1
|
Han Y, Zhang X, Miao L, Lin H, Zhuo Z, He J, Fu W. Biological function and mechanism of NAT10 in cancer. CANCER INNOVATION 2025; 4:e154. [PMID: 39817252 PMCID: PMC11732740 DOI: 10.1002/cai2.154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 06/11/2024] [Accepted: 06/24/2024] [Indexed: 01/18/2025]
Abstract
N-acetyltransferase 10 (NAT10) is a nucleolar acetyltransferase with an acetylation catalytic function and can bind various protein and RNA molecules. As the N4-acetylcytidine (ac4C) "writer" enzyme, NAT10 is reportedly involved in a variety of physiological and pathological activities. Currently, the NAT10-related molecular mechanisms in various cancers are not fully understood. In this review, we first describe the cellular localization of NAT10 and then summarize its numerous biological functions. NAT10 is involved in various biological processes by mediating the acetylation of different proteins and RNAs. These biological functions are also associated with cancer progression and patient prognosis. We also review the mechanisms by which NAT10 plays roles in various cancer types. NAT10 can affect tumor cell proliferation, metastasis, and stress tolerance through its acetyltransferase properties. Further research into NAT10 functions and expression regulation in tumors will help explore its future potential in cancer diagnosis, treatment, and prognosis.
Collapse
Affiliation(s)
- Yufeng Han
- Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Institute of PediatricsGuangzhou Medical UniversityGuangzhouGuangdongChina
| | - Xinxin Zhang
- Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Institute of PediatricsGuangzhou Medical UniversityGuangzhouGuangdongChina
| | - Lei Miao
- Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Institute of PediatricsGuangzhou Medical UniversityGuangzhouGuangdongChina
| | - Huiran Lin
- Faculty of MedicineMacau University of Science and TechnologyMacauChina
| | - Zhenjian Zhuo
- Laboratory Animal Center, School of Chemical Biology and BiotechnologyPeking University Shenzhen Graduate SchoolShenzhenGuangdongChina
- State Key Laboratory of Chemical OncogenomicsPeking University Shenzhen Graduate SchoolShenzhenGuangdongChina
| | - Jing He
- Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Institute of PediatricsGuangzhou Medical UniversityGuangzhouGuangdongChina
| | - Wen Fu
- Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Institute of PediatricsGuangzhou Medical UniversityGuangzhouGuangdongChina
| |
Collapse
|
2
|
Zhou B, Liu Y, Ma H, Zhang B, Lu B, Li S, Liu T, Qi Y, Wang Y, Zhang M, Qiu J, Fu R, Li W, Lu L, Tian S, Liu Q, Gu Y, Huang R, Lawrence T, Kong E, Zhang L, Li T, Liang Y. Zdhhc1 deficiency mitigates foam cell formation and atherosclerosis by inhibiting PI3K-Akt-mTOR signaling pathway through facilitating the nuclear translocation of p110α. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167577. [PMID: 39566590 DOI: 10.1016/j.bbadis.2024.167577] [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: 03/19/2024] [Revised: 10/17/2024] [Accepted: 11/11/2024] [Indexed: 11/22/2024]
Abstract
Monocyte-to-macrophage differentiation and subsequent foam cell formation are key processes that contribute to plaque build-up during the progression of atherosclerotic lesions. Palmitoylation enzymes are known to play pivotal roles in the development and progression of inflammatory diseases. However, their specific impact on atherosclerosis development remains unclear. In this study, we discovered that the knockout of zDHHC1 in THP-1 cells, as well as Zdhhc1 in mice, markedly reduces the uptake of oxidized low-density lipoprotein (ox-LDL) by macrophages, thereby inhibiting foam cell formation. Moreover, the absence of Zdhhc1 in ApoE-/- mice significantly suppresses atherosclerotic plaque formation. Mass spectrometry coupled with bioinformatic analysis revealed an enrichment of the PI3K-Akt-mTOR signaling pathway. Consistent with this, we observed that knockout of zDHHC1 significantly decreases the palmitoylation levels of p110α, a crucial subunit of PI3K. Notably, the deletion of Zdhhc1 facilitates the nuclear translocation of p110α in macrophages, leading to a significant reduction in the downstream phosphorylation of Akt at Ser473 and mTOR at Ser2448. This cascade results in a decreased number of macrophages within plaques and ultimately mitigates the severity of atherosclerosis. These findings unveil a novel role for zDHHC1 in regulating foam cell formation and the progression of atherosclerosis, suggesting it as a promising target for clinical intervention in atherosclerosis therapy.
Collapse
Affiliation(s)
- Binhui Zhou
- Laboratory of Genetic Regulators in the Immune System, School of Medical Technology, Xinxiang Medical University, Xinxiang 453003, China; The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, China; Laboratory of Mouse Genetics, Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang 453003, China.
| | - Yang Liu
- Laboratory of Genetic Regulators in the Immune System, School of Medical Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Haoyuan Ma
- Laboratory of Genetic Regulators in the Immune System, School of Medical Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Bowen Zhang
- Laboratory of Genetic Regulators in the Immune System, School of Medical Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Beijia Lu
- Laboratory of Mouse Genetics, Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang 453003, China
| | - Sainan Li
- Laboratory of Mouse Genetics, Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang 453003, China
| | - Tingting Liu
- Laboratory of Genetic Regulators in the Immune System, School of Medical Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Yingcheng Qi
- Laboratory of Genetic Regulators in the Immune System, School of Medical Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Ying Wang
- The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, China; Laboratory of Mouse Genetics, Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang 453003, China
| | - Mengjie Zhang
- Laboratory of Genetic Regulators in the Immune System, School of Medical Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Juanjuan Qiu
- Laboratory of Genetic Regulators in the Immune System, School of Medical Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Rui Fu
- Laboratory of Genetic Regulators in the Immune System, School of Medical Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Wushan Li
- Laboratory of Genetic Regulators in the Immune System, School of Medical Technology, Xinxiang Medical University, Xinxiang 453003, China; Laboratory of Mouse Genetics, Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang 453003, China
| | - Liaoxun Lu
- Laboratory of Genetic Regulators in the Immune System, School of Medical Technology, Xinxiang Medical University, Xinxiang 453003, China; Laboratory of Mouse Genetics, Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang 453003, China
| | - Shuanghua Tian
- Laboratory of Genetic Regulators in the Immune System, School of Medical Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Qiaoli Liu
- Laboratory of Genetic Regulators in the Immune System, School of Medical Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Yanrong Gu
- Laboratory of Genetic Regulators in the Immune System, School of Medical Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Rong Huang
- Laboratory of Genetic Regulators in the Immune System, School of Medical Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Toby Lawrence
- Centre for Inflammation Biology and Cancer Immunology, King's College London, London SE1 1UL, UK; Centre de Immunologie Marseille-Luminy, CNRS, INSERM, Aix-Marseille Universite, 13009 Marseille, France
| | - Eryan Kong
- The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, China; Laboratory of Mouse Genetics, Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang 453003, China.
| | - Lichen Zhang
- Laboratory of Genetic Regulators in the Immune System, School of Medical Technology, Xinxiang Medical University, Xinxiang 453003, China.
| | - Tianhan Li
- Laboratory of Genetic Regulators in the Immune System, School of Medical Technology, Xinxiang Medical University, Xinxiang 453003, China; Basic Medical College, Xinxiang Medical University, Xinxiang 453003, China.
| | - Yinming Liang
- Laboratory of Genetic Regulators in the Immune System, School of Medical Technology, Xinxiang Medical University, Xinxiang 453003, China; Laboratory of Mouse Genetics, Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang 453003, China; Center of Disease Model and Immunology, Hunan Academy of Chinese Medicine, Changsha 410013, China.
| |
Collapse
|
3
|
Peng B, Li Z, Cheng Y, Jiang H, Ye Q, Han G. Dental pulp stem cell-derived intracellular vesicles prevent orthodontic relapse by inhibiting PI3K/Akt/NF-κB-mediated osteoclast activity. Stem Cell Res Ther 2025; 16:22. [PMID: 39849611 PMCID: PMC11756174 DOI: 10.1186/s13287-025-04146-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 01/13/2025] [Indexed: 01/25/2025] Open
Abstract
BACKGROUND Orthodontic relapse, the undesired deviation of teeth from their corrected positions, remains a significant challenge in clinical orthodontics. Incomplete periodontal bone remodeling has been identified as a key factor in this process. Despite decades of research, currently there are no effective strategies to prevent relapse. METHODS We isolated and identified dental pulp stem cell-derived intracellular vesicles (DPSC-IV) from human dental pulp tissue. To investigate its effect, DPSC-IV was added to osteoblast or osteoclast differentiation medium. During the orthodontic retention period, DPSC-IV was administrated to rats by subgingival injection. Relapse distance and relapse rate were calculated to evaluate DPSC-IV's ability to prevent relapse. Additionally, Western blot analysis were used to examine DPSC-IV's inhibitory effect on osteoclast differentiation. RESULTS DPSC-IV significantly promoted osteoblast differentiation and inhibited osteoclast differentiation. Application of DPSC-IV during retention resulted in a significant reduction in both relapse distance and relapse rate, with improved periodontal structure and decreased osteoclast activity. This effect was mediated by the PI3K/Akt/NF-κB signaling pathway and could be reversed by the PI3K activator insulin-like growth factor-1 (IGF-1). CONCLUSION This study highlights the potential of DPSC-IV as a novel preventive approach against orthodontic relapse, offering a novel strategy for maintaining long-term orthodontic stability.
Collapse
Affiliation(s)
- Boyuan Peng
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, NO.237, Luo Yu Road, Hongshan District, Wuhan City, 430079, China
| | - Ziwei Li
- Center of Regenerative Medicine, Department of Stomatology, Renmin Hospital of Wuhan University, Gaoxin 6th Road, Jiangxia, Wuhan, Hubei, 430000, People's Republic of China
| | - Yong Cheng
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, NO.237, Luo Yu Road, Hongshan District, Wuhan City, 430079, China
| | - Henghua Jiang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, NO.237, Luo Yu Road, Hongshan District, Wuhan City, 430079, China
- Department of Orthodontics Division II, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Qingsong Ye
- Center of Regenerative Medicine, Department of Stomatology, Renmin Hospital of Wuhan University, Gaoxin 6th Road, Jiangxia, Wuhan, Hubei, 430000, People's Republic of China.
- Sydney School of Dentistry, The University of Sydney, Camperdown, Sydney, NSW, Australia.
| | - Guangli Han
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, NO.237, Luo Yu Road, Hongshan District, Wuhan City, 430079, China.
