1
|
Zong D, Sun B, Ye Q, Cao H, Guan H. Circadian Gene BMAL1 Regulation of Cellular Senescence in Thyroid Aging. Aging Cell 2025:e70119. [PMID: 40434135 DOI: 10.1111/acel.70119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 04/18/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025] Open
Abstract
As global aging accelerates, the incidence of thyroid diseases, particularly hypothyroidism, is rising in the elderly. The thyroid-stimulating hormone (TSH) levels increase in healthy elderly populations. However, whether the thyroid undergoes cellular senescence and how this relates to thyroid hormone (TH) synthesis remain unclear. To investigate the molecular and functional characteristics of thyroid aging, we performed scRNA-seq on human thyroids from young, middle-aged, and old groups, identifying thousands of aging-related differentially expressed genes and revealing the early onset of aging in the middle-aged group. As aging progresses, the expression levels of genes related to TH synthesis increase, suggesting that epithelial cells (EPI) adjust their gene expression in response to elevated TSH levels. Additionally, the senescence-associated secretory phenotype (SASP) in EPI cells is progressively enhanced with aging. We identified a subgroup of epithelial cells (CDKN1A_EPI) characterized by reduced functionality and significantly elevated levels of cellular senescence. We found that the core circadian rhythm gene BMAL1 (ARNTL) is downregulated during aging. We further validated this finding using the thyroid-specific Bmal1 knockout mouse model, showing that the downregulation of Bmal1 inhibits the expression of Nfkbia (NF-κB inhibitor alpha), thereby accelerating cellular senescence and impairing hormone synthesis. Finally, through cell line experiments and transcriptome sequencing, we confirmed that BMAL1 knockout leads to decreased NFKBIA expression, promoting thyroid cellular senescence. Our study demonstrates that circadian rhythm disruption accelerates cellular senescence in the thyroid and exacerbates the decline of thyroid function, providing a novel theoretical foundation for understanding thyroid aging mechanisms and maintaining thyroid function stability.
Collapse
Affiliation(s)
- Dandan Zong
- Department of Endocrinology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Baihui Sun
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qiting Ye
- Department of Endocrinology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Hongxin Cao
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province and Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Haixia Guan
- Department of Endocrinology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
2
|
Jiang H, Yang H, Li H, Wang Y, Shen L, Adu X, Zhang J, Zhou H, Pu L, Zhang S, Guo J, Tong A, Yan H. Anti-Herpes simplex virus type 1 activity of the Rohdea chinensis (Baker) N. Tanaka aqueous extracts. JOURNAL OF ETHNOPHARMACOLOGY 2025:120024. [PMID: 40412773 DOI: 10.1016/j.jep.2025.120024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 05/21/2025] [Accepted: 05/21/2025] [Indexed: 05/27/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Herpes simplex virus type 1 (HSV-1) is a prevalent neurotropic pathogen that establishes lifelong latency in the peripheral nervous system, posing substantial clinical challenges due to its recurrent infections and emerging drug resistance. Rohdea chinensis (Baker) N. Tanaka, a traditional Chinese herbal medicine, has been historically used for heat-clearing, detoxifying, wind-dampness dispelling, blood-stasis removing, and pain-relieving properties. Despite its common folk use in Leshan, Sichuan Province, for treating herpes simplex infections, the chemical composition and antiviral activity of Rohdea chinensis remain largely uninvestigated. AIM OF THE STUDY The aim of this study was to clarify the chemical composition and antiviral activity of the Rohdea chinensis aqueous extract (RCAE), and preliminarily speculate the possible mechanism of its antiviral effect. MATERIALS AND METHODS The chemical profile of RCAE was characterized using UPLC-Q-TOF-MS/MS, identifying 9 major compounds. The antiviral effects of RCAE were assessed through in vitro and in vivo models, including direct virucidal assays, HSV-1 adhesion inhibition, and a mouse model of flank scratch HSV-1 infection. Immunomodulatory effects were evaluated by analyzing dendritic cell (DC) maturation, CD8+ T cell differentiation, and cytokine expression. Additionally, HSV-1 latent reactivation models were established to assess RCAE's impact on viral persistence in the dorsal root ganglion (DRG). Molecular docking studies were conducted to determine potential interactions between RCAE bioactive compounds and HSV-1 glycoprotein D (gD). RESULTS The overall anti-HSV-1 effect of RCAE on Vero cells was determined and the EC50 value was 7.994 μg/mL. RCAE directly killed HSV-1 and inhibited HSV-1 adhesion in vitro (P<0.001, 30 μg/mL RCAE treated groups vs HSV-1 group), and alleviated viral infection and inflammation of the skin involving the NF-κB pathway (P<0.001, different doses of RCAE treated groups vs HSV-1 group), as well as anti-viral infection of the brain and DRG in HSV-1-induced mice (P<0.05, 0.6 g/mL RCAE treated groups vs HSV-1 group). Interestingly, RCAE played an antiviral role by increasing the maturation of DC in mice, enhancing the antigen presentation ability of DC, promoting the differentiation of CD8+T cells and the secretion of CXCL1 and IFN-γ, and inhibiting the immune escape of HSV-1 virus (P<0.05, 0.6 g/mL RCAE treated groups vs HSV-1 group). Besides, RCAE prevented the reactivation of HSV-1 in vitro (P<0.001, 0.6 g/mL RCAE treated groups vs HSV-1 group). HSV gD is a critical viral glycoprotein that plays a central role in viral attachment to host cells, intercellular spread, and entry into host cells. To clarify the anti-HSV-1 mechanism of RCAE, molecular docking was employed to preliminarily confirm stable interactions between RCAE compounds (notably compound 9, with a binding energy of -10.5 kcal/mol) and HSV-1 gD. In vitro and in vivo experiments showed that RCAE reduced the expression of gD gene (P<0.001, different doses of RCAE treated groups vs HSV-1 group) and protein in skin tissues of HSV-1-infected mice, as well as the gD mRNA levels (P<0.001, different doses of RCAE treated groups vs HSV-1 group) in HSV-1-treated HaCaT cells. These findings suggest that the anti-HSV-1 effect of RCAE may be associated with the inhibition of gD function. CONCLUSIONS Our results indicate that RCAE has multi-target antiviral effects, including direct virus-killing activity, inhibition of viral replication, immune regulation, and inhibition of latent reactivation of HSV-1 in preclinical models. Integrating traditional Chinese medicine principles (heat-clearing, detoxifying, and blood-stasis-removing properties) with modern pharmacological activities (antiviral, anti-inflammatory, and immunomodulatory effects), RCAE demonstrates potential as a natural product candidate for addressing HSV-1 infection. Given RCAE's complex and undefined composition, future studies are needed to identify key antiviral components and their in vivo efficacy and mechanisms.
Collapse
Affiliation(s)
- Huiling Jiang
- Immunotherapy Laboratory, College of Pharmacy and Food, Southwest Minzu University, Chengdu, Sichuan, China
| | - Huantao Yang
- Immunotherapy Laboratory, College of Grassland Resources, Southwest Minzu University, Chengdu, Sichuan, China
| | - Hui Li
- Immunotherapy Laboratory, College of Pharmacy and Food, Southwest Minzu University, Chengdu, Sichuan, China
| | - Yujuan Wang
- Immunotherapy Laboratory, College of Grassland Resources, Southwest Minzu University, Chengdu, Sichuan, China
| | - Lidu Shen
- Immunotherapy Laboratory, College of Pharmacy and Food, Southwest Minzu University, Chengdu, Sichuan, China
| | - Xiaobi Adu
- Immunotherapy Laboratory, College of Pharmacy and Food, Southwest Minzu University, Chengdu, Sichuan, China
| | - Junxiong Zhang
- Immunotherapy Laboratory, College of Pharmacy and Food, Southwest Minzu University, Chengdu, Sichuan, China
| | - Hang Zhou
- Immunotherapy Laboratory, College of Pharmacy and Food, Southwest Minzu University, Chengdu, Sichuan, China
| | - Likai Pu
- Immunotherapy Laboratory, College of Pharmacy and Food, Southwest Minzu University, Chengdu, Sichuan, China
| | - Shiying Zhang
- Immunotherapy Laboratory, College of Grassland Resources, Southwest Minzu University, Chengdu, Sichuan, China
| | - Jiacheng Guo
- Immunotherapy Laboratory, College of Grassland Resources, Southwest Minzu University, Chengdu, Sichuan, China
| | - Aiping Tong
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, 610041, Chengdu, China.
| | - Hengxiu Yan
- Immunotherapy Laboratory, College of Pharmacy and Food, Southwest Minzu University, Chengdu, Sichuan, China.
| |
Collapse
|
3
|
Park B, Lee S, Jo I, Kang D, Kim T, Ryu J, Kong H, Baek M, Ahn T. Mechanistic Insights into Radiation Resistance in Colorectal Cancer: Gene Exploration Study. Int J Mol Sci 2025; 26:3849. [PMID: 40332515 PMCID: PMC12027542 DOI: 10.3390/ijms26083849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Revised: 04/10/2025] [Accepted: 04/12/2025] [Indexed: 05/08/2025] Open
Abstract
Radiotherapy is a cornerstone of colorectal cancer (CRC) treatment; however, its therapeutic efficacy is often compromised by both intrinsic and acquired resistance in CRC cells. This study employed small interfering RNA (siRNA) technology to elucidate the functional roles of BAMBI, GADD34, NFKBIA, and NFKBID in CRC cell lines SW480 and HCT116. We assessed their impact on key cellular processes and radiation sensitivity. Gene silencing of all four target genes significantly suppressed CRC cell proliferation, migration, and invasion. Moreover, siRNA-mediated knockdown enhanced radiation sensitivity, as evidenced by a substantial increase in apoptosis and a marked reduction in cell viability compared with controls. These findings suggest that BAMBI, GADD34, NFKBIA, and NFKBID serve as critical regulators of CRC progression and radiation resistance. Overall, this study provides a mechanistic foundation for further exploration into the pathways underlying radiation resistance and underscores the potential for developing personalized radiotherapy strategies guided by molecular profiling.
Collapse
Affiliation(s)
- Beamjun Park
- Department of Medical Life Science, Soonchunhyang University, Asan 31538, Republic of Korea; (B.P.); (T.K.); (J.R.); (H.K.)
| | - Soohyeon Lee
- Department of Colorectal Surgery, College of Medicine, Soonchunhyang University Cheonan Hospital, Cheonan 31151, Republic of Korea; (S.L.); (D.K.)
| | - Inyoung Jo
- Department of Radiation Oncology, Soonchunhyang University, Asan 31538, Republic of Korea;
| | - Donghyun Kang
- Department of Colorectal Surgery, College of Medicine, Soonchunhyang University Cheonan Hospital, Cheonan 31151, Republic of Korea; (S.L.); (D.K.)
| | - Taewan Kim
- Department of Medical Life Science, Soonchunhyang University, Asan 31538, Republic of Korea; (B.P.); (T.K.); (J.R.); (H.K.)
| | - Jaesung Ryu
- Department of Medical Life Science, Soonchunhyang University, Asan 31538, Republic of Korea; (B.P.); (T.K.); (J.R.); (H.K.)
| | - Hyejeong Kong
- Department of Medical Life Science, Soonchunhyang University, Asan 31538, Republic of Korea; (B.P.); (T.K.); (J.R.); (H.K.)
| | - Moojun Baek
- Department of Colorectal Surgery, College of Medicine, Soonchunhyang University Cheonan Hospital, Cheonan 31151, Republic of Korea; (S.L.); (D.K.)
| | - Taesung Ahn
- Department of Colorectal Surgery, College of Medicine, Soonchunhyang University Cheonan Hospital, Cheonan 31151, Republic of Korea; (S.L.); (D.K.)
| |
Collapse
|
4
|
Yu J, Zhang C, Zhang Q, Lu B, Lu G, Zhang C, Qiu R, Wang X, Zou C, Chu J, Li H, Zhao W. AZIN1-dependent polyamine synthesis accelerates tumor cell cycle progression and impairs effector T-cell function in osteosarcoma. Cell Death Dis 2025; 16:310. [PMID: 40246846 PMCID: PMC12006533 DOI: 10.1038/s41419-025-07640-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 04/01/2025] [Accepted: 04/04/2025] [Indexed: 04/19/2025]
Abstract
Osteosarcoma, the most prevalent malignant bone tumor among adolescents, frequently exhibits limited responsiveness to immunotherapy, a challenge attributed to poorly understood underlying mechanisms. Here, we identify enhanced polyamine biosynthesis as a key driver of osteosarcoma progression and immunotherapy resistance. We show that osteosarcoma cell proliferation and tumor growth rely on polyamine availability and that disruption of polyamine synthesis significantly boosts the cytotoxic efficacy of TCR-engineered T cells against osteosarcoma cells. Mechanistically, we reveal that the knockdown of antizyme inhibitor 1 (AZIN1) or suppression of polyamine production reduces MYC expression, leading to diminished tumor cell viability via the downregulation of cell cycle-related genes. Furthermore, reduced MYC levels are associated with changes in the expression of immunomodulatory cytokines and human leukocyte antigen molecules, pointing to a potential link with enhanced T-cell-mediated cytotoxicity. Collectively, our findings establish a pivotal role for the AZIN1-polyamine axis in osteosarcoma proliferation and immune evasion, and support the development of novel immunotherapeutic strategies targeting polyamine biosynthesis to combat this aggressive cancer.
Collapse
Affiliation(s)
- Jiaming Yu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Chuanxia Zhang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Qinkai Zhang
- Department of Breast and Thyroid Surgery, Guangzhou Women and Children's Medical Center, Guangzhou, 510623, China
| | - Bing Lu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Guohao Lu
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510000, China
| | - Chunxiao Zhang
- School of Laboratory Medicine, Guangzhou Health Science College, Guangzhou, 510450, China
| | - Ru Qiu
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510000, China
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510000, China
| | - Xinyue Wang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510000, China
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510000, China
| | - Changye Zou
- Musculoskeletal Oncology Center, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Junjun Chu
- HRYZ Biotech Co., Guangzhou, 510507, China.
| | - Haizhou Li
- Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, Ganzhou, 341000, China.
| | - Wei Zhao
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China.
