1038/s41388-017-0112-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 11/23/2017] [Accepted: 12/14/2017] [Indexed: 01/17/2023]
Abstract
Phosphorylation of Ser/Thr residues is a well-established modulating mechanism of the pro-apoptotic function of the BH3-only protein Bim. However, nothing is known about the putative tyrosine phosphorylation of this Bcl-2 family member and its potential impact on Bim function and subsequent Bax/Bak-mediated cytochrome c release and apoptosis. As we have previously shown that the tyrosine kinase Lyn could behave as an anti-apoptotic molecule, we investigated whether this Src family member could directly regulate the pro-apoptotic function of Bim. In the present study, we show that Bim is phosphorylated onto tyrosine residues 92 and 161 by Lyn, which results in an inhibition of its pro-apoptotic function. Mechanistically, we show that Lyn-dependent tyrosine phosphorylation of Bim increases its interaction with anti-apoptotic members such as Bcl-xL, therefore limiting mitochondrial outer membrane permeabilization and subsequent apoptosis. Collectively, our data uncover one molecular mechanism through which the oncogenic tyrosine kinase Lyn negatively regulates the mitochondrial apoptotic pathway, which may contribute to the transformation and/or the chemotherapeutic resistance of cancer cells.
Collapse
Affiliation(s)
| | - Elodie Villa
- Université Côte d'Azur, INSERM, C3M, Nice, France
| | | | | | | | | | - Emma Proics
- Université Côte d'Azur, INSERM, C3M, Nice, France
| | - Fabien Gautier
- CRCINA, UMR 1232 INSERM, Université de Nantes, Université d'Angers, Institut de Recherche en Santé-Université de Nantes, 8 Quai Moncousu - BP 70721, 44007, Nantes Cedex 1, France.,Institut de Cancérologie de l'Ouest, Bvd J Monod, Site René Gauducheau, 44805, Saint-Herblain, France
| | | | | | | | | | - Philippe P Juin
- CRCINA, UMR 1232 INSERM, Université de Nantes, Université d'Angers, Institut de Recherche en Santé-Université de Nantes, 8 Quai Moncousu - BP 70721, 44007, Nantes Cedex 1, France.,Institut de Cancérologie de l'Ouest, Bvd J Monod, Site René Gauducheau, 44805, Saint-Herblain, France
| | | | | | | |
Collapse
|
50
|
Müller L, Hainberger D, Stolz V, Hamminger P, Hassan H, Preglej T, Boucheron N, Sakaguchi S, Wiegers GJ, Villunger A, Auwerx J, Ellmeier W. The corepressor NCOR1 regulates the survival of single-positive thymocytes. Sci Rep 2017; 7:15928. [PMID: 29162920 PMCID: PMC5698297 DOI: 10.1038/s41598-017-15918-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 11/03/2017] [Indexed: 01/09/2023] Open
Abstract
Nuclear receptor corepressor 1 (NCOR1) is a transcriptional regulator bridging repressive chromatin modifying enzymes with transcription factors. NCOR1 regulates many biological processes, however its role in T cells is not known. Here we show that Cd4-Cre-mediated deletion of NCOR1 (NCOR1 cKOCd4) resulted in a reduction of peripheral T cell numbers due to a decrease in single-positive (SP) thymocytes. In contrast, double-positive (DP) thymocyte numbers were not affected in the absence of NCOR1. The reduction in SP cells was due to diminished survival of NCOR1-null postselection TCRβhiCD69+ and mature TCRβhiCD69- thymocytes. NCOR1-null thymocytes expressed elevated levels of the pro-apoptotic factor BIM and showed a higher fraction of cleaved caspase 3-positive cells upon TCR stimulation ex vivo. However, staphylococcal enterotoxin B (SEB)-mediated deletion of Vβ8+ CD4SP thymocytes was normal, suggesting that negative selection is not altered in the absence of NCOR1. Finally, transgenic expression of the pro-survival protein BCL2 restored the population of CD69+ thymocytes in NCOR1 cKOCd4 mice to a similar percentage as observed in WT mice. Together, these data identify NCOR1 as a crucial regulator of the survival of SP thymocytes and revealed that NCOR1 is essential for the proper generation of the peripheral T cell pool.
Collapse
Affiliation(s)
- Lena Müller
- Division of Immunobiology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090, Vienna, Austria
| | - Daniela Hainberger
- Division of Immunobiology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090, Vienna, Austria
| | - Valentina Stolz
- Division of Immunobiology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090, Vienna, Austria
| | - Patricia Hamminger
- Division of Immunobiology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090, Vienna, Austria
| | - Hammad Hassan
- Division of Immunobiology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090, Vienna, Austria
- Department of Biochemistry (Shankar Campus), Abdul Wali Khan University (AWKUM) Mardan, KPK, Pakistan
| | - Teresa Preglej
- Division of Immunobiology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090, Vienna, Austria
| | - Nicole Boucheron
- Division of Immunobiology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090, Vienna, Austria
| | - Shinya Sakaguchi
- Division of Immunobiology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090, Vienna, Austria
| | - G Jan Wiegers
- Innsbruck Medical University, Biocenter, Division of Developmental Immunology, Innsbruck, Austria
| | - Andreas Villunger
- Innsbruck Medical University, Biocenter, Division of Developmental Immunology, Innsbruck, Austria
| | - Johan Auwerx
- Ecole Polytechnique Fédérale de Lausanne, Laboratory of Integrative and Systems Physiology, Lausanne, Switzerland
| | - Wilfried Ellmeier
- Division of Immunobiology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090, Vienna, Austria.
| |
Collapse
|