- Department of Orthodontics Division II, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China.
| |
Collapse
|
4
|
Wang R, Yin J, Li J, Bai X, Liu H, Cheng M, Wang L, Chen Y, Wei S, Liu X. Clinical utility of the neutrophil elastase inhibitor sivelestat for the treatment of ALI/ARDS patients with COVID-19. Heliyon 2024; 10:e36337. [PMID: 39296066 PMCID: PMC11408777 DOI: 10.1016/j.heliyon.2024.e36337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 07/31/2024] [Accepted: 08/13/2024] [Indexed: 09/21/2024] Open
Abstract
Background Sivelestat, a neutrophil elastase inhibitor, is postulated to mitigate acute lung injury in patients following emergency surgery. However, its efficacy in patients with acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) induced by coronavirus disease 2019 (COVID-19) remains uncertain. This study aims to evaluate the pulmonary protective effects of sivelestat in COVID-19 patients with ALI/ARDS. Methods A retrospective study was conducted involving 2454 COVID-19 patients between October 5, 2022, and February 1, 2023. Of these, 102 patients received sivelestat (0.2 mg/kg/h), while 2352 age- and sex-matched controls were identified. Propensity score matching (PSM) analysis was used to match sivelestat and non-sivelestat subgroups in ratios of 1:1 and 1:3 for sensitivity analysis. The primary outcome was a composite of effective outcomes, including 30-day mortality. Secondary outcomes included changes in partial pressure of arterial oxygen (PaO2), the ratio of PaO2 to the fraction of inspired oxygen (PaO2/FiO2), and various cytokine levels. Safety evaluations included assessments of liver function, kidney function, and leukopenia. Results In the propensity score-matched analysis, the sivelestat group had a higher proportion of severe/critical patients (87.26 % vs. 51.02 %, P < 0.001), more ARDS patients (4.9 % vs. 0.43 %, P < 0.001), and more patients with interstitial lung disease (4.9 % vs. 1.49 %, P = 0.023), but fewer patients with stroke (17.65 % vs. 19.86 %, P < 0.001). Oxygen therapy rates were similar between the groups (79.41 % vs. 80.95 %, P = 0.9). The relative risk reduction in 30-day mortality was 88.45 % (95 % confidence interval [CI] 81.23%-93.21 %) for severe/critical COVID-19 patients treated with sivelestat. Sivelestat significantly decreased cytokine levels of interferon alpha (IFNα), interleukin-1 beta (IL-1β), and interleukin-2 (IL-2).In the sivelestat group, the mortality rate was significantly reduced with standard oxygenation and HFNC therapy(P < 0.05). The treatment with sivelestat did not increase side effects. Conclusion The administration of the neutrophil elastase inhibitor sivelestat may improve clinical outcomes in COVID-19 patients with ALI/ARDS. These findings suggest that sivelestat could be considered an effective treatment option to alleviate pulmonary inflammatory injury caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2).
Collapse
Affiliation(s)
- Ruiying Wang
- Department of Pulmonary and Critical Care Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Sino-German Joint Oncological Research Laboratory, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, Shanxi, China
| | - Junping Yin
- Institute of Molecular Medicine and Experimental Immunology, University Clinic of Rheinische Friedrich-Wilhelms-University, Bonn, Germany
| | - Jian Li
- Institute of Molecular Medicine and Experimental Immunology, University Clinic of Rheinische Friedrich-Wilhelms-University, Bonn, Germany
| | - Xueli Bai
- Department of Pulmonary and Critical Care Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Hu Liu
- Department of Pulmonary and Critical Care Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Mengyu Cheng
- Department of Pulmonary and Critical Care Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Lei Wang
- Department of Pulmonary and Critical Care Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yuan Chen
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1096 Jiefang Road, Wuhan, Hubei, China
| | - Shuang Wei
- Department of Pulmonary and Critical Care Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Sino-German Joint Oncological Research Laboratory, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, Shanxi, China
- Department of Pulmonary and Critical Care Medicine,Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiansheng Liu
- Department of Pulmonary and Critical Care Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Sino-German Joint Oncological Research Laboratory, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, Shanxi, China
- Department of Pulmonary and Critical Care Medicine,Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
5
|
Mol BA, Wasinda JJ, Xu YF, Gentle NL, Meyer V. 1,25-dihydroxyvitamin D 3 augments low-dose PMA-based monocyte-to-macrophage differentiation in THP-1 cells. J Immunol Methods 2024; 532:113716. [PMID: 38960065 DOI: 10.1016/j.jim.2024.113716] [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: 03/18/2024] [Revised: 06/24/2024] [Accepted: 06/29/2024] [Indexed: 07/05/2024]
Abstract
The human monocytic THP-1 cell line is the most routinely employed in vitro model for studying monocyte-to-macrophage differentiation. Despite the wide use of this model, differentiation protocols using phorbol 12-myristate-13-acetate (PMA) or 1,25-dihydroxyvitamin D3 (1,25D3) vary drastically between studies. Given that differences in differentiation protocols have the potential to impact the characteristics of the macrophages produced, we aimed to assess the efficacy of three different THP-1 differentiation protocols by assessing changes in morphology and gene- and cell surface macrophage marker expression. THP-1 cells were differentiated with either 5 nM PMA, 10 nM 1,25D3, or a combination thereof, followed by a rest period. The results indicated that all three protocols significantly increased the expression of the macrophage markers, CD11b (p < 0.001) and CD14 (p < 0.010). Despite this, THP-1 cells exposed to 1,25D3 alone did not adopt the morphological and expression characteristics associated with macrophages. PMA was required to produce these characteristics, which were found to be more pronounced in the presence of 1,25D3. Both PMA- and PMA with 1,25D3-differentiated THP-1 cells were capable of M1 and M2 macrophage polarization, though the gene expression of polarization-associated markers was most pronounced in PMA with 1,25D3-differentiated THP-1 cells. Moreover, the combination of PMA with 1,25D3 appeared to support the process of commitment to a particular polarization state.
Collapse
Affiliation(s)
- Bronwyn A Mol
- School of Molecular and Cell Biology, University of the Witwatersrand, Private Bag 3, WITS, Johannesburg 2050, South Africa
| | - Janet J Wasinda
- School of Molecular and Cell Biology, University of the Witwatersrand, Private Bag 3, WITS, Johannesburg 2050, South Africa
| | - Yi F Xu
- School of Molecular and Cell Biology, University of the Witwatersrand, Private Bag 3, WITS, Johannesburg 2050, South Africa
| | - Nikki L Gentle
- School of Molecular and Cell Biology, University of the Witwatersrand, Private Bag 3, WITS, Johannesburg 2050, South Africa.
| | - Vanessa Meyer
- School of Molecular and Cell Biology, University of the Witwatersrand, Private Bag 3, WITS, Johannesburg 2050, South Africa.
| |
Collapse
|
6
|
Ryan CB, Choi JS, Kang B, Herr S, Pereira C, Moraes CT, Al-Ali H, Lee JK. PI3K signaling promotes formation of lipid-laden foamy macrophages at the spinal cord injury site. Neurobiol Dis 2024; 190:106370. [PMID: 38049013 PMCID: PMC10804283 DOI: 10.1016/j.nbd.2023.106370] [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: 09/02/2023] [Revised: 11/29/2023] [Accepted: 12/01/2023] [Indexed: 12/06/2023] Open
Abstract
After spinal cord injury (SCI), infiltrating macrophages undergo excessive phagocytosis of myelin and cellular debris, forming lipid-laden foamy macrophages. To understand their role in the cellular pathology of SCI, investigation of the foamy macrophage phenotype in vitro revealed a pro-inflammatory profile, increased reactive oxygen species (ROS) production, and mitochondrial dysfunction. Bioinformatic analysis identified PI3K as a regulator of inflammation in foamy macrophages, and inhibition of this pathway decreased their lipid content, inflammatory cytokines, and ROS production. Macrophage-specific inhibition of PI3K using liposomes significantly decreased foamy macrophages at the injury site after a mid-thoracic contusive SCI in mice. RNA sequencing and in vitro analysis of foamy macrophages revealed increased autophagy and decreased phagocytosis after PI3K inhibition as potential mechanisms for reduced lipid accumulation. Together, our data suggest that the formation of pro-inflammatory foamy macrophages after SCI is due to the activation of PI3K signaling, which increases phagocytosis and decreases autophagy.
Collapse
Affiliation(s)
- Christine B Ryan
- Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, United States of America
| | - James S Choi
- Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, United States of America
| | - Brian Kang
- Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, United States of America
| | - Seth Herr
- Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, United States of America
| | - Claudia Pereira
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Carlos T Moraes
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Hassan Al-Ali
- Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, United States of America; Department of Medicine Katz Division of Nephrology and Hypertension, University of Miami, Miller School of Medicine, Miami, FL, United States of America; Peggy and Harold Katz Family Drug Discovery Center, University of Miami, Miller School of Medicine, Miami, FL, United States of America; Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, FL, United States of America
| | - Jae K Lee
- Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, United States of America.
| |
Collapse
|
7
|
Shaik MG, Joshi SV, Akunuri R, Rana P, Rahman Z, Polomoni A, Yaddanapudi VM, Dandekar MP, Srinivas N. Small molecule inhibitors of NLRP3 inflammasome and GSK-3β in the management of traumatic brain injury: A review. Eur J Med Chem 2023; 259:115718. [PMID: 37573828 DOI: 10.1016/j.ejmech.2023.115718] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 07/28/2023] [Accepted: 08/08/2023] [Indexed: 08/15/2023]
Abstract
Traumatic brain injury (TBI) is a debilitating mental condition which causes physical disability and morbidity worldwide. TBI may damage the brain by direct injury that subsequently triggers a series of neuroinflammatory events. The activation of NLRP3 inflammasome and dysregulated host immune system has been documented in various neurological disorders such as TBI, ischemic stroke and multiple sclerosis. The activation of NLRP3 post-TBI increases the production of pro-inflammatory cytokines and caspase-1, which are major drivers of neuroinflammation and apoptosis. Similarly, GSK-3β regulates apoptosis through tyrosine kinase and canonical Wnt signalling pathways. Thus, therapeutic targeting of NLRP3 inflammasome and GSK-3β has emerged as promising strategies for regulating the post-TBI neuroinflammation and neurobehavioral disturbances. In this review, we discuss the identification & development of several structurally diverse and pharmacologically interesting small molecule inhibitors for targeting the NLRP3 inflammasome and GSK-3β in the management of TBI.
Collapse
Affiliation(s)
- Mahammad Ghouse Shaik
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500 037, India
| | - Swanand Vinayak Joshi
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500 037, India
| | - Ravikumar Akunuri
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500 037, India; Ellen and Ronald Caplan Cancer Center, The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Preeti Rana
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500 037, India
| | - Ziaur Rahman
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, 500 037, India
| | - Anusha Polomoni
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500 037, India
| | - Venkata Madhavi Yaddanapudi
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500 037, India
| | - Manoj P Dandekar
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, 500 037, India.
| | - Nanduri Srinivas
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500 037, India.
| |
Collapse
|
8
|
De-Leon-Lopez YS, Thompson ME, Kean JJ, Flaherty RA. The PI3K-Akt pathway is a multifaceted regulator of the macrophage response to diverse group B Streptococcus isolates. Front Cell Infect Microbiol 2023; 13:1258275. [PMID: 37928185 PMCID: PMC10622663 DOI: 10.3389/fcimb.2023.1258275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 09/25/2023] [Indexed: 11/07/2023] Open
Abstract
Group B Streptococcus (GBS), also known as Streptococcus agalactiae, is a common member of the microbial flora in healthy individuals. However, problems may arise when GBS-colonized mothers become pregnant. GBS may be transferred from a colonized mother to her newborn or developing fetus, which may result in complications such as miscarriage, pre-term birth, meningitis, pneumonia, or sepsis. Macrophages play an especially important role in the fetal and newborn response to GBS due to the limited development of the adaptive immune system early in life. The goal of this study was to expand what is currently known about how GBS manipulates macrophage cell signaling to evade the immune system and cause disease. To this end, we investigated whether the PI3K-Akt pathway was involved in several key aspects of the macrophage response to GBS. We explored whether certain GBS strains, such as sequence type (ST)-17 strains, rely on this pathway for the more rapid macrophage uptake they induce compared to other GBS strains. Our findings suggest that this pathway is, indeed, important for macrophage uptake of GBS. Consistent with these findings, we used immunofluorescence microscopy to demonstrate that more virulent strains of GBS induce more actin projections in macrophages than less virulent strains. Additionally, we explored whether PI3K-Akt signaling impacted the ability of GBS to survive within macrophages after phagocytosis and whether this pathway influenced the survival rate of macrophages themselves following GBS infection. The PI3K-Akt pathway was found to promote the survival of both macrophages and intracellular GBS following infection. We also observed that inhibition of the PI3K-Akt pathway significantly reduced GBS-mediated activation of NFκB, which is a key regulator of cell survival and inflammatory responses. Overall, these insights into strain-dependent GBS-mediated manipulation of the PI3K-Akt pathway and its downstream targets in infected macrophages may provide new insights for the development of diagnostic and therapeutic tools to combat severe GBS disease.