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510000, China.
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510000, China.
| |
Collapse
|
5
|
Bournique E, Sanchez A, Oh S, Ghazarian D, Mahieu AL, Manjunath L, Ednacot E, Ortega P, Masri S, Marazzi I, Buisson R. ATM and IRAK1 orchestrate two distinct mechanisms of NF-κB activation in response to DNA damage. Nat Struct Mol Biol 2025; 32:740-755. [PMID: 39753776 PMCID: PMC11997730 DOI: 10.1038/s41594-024-01417-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 10/02/2024] [Indexed: 01/25/2025]
Abstract
DNA damage in cells induces the expression of inflammatory genes. However, the mechanism by which cells initiate an innate immune response in the presence of DNA lesions blocking transcription remains unknown. Here we find that genotoxic stresses lead to an acute activation of the transcription factor NF-κB through two distinct pathways, each triggered by different types of DNA lesions and coordinated by either ataxia-telangiectasia mutated (ATM) or IRAK1 kinases. ATM stimulates NF-κB in cells with DNA double-strand breaks. By contrast, IRAK1-induced NF-κB signaling occurs in neighboring cells through IL-1α secretion from transcriptionally stressed cells caused by DNA lesions blocking RNA polymerases. Subsequently, both pathways stimulate TRAF6 and the IKK complex to promote NF-κB-mediated inflammatory gene expression. These findings provide an alternative mechanism for damaged cells with impaired transcription to initiate an inflammatory response without relying on their own gene expression, a necessary step that injured cells depend on during canonical innate immune responses.
Collapse
Affiliation(s)
- Elodie Bournique
- Department of Biological Chemistry, School of Medicine, University of California Irvine, Irvine, CA, USA
- Center for Epigenetics and Metabolism, Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA
- Center for Virus Research, University of California Irvine, Irvine, CA, USA
| | - Ambrocio Sanchez
- Department of Biological Chemistry, School of Medicine, University of California Irvine, Irvine, CA, USA
- Center for Epigenetics and Metabolism, Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA
- Center for Virus Research, University of California Irvine, Irvine, CA, USA
| | - Sunwoo Oh
- Department of Biological Chemistry, School of Medicine, University of California Irvine, Irvine, CA, USA
- Center for Epigenetics and Metabolism, Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA
- Center for Virus Research, University of California Irvine, Irvine, CA, USA
| | - Daniel Ghazarian
- Department of Biological Chemistry, School of Medicine, University of California Irvine, Irvine, CA, USA
- Center for Epigenetics and Metabolism, Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA
- Center for Virus Research, University of California Irvine, Irvine, CA, USA
| | - Alisa L Mahieu
- Department of Biological Chemistry, School of Medicine, University of California Irvine, Irvine, CA, USA
- Center for Epigenetics and Metabolism, Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA
| | - Lavanya Manjunath
- Department of Biological Chemistry, School of Medicine, University of California Irvine, Irvine, CA, USA
- Center for Epigenetics and Metabolism, Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA
- Center for Virus Research, University of California Irvine, Irvine, CA, USA
| | - Eirene Ednacot
- Department of Biological Chemistry, School of Medicine, University of California Irvine, Irvine, CA, USA
- Center for Epigenetics and Metabolism, Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA
- Department of Pharmaceutical Sciences, School of Pharmacy & Pharmaceutical Sciences, University of California Irvine, Irvine, CA, USA
| | - Pedro Ortega
- Department of Biological Chemistry, School of Medicine, University of California Irvine, Irvine, CA, USA
- Center for Epigenetics and Metabolism, Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA
- Center for Virus Research, University of California Irvine, Irvine, CA, USA
| | - Selma Masri
- Department of Biological Chemistry, School of Medicine, University of California Irvine, Irvine, CA, USA
- Center for Epigenetics and Metabolism, Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA
| | - Ivan Marazzi
- Department of Biological Chemistry, School of Medicine, University of California Irvine, Irvine, CA, USA
- Center for Epigenetics and Metabolism, Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA
- Center for Virus Research, University of California Irvine, Irvine, CA, USA
| | - Rémi Buisson
- Department of Biological Chemistry, School of Medicine, University of California Irvine, Irvine, CA, USA.
- Center for Epigenetics and Metabolism, Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA.
- Center for Virus Research, University of California Irvine, Irvine, CA, USA.
- Department of Pharmaceutical Sciences, School of Pharmacy & Pharmaceutical Sciences, University of California Irvine, Irvine, CA, USA.
| |
Collapse
|
6
|
Tomimatsu N, Di Cristofaro LFM, Kanji S, Samentar L, Jordan BR, Kittler R, Habib AA, Espindola-Netto JM, Tchkonia T, Kirkland JL, Burns TC, Sarkaria JN, Gilbert A, Floyd JR, Hromas R, Zhao W, Zhou D, Sung P, Mukherjee B, Burma S. Targeting cIAP2 in a novel senolytic strategy prevents glioblastoma recurrence after radiotherapy. EMBO Mol Med 2025; 17:645-678. [PMID: 39972068 PMCID: PMC11982261 DOI: 10.1038/s44321-025-00201-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 01/28/2025] [Accepted: 02/05/2025] [Indexed: 02/21/2025] Open
Abstract
Glioblastomas (GBM) are routinely treated with high doses of ionizing radiation (IR), yet these tumors recur quickly, and the recurrent tumors are highly therapy resistant. Here, we report that IR-induced senescence of tumor cells counterintuitively spurs GBM recurrence, driven by the senescence-associated secretory phenotype (SASP). We find that irradiated GBM cell lines and patient derived xenograft (PDX) cultures senesce rapidly in a p21-dependent manner. Senescent glioma cells upregulate SASP genes and secrete a panoply of SASP factors, prominently interleukin IL-6, an activator of the JAK-STAT3 pathway. These SASP factors collectively activate the JAK-STAT3 and NF-κB pathways in non-senescent GBM cells, thereby promoting tumor cell proliferation and SASP spreading. Transcriptomic analyses of irradiated GBM cells and the TCGA database reveal that the cellular inhibitor of apoptosis protein 2 (cIAP2), encoded by the BIRC3 gene, is a potential survival factor for senescent glioma cells. Senescent GBM cells not only upregulate BIRC3 but also induce BIRC3 expression and promote radioresistance in non-senescent tumor cells. We find that second mitochondria-derived activator of caspases (SMAC) mimetics targeting cIAP2 act as novel senolytics that trigger apoptosis of senescent GBM cells with minimal toxicity towards normal brain cells. Finally, using both PDX and immunocompetent mouse models of GBM, we show that the SMAC mimetic birinapant, administered as an adjuvant after radiotherapy, can eliminate senescent GBM cells and prevent the emergence of recurrent tumors. Taken together, our results clearly indicate that significant improvement in GBM patient survival may become possible in the clinic by eliminating senescent cells arising after radiotherapy.
Collapse
Affiliation(s)
- Nozomi Tomimatsu
- Department of Neurosurgery, University of Texas Health, San Antonio, TX, USA
| | | | - Suman Kanji
- Department of Neurosurgery, University of Texas Health, San Antonio, TX, USA
| | - Lorena Samentar
- Department of Neurosurgery, University of Texas Health, San Antonio, TX, USA
| | - Benjamin Russell Jordan
- Department of Neurosurgery, University of Texas Health, San Antonio, TX, USA
- Department of Biochemistry and Structural Biology, University of Texas Health, San Antonio, TX, USA
| | - Ralf Kittler
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Amyn A Habib
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | - Tamara Tchkonia
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | | | - Terry C Burns
- Department of Neurological Surgery, Mayo Clinic, Rochester, MN, USA
| | - Jann N Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, USA
| | - Andrea Gilbert
- Department of Pathology, University of Texas Health, San Antonio, TX, USA
| | - John R Floyd
- Department of Neurosurgery, University of Texas Health, San Antonio, TX, USA
| | - Robert Hromas
- Department of Medicine, University of Texas Health, San Antonio, TX, USA
| | - Weixing Zhao
- Department of Biochemistry and Structural Biology, University of Texas Health, San Antonio, TX, USA
| | - Daohong Zhou
- Department of Biochemistry and Structural Biology, University of Texas Health, San Antonio, TX, USA
| | - Patrick Sung
- Department of Biochemistry and Structural Biology, University of Texas Health, San Antonio, TX, USA
| | - Bipasha Mukherjee
- Department of Neurosurgery, University of Texas Health, San Antonio, TX, USA.
| | - Sandeep Burma
- Department of Neurosurgery, University of Texas Health, San Antonio, TX, USA.
- Department of Biochemistry and Structural Biology, University of Texas Health, San Antonio, TX, USA.
| |
Collapse
|
7
|
Wei J, Tan Z, Huang G, Zeng Y, Chen S, Yuan G, He S, Zhou Y. Anthocyanins Delay D-Galactose-Induced Mouse Liver Aging by Regulating the NF-κB/IKK Signaling Pathway. Food Sci Nutr 2025; 13:e70161. [PMID: 40255558 PMCID: PMC12005982 DOI: 10.1002/fsn3.70161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 02/28/2025] [Accepted: 04/01/2025] [Indexed: 04/22/2025] Open
Abstract
Aging is an intricate pathophysiological phenotype. It is the result of the combined action of various inflammatory factors and cytokines. Aging is closely related to inflammation, apoptosis, tumors, and other diseases. Anthocyanins are a kind of natural flavonoid, mainly contained in plant fruits such as bilberry, grape, purple sweet potato, and so on. These flavonoids have antioxidation, antiaging, and anti-inflammatory properties. It has been found that anthocyanins can effectively delay liver, ovary, and other organ aging. However, the biological mechanism by which anthocyanins alleviate aging phenotypes is still poorly understood. To simulate liver aging in mice, D-galactose was injected daily at 800 mg/kg to accelerate aging, and anthocyanins at 20 or 40 mg/kg were given as intervention treatments. The antiaging effect of anthocyanins was evaluated by body weight, inflammatory markers, and aging markers. Serum ALT and AST levels were measured, and liver histology was assessed using hematoxylin-eosin staining. In addition, we explored the molecular mechanism of anthocyanins delaying liver aging by detecting the expression levels of NF-κB/IKK signaling protein molecules. Our results indicate that anthocyanins can effectively delay mouse liver senescence induced by D-galactose. Analyses by Western blot demonstrated that anthocyanins inhibited the NF-κB/IKK signaling pathway, thereby inhibiting inflammation. In vitro, anthocyanins attenuate the D-galactose (D-gal)-induced aging in AML12 cells, as indicated by reduced aging-associated p21 and p16. Anthocyanins can similarly inhibit the NF-κB/IKK signal pathway in D-gal-induced aging in AML12 cells. Based on these findings, anthocyanins reduce liver aging in mice by regulating the NF-κB/IKK pathway.
Collapse
Affiliation(s)
- Jie Wei
- Division of Hepatobiliary SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
- Key Laboratory of Basic and Clinical Application Research for Hepatobiliary Diseases of GuangxiNanningGuangxiChina
- Guangxi Key Laboratory of Immunology and Metabolism for Liver DiseasesNanningGuangxiChina
| | - Zhi Tan
- Division of Hepatobiliary SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
- Key Laboratory of Basic and Clinical Application Research for Hepatobiliary Diseases of GuangxiNanningGuangxiChina
- Guangxi Key Laboratory of Immunology and Metabolism for Liver DiseasesNanningGuangxiChina
| | - Guozhen Huang
- Division of Hepatobiliary SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
- Key Laboratory of Basic and Clinical Application Research for Hepatobiliary Diseases of GuangxiNanningGuangxiChina
- Guangxi Key Laboratory of Immunology and Metabolism for Liver DiseasesNanningGuangxiChina
| | - Yonglian Zeng
- Division of Hepatobiliary SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
- Key Laboratory of Basic and Clinical Application Research for Hepatobiliary Diseases of GuangxiNanningGuangxiChina
- Guangxi Key Laboratory of Immunology and Metabolism for Liver DiseasesNanningGuangxiChina
| | - Shilian Chen
- Division of Hepatobiliary SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
- Key Laboratory of Basic and Clinical Application Research for Hepatobiliary Diseases of GuangxiNanningGuangxiChina
- Guangxi Key Laboratory of Immunology and Metabolism for Liver DiseasesNanningGuangxiChina
| | - Guandou Yuan
- Division of Hepatobiliary SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
- Key Laboratory of Basic and Clinical Application Research for Hepatobiliary Diseases of GuangxiNanningGuangxiChina
- Guangxi Key Laboratory of Immunology and Metabolism for Liver DiseasesNanningGuangxiChina
| | - Songqing He
- Division of Hepatobiliary SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
- Key Laboratory of Basic and Clinical Application Research for Hepatobiliary Diseases of GuangxiNanningGuangxiChina
- Guangxi Key Laboratory of Immunology and Metabolism for Liver DiseasesNanningGuangxiChina
| | - Yi Zhou
- Division of Hepatobiliary SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
- Key Laboratory of Basic and Clinical Application Research for Hepatobiliary Diseases of GuangxiNanningGuangxiChina
- Guangxi Key Laboratory of Immunology and Metabolism for Liver DiseasesNanningGuangxiChina
| |
Collapse
|
8
|
Song J, Li Z, Yang J, Ma R, Wang D, Quan R, Wen X, Liu J. Seneca Valley virus infection exploits DNA damage response to facilitate viral replication. J Virol 2025; 99:e0221124. [PMID: 40008889 PMCID: PMC11915816 DOI: 10.1128/jvi.02211-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Accepted: 01/26/2025] [Indexed: 02/27/2025] Open
Abstract
Seneca Valley virus (SVV) is an emerging pathogen that causes severe vesicular diseases in swine, posing a significant threat to the global pork industry. DNA and RNA viruses manipulate the host DNA damage response (DDR) to modulate cellular machinery and facilitate their life cycles. However, the interaction between the host DDR and SVV infection remains unexplored. Here, we aimed to comprehensively investigate the DDR and DNA repair signaling pathways during SVV infection. We found that SVV infection causes DNA damage and triggers distinct DDR signaling pathways, including ataxia telangiectasia-mutated (ATM) kinase, ATM-Rad3-related kinase, and DNA-dependent protein kinase. However, it failed to induce the formation of γH2AX and 53BP1 foci, resulting in unrepaired DNA damage. Furthermore, we found that SVV 2B and 2C proteins can activate DDR signaling pathways and impair DNA repair. SVV-induced DDR triggered NF-κB signaling accompanied by upregulation of pro-inflammatory cytokines, as evidenced by the inhibition of ATM kinase, abolished SVV-induced NF-κB activation. Inhibition of the ATM pathway attenuated SVV replication. These findings expand our understanding of host DDR manipulation during viral infection and provide crucial insights into a novel mechanism exploited by SVV to regulate the inflammatory response for efficient replication.IMPORTANCEDDR is a cellular machinery that senses and repairs host DNA lesions to maintain genome integrity. Viruses have evolved diverse strategies to manipulate host DDR for replicative efficiency. SVV is an emerging virus that causes vesicular diseases in pigs and severely threatens the swine industry. However, the interaction between SVV and DDR remains unclear. Here, we found that SVV modulates host DDR pathways to facilitate viral replication. Our results demonstrated that SVV infection causes DNA damage, activates ATM-mediated DNA double-strand break response, and impedes DNA repair. SVV 2B and 2C proteins induced DNA damage and activated the DDR pathway while impairing repair mechanisms. This study revealed a fine-tuned molecular mechanism of SVV-modulated DDR that contributes to viral replication, facilitating deeper insight into SVV replication.
Collapse
Affiliation(s)
- Jiangwei Song
- Beijing Key Laboratory for Prevention and Control of Infectious Diseases in Livestock and Poultry, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| | - Zijian Li
- College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China
| | - Jingjing Yang
- College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China
| | - Ruiyi Ma
- Beijing Key Laboratory for Prevention and Control of Infectious Diseases in Livestock and Poultry, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| | - Dan Wang
- Beijing Key Laboratory for Prevention and Control of Infectious Diseases in Livestock and Poultry, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| | - Rong Quan
- Beijing Key Laboratory for Prevention and Control of Infectious Diseases in Livestock and Poultry, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| | - Xuexia Wen
- College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China
| | - Jue Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| |
Collapse
|
9
|
Nihira NT, Wu W, Hosoi M, Togashi Y, Sunada S, Miyoshi Y, Miki Y, Ohta T. Nuclear PD-L1 triggers tumour-associated inflammation upon DNA damage. EMBO Rep 2025; 26:635-655. [PMID: 39747659 PMCID: PMC11811057 DOI: 10.1038/s44319-024-00354-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 12/02/2024] [Accepted: 12/08/2024] [Indexed: 01/04/2025] Open
Abstract
Immune checkpoint inhibitors against PD-1/PD-L1 are highly effective in immunologically hot tumours such as triple-negative breast cancer, wherein constitutive DNA damage promotes inflammation, while inducing PD-L1 expression to avoid attack by cytotoxic T cells. However, whether and how PD-L1 regulates the DNA damage response and inflammation remains unclear. Here, we show that nuclear PD-L1 activates the ATR-Chk1 pathway and induces proinflammatory chemocytokines upon genotoxic stress. PD-L1 interacts with ATR and is essential for Chk1 activation and chromatin binding. cGAS-STING and NF-κB activation in the late phase of the DNA damage response is inhibited by PD-L1 deletion or by inhibitors of ATR and Chk1. Consequently, the induction of proinflammatory chemocytokines at this stage is inhibited by deletion of PD-L1, but restored by the ATR activator Garcinone C. Inhibition of nuclear localisation by PD-L1 mutations or the HDAC2 inhibitor Santacruzamate A inhibits chemocytokine induction. Conversely, the p300 inhibitor C646, which accelerates PD-L1 nuclear localisation, promotes chemocytokine induction. These findings suggest that nuclear PD-L1 strengthens the properties of hot tumours and contributes to shaping the tumour microenvironment.
Collapse
Affiliation(s)
- Naoe T Nihira
- Department of Translational Oncology, St. Marianna University Graduate School of Medicine, Kawasaki, 216-8511, Japan
| | - Wenwen Wu
- Department of Translational Oncology, St. Marianna University Graduate School of Medicine, Kawasaki, 216-8511, Japan
| | - Mitsue Hosoi
- Department of Translational Oncology, St. Marianna University Graduate School of Medicine, Kawasaki, 216-8511, Japan
| | - Yukiko Togashi
- Department of Translational Oncology, St. Marianna University Graduate School of Medicine, Kawasaki, 216-8511, Japan
| | - Shigeaki Sunada
- Juntendo Advanced Research Institute for Health Science, Juntendo University, Tokyo, 113-8421, Japan
| | - Yasuo Miyoshi
- Department of Surgery, Division of Breast and Endocrine Surgery, School of Medicine, Hyogo Medical University, Nishinomiya City, Hyogo, Japan
| | - Yoshio Miki
- Research and Development Center for Precision Medicine, University of Tsukuba, Ibaraki, 305-8550, Japan
| | - Tomohiko Ohta
- Department of Translational Oncology, St. Marianna University Graduate School of Medicine, Kawasaki, 216-8511, Japan.
| |
Collapse
|
10
|
Hou P, Zhu H, Chu F, Gao Y, Sun X, Zhang F, Wang X, Feng Y, Li X, Liu Y, Wang J, Wang X, He DC, Wang H, He H. Phenazine biosynthesis-like domain-containing protein (PBLD) and Cedrelone promote antiviral immune response by activating NF-ĸB. Nat Commun 2025; 16:496. [PMID: 39779683 PMCID: PMC11711403 DOI: 10.1038/s41467-024-54882-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 11/21/2024] [Indexed: 01/30/2025] Open
Abstract
Phenazine biosynthesis-like domain-containing protein (PBLD) and Cedrelone have been identified as tumor suppressors. However, their roles in virus infection remain unclear. Here, we demonstrate that PBLD upregulates the type I interferon (IFN-I) response through activating NF-kappaB (NF-κB) signaling pathway to resist viral infection in cells and mice. Mechanistically, PBLD activates NF-κB signaling pathway during viral infection via blocking tripartite motif containing 21 (TRIM21)-mediated phosphorylated inhibitory kappa B kinase beta (IKKβ) degradation. Furthermore, we show Cedrelone inhibits viral replication by increasing the PBLD protein expression and subsequently activating NF-κB-mediated IFN-I response. Furthermore, the therapeutic potential of Cedrelone lies in its ability to enhance antiviral immunity in primary macrophages and to promote survival and reduce lung tissue damage in HSV-1-infected mice in a PBLD-dependent manner. Consequently, our findings provide a potential combination model that targets PBLD for Cedrelone antiviral drug therapy, potentially paving the way for the development of broad-spectrum antiviral agents.
Collapse
Affiliation(s)
- Peili Hou
- Ruminant Diseases Research Center, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Taian, Shandong, China
| | - Hongchao Zhu
- Ruminant Diseases Research Center, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
| | - Fengyun Chu
- Ruminant Diseases Research Center, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
| | - Yan Gao
- Ruminant Diseases Research Center, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
| | - Xiaonan Sun
- Ruminant Diseases Research Center, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
| | - Fuzhen Zhang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Taian, Shandong, China
| | - Xiaomeng Wang
- Ruminant Diseases Research Center, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
| | - Yueyue Feng
- Ruminant Diseases Research Center, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
| | - Xingyu Li
- Ruminant Diseases Research Center, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
| | - Yu Liu
- Ruminant Diseases Research Center, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
| | - Jun Wang
- Ruminant Diseases Research Center, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
| | - Xiaoyun Wang
- Ruminant Diseases Research Center, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
| | - Daniel Chang He
- The College of Arts and Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Hongmei Wang
- Ruminant Diseases Research Center, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China.
| | - Hongbin He
- Ruminant Diseases Research Center, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China.
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Taian, Shandong, China.
| |
Collapse
|
11
|
Wang B, Han J, Elisseeff JH, Demaria M. The senescence-associated secretory phenotype and its physiological and pathological implications. Nat Rev Mol Cell Biol 2024; 25:958-978. [PMID: 38654098 DOI: 10.1038/s41580-024-00727-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/21/2024] [Indexed: 04/25/2024]
Abstract
Cellular senescence is a state of terminal growth arrest associated with the upregulation of different cell cycle inhibitors, mainly p16 and p21, structural and metabolic alterations, chronic DNA damage responses, and a hypersecretory state known as the senescence-associated secretory phenotype (SASP). The SASP is the major mediator of the paracrine effects of senescent cells in their tissue microenvironment and of various local and systemic biological functions. In this Review, we discuss the composition, dynamics and heterogeneity of the SASP as well as the mechanisms underlying its induction and regulation. We describe the various biological properties of the SASP, its beneficial and detrimental effects in different physiological and pathological settings, and its impact on overall health span. Finally, we discuss the use of the SASP as a biomarker and of SASP inhibitors as senomorphic interventions to treat cancer and other age-related conditions.
Collapse
Affiliation(s)
- Boshi Wang
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen (UMCG), University of Groningen (RUG), Groningen, Netherlands
| | - Jin Han
- Translational Tissue Engineering Center, Wilmer Eye Institute, and Department of Biomedical Engineering, John Hopkins University School of Medicine, Baltimore MD, MD, USA
| | - Jennifer H Elisseeff
- Translational Tissue Engineering Center, Wilmer Eye Institute, and Department of Biomedical Engineering, John Hopkins University School of Medicine, Baltimore MD, MD, USA
| | - Marco Demaria
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen (UMCG), University of Groningen (RUG), Groningen, Netherlands.
| |
Collapse
|
12
|
Kment J, Newsted D, Young S, Vermeulen MC, Laight BJ, Greer PA, Lan Y, Craig AW. Blockade of TGF-β and PD-L1 by bintrafusp alfa promotes survival in preclinical ovarian cancer models by promoting T effector and NK cell responses. Br J Cancer 2024; 130:2003-2015. [PMID: 38622286 PMCID: PMC11183086 DOI: 10.1038/s41416-024-02677-9] [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: 10/03/2023] [Revised: 03/27/2024] [Accepted: 04/02/2024] [Indexed: 04/17/2024] Open
Abstract
BACKGROUND Failure of immunotherapy in high-grade serous ovarian cancer (HGSC) may be due to high levels of transforming growth factor-β (TGF-β) in ascites or tumour immune microenvironment (TIME). Here, we test whether coordinated blockade of TGF-β and PD-L1 with bintrafusp alfa (BA) can provoke anti-tumour immune responses in preclinical HGSC models. METHODS BA is a first-in-class bifunctional inhibitor of TGF-β and PD-L1, and was tested for effects on overall survival and altered TIME in syngeneic HGSC models. RESULTS Using a mouse ID8-derived HGSC syngeneic model with IFNγ-inducible PD-L1 expression, BA treatments significantly reduced ascites development and tumour burden. BA treatments depleted TGF-β and VEGF in ascites, and skewed the TIME towards cytotoxicity compared to control. In the BR5 HGSC syngeneic model, BA treatments increased tumour-infiltrating CD8 T cells with effector memory and cytotoxic markers, as well as cytolytic NK cells. Extended BA treatments in the BR5 model produced ∼50% BA-cured mice that were protected from re-challenge. These BA-cured mice had increased peritoneal T-effector memory and NK cells compared to controls. CONCLUSIONS Our preclinical studies of BA in advanced ovarian cancer models support further testing of BA as an improved immunotherapy option for patients with advanced ovarian cancer.
Collapse
Affiliation(s)
- Jacob Kment
- Cancer Biology & Genetics division, Queen's Cancer Research Institute, Kingston, ON, Canada
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Daniel Newsted
- Cancer Biology & Genetics division, Queen's Cancer Research Institute, Kingston, ON, Canada
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Stephanie Young
- Cancer Biology & Genetics division, Queen's Cancer Research Institute, Kingston, ON, Canada
| | - Michael C Vermeulen
- Cancer Biology & Genetics division, Queen's Cancer Research Institute, Kingston, ON, Canada
| | - Brian J Laight
- Cancer Biology & Genetics division, Queen's Cancer Research Institute, Kingston, ON, Canada
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| | - Peter A Greer
- Cancer Biology & Genetics division, Queen's Cancer Research Institute, Kingston, ON, Canada
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| | - Yan Lan
- EMD Serono Research & Development Institute, Inc., Billerica, MA, USA
| | - Andrew W Craig
- Cancer Biology & Genetics division, Queen's Cancer Research Institute, Kingston, ON, Canada.