Collapse
Affiliation(s)
| | | | | | - Rebecca A. Flaherty
- Department of Biology and Health Science, Aquinas College, Grand Rapids, MI, United States
| |
Collapse
|
9
|
Li X, Li J, Zheng Y, Lee SJ, Zhou J, Giobbie-Hurder A, Butterfield LH, Dranoff G, Hodi FS. Granulocyte-Macrophage Colony-Stimulating Factor Influence on Soluble and Membrane-Bound ICOS in Combination with Immune Checkpoint Blockade. Cancer Immunol Res 2023; 11:1100-1113. [PMID: 37262321 PMCID: PMC10398357 DOI: 10.1158/2326-6066.cir-22-0702] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 01/03/2023] [Accepted: 05/23/2023] [Indexed: 06/03/2023]
Abstract
With the successful development of immune checkpoint blockade, there remains the continued need to improve efficacy and decrease toxicities. The addition of granulocyte-macrophage colony-stimulating factor (GM-CSF) to ipilimumab has previously demonstrated both an improvement in efficacy and decrease in the incidence of high-grade adverse events. ICOS+CD4+ or ICOS+CD8+ peripheral blood T cells are significantly greater in the patients treated with ipilimumab plus GM-CSF than in the patients treated with ipilimumab alone. To better understand the effects of GM-CSF on inducible T-cell costimulator (ICOS) and clinical outcomes, the relative roles of identified soluble ICOS and membrane-bound ICOS were evaluated. The ICOS splice variant was secreted and found to have immunologic suppressive effects. Changes in soluble ICOS splice variant levels in treated patients correlated with clinical outcomes. GM-CSF enhanced membrane-bound ICOS in an IL12-dependent manner but did not increase soluble ICOS levels. Whereas soluble ICOS plays a role in immune suppression, GM-CSF efficacy involves increasing membrane-bound ICOS and induction of dendritic cell development. Thus, soluble ICOS splice variants may be used as a biomarker for GM-CSF and immune checkpoint blockade-based therapies.
Collapse
Affiliation(s)
- Xiaoyu Li
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Melanoma Division, Dana-Farber Cancer Institute, Boston, Massachusetts
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Jingjing Li
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Melanoma Division, Dana-Farber Cancer Institute, Boston, Massachusetts
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Yue Zheng
- Department of Data Sciences, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Sandra J. Lee
- Department of Data Sciences, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Jun Zhou
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Melanoma Division, Dana-Farber Cancer Institute, Boston, Massachusetts
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Anita Giobbie-Hurder
- Melanoma Division, Dana-Farber Cancer Institute, Boston, Massachusetts
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Data Sciences, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Lisa H. Butterfield
- Parker Institute for Cancer Immunotherapy and University of California San Francisco, San Francisco, California
| | - Glenn Dranoff
- Immuno-Oncology, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - F. Stephen Hodi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Melanoma Division, Dana-Farber Cancer Institute, Boston, Massachusetts
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| |
Collapse
|
10
|
Chaintreuil P, Kerreneur E, Bourgoin M, Savy C, Favreau C, Robert G, Jacquel A, Auberger P. The generation, activation, and polarization of monocyte-derived macrophages in human malignancies. Front Immunol 2023; 14:1178337. [PMID: 37143666 PMCID: PMC10151765 DOI: 10.3389/fimmu.2023.1178337] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 04/04/2023] [Indexed: 05/06/2023] Open
Abstract
Macrophages are immune cells that originate from embryogenesis or from the differentiation of monocytes. They can adopt numerous phenotypes depending on their origin, tissue distribution and in response to different stimuli and tissue environment. Thus, in vivo, macrophages are endowed with a continuum of phenotypes that are rarely strictly pro-inflammatory or anti-inflammatory and exhibit a broad expression profile that sweeps over the whole polarization spectrum. Schematically, three main macrophage subpopulations coexist in human tissues: naïve macrophages also called M0, pro-inflammatory macrophages referred as M1 macrophages, and anti-inflammatory macrophages also known as M2 macrophages. Naïve macrophages display phagocytic functions, recognize pathogenic agents, and rapidly undergo polarization towards pro or anti-inflammatory macrophages to acquire their full panel of functions. Pro-inflammatory macrophages are widely involved in inflammatory response, during which they exert anti-microbial and anti-tumoral functions. By contrast, anti-inflammatory macrophages are implicated in the resolution of inflammation, the phagocytosis of cell debris and tissue reparation following injuries. Macrophages also play important deleterious or beneficial roles in the initiation and progression of different pathophysiological settings including solid and hematopoietic cancers. A better understanding of the molecular mechanisms involved in the generation, activation and polarization of macrophages is a prerequisite for the development of new therapeutic strategies to modulate macrophages functions in pathological situations.
Collapse
Affiliation(s)
- Paul Chaintreuil
- Université Côte d’Azur, Institut National de la Santé et de la Recherche Médicale, Nice, France
- Inserm U1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France
| | - Emeline Kerreneur
- Université Côte d’Azur, Institut National de la Santé et de la Recherche Médicale, Nice, France
- Inserm U1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France
| | - Maxence Bourgoin
- Université Côte d’Azur, Institut National de la Santé et de la Recherche Médicale, Nice, France
- Inserm U1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France
| | - Coline Savy
- Université Côte d’Azur, Institut National de la Santé et de la Recherche Médicale, Nice, France
- Inserm U1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France
| | - Cécile Favreau
- Université Côte d’Azur, Institut National de la Santé et de la Recherche Médicale, Nice, France
- Inserm U1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France
| | - Guillaume Robert
- Université Côte d’Azur, Institut National de la Santé et de la Recherche Médicale, Nice, France
- Inserm U1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France
| | - Arnaud Jacquel
- Université Côte d’Azur, Institut National de la Santé et de la Recherche Médicale, Nice, France
- Inserm U1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France
- *Correspondence: Arnaud Jacquel, ; Patrick Auberger,
| | - Patrick Auberger
- Université Côte d’Azur, Institut National de la Santé et de la Recherche Médicale, Nice, France
- Inserm U1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France
- *Correspondence: Arnaud Jacquel, ; Patrick Auberger,
| |
Collapse
|
11
|
Ding Q, Wang Y, Yang C, Li X, Yu X. Clinical Utility of the Sivelestat for the Treatment of ALI/ARDS: Moving on in the Controversy? INTENSIVE CARE RESEARCH 2023; 3:12-17. [PMID: 36911225 PMCID: PMC9412801 DOI: 10.1007/s44231-022-00012-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 07/27/2022] [Indexed: 01/08/2023]
Abstract
Acute respiratory distress syndrome (ARDS) is a serious condition that can arise following direct or indirect acute lung injury (ALI). It is heterogeneous and has a high mortality rate. Supportive care is the mainstay of treatment and there is no definitive pharmacological treatment as yet. In nonclinical studies, neutrophil elastase inhibitor sivelestat appears to show benefit in ARDS without inhibiting the host immune defense in cases of infection. In clinical studies, the efficacy of sivelestat in the treatment of ARDS remains controversial. The currently available evidence suggests that sivelestat may show some benefit in the treatment of ARDS, although large, randomized controlled trials are needed in specific pathophysiological conditions to explore these potential benefits.
Collapse
Affiliation(s)
- Qiongli Ding
- Critical Medicine Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054 Xinjiang Uygur Autonomous Region China.,Xinjiang Medical University, Urumqi, 830054 Xinjiang Uygur Autonomous Region China
| | - Yi Wang
- Critical Medicine Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054 Xinjiang Uygur Autonomous Region China.,Xinjiang Medical University, Urumqi, 830054 Xinjiang Uygur Autonomous Region China.,Xinjiang Uygur Autonomous Region Institute of Critical Medicine, Urumqi, 830054 Xinjiang Uygur Autonomous Region China
| | - Chunbo Yang
- Critical Medicine Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054 Xinjiang Uygur Autonomous Region China.,Xinjiang Medical University, Urumqi, 830054 Xinjiang Uygur Autonomous Region China.,Xinjiang Uygur Autonomous Region Institute of Critical Medicine, Urumqi, 830054 Xinjiang Uygur Autonomous Region China
| | - Xiang Li
- Critical Medicine Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054 Xinjiang Uygur Autonomous Region China.,Xinjiang Uygur Autonomous Region Institute of Critical Medicine, Urumqi, 830054 Xinjiang Uygur Autonomous Region China
| | - Xiangyou Yu
- Critical Medicine Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054 Xinjiang Uygur Autonomous Region China.,Xinjiang Medical University, Urumqi, 830054 Xinjiang Uygur Autonomous Region China.,Xinjiang Uygur Autonomous Region Institute of Critical Medicine, Urumqi, 830054 Xinjiang Uygur Autonomous Region China
| |
Collapse
|
12
|
Sun J, Zhang Q, Yang G, Li Y, Fu Y, Zheng Y, Jiang X. The licorice flavonoid isoliquiritigenin attenuates Mycobacterium tuberculosis-induced inflammation through Notch1/NF-κB and MAPK signaling pathways. JOURNAL OF ETHNOPHARMACOLOGY 2022; 294:115368. [PMID: 35589023 DOI: 10.1016/j.jep.2022.115368] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/24/2022] [Accepted: 05/07/2022] [Indexed: 06/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The genus Glycyrrhiza is a small perennial herb that has been traditionally used to treat many diseases across the world. Licorice (Gancao in Chinese) is the dried root and rhizome of G. glabra, G. uralensis or G. inflata. Licorice plays an important role in traditional Chinese medicine (TCM), and is the most frequently used in Chinese herbal formulas. Isoliquiritigenin (ISL) is a flavonoid extracted from licorice, and has been evaluated for its various biological activities, including anti-inflammatory, anti-tumor and anti-oxidant activities. Excessive and persistent inflammation in the Mycobacterium tuberculosis (Mtb) infection is not conducive to the elimination of Mtb, but contributes to serious pulmonary dysfunction. AIM OF THE STUDY This study aimed to examine the anti-inflammatory effects of ISL in the Mtb infection. METHODS In vitro models of Mtb-infected macrophages were established. Murine macrophage Raw 264.7 cells and primary peritoneal macrophages were used in this study. Cell viability was determined by the cell counting kit-8 (CCK-8) assay. The effects of ISL on the secretion levels of interleukin -1β (IL-1β), tumor necrosis factor -α (TNF-α), and interleukin -6 (IL-6) were detected by the enzyme-linked immunosorbent assay (ELISA). The expression levels of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX2) were measured by the real time quantitative reverse transcription polymerase chain reaction (RT-qPCR) and Western blot. Western blot was used to assess the effects of ISL on the activation of NLRP3 inflammasome and Notch1/NF-κB and MAPK signaling pathways. Immunofluorescence assays was used to detected the translocation of phosphorylation of p65 subunit of NF-κB. RESULTS It was revealed that ISL inhibited the secretion of IL-1β and the activation of pore-forming protein (gasdermin D, GSDMD) by suppressing the activation of NLPR3 inflammasome induced by Mtb infection. ISL was also shown to have promising inhibitory effects on inflammatory factors, such as TNF-α, IL-6, iNOS and COX2. Regarding the anti-inflammatory mechanism of ISL, it was found that ISL exerted its anti-inflammatory effects by inhibiting the activation of Notch1/NF-κB and MAPK signaling pathways. CONCLUSION ISL reduced Mtb-induced inflammation through the Notch1/NF-κB and MAPK signaling pathways. ISL might be used as a potential adjuvant drug to treat tuberculosis by adjusting host immune responses.