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada.
| |
Collapse
|
13
|
Wang B, Sun D, Li H, Chen J. A bird's eye view of the potential role of NFKBIA in pan-cancer. Heliyon 2024; 10:e31204. [PMID: 38813139 PMCID: PMC11133827 DOI: 10.1016/j.heliyon.2024.e31204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 05/07/2024] [Accepted: 05/13/2024] [Indexed: 05/31/2024] Open
Abstract
In the 21st century, cancer remains a serious threat to people's health and has become a prominent public health problem. NFKBIA is involved in the pathological process of many diseases including cancer, but its specific role in pan-cancer has not yet been fully elucidated. This study aims to deepen the understanding of cancer pathology by analyzing the potential functions of NFKBIA in pan-cancer. We used TCGA data to analyze differences of expression of NFKBIA in pan-cancer. We explored the prognostic value, clinical relevance, immune relevance, potential biological function, and diagnosis and treatment value of NFKBIA in pan-cancer through bioinformatics analysis. This study found that in pan-cancer, NFKBIA exhibits differences in expression, which correlate with the prognosis, diagnosis, treatment value and clinical and immune parameters. We have identified that Aspirin, Astaxanthin and Bardoxolone methyl are expected to play a potential therapeutic role in pan-cancer. The results of this study will help to improve our understanding of the role and potential mechanism of NFKBIA in cancer pathology, which may provide guidance for cancer-related research and clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Bin Wang
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Difang Sun
- Department of Ophthalmology, Qingdao Eye Hospital of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao, China
| | - Haifeng Li
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Jinli Chen
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| |
Collapse
|
14
|
Tao W, Yu Z, Han JDJ. Single-cell senescence identification reveals senescence heterogeneity, trajectory, and modulators. Cell Metab 2024; 36:1126-1143.e5. [PMID: 38604170 DOI: 10.1016/j.cmet.2024.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 12/15/2023] [Accepted: 03/13/2024] [Indexed: 04/13/2024]
Abstract
Cellular senescence underlies many aging-related pathologies, but its heterogeneity poses challenges for studying and targeting senescent cells. We present here a machine learning program senescent cell identification (SenCID), which accurately identifies senescent cells in both bulk and single-cell transcriptome. Trained on 602 samples from 52 senescence transcriptome datasets spanning 30 cell types, SenCID identifies six major senescence identities (SIDs). Different SIDs exhibit different senescence baselines, stemness, gene functions, and responses to senolytics. SenCID enables the reconstruction of senescent trajectories under normal aging, chronic diseases, and COVID-19. Additionally, when applied to single-cell Perturb-seq data, SenCID helps reveal a hierarchy of senescence modulators. Overall, SenCID is an essential tool for precise single-cell analysis of cellular senescence, enabling targeted interventions against senescent cells.
Collapse
Affiliation(s)
- Wanyu Tao
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Center for Quantitative Biology (CQB), Peking University, Beijing, China
| | - Zhengqing Yu
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Center for Quantitative Biology (CQB), Peking University, Beijing, China
| | - Jing-Dong J Han
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Center for Quantitative Biology (CQB), Peking University, Beijing, China; Peking University Chengdu Academy for Advanced Interdisciplinary Biotechnologies, Chengdu, China.
| |
Collapse
|
15
|
Ma Y, Liu Y, Luo D, Guo Z, Xiang H, Chen B, Wu X. Identification of biomarkers and immune infiltration characterization of lipid metabolism-associated genes in osteoarthritis based on machine learning algorithms. Aging (Albany NY) 2024; 16:7043-7059. [PMID: 38637111 PMCID: PMC11087088 DOI: 10.18632/aging.205740] [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: 11/24/2023] [Accepted: 03/18/2024] [Indexed: 04/20/2024]
Abstract
Osteoarthritis (OA) is a prevalent degenerative condition commonly observed in the elderly, leading to consequential disability. Despite notable advancements made in clinical strategies for OA, its pathogenesis remains uncertain. The intricate association between OA and metabolic processes has yet to receive comprehensive exploration. In our investigation, we leveraged public databases and applied machine learning algorithms, including WGCNA, LASSO, RF, immune infiltration analysis, and pathway enrichment analysis, to scrutinize the role of lipid metabolism-associated genes (LAGs) in the OA. Our findings identified three distinct biomarkers, and evaluated their expression to assess their diagnostic value in the OA patients. The exploration of immune infiltration in these patients revealed an intricate relationship between immune cells and the identified biomarkers. In addition, in vitro experiments, including qRT-PCR, Western blot, chondrocyte lipid droplets detection and mitochondrial fatty acid oxidation measurement, further verified abnormal expressions of selected LAGs in OA cartilage and confirmed the correlation between lipid metabolism and OA.
Collapse
Affiliation(s)
- Yuanye Ma
- Department of Orthopedics, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266003, China
| | - Yang Liu
- Department of Orthopedics, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266003, China
| | - Dan Luo
- Department of Pathology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, China
| | - Zhu Guo
- Department of Orthopedics, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266003, China
| | - Hongfei Xiang
- Department of Orthopedics, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266003, China
| | - Bohua Chen
- Department of Orthopedics, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266003, China
| | - Xiaolin Wu
- Department of Orthopedics, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266003, China
- Cancer Institute, Qingdao University, Qingdao 266071, China
| |
Collapse
|
16
|
Reimann M, Lee S, Schmitt CA. Cellular senescence: Neither irreversible nor reversible. J Exp Med 2024; 221:e20232136. [PMID: 38385946 PMCID: PMC10883852 DOI: 10.1084/jem.20232136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 01/10/2024] [Accepted: 02/06/2024] [Indexed: 02/23/2024] Open
Abstract
Cellular senescence is a critical stress response program implicated in embryonic development, wound healing, aging, and immunity, and it backs up apoptosis as an ultimate cell-cycle exit mechanism. In analogy to replicative exhaustion of telomere-eroded cells, premature types of senescence-referring to oncogene-, therapy-, or virus-induced senescence-are widely considered irreversible growth arrest states as well. We discuss here that entry into full-featured senescence is not necessarily a permanent endpoint, but dependent on essential maintenance components, potentially transient. Unlike a binary state switch, we view senescence with its extensive epigenomic reorganization, profound cytomorphological remodeling, and distinctive metabolic rewiring rather as a journey toward a full-featured arrest condition of variable strength and depth. Senescence-underlying maintenance-essential molecular mechanisms may allow cell-cycle reentry if not continuously provided. Importantly, senescent cells that resumed proliferation fundamentally differ from those that never entered senescence, and hence would not reflect a reversion but a dynamic progression to a post-senescent state that comes with distinct functional and clinically relevant ramifications.
Collapse
Affiliation(s)
- Maurice Reimann
- Medical Department of Hematology, Oncology and Tumor Immunology, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, and Molekulares Krebsforschungszentrum-MKFZ, Campus Virchow Klinikum, Charité-Universitätsmedizin, Berlin, Germany
| | - Soyoung Lee
- Medical Department of Hematology, Oncology and Tumor Immunology, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, and Molekulares Krebsforschungszentrum-MKFZ, Campus Virchow Klinikum, Charité-Universitätsmedizin, Berlin, Germany
- Johannes Kepler University , Linz, Austria
| | - Clemens A Schmitt
- Medical Department of Hematology, Oncology and Tumor Immunology, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, and Molekulares Krebsforschungszentrum-MKFZ, Campus Virchow Klinikum, Charité-Universitätsmedizin, Berlin, Germany
- Johannes Kepler University , Linz, Austria
- Department of Hematology and Oncology, Kepler University Hospital, Linz, Austria
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association , Berlin, Germany
| |
Collapse
|
17
|
Xiao HH, Zhang FR, Li S, Guo FF, Hou JL, Wang SC, Yu J, Li XY, Yang HJ. Xinshubao tablet rescues cognitive dysfunction in a mouse model of vascular dementia: Involvement of neurogenesis and neuroinflammation. Biomed Pharmacother 2024; 172:116219. [PMID: 38310654 DOI: 10.1016/j.biopha.2024.116219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 02/06/2024] Open
Abstract
Vascular dementia (VaD) represents a severe cognitive dysfunction syndrome closed linked to cardiovascular function. In the present study, we assessed the potential of Xinshubao tablet (XSB), a traditional Chinese prescription widely used for cardiovascular diseases, to mitigate neuropathological damage in a mouse model of VaD and elucidated the underlying mechanisms. Our findings revealed that oral administration of XSB rescued the cardiac dysfunction resulting from bilateral common carotid artery stenosis (BCAS), improved the cerebral blood flow (CBF) and cognitive function, reduced white matter injury, inhibited excessive microglial and astrocytic activation, stimulated hippocampal neurogenesis, and reduced neural apoptosis in the brains of BCAS mice. Mechanistically, RNA-seq analysis indicated that XSB treatment was significantly associated with neuroinflammation, vasculature development, and synaptic transmission, which were further confirmed by q-PCR assays. Western blot results revealed that XSB treatment hindered the nuclear translocation of nuclear factor-κB (NF-κB), thereby suppressing the NF-κB signaling pathway. These results collectively demonstrated that XSB could ameliorate cognitive dysfunction caused by BCAS through regulating CBF, reducing white matter lesions, suppressing glial activation, promoting neurogenesis, and mitigating neuroinflammation. Notably, the NF-κB signaling pathway emerged as a pivotal player in this mechanism.
Collapse
Affiliation(s)
- Hong-He Xiao
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China; School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China; Fujian Pien Tze Huang Enterprise Key Laboratory of Natural Medicine Research and Development, Zhangzhou Pien Tze Huang Pharmaceutical Co., Ltd., Zhangzhou, Fujian Province 363099, China
| | - Feng-Rong Zhang
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China; Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Sen Li
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Fei-Fei Guo
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Jin-Li Hou
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Shi-Cong Wang
- Fujian Pien Tze Huang Enterprise Key Laboratory of Natural Medicine Research and Development, Zhangzhou Pien Tze Huang Pharmaceutical Co., Ltd., Zhangzhou, Fujian Province 363099, China
| | - Juan Yu
- Fujian Pien Tze Huang Enterprise Key Laboratory of Natural Medicine Research and Development, Zhangzhou Pien Tze Huang Pharmaceutical Co., Ltd., Zhangzhou, Fujian Province 363099, China.
| | - Xian-Yu Li
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Hong-Jun Yang
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China; China Academy of Chinese Medical Sciences, Beijing 100700, China.
| |
Collapse
|
18
|
Harasawa A, Ishiyama S, Mochizuki K. Fructo-oligosaccharide-mediated alteration in claudin expression in small intestinal absorptive Caco-2 cells is positively associated with the induction of inflammatory genes and the glucan receptor gene CLEC7A. Nutrition 2023; 115:112140. [PMID: 37481839 DOI: 10.1016/j.nut.2023.112140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 06/02/2023] [Accepted: 06/18/2023] [Indexed: 07/25/2023]
Abstract
OBJECTIVES Indigestible carbohydrates may strengthen tight junctions (TJs) independently of intestinal bacteria. This study investigated whether indigestible carbohydrates (i.e., fructo-oligosaccharides [FOS]) promote TJs directly to intestinal absorptive Caco-2 cells and examined the association between the expression of genes constructing TJs and other genes using mRNA microarray analysis. METHODS Caco-2 cells at 1.0 × 105/mL were seeded in a type I collagen plate and cultured in high-glucose Dulbecco's modified Eagle medium (DMEM) supplemented with 10% fetal calf serum (FCS); the cells reached confluence at 7 d after seeding. Ten days after the cells reached confluency, they were cultured for 24 h in 10% FCS-containing DMEM medium supplemented with 0%, 5%, or 10% FOS. We performed mRNA microarray to identify the genes whose expression was altered by FOS. Subsequently, quantitative reverse transcription polymerase chain reaction was performed for these altered genes, including CLEC7A encoding the glucan receptor, and for the claudin (CLDN) family genes. The expression of CLDN2, CLDN4, and CLEC7A proteins was assessed using western blot analysis. RESULTS FOS decreased the mRNA and protein expression of CLDN2, which weakens TJs, and increased those of CLDN4, which strengthens TJs, in Caco-2 cells. FOS treatment (10%) reduced the mRNA expression of antioxidative genes and induced the expression of immune response-related genes, including CLEC7A, CCL2, and ITGA2. Furthermore, the expression of CLEC7A protein was enhanced by FOS. CONCLUSIONS Induction of TJ-strengthening CLDN4 and reduction of TJ-weakening CLDN2 by FOS treatment in small intestinal absorptive Caco-2 cells is positively associated with the induction of inflammatory genes, including CLEC7A.
Collapse
Affiliation(s)
- Aya Harasawa
- Department of Integrated Applied Life Science, Integrated Graduate School of Medicine, Engineering, and Agricultural Sciences, University of Yamanashi, Kofu, Japan
| | - Shiori Ishiyama
- Laboratory of Food and Nutritional Sciences, Department of Local Produce and Food Sciences, Faculty of Life and Environmental Sciences, University of Yamanashi, Kofu, Japan
| | - Kazuki Mochizuki
- Laboratory of Food and Nutritional Sciences, Department of Local Produce and Food Sciences, Faculty of Life and Environmental Sciences, University of Yamanashi, Kofu, Japan.
| |
Collapse
|
19
|
Mucka P, Lindemann P, Bosco B, Willenbrock M, Radetzki S, Neuenschwander M, Brischetto C, Peter von Kries J, Nazaré M, Scheidereit C. CLK2 and CLK4 are regulators of DNA damage-induced NF-κB targeted by novel small molecule inhibitors. Cell Chem Biol 2023; 30:1303-1312.e3. [PMID: 37506701 DOI: 10.1016/j.chembiol.2023.06.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 04/20/2023] [Accepted: 06/29/2023] [Indexed: 07/30/2023]
Abstract
Transcription factor NF-κB potently activates anti-apoptotic genes, and its inactivation significantly reduces tumor cell survival following genotoxic stresses. We identified two structurally distinct lead compounds that selectively inhibit NF-κB activation by DNA double-strand breaks, but not by other stimuli, such as TNFα. Our compounds do not directly inhibit previously identified regulators of this pathway, most critically including IκB kinase (IKK), but inhibit signal transmission in-between ATM, PARP1, and IKKγ. Deconvolution strategies, including derivatization and in vitro testing in multi-kinase panels, yielded shared targets, cdc-like kinase (CLK) 2 and 4, as essential regulators of DNA damage-induced IKK and NF-κB activity. Both leads sensitize to DNA damaging agents by increasing p53-induced apoptosis, thereby reducing cancer cell viability. We propose that our lead compounds and derivatives can be used in context of genotoxic therapy-induced or ongoing DNA damage to increase tumor cell apoptosis, which may be beneficial in cancer treatment.
Collapse
Affiliation(s)
- Patrick Mucka
- Laboratory of Signal Transduction in Tumor Cells, Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| | - Peter Lindemann
- Laboratory of Medicinal Chemistry, Leibniz Forschungsinstitut für Molekulare Pharmakologie, 13125 Berlin, Germany
| | - Bartolomeo Bosco
- Laboratory of Signal Transduction in Tumor Cells, Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| | - Michael Willenbrock
- Laboratory of Signal Transduction in Tumor Cells, Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| | - Silke Radetzki
- Screening Unit, Leibniz Forschungsinstitut für Molekulare Pharmakologie, 13125 Berlin, Germany
| | - Martin Neuenschwander
- Screening Unit, Leibniz Forschungsinstitut für Molekulare Pharmakologie, 13125 Berlin, Germany
| | - Cristina Brischetto
- Laboratory of Signal Transduction in Tumor Cells, Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| | - Jens Peter von Kries
- Screening Unit, Leibniz Forschungsinstitut für Molekulare Pharmakologie, 13125 Berlin, Germany
| | - Marc Nazaré
- Laboratory of Medicinal Chemistry, Leibniz Forschungsinstitut für Molekulare Pharmakologie, 13125 Berlin, Germany.
| | - Claus Scheidereit
- Laboratory of Signal Transduction in Tumor Cells, Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany.
| |
Collapse
|
20
|
Keenum MC, Chatterjee P, Atalis A, Pandey B, Jimenez A, Roy K. Single-cell epitope-transcriptomics reveal lung stromal and immune cell response kinetics to nanoparticle-delivered RIG-I and TLR4 agonists. Biomaterials 2023; 297:122097. [PMID: 37001347 PMCID: PMC10192313 DOI: 10.1016/j.biomaterials.2023.122097] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 03/14/2023] [Accepted: 03/20/2023] [Indexed: 03/31/2023]
Abstract
Lung-resident and circulatory lymphoid, myeloid, and stromal cells, expressing various pattern recognition receptors (PRRs), detect pathogen- and danger-associated molecular patterns (PAMPs/DAMPs), and defend against respiratory pathogens and injuries. Here, we report the early responses of murine lungs to nanoparticle-delivered PAMPs, specifically the retinoic acid-inducible gene I (RIG-I) agonist poly-U/UC (PUUC), with or without the TLR4 agonist monophosphoryl lipid A (MPLA). Using cellular indexing of transcriptomes and epitopes by sequencing (CITE-seq), we characterized the responses at 4 and 24 h after intranasal administration. Within 4 h, ribosome-associated transcripts decreased in both stromal and immune cells, followed by widespread interferon-stimulated gene (ISG) expression. Using RNA velocity, we show that lung-neutrophils dynamically regulate the synthesis of cytokines like CXCL-10, IL-1α, and IL-1β. Co-delivery of MPLA and PUUC increased chemokine synthesis and upregulated antimicrobial binding proteins targeting iron, manganese, and zinc in many cell types, including fibroblasts, endothelial cells, and epithelial cells. Overall, our results elucidate the early PAMP-induced cellular responses in the lung and demonstrate that stimulation of the RIG-I pathway, with or without TLR4 agonists, induces a ubiquitous microbial defense state in lung stromal and immune cells. Nanoparticle-delivered combination PAMPs may have applications in intranasal antiviral and antimicrobial therapies and prophylaxis.
Collapse
Affiliation(s)
- M Cole Keenum
- Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology and Emory University Atlanta, GA, USA
| | - Paramita Chatterjee
- Marcus Center for Therapeutic Cell Characterization and Manufacturing Georgia Institute of Technology, Atlanta, GA, USA
| | - Alexandra Atalis
- Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology and Emory University Atlanta, GA, USA
| | - Bhawana Pandey
- Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology and Emory University Atlanta, GA, USA
| | - Angela Jimenez
- Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology and Emory University Atlanta, GA, USA
| | - Krishnendu Roy
- Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology and Emory University Atlanta, GA, USA; Marcus Center for Therapeutic Cell Characterization and Manufacturing Georgia Institute of Technology, Atlanta, GA, USA; The Parker H. Petit Institute for Bioengineering and Biosciences Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
21
|
Koslow M, Zhu P, McCabe C, Xu X, Lin X. Kidney transcriptome and cystic kidney disease genes in zebrafish. Front Physiol 2023; 14:1184025. [PMID: 37256068 PMCID: PMC10226271 DOI: 10.3389/fphys.2023.1184025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 04/20/2023] [Indexed: 06/01/2023] Open
Abstract
Introduction: Polycystic kidney disease (PKD) is a condition where fluid filled cysts form on the kidney which leads to overall renal failure. Zebrafish has been recently adapted to study polycystic kidney disease, because of its powerful embryology and genetics. However, there are concerns on the conservation of this lower vertebrate in modeling polycystic kidney disease. Methods: Here, we aim to assess the molecular conservation of zebrafish by searching homologues polycystic kidney disease genes and carrying transcriptome studies in this animal. Results and Discussion: We found that out of 82 human cystic kidney disease genes, 81 have corresponding zebrafish homologs. While 75 of the genes have a single homologue, only 6 of these genes have two homologs. Comparison of the expression level of the transcripts enabled us to identify one homolog over the other homolog with >70% predominance, which would be prioritized for future experimental studies. Prompted by sexual dimorphism in human and rodent kidneys, we studied transcriptome between different sexes and noted significant differences in male vs. female zebrafish, indicating that sex dimorphism also occurs in zebrafish. Comparison between zebrafish and mouse identified 10% shared genes and 38% shared signaling pathways. String analysis revealed a cluster of genes differentially expressed in male vs. female zebrafish kidneys. In summary, this report demonstrated remarkable molecular conservation, supporting zebrafish as a useful animal model for cystic kidney disease.
Collapse
Affiliation(s)
- Matthew Koslow
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States
| | - Ping Zhu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States
| | - Chantal McCabe
- Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, United States
| | - Xiaolei Xu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
| | - Xueying Lin
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
22
|
Liu H, Yang R, Zhao S, Zhou F, Liu Y, Zhou Z, Chen L, Xie J. Collagen scaffolds derived from bovine skin loaded with MSC optimized M1 macrophages remodeling and chronic diabetic wounds healing. Bioeng Transl Med 2023; 8:e10467. [PMID: 37206210 PMCID: PMC10189465 DOI: 10.1002/btm2.10467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 10/25/2022] [Accepted: 11/23/2022] [Indexed: 12/13/2022] Open
Abstract
Owing to the persistent inflammatory microenvironment and unsubstantial dermal tissues, chronic diabetic wounds do not heal easily and their recurrence rate is high. Therefore, a dermal substitute that can induce rapid tissue regeneration and inhibit scar formation is urgently required to address this concern. In this study, we established biologically active dermal substitutes (BADS) by combining novel animal tissue-derived collagen dermal-replacement scaffolds (CDRS) and bone marrow mesenchymal stem cells (BMSCs) for the healing and recurrence treatments of chronic diabetic wounds. The collagen scaffolds derived from bovine skin (CBS) displayed good physicochemical properties and superior biocompatibility. CBS loaded with BMSCs (CBS-MCSs) could inhibit M1 macrophage polarization in vitro. Decreased MMP-9 and increased Col3 at the protein level were detected in CBS-MSCs-treated M1 macrophages, which may be attributed to the suppression of the TNF-α/NF-κB signaling pathway (downregulating phospho-IKKα/β/total IKKα/β, phospho-IκB/total IκB, and phospho-NFκB/total NFκB) in M1 macrophages. Moreover, CBS-MSCs could benefit the transformation of M1 (downregulating iNOS) to M2 (upregulating CD206) macrophages. Wound-healing evaluations demonstrated that CBS-MSCs regulated the polarization of macrophages and the balance of inflammatory factors (pro-inflammatory: IL-1β, TNF-α, and MMP-9; anti-inflammatory: IL-10 and TGF-β3) in db/db mice. Furthermore, CBS-MSCs facilitated the noncontractile and re-epithelialized processes, granulation tissue regeneration, and neovascularization of chronic diabetic wounds. Thus, CBS-MSCs have a potential value for clinical application in promoting the healing of chronic diabetic wounds and preventing the recurrence of ulcers.
Collapse
Affiliation(s)
- Hengdeng Liu
- Department of Burns, Laboratory of General SurgeryThe First Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Engineering Technology Research Center of Burn and Wound Accurate Diagnosis and Treatment Key Technology and Series of ProductsSun Yat‐Sen UniversityGuangzhouGuangdongChina
- Institute of Precision Medicine, The First Affiliated HospitalSun Yat‐Sen UniversityGuangzhouGuangdongChina
| | - Ronghua Yang
- Department of Burn and Plastic SurgeryGuangzhou First People's Hospital, South China University of TechnologyGuangzhouGuangdongChina
| | - Shixin Zhao
- Department of Burns, Laboratory of General SurgeryThe First Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Engineering Technology Research Center of Burn and Wound Accurate Diagnosis and Treatment Key Technology and Series of ProductsSun Yat‐Sen UniversityGuangzhouGuangdongChina
- Institute of Precision Medicine, The First Affiliated HospitalSun Yat‐Sen UniversityGuangzhouGuangdongChina
| | - Fei Zhou
- Department of Burns, Laboratory of General SurgeryThe First Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Engineering Technology Research Center of Burn and Wound Accurate Diagnosis and Treatment Key Technology and Series of ProductsSun Yat‐Sen UniversityGuangzhouGuangdongChina
- Institute of Precision Medicine, The First Affiliated HospitalSun Yat‐Sen UniversityGuangzhouGuangdongChina
| | - Yiling Liu
- Department of Burns, Laboratory of General SurgeryThe First Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Engineering Technology Research Center of Burn and Wound Accurate Diagnosis and Treatment Key Technology and Series of ProductsSun Yat‐Sen UniversityGuangzhouGuangdongChina
- Institute of Precision Medicine, The First Affiliated HospitalSun Yat‐Sen UniversityGuangzhouGuangdongChina
| | - Ziheng Zhou
- Department of Burns, Laboratory of General SurgeryThe First Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Engineering Technology Research Center of Burn and Wound Accurate Diagnosis and Treatment Key Technology and Series of ProductsSun Yat‐Sen UniversityGuangzhouGuangdongChina
- Institute of Precision Medicine, The First Affiliated HospitalSun Yat‐Sen UniversityGuangzhouGuangdongChina
| | - Lei Chen
- Department of Burns, Laboratory of General SurgeryThe First Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Engineering Technology Research Center of Burn and Wound Accurate Diagnosis and Treatment Key Technology and Series of ProductsSun Yat‐Sen UniversityGuangzhouGuangdongChina
- Institute of Precision Medicine, The First Affiliated HospitalSun Yat‐Sen UniversityGuangzhouGuangdongChina
| | - Julin Xie
- Department of Burns, Laboratory of General SurgeryThe First Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Engineering Technology Research Center of Burn and Wound Accurate Diagnosis and Treatment Key Technology and Series of ProductsSun Yat‐Sen UniversityGuangzhouGuangdongChina
- Institute of Precision Medicine, The First Affiliated HospitalSun Yat‐Sen UniversityGuangzhouGuangdongChina
| |
Collapse
|
23
|
Zhang Y, Zhu M, Pan J, Qiu Q, Tong X, Hu X, Gong C. BmCPV replication is suppressed by the activation of the NF-κB/autophagy pathway through the interaction of vsp21 translated by vcircRNA_000048 with ubiquitin carboxyl-terminal hydrolase. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2023; 156:103947. [PMID: 37086910 DOI: 10.1016/j.ibmb.2023.103947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 04/16/2023] [Accepted: 04/16/2023] [Indexed: 05/03/2023]
Abstract
Bombyx mori cypovirus (BmCPV), a typical double-stranded RNA virus, was demonstrated to generate a viral circRNA, vcircRNA_000048, which encodes a vsp21 with 21 amino acid residues to suppress viral replication. However, the regulatory mechanism of vsp21 on virus infection remained unclear. This study discovered that vsp21 induces reactive oxygen species (ROS) generation, activates autophagy, and attenuates virus replication by inducing autophagy. Then we confirmed that the effect of vsp21-induced autophagy on viral replication was attributed to the activation of the NF-κB signaling pathway. Furthermore, we clarified that vsp21 interacted with ubiquitin carboxyl-terminal hydrolase (UCH) and that ubiquitination and degradation of phospho-IκB-α were enhanced by vsp21 via competitive binding to UCH. Finally, we validated that vsp21 activates the NF-κB/autophagy pathway to suppress viral replication by interacting with UCH. These findings provided new insights into regulating viral multiplication and reovirus-host interaction.
Collapse
Affiliation(s)
- Yunshan Zhang
- School of Biology & Basic Medical Science, Soochow University, Suzhou, 215123, China
| | - Min Zhu
- School of Biology & Basic Medical Science, Soochow University, Suzhou, 215123, China
| | - Jun Pan
- School of Biology & Basic Medical Science, Soochow University, Suzhou, 215123, China
| | - Qunnan Qiu
- School of Biology & Basic Medical Science, Soochow University, Suzhou, 215123, China
| | - Xinyu Tong
- School of Biology & Basic Medical Science, Soochow University, Suzhou, 215123, China
| | - Xiaolong Hu
- School of Biology & Basic Medical Science, Soochow University, Suzhou, 215123, China; Institute of Agricultural Biotechnology and Ecological Research, Soochow University, Suzhou, 215123, China
| | - Chengliang Gong
- School of Biology & Basic Medical Science, Soochow University, Suzhou, 215123, China; Institute of Agricultural Biotechnology and Ecological Research, Soochow University, Suzhou, 215123, China.
| |
Collapse
|
24
|
Yang X, Guo T, Du Z, Qin X, Wang K, Kebreab E, Wang D, Lyu L. Protective effects of MNQ against Lipopolysaccharide-induced inflammatory damage in bovine ovarian follicular granulosa cells in Vitro. J Steroid Biochem Mol Biol 2023; 230:106274. [PMID: 36813140 DOI: 10.1016/j.jsbmb.2023.106274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 02/18/2023] [Accepted: 02/19/2023] [Indexed: 02/23/2023]
Abstract
Inflammation of the reproductive tract in dairy cows lead to functional disorders of follicular granulosa cells (GCs) in mammalian ovaries resulting in infertility and serious losses to the livestock industry. Lipopolysaccharide (LPS) can induce an inflammatory response in follicular granulosa cells in vitro. The aim of this study was to investigate the cellular regulatory mechanism of MNQ (2-methoxy-1,4-naphthoquinone) on eliminating the inflammatory response and restoring normal functions for bovine ovarian follicular GCs cultured in vitro exposed to LPS. The cytotoxicity of MNQ and LPS on GCs were detected by MTT method to determine the safe concentration. The relative expression of inflammatory factors and steroid synthesis-related genes were detected by qRT-PCR. The concentration of steroid hormones in the culture broth were detected by ELISA. Differential gene expressions were analyzed by RNA-seq. There were no toxic effects on GCs at MNQ and LPS concentrations of less than 3 µM and 10 µg/mL, respectively and treated in 12 h. The relative expressions of IL-6, IL-1β and TNF-α were significantly higher in the LPS group compared with the CK group when GCs cultured in vitro were treated with the above concentrations and times (P < 0.05), but significantly lower in the MNQ+LPS group compared with the LPS group (P < 0.05). The levels of E2 and P4 in the culture solution were significantly reduced in the LPS group compared to the CK group (P < 0.05), and restored in the MNQ+LPS group. The relative expressions of CYP19A1, CYP11A1, 3β-HSD, and STAR were significantly decreased in the LPS group compared with the CK group (P < 0.05), while the MNQ+LPS group also recovered to some extent. There were 407 differential genes shared by LPS vs CK and MNQ+LPS vs LPS by RNA-seq analysis, which were mainly enriched in steroid biosynthesis and TNF signaling pathway. We screened 10 genes for analysis and found consistent results for RNA-seq and qRT-PCR. In this study, we confirmed the protective effect of MNQ, an extract from Impatiens balsamina L, on LPS-induced inflammatory responses in bovine follicular granulosa cells in vitro as well as functional damage, and acted through steroid biosynthesis and TNF signaling pathways.