Collapse
Affiliation(s)
- Jinxia Sun
- Center for Traditional Chinese Medicine and Immunology Research, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, 201203, Shanghai, PR China
| | - Qingwen Zhang
- Center for Traditional Chinese Medicine and Immunology Research, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, 201203, Shanghai, PR China; Department of Inspection and Quarantine, School of Medical Technology, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Guizhen Yang
- Center for Traditional Chinese Medicine and Immunology Research, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, 201203, Shanghai, PR China
| | - Yinhong Li
- Center for Traditional Chinese Medicine and Immunology Research, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, 201203, Shanghai, PR China
| | - Yan Fu
- Center for Traditional Chinese Medicine and Immunology Research, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, 201203, Shanghai, PR China
| | - Yuejuan Zheng
- Center for Traditional Chinese Medicine and Immunology Research, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, 201203, Shanghai, PR China.
| | - Xin Jiang
- Center for Traditional Chinese Medicine and Immunology Research, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, 201203, Shanghai, PR China.
| |
Collapse
|
13
|
Zhang Y, Deng Z, Sun S, Xie S, Jiang M, Chen B, Gu C, Yang Y. NAT10 acetylates BCL-XL mRNA to promote the proliferation of multiple myeloma cells through PI3K-AKT pathway. Front Oncol 2022; 12:967811. [PMID: 35978804 PMCID: PMC9376478 DOI: 10.3389/fonc.2022.967811] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 07/07/2022] [Indexed: 11/13/2022] Open
Abstract
Multiple myeloma (MM) is a clinically distinctive plasma cell malignancy in the bone marrow (BM), in which epigenetic abnormalities are featured prominently. Epigenetic modifications including acetylation have been deemed to contribute to tumorigenesis. N-acetyltransferase 10 (NAT10) is an important regulator of mRNA acetylation in many cancers, however its function in MM is poorly studied. We first analyzed MM clinical databases and found that elevated NAT10 expression conferred a poor prognosis in MM patients. Furthermore, overexpression of NAT10 promoted MM cell proliferation. The correlation analysis of acRIP-seq screened BCL-XL (BCL2L1) as a significant downstream target of NAT10. Further RNA decay assay showed that increased NAT10 improved the stability of BCL-XL mRNA and promoted protein translation to suppress cell apoptosis. NAT10 activated PI3K-AKT pathway and upregulated CDK4/CDK6 to accelerate cellular proliferation. Importantly, inhibition of NAT10 by Remodelin suppressed MM cell growth and induced cell apoptosis. Our findings show the important role of NAT10/BCL-XL axis in promoting MM cell proliferation. Further explorations are needed to fully define the potential of targeting NAT10 therapy in MM treatment.
Collapse
Affiliation(s)
- Yuanjiao Zhang
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhendong Deng
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shanliang Sun
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Siyuan Xie
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Mingmei Jiang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Bing Chen
- Department of Hematology, The Affiliated Drum Tower Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Chunyan Gu
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- Department of Hematology, The Affiliated Drum Tower Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Ye Yang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
14
|
Wang Z, Zhang Q, Qi C, Bai Y, Zhao F, Chen H, Li Z, Wang X, Chen M, Gong J, Peng Z, Zhang X, Cai J, Chen S, Zhao X, Shen L, Li J. Combination of AKT1 and CDH1 mutations predicts primary resistance to immunotherapy in dMMR/MSI-H gastrointestinal cancer. J Immunother Cancer 2022; 10:jitc-2022-004703. [PMID: 35705314 PMCID: PMC9204428 DOI: 10.1136/jitc-2022-004703] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/19/2022] [Indexed: 12/13/2022] Open
Abstract
Background Gastrointestinal (GI) cancer is the second most common cancer type with mismatch repair-deficient (dMMR)/microsatellite instability-high (MSI-H) phenotype that is expected to respond to immune-checkpoint inhibitors (ICIs). However, approximately half of the patients with dMMR/MSI-H GI cancer derive no benefit from ICIs. We sought to identify the predictors of primary resistance to ICIs in dMMR/MSI-H GI cancer. Methods Three independent cohorts were included: (1) the discovery cohort (65 patients with dMMR/MSI-H GI cancer) with ICI efficacy data and pre-ICIs tissue samples for genomic profile and tumor immune infiltration; (2) the validation cohort (22 patients with dMMR/MSI-H GI cancer) with ICI efficacy data and pre-ICIs plasma samples for genomic profile; and (3) the TCGA (The Cancer Genome Atlas) cohort not receiving ICIs (152 patients with MSI-H GI cancer) with genomic profile and survival data. Results AKT1 and CDH1 mutations were identified as independent predictors of poor progression-free survival (PFS) and primary resistance to ICIs in dMMR/MSI-H GI cancer. We combined these two genes as an immuno-oncology therapy predictor (IOpred), which could recognize 52.4% (11/21) of dMMR/MSI-H patients with primary resistance to ICIs with a positive predictive value (PPV) of 91.7% (11/12). Receiver operating characteristic analysis demonstrated IOpred with a good performance in predicting primary resistance (area under the curve 0.751). Patients with IOpred-Mut (mutant AKT1 or CDH1) GI cancer had significantly shorter PFS (HR=8.36, p<0.001) and overall survival (OS, HR=5.17, p<0.001) than IOpred-WT (wild-type for both AKT1 and CDH1) cases upon ICI treatment. The validation cohort also confirmed the correlation between IOpred-mutation and poorer prognosis (PFS, HR=4.68, p=0.004; OS, HR=15.98, p<0.001) in dMMR/MSI-H patients after ICIs. The PPV of IOpred in identifying primary resistance to ICIs was 80% (4/5) in the validation cohort. Additionally, IOpred-WT patients could be further stratified by tumor mutational burden (TMB), wherein TMB-low patients (TMB ≤26.19 mutations per megabase (Mb)) had a significantly higher primary resistance rate to ICIs (34.8% vs 6.7%, p=0.014) and poorer PFS (HR=3.46, p=0.008) and OS (HR=4.42, p=0.047) than TMB-high patients (TMB >26.19 mutations/Mb). Conclusions IOpred was identified as a powerful predictor of primary resistance to ICIs in dMMR/MSI-H GI cancer, which might serve as a promising biomarker to help guide immunotherapy decision-making.
Collapse
Affiliation(s)
- Zhenghang Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Qi Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Changsong Qi
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yuezong Bai
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Feilong Zhao
- Medical Affairs, 3D Medicines Inc, Shanghai, China
| | - Hui Chen
- Medical Affairs, 3D Medicines Inc, Shanghai, China
| | - Zhongwu Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xicheng Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Mifen Chen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jifang Gong
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Zhi Peng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiaotian Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jinping Cai
- Medical Affairs, 3D Medicines Inc, Shanghai, China
| | - Shiqing Chen
- Medical Affairs, 3D Medicines Inc, Shanghai, China
| | | | - Lin Shen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jian Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| |
Collapse
|
15
|
Zehnle PMA, Wu Y, Pommerening H, Erlacher M. Stayin‘ alive: BCL-2 proteins in the hematopoietic system. Exp Hematol 2022; 110:1-12. [PMID: 35315320 DOI: 10.1016/j.exphem.2022.03.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/09/2022] [Accepted: 03/10/2022] [Indexed: 11/04/2022]
|
16
|
Caballero RE, Dong SXM, Gajanayaka N, Ali H, Cassol E, Cameron WD, Korneluk R, Tremblay MJ, Angel JB, Kumar A. Role of RIPK1 in SMAC mimetics-induced apoptosis in primary human HIV-infected macrophages. Sci Rep 2021; 11:22901. [PMID: 34824340 PMCID: PMC8617210 DOI: 10.1038/s41598-021-02146-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 11/09/2021] [Indexed: 11/09/2022] Open
Abstract
Macrophages serve as viral reservoirs due to their resistance to apoptosis and HIV-cytopathic effects. We have previously shown that inhibitor of apoptosis proteins (IAPs) confer resistance to HIV-Vpr-induced apoptosis in normal macrophages. Herein, we show that second mitochondrial activator of caspases (SMAC) mimetics (SM) induce apoptosis of monocyte-derived macrophages (MDMs) infected in vitro with a R5-tropic laboratory strain expressing heat stable antigen, chronically infected U1 cells, and ex-vivo derived MDMs from HIV-infected individuals. To understand the mechanism governing SM-induced cell death, we show that SM-induced cell death of primary HIV-infected macrophages was independent of the acquisition of M1 phenotype following HIV infection of macrophages. Instead, SM-induced cell death was found to be mediated by IAPs as downregulation of IAPs by siRNAs induced cell death of HIV-infected macrophages. Moreover, HIV infection caused receptor interacting protein kinase-1 (RIPK1) degradation which in concert with IAP1/2 downregulation following SM treatment may result in apoptosis of macrophages. Altogether, our results show that SM selectively induce apoptosis in primary human macrophages infected in vitro with HIV possibly through RIPK1. Moreover, modulation of the IAP pathways may be a potential strategy for selective killing of HIV-infected macrophages in vivo.