Collapse
Affiliation(s)
- Xiaofeng Yang
- College of Animal Science, Shanxi Agricultural University, Taigu, Shanxi 030801, China; Department of Biology, Xinzhou Normal University, Xinzhou, Shanxi 034000, China
| | - Tong Guo
- Department of Animal Husbandry and Veterinary Medicine, Beijing Vocational College of Agriculture, Beijing 102442, China
| | - Zhangsheng Du
- College of Animal Science, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - Xiaowei Qin
- College of Animal Science, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - Kai Wang
- College of Animal Science, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - Ermias Kebreab
- Department of Animal Science, University of California Davis, Davis, CA 95616, USA
| | - Dong Wang
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Lihua Lyu
- College of Animal Science, Shanxi Agricultural University, Taigu, Shanxi 030801, China.
| |
Collapse
|
25
|
Transcriptome Analysis Reveals Effect of Dietary Probiotics on Immune Response Mechanism in Southern Catfish ( Silurus meridionalis) in Response to Plesiomonas shigelloides. Animals (Basel) 2023; 13:ani13030449. [PMID: 36766339 PMCID: PMC9913393 DOI: 10.3390/ani13030449] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 01/19/2023] [Accepted: 01/25/2023] [Indexed: 01/31/2023] Open
Abstract
To explore whether a probiotic complex composed of Lactobacillus rhamnosus, Lactobacillus plantarum, and Lactobacillus casei can prevent or inhibit the inflammatory response caused by the invasion of Plesiomonas shigelloides in the southern catfish, we screened differentially expressed genes and enriched inflammation-related pathways among a control and three experimental groups and conducted analysis by transcriptome sequencing after a 21-day breeding experiment. Compared with those in the PS (Plesiomonas shigelloides) group, southern catfish in the L-PS (Lactobacillus-Plesiomonas shigelloides) group had no obvious haemorrhages or ulcerations. The results also showed that inflammation-related genes, such as mmp9, cxcr4, nfkbia, socs3, il-8, pigr, tlr5, and tnfr1, were significantly upregulated in the PS group compared with those in the L-PS groups. In addition, we verified six DEGs (mmp9, cxcr4, nfkbia, socs3, rbp2, and calr) and three proteins (CXCR4, NFKBIA, and CALR) by qRT-PCR and ELISA, respectively. Our results were consistent with the transcriptome data. Moreover, significantly downregulated genes (p < 0.05) were enriched in inflammation-related GO terms (lymphocyte chemotaxis and positive regulation of inflammatory response) and immune-related pathways (intestinal immune network for IgA production and IL-17 signalling pathway) in the L-PS vs. the PS group. Our results indicate that the infection of P. shigelloides can produce an inflammatory response, and probiotics could inhibit the inflammatory response caused by P. shigelloides to some extent.
Collapse
|
26
|
Li Z, Huang Z, Luo Y, Yang H, Yang M. DUSP9 alleviates hepatic ischemia/reperfusion injury by restraining both mitogen-activated protein kinase and IKK in an apoptosis signal-regulating kinase 1-dependent manner. Acta Biochim Biophys Sin (Shanghai) 2022; 54:1811-1821. [PMID: 36789693 PMCID: PMC10157530 DOI: 10.3724/abbs.2022183] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 06/06/2022] [Indexed: 12/03/2022] Open
Abstract
Hepatic ischemia/reperfusion (I/R) injury occurs frequently in various liver operations and diseases, but its effective treatment remains inadequate because the key switch that leads to hepatic explosive inflammation has not been well disclosed. Dual specificity phosphatase 9 (DUSP9) is widely involved in the innate immune response of solid organs and is sometimes regulated by ubiquitin. In the present study, we find that DUSP9 is reduced in mouse hepatic I/R injury. DUSP9 enrichment attenuates hepatic inflammation both in vivo and in vitro as revealed by western blot analysis and qRT-PCR. In contrast, DUSP9 depletion leads to more severe I/R injury. Mechanistically, DUSP9 inhibits the phosphorylation of apoptosis signal-regulating kinase 1 (ASK1) by directly binding to ASK1, thereby decreasing tumor necrosis factor receptor-associated factor 6 (TRAF6), K63 ubiquitin and the phosphorylation of p38/JNK1 instead of ERK1. The present study documents a novel role of DUSP9 in hepatic I/R injury and implies the potential of targeting the DUSP9/ASK1 axis towards mitogen-activated protein kinase and TRAF6/inhibitor of κB kinase pathways.
Collapse
Affiliation(s)
- Zhongtang Li
- College of Basic MedicineChongqing Medical UniversityChongqing400016China
| | - Zuotian Huang
- Department of Hepatobiliary Pancreatic Tumor CenterChongqing University Cancer HospitalChongqing400030China
| | - Yunhai Luo
- The First Affiliated Hospital of Chongqing Medical UniversityChongqing400016
| | - Hang Yang
- The First Affiliated Hospital of Chongqing Medical UniversityChongqing400016
| | - Mei Yang
- College of Basic MedicineChongqing Medical UniversityChongqing400016China
| |
Collapse
|
27
|
Li X, Li H, Wang T, Zhao Y, Shao Y, Sun Y, Zhang Y, Liu Z. Network pharmacology-based analysis of the mechanism of Saposhnikovia divaricata for the treatment of type I allergy. PHARMACEUTICAL BIOLOGY 2022; 60:1224-1236. [PMID: 35760567 PMCID: PMC9246231 DOI: 10.1080/13880209.2022.2086583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 04/11/2022] [Accepted: 06/01/2022] [Indexed: 06/15/2023]
Abstract
CONTEXT Saposhnikovia divaricata (Turcz.) Schischk (Apiaceae) (SD) has various pharmacological activities, but its effects on type I allergy (TIA) have not been comprehensively studied. OBJECTIVE This study evaluates the treatment and molecular mechanisms of SD against TIA. MATERIALS AND METHODS The effective components and action targets of SD were screened using TCMSP database, and allergy-related targets of SD were predicted using GeneCards and OMIM database. The obtained target intersections were imported into David database for GO analysis, and used R software to perform KEGG analysis. The RBL-2H3 cells sensitised by DNP-IgE/DNP-BSA were treated with different concentrations of SD (root decoction, 0.5, 1, and 2 mg/mL), prim-O-glucosylcimifugin (POG, 10, 40, and 80 μg/mL) and the positive control drug-ketotifen fumarate (KF, 30 μM) for 12 h, then subjected to cell degranulation and qPCR analysis. RESULTS Eighteen active compounds of SD and 38 intersection targets were obtained: TIA-related signal pathways mainly include calcium signal pathway, PI3K-Akt signal pathway and MAPK signal pathway. Taking the β-Hex release rate of the model group as the base, the release rate of SD and POG in high dose groups were 43.79% and 57.01%, respectively, which were significantly lower than model group (p < 0.01), and significantly lower than KF group (63.83%, p < 0.01, p < 0.05). SD and POG could down-regulate the expression of related proteins in the Lyn/Syk, PI3K/AKT and MAPK signalling pathways. DISCUSSION AND CONCLUSION Saposhnikovia divaricata could inhibit IgE-induced degranulation of mast cells, providing a scientific basis for further research and clinical applications of SD in TIA treatment.
Collapse
Affiliation(s)
- Xiangsheng Li
- College of Pharmaceutical Sciences, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Institute of Life Science and Green Development, Hebei University, Baoding, China
| | - Hui Li
- Department of Urology, Peking University International Hospital, Beijing, China
| | - Tingting Wang
- College of Pharmaceutical Sciences, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Institute of Life Science and Green Development, Hebei University, Baoding, China
| | - Yang Zhao
- College of Pharmaceutical Sciences, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Institute of Life Science and Green Development, Hebei University, Baoding, China
| | - Yuxin Shao
- College of Pharmaceutical Sciences, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Institute of Life Science and Green Development, Hebei University, Baoding, China
| | - Yizhao Sun
- College of Pharmaceutical Sciences, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Institute of Life Science and Green Development, Hebei University, Baoding, China
| | - Yanfen Zhang
- Technology Transfer Center, Hebei University, Baoding, China
| | - Zhongcheng Liu
- College of Pharmaceutical Sciences, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Institute of Life Science and Green Development, Hebei University, Baoding, China
| |
Collapse
|
28
|
Shen X, Tao J, Wang Z, Li G, Zhang Z, Li J, Diliar A. MiR-7015-3p Targets Nuclear Factor-Kappa-B-Inhibitor Alpha to Aggravate Hypoxia/Reoxygenation Injury in Cardiomyocytes Through the NF-κB Pathway. Int Heart J 2022; 63:881-892. [DOI: 10.1536/ihj.22-036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Xin Shen
- Department of Cardiology, People's Hospital of Xinjiang Uygur Autonomous Region
| | - Jing Tao
- Department of Cardiology, People's Hospital of Xinjiang Uygur Autonomous Region
| | - Zhao Wang
- Department of Cardiology, People's Hospital of Xinjiang Uygur Autonomous Region
| | - Guoqing Li
- Department of Cardiology, People's Hospital of Xinjiang Uygur Autonomous Region
| | - Zilong Zhang
- Department of Cardiology, People's Hospital of Xinjiang Uygur Autonomous Region
| | - Jie Li
- Department of Cardiology, People's Hospital of Xinjiang Uygur Autonomous Region
| | - Adri Diliar
- Department of Cardiology, People's Hospital of Xinjiang Uygur Autonomous Region
| |
Collapse
|
29
|
Tufan AB, Lazarow K, Kolesnichenko M, Sporbert A, von Kries JP, Scheidereit C. TSG101 associates with PARP1 and is essential for PARylation and DNA damage-induced NF-κB activation. EMBO J 2022; 41:e110372. [PMID: 36124865 DOI: 10.15252/embj.2021110372] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 08/11/2022] [Accepted: 08/25/2022] [Indexed: 11/09/2022] Open
Abstract
In a genome-wide screening for components of the dsDNA-break-induced IKK-NF-κB pathway, we identified scores of regulators, including tumor susceptibility gene TSG101. TSG101 is essential for DNA damage-induced formation of cellular poly(ADP-ribose) (PAR). TSG101 binds to PARP1 and is required for PARP1 activation. This function of TSG101 is independent of its role in the ESCRT-I endosomal sorting complex. In the absence of TSG101, the PAR-dependent formation of a nuclear PARP1-IKKγ signalosome, which triggers IKK activation, is impaired. According to its requirement for PARP1 and NF-κB activation, TSG101-deficient cells are defective in DNA repair and apoptosis protection. Loss of TSG101 results in PARP1 trapping at damage sites and mimics the effect of pharmacological PARP inhibition. We also show that the loss of TSG101 in connection with inactivated tumor suppressors BRCA1/2 in breast cancer cells is lethal. Our results imply TSG101 as a therapeutic target to achieve synthetic lethality in cancer treatment.
Collapse
Affiliation(s)
- Ahmet Buğra Tufan
- Laboratory for Signal Transduction in Tumor Cells, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Katina Lazarow
- Leibniz-Forschungsinstitut for Molecular Pharmacology (FMP), Berlin, Germany
| | - Marina Kolesnichenko
- Laboratory for Signal Transduction in Tumor Cells, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany.,Department of Hepatology and Gastroenterology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Anje Sporbert
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Advanced Light Microscopy Technology Platform, Berlin, Germany
| | | | - Claus Scheidereit
- Laboratory for Signal Transduction in Tumor Cells, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| |
Collapse
|
30
|
Schmitt CA, Wang B, Demaria M. Senescence and cancer - role and therapeutic opportunities. Nat Rev Clin Oncol 2022; 19:619-636. [PMID: 36045302 PMCID: PMC9428886 DOI: 10.1038/s41571-022-00668-4] [Citation(s) in RCA: 390] [Impact Index Per Article: 130.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/18/2022] [Indexed: 01/10/2023]
Abstract
Cellular senescence is a state of stable, terminal cell cycle arrest associated with various macromolecular changes and a hypersecretory, pro-inflammatory phenotype. Entry of cells into senescence can act as a barrier to tumorigenesis and, thus, could in principle constitute a desired outcome for any anticancer therapy. Paradoxically, studies published in the past decade have demonstrated that, in certain conditions and contexts, malignant and non-malignant cells with lastingly persistent senescence can acquire pro-tumorigenic properties. In this Review, we first discuss the major mechanisms involved in the antitumorigenic functions of senescent cells and then consider the cell-intrinsic and cell-extrinsic factors that participate in their switch towards a tumour-promoting role, providing an overview of major translational and emerging clinical findings. Finally, we comprehensively describe various senolytic and senomorphic therapies and their potential to benefit patients with cancer. The entry of cells into senescence can act as a barrier to tumorigenesis; however, in certain contexts senescent malignant and non-malignant cells can acquire pro-tumorigenic properties. The authors of this Review discuss the cell-intrinsic and cell-extrinsic mechanisms involved in both the antitumorigenic and tumour-promoting roles of senescent cells, and describe the potential of various senolytic and senomorphic therapeutic approaches in oncology. Cellular senescence is a natural barrier to tumorigenesis; senescent cells are widely detected in premalignant lesions from patients with cancer. Cellular senescence is induced by anticancer therapy and can contribute to some treatment-related adverse events (TRAEs). Senescent cells exert both protumorigenic and antitumorigenic effects via cell-autonomous and paracrine mechanisms. Pharmacological modulation of senescence-associated phenotypes has the potential to improve therapy efficacy and reduce the incidence of TRAEs.
Collapse
Affiliation(s)
- Clemens A Schmitt
- Charité Universitätsmedizin Berlin, Medical Department of Hematology, Oncology and Tumour Immunology, and Molekulares Krebsforschungszentrum-MKFZ, Campus Virchow Klinikum, Berlin, Germany.,Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,Johannes Kepler University, Linz, Austria.,Kepler University Hospital, Department of Hematology and Oncology, Linz, Austria.,Deutsches Konsortium für Translationale Krebsforschung (German Cancer Consortium), Partner site Berlin, Berlin, Germany
| | - Boshi Wang
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen (UMCG), University of Groningen (RUG), Groningen, the Netherlands
| | - Marco Demaria
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen (UMCG), University of Groningen (RUG), Groningen, the Netherlands.
| |
Collapse
|
31
|
Post-Transcriptional Control of mRNA Metabolism and Protein Secretion: The Third Level of Regulation within the NF-κB System. Biomedicines 2022; 10:biomedicines10092108. [PMID: 36140209 PMCID: PMC9495616 DOI: 10.3390/biomedicines10092108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/12/2022] [Accepted: 08/21/2022] [Indexed: 11/17/2022] Open
Abstract
The NF-κB system is a key transcriptional pathway that regulates innate and adaptive immunity because it triggers the activation and differentiation processes of lymphocytes and myeloid cells during immune responses. In most instances, binding to cytoplasmic inhibitory IκB proteins sequesters NF-κB into an inactive state, while a plethora of external triggers activate three complex signaling cascades that mediate the release and nuclear translocation of the NF-κB DNA-binding subunits. In addition to these cytosolic steps (level 1 of NF-κB regulation), NF-κB activity is also controlled in the nucleus by signaling events, cofactors and the chromatin environment to precisely determine chromatin recruitment and the specificity and timing of target gene transcription (level 2 of NF-κB regulation). Here, we discuss an additional layer of the NF-κB system that manifests in various steps of post-transcriptional gene expression and protein secretion. This less-studied regulatory level allows reduction of (transcriptional) noise and signal integration and endows time-shifted control of the secretion of inflammatory mediators. Detailed knowledge of these steps is important, as dysregulated post-transcriptional NF-κB signaling circuits are likely to foster chronic inflammation and contribute to the formation and maintenance of a tumor-promoting microenvironment.
Collapse
|
32
|
Saul D, Kosinsky RL, Atkinson EJ, Doolittle ML, Zhang X, LeBrasseur NK, Pignolo RJ, Robbins PD, Niedernhofer LJ, Ikeno Y, Jurk D, Passos JF, Hickson LJ, Xue A, Monroe DG, Tchkonia T, Kirkland JL, Farr JN, Khosla S. A new gene set identifies senescent cells and predicts senescence-associated pathways across tissues. Nat Commun 2022; 13:4827. [PMID: 35974106 PMCID: PMC9381717 DOI: 10.1038/s41467-022-32552-1] [Citation(s) in RCA: 399] [Impact Index Per Article: 133.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 08/05/2022] [Indexed: 02/01/2023] Open
Abstract
Although cellular senescence drives multiple age-related co-morbidities through the senescence-associated secretory phenotype, in vivo senescent cell identification remains challenging. Here, we generate a gene set (SenMayo) and validate its enrichment in bone biopsies from two aged human cohorts. We further demonstrate reductions in SenMayo in bone following genetic clearance of senescent cells in mice and in adipose tissue from humans following pharmacological senescent cell clearance. We next use SenMayo to identify senescent hematopoietic or mesenchymal cells at the single cell level from human and murine bone marrow/bone scRNA-seq data. Thus, SenMayo identifies senescent cells across tissues and species with high fidelity. Using this senescence panel, we are able to characterize senescent cells at the single cell level and identify key intercellular signaling pathways. SenMayo also represents a potentially clinically applicable panel for monitoring senescent cell burden with aging and other conditions as well as in studies of senolytic drugs.
Collapse
Affiliation(s)
- Dominik Saul
- Division of Endocrinology, Mayo Clinic, Rochester, MN, 55905, USA.
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, 55905, USA.
- Department of Trauma, Orthopedics and Reconstructive Surgery, Georg-August-University of Goettingen, Goettingen, Germany.
| | - Robyn Laura Kosinsky
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, 55905, USA
| | | | - Madison L Doolittle
- Division of Endocrinology, Mayo Clinic, Rochester, MN, 55905, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, 55905, USA
| | - Xu Zhang
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Nathan K LeBrasseur
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Robert J Pignolo
- Division of Endocrinology, Mayo Clinic, Rochester, MN, 55905, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Paul D Robbins
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Laura J Niedernhofer
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Yuji Ikeno
- Department of Pathology, University of Texas Health, San Antonio, TX, USA
| | - Diana Jurk
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - João F Passos
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - LaTonya J Hickson
- Division of Nephrology and Hypertension, Mayo Clinic, Jacksonville, FL, USA
| | - Ailing Xue
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, 55905, USA
| | - David G Monroe
- Division of Endocrinology, Mayo Clinic, Rochester, MN, 55905, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, 55905, USA
| | - Tamara Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - James L Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Joshua N Farr
- Division of Endocrinology, Mayo Clinic, Rochester, MN, 55905, USA.
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, 55905, USA.
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA.
| | - Sundeep Khosla
- Division of Endocrinology, Mayo Clinic, Rochester, MN, 55905, USA.
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, 55905, USA.
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
33
|
Tang Y, Zhang P, Liu Q, Cao L, Xu J. Pyroptotic Patterns in Blood Leukocytes Predict Disease Severity and Outcome in COVID-19 Patients. Front Immunol 2022; 13:888661. [PMID: 35928821 PMCID: PMC9343985 DOI: 10.3389/fimmu.2022.888661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 06/17/2022] [Indexed: 12/04/2022] Open
Abstract
The global coronavirus disease 2019 (COVID-19) pandemic has lasted for over 2 years now and has already caused millions of deaths. In COVID-19, leukocyte pyroptosis has been previously associated with both beneficial and detrimental effects, so its role in the development of this disease remains controversial. Using transcriptomic data (GSE157103) of blood leukocytes from 126 acute respiratory distress syndrome patients (ARDS) with or without COVID-19, we found that COVID-19 patients present with enhanced leukocyte pyroptosis. Based on unsupervised clustering, we divided 100 COVID-19 patients into two clusters (PYRcluster1 and PYRcluster2) according to the expression of 35 pyroptosis-related genes. The results revealed distinct pyroptotic patterns associated with different leukocytes in these PYRclusters. PYRcluster1 patients were in a hyperinflammatory state and had a worse prognosis than PYRcluster2 patients. The hyperinflammation of PYRcluster1 was validated by the results of gene set enrichment analysis (GSEA) of proteomic data (MSV000085703). These differences in pyroptosis between the two PYRclusters were confirmed by the PYRscore. To improve the clinical treatment of COVID-19 patients, we used least absolute shrinkage and selection operator (LASSO) regression to construct a prognostic model based on differentially expressed genes between PYRclusters (PYRsafescore), which can be applied as an effective prognosis tool. Lastly, we explored the upstream transcription factors of different pyroptotic patterns, thereby identifying 112 compounds with potential therapeutic value in public databases.
Collapse
Affiliation(s)
- Yingkui Tang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Peidong Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Qiuyu Liu
- Department of Critical Care Medicine, Yongchuan Hospital, Chongqing Medical University, Chongqing, China
| | - Luyang Cao
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory (GRMH-GDL), Guangzhou, China
- *Correspondence: Jingsong Xu, ; Luyang Cao,
| | - Jingsong Xu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory (GRMH-GDL), Guangzhou, China
- *Correspondence: Jingsong Xu, ; Luyang Cao,
| |
Collapse
|
34
|
Peng Z, Liang X, Lin X, Lin W, Lin Z, Wei S. Exploration of the molecular mechanisms, shared gene signatures, and MicroRNAs between systemic lupus erythematosus and diffuse large B cell lymphoma by bioinformatics analysis. Lupus 2022; 31:1317-1327. [PMID: 35817571 DOI: 10.1177/09612033221114578] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) is a complex heterogeneous systemic autoimmune disease. Previous studies have shown that SLE may be related to diffuse large B cell lymphoma (DLBCL), but the mechanism of their relationship is still unclear. The present study aimed to explore the common genetic molecular mechanisms, core shared genes, and miRNAs between SLE and DLBCL as well as to investigate the diagnostic markers of DLBCL. METHODS The SLE and DLBCL microarray data were downloaded from the comprehensive Gene Expression Omnibus (GEO) database. Weighted gene co-expression network analysis (WGCNA) was used to identify co-expression modules. Four core shared genes were screened out by various algorithms and validated in other cohorts. Finally, we constructed a common core gene-miRNA network using the human microRNA disease database (HMDD) and TarBase. RESULTS Using WGCNA, four modules were identified as important modules for SLE and DLBCL. Enrichment analysis of the shared genes showed that the highly activated NF-κB pathway was a common feature of the pathophysiology. Four core shared genes, namely, PSMB10, PSMB4, TAF10, and NFΚBIA, were screened out. These core shared genes were significantly upregulated in both diseases, and they may be potential diagnostic markers of DLBCL. The core gene-miRNA network showed that miR-155-5p, regulating the shared NF-κB pathway, may play an important role in the susceptibility of SLE patients to DLBCL. CONCLUSION The present study revealed that NF-κB pathway in SLE may be a crucial susceptible factor for DLBCL. In addition, we identified PSMB10, PSMB4, TAF10, NFΚBIA and miR-155 involved in the common pathogenesis as potential biomarkers and therapeutic targets for DLBCL.
Collapse
Affiliation(s)
- Zhishen Peng
- Zhujiang Hospital, The Second School of Clinical Medicine70570,Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Xiaofeng Liang
- Zhujiang Hospital, The Second School of Clinical Medicine70570,Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Xiaobing Lin
- Zhujiang Hospital, The Second School of Clinical Medicine70570,Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Weiyi Lin
- Zhujiang Hospital, The Second School of Clinical Medicine70570,Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Zien Lin
- Zhujiang Hospital, The Second School of Clinical Medicine70570,Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Shanshan Wei
- Department of Dermatology, 70570Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| |
Collapse
|
35
|
Wizenty J, Müllerke S, Kolesnichenko M, Heuberger J, Lin M, Fischer AS, Mollenkopf HJ, Berger H, Tacke F, Sigal M. Gastric stem cells promote inflammation and gland remodeling in response to Helicobacter pylori via Rspo3-Lgr4 axis. EMBO J 2022; 41:e109996. [PMID: 35767364 PMCID: PMC9251867 DOI: 10.15252/embj.2021109996] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 05/17/2022] [Indexed: 12/11/2022] Open
Abstract
Helicobacter pylori is a pathogen that colonizes the stomach and causes chronic gastritis. Helicobacter pylori can colonize deep inside gastric glands, triggering increased R‐spondin 3 (Rspo3) signaling. This causes an expansion of the “gland base module,” which consists of self‐renewing stem cells and antimicrobial secretory cells and results in gland hyperplasia. The contribution of Rspo3 receptors Lgr4 and Lgr5 is not well explored. Here, we identified that Lgr4 regulates Lgr5 expression and is required for H. pylori‐induced hyperplasia and inflammation, while Lgr5 alone is not. Using conditional knockout mice, we reveal that R‐spondin signaling via Lgr4 drives proliferation of stem cells and also induces NF‐κB activity in the proliferative stem cells. Upon exposure to H. pylori, the Lgr4‐driven NF‐κB activation is responsible for the expansion of the gland base module and simultaneously enables chemokine expression in stem cells, resulting in gland hyperplasia and neutrophil recruitment. This demonstrates a connection between R‐spondin‐Lgr and NF‐κB signaling that links epithelial stem cell behavior and inflammatory responses to gland‐invading H. pylori.