Collapse
Affiliation(s)
- Ramon Edwin Caballero
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada. .,Division of Virology, Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, 401 Smyth Road, Research Building 2, University of Ottawa, Ottawa, ON, K1H 8L1, Canada.
| | - Simon Xin Min Dong
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Niranjala Gajanayaka
- Division of Virology, Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, 401 Smyth Road, Research Building 2, University of Ottawa, Ottawa, ON, K1H 8L1, Canada
| | - Hamza Ali
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.,Division of Virology, Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, 401 Smyth Road, Research Building 2, University of Ottawa, Ottawa, ON, K1H 8L1, Canada
| | - Edana Cassol
- Department of Health Sciences, Carleton University, Ottawa, ON, Canada
| | - William D Cameron
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.,Division of Infectious Diseases, The Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Robert Korneluk
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.,Division of Virology, Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, 401 Smyth Road, Research Building 2, University of Ottawa, Ottawa, ON, K1H 8L1, Canada
| | - Michel J Tremblay
- Centre de recherche du CHU de Québec-Université Laval, Université Laval, Québec City, QC, Canada
| | - Jonathan B Angel
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.,Division of Infectious Diseases, The Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Ashok Kumar
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada. .,Division of Virology, Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, 401 Smyth Road, Research Building 2, University of Ottawa, Ottawa, ON, K1H 8L1, Canada. .,Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
17
|
Gajanayaka N, Dong SXM, Ali H, Iqbal S, Mookerjee A, Lawton DA, Caballero RE, Cassol E, Cameron DW, Angel JB, Crawley AM, Kumar A. TLR-4 Agonist Induces IFN-γ Production Selectively in Proinflammatory Human M1 Macrophages through the PI3K-mTOR- and JNK-MAPK-Activated p70S6K Pathway. THE JOURNAL OF IMMUNOLOGY 2021; 207:2310-2324. [PMID: 34551966 DOI: 10.4049/jimmunol.2001191] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 08/18/2021] [Indexed: 12/18/2022]
Abstract
IFN-γ, a proinflammatory cytokine produced primarily by T cells and NK cells, activates macrophages and engages mechanisms to control pathogens. Although there is evidence of IFN-γ production by murine macrophages, IFN-γ production by normal human macrophages and their subsets remains unknown. Herein, we show that human M1 macrophages generated by IFN-γ and IL-12- and IL-18-stimulated monocyte-derived macrophages (M0) produce significant levels of IFN-γ. Further stimulation of IL-12/IL-18-primed macrophages or M1 macrophages with agonists for TLR-2, TLR-3, or TLR-4 significantly enhanced IFN-γ production in contrast to the similarly stimulated M0, M2a, M2b, and M2c macrophages. Similarly, M1 macrophages generated from COVID-19-infected patients' macrophages produced IFN-γ that was enhanced following LPS stimulation. The inhibition of M1 differentiation by Jak inhibitors reversed LPS-induced IFN-γ production, suggesting that differentiation with IFN-γ plays a key role in IFN-γ induction. We subsequently investigated the signaling pathway(s) responsible for TLR-4-induced IFN-γ production in M1 macrophages. Our results show that TLR-4-induced IFN-γ production is regulated by the ribosomal protein S6 kinase (p70S6K) through the activation of PI3K, the mammalian target of rapamycin complex 1/2 (mTORC1/2), and the JNK MAPK pathways. These results suggest that M1-derived IFN-γ may play a key role in inflammation that may be augmented following bacterial/viral infections. Moreover, blocking the mTORC1/2, PI3K, and JNK MAPKs in macrophages may be of potential translational significance in preventing macrophage-mediated inflammatory diseases.
Collapse
Affiliation(s)
- Niranjala Gajanayaka
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Simon Xin Min Dong
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Hamza Ali
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada.,Faculty of Applied Medical Sciences, Taibah University, Medina, Kingdom of Saudi Arabia
| | - Salma Iqbal
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Ananda Mookerjee
- Apoptosis Research Center, Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada
| | - David A Lawton
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Ramon Edwin Caballero
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada.,Apoptosis Research Center, Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada
| | - Edana Cassol
- Department of Biology, Carleton University, Ottawa, Ontario, Canada
| | - Donald William Cameron
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada.,Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Department of Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Jonathan B Angel
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada.,Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Department of Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Angela M Crawley
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada.,Department of Biology, Carleton University, Ottawa, Ontario, Canada.,Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Center for Infection, Immunity, and Inflammation, Faculty of Medicine, University of Ottawa, Ontario, Canada; and
| | - Ashok Kumar
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada; .,Apoptosis Research Center, Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada.,Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
18
|
Ali H, Dong SXM, Gajanayaka N, Cassol E, Angel JB, Kumar A. Selective Induction of Cell Death in Human M1 Macrophages by Smac Mimetics Is Mediated by cIAP-2 and RIPK-1/3 through the Activation of mTORC. THE JOURNAL OF IMMUNOLOGY 2021; 207:2359-2373. [PMID: 34561230 DOI: 10.4049/jimmunol.2100108] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 08/28/2021] [Indexed: 12/22/2022]
Abstract
Inflammatory macrophages have been implicated in many diseases, including rheumatoid arthritis and inflammatory bowel disease. Therefore, targeting macrophage function and activation may represent a potential strategy to treat macrophage-associated diseases. We have previously shown that IFN-γ-induced differentiation of human M0 macrophages toward proinflammatory M1 state rendered them highly susceptible to the cytocidal effects of second mitochondria-derived activator of caspases mimetics (SMs), antagonist of the inhibitors of apoptosis proteins (IAPs), whereas M0 and anti-inflammatory M2c macrophages were resistant. In this study, we investigated the mechanism governing SM-induced cell death during differentiation into M1 macrophages and in polarized M1 macrophages. IFN-γ stimulation conferred on M0 macrophages the sensitivity to SM-induced cell death through the Jak/STAT, IFN regulatory factor-1, and mammalian target of rapamycin complex-1 (mTORC-1)/ribosomal protein S6 kinase pathways. Interestingly, mTORC-1 regulated SM-induced cell death independent of M1 differentiation. In contrast, SM-induced cell death in polarized M1 macrophages is regulated by the mTORC-2 pathway. Moreover, SM-induced cell death is regulated by cellular IAP (cIAP)-2, receptor-interacting protein kinase (RIPK)-1, and RIPK-3 degradation through mTORC activation during differentiation into M1 macrophages and in polarized M1 macrophages. In contrast to cancer cell lines, SM-induced cell death in M1 macrophages is independent of endogenously produced TNF-α, as well as the NF-κB pathway. Collectively, selective induction of cell death in human M1 macrophages by SMs may be mediated by cIAP-2, RIPK-1, and RIPK-3 degradation through mTORC activation. Moreover, blocking cIAP-1/2, mTORC, or IFN regulatory factor-1 may represent a promising therapeutic strategy to control M1-associated diseases.
Collapse
Affiliation(s)
- Hamza Ali
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ontario, Canada; .,Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, Ontario, Canada.,Faculty of Applied Medical Sciences, Taibah University, Medina, Kingdom of Saudi Arabia
| | - Simon Xin Min Dong
- Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Niranjala Gajanayaka
- Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Edana Cassol
- Department of Health Sciences, Carleton University, Ottawa, Ontario, Canada
| | - Jonathan B Angel
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ontario, Canada.,Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Department of Medicine, Faculty of Medicine, University of Ottawa, Ontario, Canada; and
| | - Ashok Kumar
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ontario, Canada; .,Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, Ontario, Canada.,Department of Pathology and Laboratory Medicine, University of Ottawa, Ontario, Canada
| |
Collapse
|
19
|
Hussain SM, Kansal RG, Alvarez MA, Hollingsworth TJ, Elahi A, Miranda-Carboni G, Hendrick LE, Pingili AK, Albritton LM, Dickson PV, Deneve JL, Yakoub D, Hayes DN, Kurosu M, Shibata D, Makowski L, Glazer ES. Role of TGF-β in pancreatic ductal adenocarcinoma progression and PD-L1 expression. Cell Oncol (Dordr) 2021; 44:673-687. [PMID: 33694102 DOI: 10.1007/s13402-021-00594-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/04/2021] [Indexed: 02/07/2023] Open
Abstract
PURPOSE The transforming growth factor-beta (TGF-β) pathway plays a paradoxical, context-dependent role in pancreatic ductal adenocarcinoma (PDAC): a tumor-suppressive role in non-metastatic PDAC and a tumor-promotive role in metastatic PDAC. We hypothesize that non-SMAD-TGF-β signaling induces PDAC progression. METHODS We investigated the expression of non-SMAD-TGF-β signaling proteins (pMAPK14, PD-L1, pAkt and c-Myc) in patient-derived tissues, cell lines and an immunocompetent mouse model. Experimental models were complemented by comparing the signaling proteins in PDAC specimens from patients with various survival intervals. We manipulated models with TGF-β, gemcitabine (DNA synthesis inhibitor), galunisertib (TGF-β receptor inhibitor) and MK-2206 (Akt inhibitor) to investigate their effects on NF-κB, β-catenin, c-Myc and PD-L1 expression. PD-L1 expression was also investigated in cancer cells and tumor associated macrophages (TAMs) in a mouse model. RESULTS We found that tumors from patients with aggressive PDAC had higher levels of the non-SMAD-TGF-β signaling proteins pMAPK14, PD-L1, pAkt and c-Myc. In PDAC cells with high baseline β-catenin expression, TGF-β increased β-catenin expression while gemcitabine increased PD-L1 expression. Gemcitabine plus galunisertib decreased c-Myc and NF-κB expression, but induced PD-L1 expression in some cancer models. In mice, gemcitabine plus galunisertib treatment decreased metastases (p = 0.018), whereas galunisertib increased PD-L1 expression (p < 0.0001). In the mice, liver metastases contained more TAMs compared to the primary pancreatic tumors (p = 0.001), and TGF-β increased TAM PD-L1 expression (p < 0.05). CONCLUSIONS In PDAC, the non-SMAD-TGF-β signaling pathway leads to more aggressive phenotypes, TAM-induced immunosuppression and PD-L1 expression. The divergent effects of TGF-β ligand versus receptor inhibition in tumor cells versus TAMs may explain the TGF-β paradox. Further evaluation of each mechanism is expected to lead to the development of targeted therapies.
Collapse
Affiliation(s)
- S Mazher Hussain
- Department of Surgery, College of Medicine, 910 Madison Ave., Suite 300, Memphis, TN, 38163, USA
| | - Rita G Kansal
- Department of Surgery, College of Medicine, 910 Madison Ave., Suite 300, Memphis, TN, 38163, USA
| | - Marcus A Alvarez
- Department of Surgery, College of Medicine, 910 Madison Ave., Suite 300, Memphis, TN, 38163, USA
| | - T J Hollingsworth
- Department of Surgery, College of Medicine, 910 Madison Ave., Suite 300, Memphis, TN, 38163, USA
| | - Abul Elahi
- Department of Surgery, College of Medicine, 910 Madison Ave., Suite 300, Memphis, TN, 38163, USA
| | | | - Leah E Hendrick
- Department of Surgery, College of Medicine, 910 Madison Ave., Suite 300, Memphis, TN, 38163, USA
| | | | | | - Paxton V Dickson
- Department of Surgery, College of Medicine, 910 Madison Ave., Suite 300, Memphis, TN, 38163, USA
| | - Jeremiah L Deneve
- Department of Surgery, College of Medicine, 910 Madison Ave., Suite 300, Memphis, TN, 38163, USA
| | - Danny Yakoub
- Department of Surgery, College of Medicine, 910 Madison Ave., Suite 300, Memphis, TN, 38163, USA
| | - D Neil Hayes
- Department of Medicine, College of Medicine, Memphis, TN, USA
| | - Michio Kurosu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, USA
| | - David Shibata
- Department of Surgery, College of Medicine, 910 Madison Ave., Suite 300, Memphis, TN, 38163, USA
| | - Liza Makowski
- Department of Medicine, College of Medicine, Memphis, TN, USA.,Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Evan S Glazer
- Department of Surgery, College of Medicine, 910 Madison Ave., Suite 300, Memphis, TN, 38163, USA.
| |
Collapse
|
20
|
Wang K, Zhang Z, Meng D, Li J. Investigating genetic drivers of juvenile dermatomyositis pathogenesis using bioinformatics methods. J Dermatol 2021; 48:1007-1020. [PMID: 33891717 DOI: 10.1111/1346-8138.15856] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 03/03/2021] [Accepted: 03/04/2021] [Indexed: 11/30/2022]
Abstract
Juvenile dermatomyositis (JDM) is a chronic autoimmune disease. The pathogenic mechanisms remain ill-defined. The purpose of this study was to identify key genes related to JDM. Microarray datasets were downloaded from the Gene Expression Omnibus database. The differentially expressed genes (DEG) were identified. Then, Gene Ontology, Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis, and protein-protein interaction (PPI) network were carried out. In addition, the hub genes were selected by cytoHubba. The expression profile and diagnostic capacity (receiver-operator curve [ROC]) of interested hub genes were verified. Gene set enrichment analysis (GSEA) was also carried out. Moreover, the signature of hub genes was then used as a search query to explore the Connectivity Map (CMAP). A total of 128 DEG were identified. The enriched functions and pathways of the DEG include response to virus, negative regulation of cell migration, cadmium ion transmembrane transport, defense response to Gram-negative bacterium, positive regulation of megakaryocyte differentiation, and negative regulation of angiogenesis. Twenty-one hub genes were identified. The expression levels of the interested genes were also confirmed. ROC analysis confirmed that the expression of these genes can distinguish JDM from controls. GSEA showed that these genes are mainly related to "inflammatory response", "complement", "interferon-α response", "IL6/JAK/STAT3 signaling", "TGF-β signaling", "IL2/STAT5 signaling" and "TNF-α signaling via NF-κB". The CMAP research found some compounds with the potential to counteract the effects of the dysregulated molecular signature in JDM. In this study, bioinformatics methods were used to identify DEG, which helps us understand the molecular mechanisms of JDM and provide candidate targets for diagnosis and treatment of JDM.
Collapse
Affiliation(s)
- Kai Wang
- Department of Rheumatology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huaian, China
| | - Zhongyuan Zhang
- Department of Rheumatology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huaian, China
| | - Deqian Meng
- Department of Rheumatology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huaian, China
| | - Ju Li
- Department of Rheumatology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huaian, China
| |
Collapse
|
21
|
Vijayan M, Lee CL, Wong VHH, Wang X, Bai K, Wu J, Koistinen H, Seppälä M, Lee KF, Yeung WSB, Ng EHY, Chiu PCN. Decidual glycodelin-A polarizes human monocytes into a decidual macrophage-like phenotype through Siglec-7. J Cell Sci 2020; 133:jcs244400. [PMID: 32513821 DOI: 10.1242/jcs.244400] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 05/28/2020] [Indexed: 12/20/2022] Open
Abstract
Decidual macrophages constitute 20-30% of the total leukocytes in the uterus of pregnant women, regulating the maternal immune tolerance and placenta development. Abnormal number or activities of decidual macrophages (dMs) are associated with fetal loss and pregnancy complications, such as preeclampsia. Monocytes differentiate into dMs in a decidua-specific microenvironment. Despite their important roles in pregnancy, the exact factors that regulate the differentiation into dMs remain unclear. Glycodelin-A (PAEP, hereafter referred to as GdA) is a glycoprotein that is abundantly present in the decidua, and plays an important role in fetomaternal defense and placental development. It modulates the differentiation and activity of several immune cell types residing in the decidua. In this study, we demonstrated that GdA induces the differentiation of human monocytes into dM-like phenotypes in terms of transcriptome, cell surface marker expression, secretome, and regulation of trophoblast and endothelial cell functions. We found that Sialic acid-binding Ig-like lectin 7 (Siglec-7) mediates the binding and biological actions of GdA in a sialic acid-dependent manner. We, therefore, suggest that GdA, induces the polarization of monocytes into dMs to regulate fetomaternal tolerance and placental development.
Collapse
Affiliation(s)
- Madhavi Vijayan
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong S.A.R
| | - Cheuk-Lun Lee
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong S.A.R
- The University of Hong Kong Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Vera H H Wong
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong S.A.R
| | - Xia Wang
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong S.A.R
| | - Kungfeng Bai
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong S.A.R
- The University of Hong Kong Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Jian Wu
- The University of Hong Kong Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Hannu Koistinen
- Department of Clinical Chemistry and Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, 00029 HUS Helsinki, Finland
| | - Markku Seppälä
- Department of Clinical Chemistry and Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, 00029 HUS Helsinki, Finland
| | - Kai-Fai Lee
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong S.A.R
- The University of Hong Kong Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - William S B Yeung
- The University of Hong Kong Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Ernest H Y Ng
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong S.A.R
- The University of Hong Kong Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Philip C N Chiu
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong S.A.R
- The University of Hong Kong Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
22
|
Imidazo[1,2- b]pyrazole-7-Carboxamide Derivative Induces Differentiation-Coupled Apoptosis of Immature Myeloid Cells Such as Acute Myeloid Leukemia and Myeloid-Derived Suppressor Cells. Int J Mol Sci 2020; 21:ijms21145135. [PMID: 32698503 PMCID: PMC7404197 DOI: 10.3390/ijms21145135] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 07/15/2020] [Accepted: 07/17/2020] [Indexed: 12/29/2022] Open
Abstract
Chemotherapy-induced differentiation of immature myeloid progenitors, such as acute myeloid leukemia (AML) cells or myeloid-derived suppressor cells (MDSCs), has remained a challenge for the clinicians. Testing our imidazo[1,2-b]pyrazole-7-carboxamide derivative on HL-60 cells, we obtained ERK phosphorylation as an early survival response to treatment followed by the increase of the percentage of the Bcl-xlbright and pAktbright cells. Following the induction of Vav1 and the AP-1 complex, a driver of cellular differentiation, FOS, JUN, JUNB, and JUND were elevated on a concentration and time-dependent manner. As a proof of granulocytic differentiation, the cells remained non-adherent, the expression of CD33 decreased; the granularity, CD11b expression, and MPO activity of HL-60 cells increased upon treatment. Finally, viability of HL-60 cells was hampered shown by the depolarization of mitochondria, activation of caspase-3, cleavage of Z-DEVD-aLUC, appearance of the sub-G1 population, and the leakage of the lactate-dehydrogenase into the supernatant. We confirmed the differentiating effect of our drug candidate on human patient-derived AML cells shown by the increase of CD11b and decrease of CD33+, CD7+, CD206+, and CD38bright cells followed apoptosis (IC50: 80 nM) after treatment ex vivo. Our compound reduced both CD11b+/Ly6C+ and CD11b+/Ly6G+ splenic MDSCs from the murine 4T1 breast cancer model ex vivo.
Collapse
|
23
|
Human Cytomegalovirus Mediates Unique Monocyte-to-Macrophage Differentiation through the PI3K/SHIP1/Akt Signaling Network. Viruses 2020; 12:v12060652. [PMID: 32560319 PMCID: PMC7354488 DOI: 10.3390/v12060652] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 05/29/2020] [Accepted: 06/09/2020] [Indexed: 12/11/2022] Open
Abstract
Blood monocytes mediate the hematogenous dissemination of human cytomegalovirus (HCMV) in the host. However, monocytes have a short 48-hour (h) lifespan and are not permissive for viral replication. We previously established that HCMV infection drives differentiation of monocytes into long-lived macrophages to mediate viral dissemination, though the mechanism was unclear. Here, we found that HCMV infection promoted monocyte polarization into distinct macrophages by inducing select M1 and M2 differentiation markers and that Akt played a central role in driving differentiation. Akt's upstream positive regulators, PI3K and SHIP1, facilitated the expression of the M1/M2 differentiation markers with p110δ being the predominant PI3K isoform inducing differentiation. Downstream of Akt, M1/M2 differentiation was mediated by caspase 3, whose activity was tightly regulated by Akt in a temporal manner. Overall, this study highlights that HCMV employs the PI3K/SHIP1/Akt pathway to regulate caspase 3 activity and drive monocyte differentiation into unique macrophages, which is critical for viral dissemination.
Collapse
|
24
|
Yuan S, Li H, Yang C, Xie W, Wang Y, Zhang J, Cai Z, Mao Z, Xie W, Lü T. DHA attenuates Aβ-induced necroptosis through the RIPK1/RIPK3 signaling pathway in THP-1 monocytes. Biomed Pharmacother 2020; 126:110102. [PMID: 32199223 DOI: 10.1016/j.biopha.2020.110102] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 03/11/2020] [Accepted: 03/11/2020] [Indexed: 01/14/2023] Open
Abstract
Monocytes play a crucial role in Alzheimer's disease (AD), and docosahexaenoic acid (DHA) has a neuroprotective effect for many neurodegenerative diseases. However, mechanisms that regulate monocyte and Aβ protein interaction in AD and the effects of DHA on monocytes in the context of AD are not fully understood. The experiments were designed to further explore possible mechanisms of interaction between monocytes and Aβ plaques. Another objective of this study was to investigate a potential mechanism for Aβ-induced necroptosis involving the activation of MAPK and NF-kB signaling pathways in human THP-1 monocytes, as well as how these pathways might be modulated by DHA. Our findings indicate that Aβ25-35 has a "Hormesis" effect on cell viability and necroptosis in THP-1 cells, and Aβ25-35 influences THP-1 cells differentiation as analyzed by flow cytometry. Pretreatment of THP-1 monocytes with DHA effectively inhibited Aβ-induced activation and markedly suppressed protein expression of necroptosis (RIPK1, RIPK3, MLKL) and pro-inflammatory cytokines (TNF-α, IL-1β, IL-6). Moreover, our findings indicate that Aβ25-35 activated the ERK1/2 and p38 signaling pathways, but not NF-κB/p65 signaling, while pre-treatment with DHA followed by Aβ25-35 treatment suppressed only ERK1/2 signaling. Further study revealed that the expression level of RIPK3 is reduced much more during coadministration with DHA and necrostatin-1 (NEC-1) than administration alone with either of them, indicating that DHA may have additional targets. Meanwhile, this finding indicates that DHA can prevent Aβ-induced necroptosis of THP-1 cells via the RIPK1/RIPK3 signaling pathway. Our results also indicate that DHA treatment restored migration of THP-1 monocytes induced by Aβ25-35, and DHA treatment could be a promising new therapy for AD management.
Collapse
Affiliation(s)
- Shiqi Yuan
- Department of Neurology, The Third Affiliated Hospital of Southern Medical University, No. 183, Zhongshan Road West, Guangzhou 510630, PR China
| | - Huan Li
- Department of Neurology, The Third Affiliated Hospital of Southern Medical University, No. 183, Zhongshan Road West, Guangzhou 510630, PR China
| | - Canhong Yang
- Department of Neurology, The Third Affiliated Hospital of Southern Medical University, No. 183, Zhongshan Road West, Guangzhou 510630, PR China
| | - Wenyi Xie
- Department of Neurology, The Third Affiliated Hospital of Southern Medical University, No. 183, Zhongshan Road West, Guangzhou 510630, PR China
| | - Yuanyuan Wang
- Department of Neurology, The Third Affiliated Hospital of Southern Medical University, No. 183, Zhongshan Road West, Guangzhou 510630, PR China
| | - Jiafa Zhang
- Department of Neurology, The Third Affiliated Hospital of Southern Medical University, No. 183, Zhongshan Road West, Guangzhou 510630, PR China
| | - Zibo Cai
- Department of Neurology, The Third Affiliated Hospital of Southern Medical University, No. 183, Zhongshan Road West, Guangzhou 510630, PR China
| | - Zhenlin Mao
- Department of Neurology, The Third Affiliated Hospital of Southern Medical University, No. 183, Zhongshan Road West, Guangzhou 510630, PR China
| | - Weibing Xie
- Judicial Identification Center of Southern Medical University, No.1023-1063, Shatai Road South, Guangzhou 510515, PR China
| | - Tianming Lü
- Department of Neurology, The Third Affiliated Hospital of Southern Medical University, No. 183, Zhongshan Road West, Guangzhou 510630, PR China.
| |
Collapse
|
25
|
LncRNA NEAT1 reversed the hindering effects of miR-495-3p/STAT3 axis and miR-211/PI3K/AKT axis on sepsis-relevant inflammation. Mol Immunol 2020; 117:168-179. [DOI: 10.1016/j.molimm.2019.10.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 10/08/2019] [Accepted: 10/10/2019] [Indexed: 01/05/2023]
|
26
|
Sharifpanah F, Ali EH, Wartenberg M, Sauer H. The milk thistle (Silybum marianum) compound Silibinin stimulates leukopoiesis from mouse embryonic stem cells. Phytother Res 2019; 33:452-460. [PMID: 30548344 DOI: 10.1002/ptr.6241] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 10/30/2018] [Accepted: 11/05/2018] [Indexed: 01/02/2023]
Abstract
The milk thistle compound Silibinin (i.e., a 1:1 mixture of Silybin A and Silybin B) stimulates vasculogenesis of mouse embryonic stem (ES) cells. Because vasculogenesis and leukopoiesis are interrelated, the effect of Silibinin on leukopoiesis of ES cells was investigated. Treatment of differentiating ES cells with hydrosoluble Silibinin-C-2',3-dihydrogen succinate dose-dependent increased the number of CD18+ , CD45+ , and CD68+ cells, indicating leukocyte/macrophage differentiation. Silibinin treatment activated phosphoinositide 3-kinase (PI3K), AKT (protein kinase B), signal transducer and activator of transcription 3 (STAT3), stimulated hypoxia-induced factor-1α (HIF-1α), and vascular endothelial growth factor receptor 2 (VEGFR2) expression and raised intracellular nitric oxide (NO). Western blot experiments showed that upon coincubation with either the PI3K inhibitor LY294002, the STAT3 inhibitor Stattic, the AKT antagonist AKT inhibitor VIII, or the NO inhibitor L-NAME, the Silibinin-induced expression of CD18, CD45, and CD68 was abolished. Moreover, the stimulation of HIF-1α and VEGFR2 expression was blunted upon STAT3 and PI3K/AKT inhibition. Treatment of differentiating ES cells with L-NAME abolished the stimulation of VEGFR2 and VE-cadherin expression achieved with Silibinin, indicating that NO is involved in vasculogenesis and leukocyte differentiation pathways. In summary, the data of the present study demonstrate that Silibinin stimulates leukocyte differentiation of ES cells, which is associated to vasculogenesis and regulated by PI3K/AKT-, STAT3-, and NO-mediated signaling.
Collapse
Affiliation(s)
- Fatemeh Sharifpanah
- Department of Physiology, Faculty of Medicine, Justus Liebig University Giessen, Giessen, Germany
| | - Enas Hussein Ali
- Department of Physiology, Faculty of Medicine, Justus Liebig University Giessen, Giessen, Germany
| | - Maria Wartenberg
- Department of Internal Medicine I, Division of Cardiology, Angiology, Pneumology and Intensive Medical Care, University Hospital Jena, Friedrich-Schiller-University Jena, Jena, Germany
| | - Heinrich Sauer
- Department of Physiology, Faculty of Medicine, Justus Liebig University Giessen, Giessen, Germany
| |
Collapse
|
27
|
Fiore A, Ugel S, De Sanctis F, Sandri S, Fracasso G, Trovato R, Sartoris S, Solito S, Mandruzzato S, Vascotto F, Hippen KL, Mondanelli G, Grohmann U, Piro G, Carbone C, Melisi D, Lawlor RT, Scarpa A, Lamolinara A, Iezzi M, Fassan M, Bicciato S, Blazar BR, Sahin U, Murray PJ, Bronte V. Induction of immunosuppressive functions and NF-κB by FLIP in monocytes. Nat Commun 2018; 9:5193. [PMID: 30518925 PMCID: PMC6281604 DOI: 10.1038/s41467-018-07654-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 11/15/2018] [Indexed: 12/19/2022] Open
Abstract
Immunosuppression is a hallmark of tumor progression, and treatments that inhibit or deplete monocytic myeloid-derived suppressive cells could promote anti-tumor immunity. c-FLIP is a central regulator of caspase-8-mediated apoptosis and necroptosis. Here we show that low-dose cytotoxic chemotherapy agents cause apoptosis linked to c-FLIP down-regulation selectively in monocytes. Enforced expression of c-FLIP or viral FLIP rescues monocytes from cytotoxicity and concurrently induces potent immunosuppressive activity, in T cell cultures and in vivo models of tumor progression and immunotherapy. FLIP-transduced human blood monocytes can suppress graft versus host disease. Neither expression of FLIP in granulocytes nor expression of other anti-apoptotic genes in monocytes conferred immunosuppression, suggesting that FLIP effects on immunosuppression are specific to monocytic lineage and distinct from death inhibition. Mechanistically, FLIP controls a broad transcriptional program, partially by NF-κB activation. Therefore, modulation of FLIP in monocytes offers a means to elicit or block immunosuppressive myeloid cells.
Collapse
Affiliation(s)
- Alessandra Fiore
- Department of Medicine, Section of Immunology, University of Verona, Verona, 37134, Italy
- Max Planck Institute of Biochemistry, Martinsried, 82152, Germany
| | - Stefano Ugel
- Department of Medicine, Section of Immunology, University of Verona, Verona, 37134, Italy.
| | - Francesco De Sanctis
- Department of Medicine, Section of Immunology, University of Verona, Verona, 37134, Italy
| | - Sara Sandri
- Department of Medicine, Section of Immunology, University of Verona, Verona, 37134, Italy
| | - Giulio Fracasso
- Department of Medicine, Section of Immunology, University of Verona, Verona, 37134, Italy
| | - Rosalinda Trovato
- Department of Medicine, Section of Immunology, University of Verona, Verona, 37134, Italy
| | - Silvia Sartoris
- Department of Medicine, Section of Immunology, University of Verona, Verona, 37134, Italy
| | - Samantha Solito
- Department of Surgery, Oncology and Gastroenterology, Section of Oncology and Immunology, University of Padova, Padova, 35124, Italy
| | - Susanna Mandruzzato
- Department of Surgery, Oncology and Gastroenterology, Section of Oncology and Immunology, University of Padova, Padova, 35124, Italy
- Istituto Oncologico Veneto IOV-IRCCS, Padova, 35124, Italy
| | - Fulvia Vascotto
- TRON-Translational Oncology, University Medical Center of Johannes Gutenberg University, Mainz, 55131, Germany
| | - Keli L Hippen
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, 55455, MN, USA
| | - Giada Mondanelli
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy
| | - Ursula Grohmann
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy
| | - Geny Piro
- Department of Medicine, Digestive Molecular Clinical Oncology Research Unit, University of Verona, Verona, 37134, Italy
- Department of Medicine, Laboratory of Oncology and Molecular Therapy, University of Verona, Verona, 37134, Italy
| | - Carmine Carbone
- Department of Medicine, Digestive Molecular Clinical Oncology Research Unit, University of Verona, Verona, 37134, Italy
| | - Davide Melisi
- Department of Medicine, Digestive Molecular Clinical Oncology Research Unit, University of Verona, Verona, 37134, Italy
| | - Rita T Lawlor
- ARC-Net Centre for Applied Research on Cancer, University and Hospital Trust of Verona, Verona, 37134, Italy
| | - Aldo Scarpa
- ARC-Net Centre for Applied Research on Cancer, University and Hospital Trust of Verona, Verona, 37134, Italy
- Department of Pathology and Diagnostics, University of Verona, Verona, 37134, Italy
| | - Alessia Lamolinara
- Department of Medicine and Aging Science, Center of Excellence on Aging and Translational Medicine (CeSi-Met), University G. D'Annunzio of Chieti-Pescara, Chieti, 66100, Italy
| | - Manuela Iezzi
- Department of Medicine and Aging Science, Center of Excellence on Aging and Translational Medicine (CeSi-Met), University G. D'Annunzio of Chieti-Pescara, Chieti, 66100, Italy
| | - Matteo Fassan
- Department of Medicine-DIMED, University of Padova, Padova, 35124, Italy
| | - Silvio Bicciato
- Department of Life Sciences, Center for Genome Research, University of Modena and Reggio Emilia, Modena, 41100, Italy
| | - Bruce R Blazar
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, 55455, MN, USA
| | - Ugur Sahin
- TRON-Translational Oncology, University Medical Center of Johannes Gutenberg University, Mainz, 55131, Germany
- University Medical Center of the Johannes Gutenberg University, Mainz, 55131, Germany
- Biopharmaceutical New Technologies (BioNTech) Corporation, Mainz, 55131, Germany
| | - Peter J Murray
- Max Planck Institute of Biochemistry, Martinsried, 82152, Germany.
| | - Vincenzo Bronte
- Department of Medicine, Section of Immunology, University of Verona, Verona, 37134, Italy.
| |
Collapse
|
28
|
Contreras JI, Robb CM, King HM, Baxter J, Crawford AJ, Kour S, Kizhake S, Sonawane YA, Rana S, Hollingsworth MA, Luo X, Natarajan A. Chemical Genetic Screens Identify Kinase Inhibitor Combinations that Target Anti-Apoptotic Proteins for Cancer Therapy. ACS Chem Biol 2018; 13:1148-1152. [PMID: 29608269 DOI: 10.1021/acschembio.8b00077] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The study presented here provides a framework for the discovery of unique inhibitor combinations that target the apoptosis network for cancer therapy. A pair of doxycycline (Dox)-inducible cell lines that specifically report on the ability of an inhibitor to induce apoptosis by targeting either the Mcl-1 arm or the Bcl-2/Bcl-xL/Bcl-w arm were used. Cell-based assays were optimized for high throughput screening (HTS) with caspase 3/7 as a read out. HTS with a 355-member kinase inhibitor library and the panel of Dox-inducible cell lines revealed that cyclin dependent kinase (CDK) inhibitors induced apoptosis by targeting the Mcl-1 arm, whereas PI3K inhibitors induced apoptosis by targeting the Bcl-2/Bcl-xL/Bcl-w arm. Validation studies identified unique combinations that synergistically inhibited growth and induced apoptosis in a panel of cancer cell lines. Since these inhibitors have been or are currently in clinical trials as single agents, the combinations can be rapidly translated to the clinics.
Collapse
|
29
|
Li Y, Pang Z, Dong X, Liao X, Deng H, Liao C, Liao Y, Chen G, Huang L. MUC1 induces M2 type macrophage influx during postpartum mammary gland involution and triggers breast cancer. Oncotarget 2017; 9:3446-3458. [PMID: 29423058 PMCID: PMC5790475 DOI: 10.18632/oncotarget.23316] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 11/19/2017] [Indexed: 01/28/2023] Open
Abstract
The microenvironment of postpartum mammary gland involution (PMI) has been linked to the increased risk of breast cancer and poor outcome of patients. Nevertheless the mechanism underlying regulates the microenvironment remains largely unknown. MUC1, which is abnormally overexpressed in most breast cancer, is physiologically expressed in PMI. Using MUC1 cytoplasm domain (MUC1-CD) transgenic mice, we reveal that the overexpression of MUC1-CD in mammary epithelial cells increases M2 type macrophage infiltration in PMI. By sustain activating p50, MUC1 upregulates M2 macrophage chemo-attractants and the anti-apoptotic protein Bcl-xL. Because of the tumor promotional microenvironments and reduced apoptosis, MUC1-CD delays PMI process and results in atypical phenotype in multiparous mice mammary. This finding is further supported by the positive association between the expression of MUC1 and p50 in Luminal A and Luminal B subtypes through analyzing breast cancer databases. Taken together, our study demonstrates that MUC1-CD plays an important role in regulating microenvironment of PMI and promoting postpartum mammary tumorigenicity, providing novel prevention and treatment strategies against postpartum breast cancer.
Collapse
Affiliation(s)
- Yuan Li
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China.,Department of Endocrinology, Huadong Hospital Affiliated to Fudan University, Shanghai, P.R. China
| | - Zhi Pang
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Xinran Dong
- Key Laboratory of Birth Defects, Children's Hospital of Fudan University, Shanghai, P.R. China
| | - Xiaodong Liao
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China.,Department of Histoembryology, Genetics and Developmental Biology, Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Huayun Deng
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China.,Department of Histoembryology, Genetics and Developmental Biology, Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Chunhua Liao
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Yahui Liao
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Guoqiang Chen
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Lei Huang
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China.,Department of Histoembryology, Genetics and Developmental Biology, Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| |
Collapse
|
30
|
Tang F, Wang Y, Hemmings BA, Rüegg C, Xue G. PKB/Akt-dependent regulation of inflammation in cancer. Semin Cancer Biol 2017; 48:62-69. [PMID: 28476657 DOI: 10.1016/j.semcancer.2017.04.018] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Revised: 04/13/2017] [Accepted: 04/28/2017] [Indexed: 12/14/2022]
Abstract
Chronic inflammation is a major cause of human cancer. Clinical cancer therapies against inflammatory risk factors are strategically determined. To rationally guide a novel drug development, an improved mechanistic understanding on the pathological connection between inflammation and carcinogenesis is essential. PI3K-PKB signaling axis has been extensively studied and shown to be one of the key oncogenic drivers in most types of cancer. Pharmacological inhibition of the components along this signaling axis is of great interest for developing novel therapies. Interestingly, emerging studies have shown a close association between PKB activation and inflammatory activity in the vicinity of the tumor, and either blockade of PKB or attenuation of para-tumoral inflammation reveals a mutual-interactive pattern through pathway crosstalk. In this review, we intend to discuss recent advances of PKB-regulated chronic inflammation and its potential impacts on tumor development.
Collapse
Affiliation(s)
- Fengyuan Tang
- Department of Biomedicine, University of Basel, 4031 Basel, Switzerland.
| | - Yuhua Wang
- Novartis Pharma AG, 4057 Basel, Switzerland
| | - Brian A Hemmings
- Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland
| | - Curzio Rüegg
- Pathology, Department of Medicine, Faculty of Sciences, University of Fribourg, 1700 Fribourg, Switzerland
| | - Gongda Xue
- Department of Biomedicine, University of Basel, 4031 Basel, Switzerland.
| |
Collapse
|
31
|
Wang MM, Xue M, Miao Y, Kou N, Xu YG, Yang L, Zhang Y, Shi DZ. Panax quinquefolium saponin combined with dual antiplatelet drugs inhibits platelet adhesion to injured HUVECs via PI3K/AKT and COX pathways. JOURNAL OF ETHNOPHARMACOLOGY 2016; 192:10-19. [PMID: 27401285 DOI: 10.1016/j.jep.2016.07.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 06/02/2016] [Accepted: 07/07/2016] [Indexed: 06/06/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Panax quinquefolium saponin (PQS) is the active component extracted from traditional Chinese medicine Panax quinquefolius L. and has been widely used as a supplement to dual antiplatelet drugs (DA) for treatment of coronary artery disease (CAD) for two decades; however, the efficacy of PQS combined with DA against platelet adhesion to endothelial cells (ECs), an essential step in thrombosis, remains unclear. AIM OF THE STUDY To compare PQS combined with DA and DA alone in inhibiting platelet adhesion to injured human umbilical vein endothelial cells (HUVECs) and to explore the possible mechanisms focusing on PI3K/AKT, COX-2/6-keto-PGF1α, and COX-1/TXB2 pathways. METHODS HUVECs injured by oxidized low-density lipoprotein (ox-LDL) were randomly allocated into control, model, DA, PQS+DA (P+DA), LY294002 (a PI3K inhibitor)+DA (L+DA), and LY294002+PQS+DA (LP+DA) groups. HUVEC apoptosis, platelet adhesion to injured HUVECs, and platelet CD62p expression were assayed by fluorescence activated cell sorting (FACS). The concentrations of 6-keto-PGF1α and TXB2 in the supernatant were measured by radioimmunoassay. Protein expression of phosphorylated-PI3K, PI3K, phosphorylated-AKT, AKT, COX-1, and COX-2 in both platelets and HUVECs was evaluated by western blot. RESULTS Compared to DA alone, PQS combined with DA reduced platelet adhesion to HUVECs and HUVEC apoptosis more potently, increased the concentration of supernatant 6-keto-PGF1α and up-regulated phospho-AKT protein in HUVECs. LY294002 mitigated the effects of PQS on HUVEC apoptosis and platelet adhesion. CONCLUSIONS These findings show that PQS as a powerful supplement to DA, attenuated HUVEC apoptosis and improved the DA-mediated reduction of platelet adhesion to injured HUVECs and the underlying mechanisms may be associated with PI3K/AKT and COX pathways in HUVECs and platelets. PQS might provide a new complementary approach to improve the prognosis of thrombotic diseases in future.
Collapse
Affiliation(s)
- Ming-Ming Wang
- Center for Cardiovascular Disease, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Mei Xue
- Center for Cardiovascular Disease, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China.
| | - Yu Miao
- Center for Cardiovascular Disease, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Na Kou
- Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yong-Gang Xu
- Center for Cardiovascular Disease, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Lin Yang
- Center for Cardiovascular Disease, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Ying Zhang
- Beijing University of Chinese Medicine, Beijing 100029, China
| | - Da-Zhuo Shi
- Center for Cardiovascular Disease, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China.
| |
Collapse
|
32
|
Kollek M, Müller A, Egle A, Erlacher M. Bcl-2 proteins in development, health, and disease of the hematopoietic system. FEBS J 2016; 283:2779-810. [DOI: 10.1111/febs.13683] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2015] [Revised: 01/29/2016] [Accepted: 02/12/2016] [Indexed: 12/20/2022]
Affiliation(s)
- Matthias Kollek
- Division of Pediatric Hematology and Oncology; Department of Pediatrics and Adolescent Medicine; University Medical Center of Freiburg; Germany
- Faculty of Biology; University of Freiburg; Germany
| | - Alexandra Müller
- Division of Pediatric Hematology and Oncology; Department of Pediatrics and Adolescent Medicine; University Medical Center of Freiburg; Germany
| | - Alexander Egle
- Laboratory for Immunological and Molecular Cancer Research; 3rd Medical Department for Hematology; Paracelsus Private Medical University Hospital; Salzburg Austria
| | - Miriam Erlacher
- Division of Pediatric Hematology and Oncology; Department of Pediatrics and Adolescent Medicine; University Medical Center of Freiburg; Germany
| |
Collapse
|
33
|
Birtolo C, Go VLW, Ptasznik A, Eibl G, Pandol SJ. Phosphatidylinositol 3-Kinase: A Link Between Inflammation and Pancreatic Cancer. Pancreas 2016; 45:21-31. [PMID: 26658038 PMCID: PMC4859755 DOI: 10.1097/mpa.0000000000000531] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Even though a strong association between inflammation and cancer has been widely accepted, the underlying precise molecular mechanisms are still largely unknown. A complex signaling network between tumor and stromal cells is responsible for the infiltration of inflammatory cells into the cancer microenvironment. Tumor stromal cells such as pancreatic stellate cells (PSCs) and immune cells create a microenvironment that protects cancer cells through a complex interaction, ultimately facilitating their local proliferation and their migration to different sites. Furthermore, PSCs have multiple functions related to local immunity, angiogenesis, inflammation, and fibrosis. Recently, many studies have shown that members of the phosphoinositol-3-phosphate kinase (PI3K) family are activated in tumor cells, PSCs, and tumor-infiltrating inflammatory cells to promote cancer growth. Proinflammatory cytokines and chemokines secreted by immune cells and fibroblasts within the tumor environment can activate the PI3K pathway both in cancer and inflammatory cells. In this review, we focus on the central role of the PI3K pathway in regulating the cross talk between immune/stromal cells and cancer cells. Understanding the role of the PI3K pathway in the development of chronic pancreatitis and cancer is crucial for the discovery of novel and efficacious treatment options.
Collapse
Affiliation(s)
- Chiara Birtolo
- Department of Medicine, Cedars Sinai Medical Center, Los Angeles, CA,Department of Internal Medicine, S. Orsola-Malpighi Hospital, University of Bologna, Italy
| | - Vay Liang W. Go
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Andrzej Ptasznik
- Department of Medicine, Cedars Sinai Medical Center, Los Angeles, CA
| | - Guido Eibl
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Stephen J. Pandol
- Department of Medicine, Cedars Sinai Medical Center, Los Angeles, CA,VA Greater Los Angeles Health Care System, Los Angeles, CA
| |
Collapse
|
34
|
Zeng C, Wang W, Yu X, Yang L, Chen S, Li Y. Pathways related to PMA-differentiated THP1 human monocytic leukemia cells revealed by RNA-Seq. SCIENCE CHINA-LIFE SCIENCES 2015; 58:1282-7. [PMID: 26582014 DOI: 10.1007/s11427-015-4967-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 08/19/2015] [Indexed: 10/22/2022]
Abstract
Previous analyses have reported that the human monocytic cell line THP1 can be differentiated into cells with macrophage-like characteristics by phorbol 12-myristate 13-acetate (PMA). However, little is known about the mechanism responsible for regulating this differentiation process. Here, we performed high-throughput RNA-Seq analysis to investigate the genes differently expressed in THP1 cells treated with and without PMA and examined those that may be responsible for the PMA-induced differentiation of monocytes into macrophages. We found 3,000 genes to be differentially expressed after PMA treatment. Gene ontology analysis revealed that genes related to cellular processes and regulation of biological processes were significantly enriched. KEGG analysis also demonstrated that the differentially expressed genes (DEGs) were significantly enriched in the PI3K/AKT signaling pathway and phagosome pathway. Importantly, we reveal an important role of the PI3K/AKT pathway in PMA-induced THP1 cell differentiation. The identified DEGs and pathways may facilitate further study of the detailed molecular mechanisms of THP1 differentiation. Thus, our results provide numerous potential therapeutic targets for modulation of the differentiation of this disease.
Collapse
Affiliation(s)
- ChengWu Zeng
- First Affiliated Hospital, Jinan University, Guangzhou, 510632, China.,Institute of Hematology, Medical College, Jinan University, Guangzhou, 510632, China.,Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, 510632, China
| | - WenTao Wang
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory for Biocontrol, School of Life Science, Sun Yat-sen University, Guangzhou, 510275, China
| | - XiBao Yu
- Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, 510632, China
| | - LiJian Yang
- Institute of Hematology, Medical College, Jinan University, Guangzhou, 510632, China
| | - ShaoHua Chen
- Institute of Hematology, Medical College, Jinan University, Guangzhou, 510632, China
| | - YangQiu Li
- First Affiliated Hospital, Jinan University, Guangzhou, 510632, China. .,Institute of Hematology, Medical College, Jinan University, Guangzhou, 510632, China. .,Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
35
|
Xue G, Zippelius A, Wicki A, Mandala M, Tang F, Massi D, Hemmings BA. Integrated Akt/PKB Signaling in Immunomodulation and Its Potential Role in Cancer Immunotherapy. J Natl Cancer Inst 2015; 107:djv171. [DOI: 10.1093/jnci/djv171] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 05/22/2015] [Indexed: 12/17/2022] Open
|