Collapse
Affiliation(s)
- Jonas Wizenty
- Division of Gastroenterology and Hepatology, Medical Department, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Stefanie Müllerke
- Division of Gastroenterology and Hepatology, Medical Department, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany
| | - Marina Kolesnichenko
- Division of Gastroenterology, Infectiology and Rheumatology, Medical Department, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Julian Heuberger
- Division of Gastroenterology and Hepatology, Medical Department, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany
| | - Manqiang Lin
- Division of Gastroenterology and Hepatology, Medical Department, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany
| | - Anne-Sophie Fischer
- Division of Gastroenterology and Hepatology, Medical Department, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany.,Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany
| | - Hans-Joachim Mollenkopf
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Hilmar Berger
- Division of Gastroenterology and Hepatology, Medical Department, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Frank Tacke
- Division of Gastroenterology and Hepatology, Medical Department, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Michael Sigal
- Division of Gastroenterology and Hepatology, Medical Department, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany.,Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany
| |
Collapse
|
36
|
CF-PPiD technology based on cell-free protein array and proximity biotinylation enzyme for in vitro direct interactome analysis. Sci Rep 2022; 12:10592. [PMID: 35732899 PMCID: PMC9217950 DOI: 10.1038/s41598-022-14872-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 06/14/2022] [Indexed: 12/04/2022] Open
Abstract
Protein–protein interaction (PPI) analysis is a key process to understand protein functions. Recently, we constructed a human protein array (20 K human protein beads array) consisting of 19,712 recombinant human proteins produced by a wheat cell-free protein production system. Here, we developed a cell-free protein array technology for proximity biotinylation-based PPI identification (CF-PPiD). The proximity biotinylation enzyme AirID-fused TP53 and -IκBα proteins each biotinylated specific interacting proteins on a 1536-well magnetic plate. In addition, AirID-fused cereblon was shown to have drug-inducible PPIs using CF-PPiD. Using the human protein beads array with AirID-IκBα, 132 proteins were biotinylated, and then selected clones showed these biological interactions in cells. Although ZBTB9 was not immunoprecipitated, it was highly biotinylated by AirID-IκBα, suggesting that this system detected weak interactions. These results indicated that CF-PPiD is useful for the biochemical identification of directly interacting proteins.
Collapse
|
37
|
Yu JT, Hu XW, Yang Q, Shan RR, Zhang Y, Dong ZH, Li HD, Wang JN, Li C, Xie SS, Dong YH, Ni WJ, Jiang L, Liu XQ, Wei B, Wen JG, Liu MM, Chen Q, Yang YR, Zhang GY, Zang HM, Jin J, Wu YG, Zhong X, Li J, Wang W, Meng XM. Insulin-like growth factor binding protein 7 promotes acute kidney injury by alleviating poly ADP ribose polymerase 1 degradation. Kidney Int 2022; 102:828-844. [PMID: 35752325 DOI: 10.1016/j.kint.2022.05.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 05/16/2022] [Accepted: 05/26/2022] [Indexed: 12/30/2022]
Abstract
The novel biomarker, insulin-like growth factor binding protein 7 (IGFBP7), is used clinically to predict different types of acute kidney injury (AKI) and has drawn significant attention as a urinary biomarker. However, as a secreted protein in the circulation of patients with AKI, it is unclear whether IGFBP7 acts as a key regulator in AKI progression, and if mechanisms underlying its upregulation still need to be determined. Here we found that IGFBP7 is highly expressed in the blood and urine of patients and mice with AKI possibly via a c-Jun-dependent mechanism, and is positively correlated with kidney dysfunction. Global knockout of IGFBP7 ameliorated kidney dysfunction, inflammatory responses, and programmed cell death in murine models of cisplatin-, kidney ischemia/reperfusion-, and lipopolysaccharide-induced AKI. IGFBP7 mainly originated from kidney tubular epithelial cells. Conditional knockout of IGFBP7 from the kidney protected against AKI. By contrast, rescue of IGFBP7 expression in IGFBP7-knockout mice restored kidney damage and inflammation. IGFBP7 function was determined in vitro using recombinant IGFBP7 protein, IGFBP7 knockdown, or overexpression. Additionally, IGFBP7 was found to bind to poly [ADP-ribose] polymerase 1 (PARP1) and inhibit its degradation by antagonizing the E3 ubiquitin ligase ring finger protein 4 (RNF4). Thus, IGFBP7 in circulation acts as a biomarker and key mediator of AKI by inhibiting RNF4/PARP1-mediated tubular injury and inflammation. Hence, over-activation of the IGFBP7/PARP1 axis represents a promising target for AKI treatment.
Collapse
Affiliation(s)
- Ju-Tao Yu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Xiao-Wei Hu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Department of clinical pharmacy, Anhui provincial children's hospital, Hefei 230051, China
| | - Qin Yang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Department of Clinical Pharmacology, Second Hospital of Anhui Medical University, 678 Furong Road, Hefei, 230601, Anhui, China
| | - Run-Run Shan
- School of Life Sciences, Anhui Medical University, Hefei 230032, China
| | - Yao Zhang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Ze-Hui Dong
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Hai-di Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Jia-Nan Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Chao Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Shuai-Shuai Xie
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Yu-Hang Dong
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Wei-Jian Ni
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Ling Jiang
- Department of Nephropathy, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Xue-Qi Liu
- Department of Nephropathy, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Biao Wei
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Jia-Gen Wen
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Ming-Ming Liu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Qi Chen
- School of Life Sciences, Anhui Medical University, Hefei 230032, China
| | - Ya-Ru Yang
- Department of Clinical Pharmacology, Second Hospital of Anhui Medical University, 678 Furong Road, Hefei, 230601, Anhui, China
| | - Gui-Yang Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China
| | - Hong-Mei Zang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Juan Jin
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China
| | - Yong-Gui Wu
- Department of Nephropathy, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Xiang Zhong
- Department of Nephrology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, 610072 Chengdu, Sichuan, China
| | - Jun Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Wei Wang
- Department of Urology, Institute of Urology, The First Affiliated Hospital of Anhui Medical University; Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei City 230032 China.
| | - Xiao-Ming Meng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
38
|
Strickland BA, Ansari SA, Dantoft W, Uhlenhaut NH. How to tame your genes: mechanisms of inflammatory gene repression by glucocorticoids. FEBS Lett 2022; 596:2596-2616. [PMID: 35612756 DOI: 10.1002/1873-3468.14409] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 04/24/2022] [Accepted: 05/18/2022] [Indexed: 01/08/2023]
Abstract
Glucocorticoids (GCs) are widely used therapeutic agents to treat a broad range of inflammatory conditions. Their functional effects are elicited by binding to the glucocorticoid receptor (GR), which regulates transcription of distinct gene networks in response to ligand. However, the mechanisms governing various aspects of undesired side effects versus beneficial immunomodulation upon GR activation remain complex and incompletely understood. In this review, we discuss emerging models of inflammatory gene regulation by GR, highlighting GR's regulatory specificity conferred by context-dependent changes in chromatin architecture and transcription factor or co-regulator dynamics. GR controls both gene activation and repression, with the repression mechanism being central to favorable clinical outcomes. We describe current knowledge about 3D genome organization and its role in spatiotemporal transcriptional control by GR. Looking beyond, we summarize the evidence for dynamics in gene regulation by GR through cooperative convergence of epigenetic modifications, transcription factor crosstalk, molecular condensate formation and chromatin looping. Further characterizing these genomic events will reframe our understanding of mechanisms of transcriptional repression by GR.
Collapse
Affiliation(s)
- Benjamin A Strickland
- Metabolic Programming, Technische Universitaet Muenchen (TUM), School of Life Sciences Weihenstephan, ZIEL - Institute for Food and Health, Gregor-Mendel-Str. 2, 85354, Freising, Germany
| | - Suhail A Ansari
- Institute for Diabetes and Endocrinology (IDE), Helmholtz Center Munich (HMGU) and German Center for Diabetes Research (DZD), Ingolstaedter Landstr. 1, 85764, Neuherberg, Germany
| | - Widad Dantoft
- Institute for Diabetes and Endocrinology (IDE), Helmholtz Center Munich (HMGU) and German Center for Diabetes Research (DZD), Ingolstaedter Landstr. 1, 85764, Neuherberg, Germany
| | - N Henriette Uhlenhaut
- Metabolic Programming, Technische Universitaet Muenchen (TUM), School of Life Sciences Weihenstephan, ZIEL - Institute for Food and Health, Gregor-Mendel-Str. 2, 85354, Freising, Germany.,Institute for Diabetes and Endocrinology (IDE), Helmholtz Center Munich (HMGU) and German Center for Diabetes Research (DZD), Ingolstaedter Landstr. 1, 85764, Neuherberg, Germany
| |
Collapse
|
39
|
Single-cell transcriptomics identifies Mcl-1 as a target for senolytic therapy in cancer. Nat Commun 2022; 13:2177. [PMID: 35449130 PMCID: PMC9023465 DOI: 10.1038/s41467-022-29824-1] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 03/24/2022] [Indexed: 01/10/2023] Open
Abstract
Cells subjected to treatment with anti-cancer therapies can evade apoptosis through cellular senescence. Persistent senescent tumor cells remain metabolically active, possess a secretory phenotype, and can promote tumor proliferation and metastatic dissemination. Removal of senescent tumor cells (senolytic therapy) has therefore emerged as a promising therapeutic strategy. Here, using single-cell RNA-sequencing, we find that senescent tumor cells rely on the anti-apoptotic gene Mcl-1 for their survival. Mcl-1 is upregulated in senescent tumor cells, including cells expressing low levels of Bcl-2, an established target for senolytic therapy. While treatment with the Bcl-2 inhibitor Navitoclax results in the reduction of metastases in tumor bearing mice, treatment with the Mcl-1 inhibitor S63845 leads to complete elimination of senescent tumor cells and metastases. These findings provide insights on the mechanism by which senescent tumor cells survive and reveal a vulnerability that can be exploited for cancer therapy. Cell senescence remains a barrier to tumor elimination in many cancers. Here, the authors use single cell RNA-seq to identify a role for Mcl-1 in senescent cell survival, and show that Mcl-1 inhibition may be an effective therapeutic strategy.
Collapse
|
40
|
Lehmann J, Narcisi R, Franceschini N, Chatzivasileiou D, Boer CG, Koevoet WJLM, Putavet D, Drabek D, van Haperen R, de Keizer PLJ, van Osch GJVM, Ten Berge D. WNT/beta-catenin signalling interrupts a senescence-induction cascade in human mesenchymal stem cells that restricts their expansion. Cell Mol Life Sci 2022; 79:82. [PMID: 35048158 PMCID: PMC8770385 DOI: 10.1007/s00018-021-04035-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 10/18/2021] [Accepted: 11/09/2021] [Indexed: 12/23/2022]
Abstract
Senescence, the irreversible cell cycle arrest of damaged cells, is accompanied by a deleterious pro-inflammatory senescence-associated secretory phenotype (SASP). Senescence and the SASP are major factors in aging, cancer, and degenerative diseases, and interfere with the expansion of adult cells in vitro, yet little is known about how to counteract their induction and deleterious effects. Paracrine signals are increasingly recognized as important senescence triggers and understanding their regulation and mode of action may provide novel opportunities to reduce senescence-induced inflammation and improve cell-based therapies. Here, we show that the signalling protein WNT3A counteracts the induction of paracrine senescence in cultured human adult mesenchymal stem cells (MSCs). We find that entry into senescence in a small subpopulation of MSCs triggers a secretome that causes a feed-forward signalling cascade that with increasing speed induces healthy cells into senescence. WNT signals interrupt this cascade by repressing cytokines that mediate this induction of senescence. Inhibition of those mediators by interference with NF-κB or interleukin 6 signalling reduced paracrine senescence in absence of WNT3A and promoted the expansion of MSCs. Our work reveals how WNT signals can antagonize senescence and has relevance not only for expansion of adult cells but can also provide new insights into senescence-associated inflammatory and degenerative diseases.
Collapse
Affiliation(s)
- Johannes Lehmann
- Department of Otorhinolaryngology and Head and Neck Surgery, Erasmus MC, University Medical Center, Rotterdam, The Netherlands.,Department of Cell Biology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands.,Center for Molecular Medicine, Section Molecular Cancer Research, Division LAB, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Roberto Narcisi
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Natasja Franceschini
- Department of Otorhinolaryngology and Head and Neck Surgery, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Danai Chatzivasileiou
- Department of Otorhinolaryngology and Head and Neck Surgery, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Cindy G Boer
- Department of Internal Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Wendy J L M Koevoet
- Department of Otorhinolaryngology and Head and Neck Surgery, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Diana Putavet
- Center for Molecular Medicine, Section Molecular Cancer Research, Division LAB, University Medical Center Utrecht, Utrecht, The Netherlands.,Department of Genetics, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Dubravka Drabek
- Department of Cell Biology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands.,Harbour Biomed, Rotterdam, the Netherlands
| | - Rien van Haperen
- Department of Cell Biology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands.,Harbour Biomed, Rotterdam, the Netherlands
| | - Peter L J de Keizer
- Center for Molecular Medicine, Section Molecular Cancer Research, Division LAB, University Medical Center Utrecht, Utrecht, The Netherlands.,Department of Genetics, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Gerjo J V M van Osch
- Department of Otorhinolaryngology and Head and Neck Surgery, Erasmus MC, University Medical Center, Rotterdam, The Netherlands.,Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Derk Ten Berge
- Department of Cell Biology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
41
|
Brischetto C, Krieger K, Klotz C, Krahn I, Kunz S, Kolesnichenko M, Mucka P, Heuberger J, Scheidereit C, Schmidt-Ullrich R. NF-κB determines Paneth versus goblet cell fate decision in the small intestine. Development 2021; 148:273388. [PMID: 34751748 PMCID: PMC8627599 DOI: 10.1242/dev.199683] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 09/28/2021] [Indexed: 12/12/2022]
Abstract
Although the role of the transcription factor NF-κB in intestinal inflammation and tumor formation has been investigated extensively, a physiological function of NF-κB in sustaining intestinal epithelial homeostasis beyond inflammation has not been demonstrated. Using NF-κB reporter mice, we detected strong NF-κB activity in Paneth cells, in ‘+4/+5’ secretory progenitors and in scattered Lgr5+ crypt base columnar stem cells of small intestinal (SI) crypts. To examine NF–κB functions in SI epithelial self-renewal, mice or SI crypt organoids (‘mini-guts’) with ubiquitously suppressed NF-κB activity were used. We show that NF-κB activity is dispensable for maintaining SI epithelial proliferation, but is essential for ex vivo organoid growth. Furthermore, we demonstrate a dramatic reduction of Paneth cells in the absence of NF-κB activity, concomitant with a significant increase in goblet cells and immature intermediate cells. This indicates that NF-κB is required for proper Paneth versus goblet cell differentiation and for SI epithelial homeostasis, which occurs via regulation of Wnt signaling and Sox9 expression downstream of NF-κB. The current study thus presents evidence for an important role for NF-κB in intestinal epithelial self-renewal. Summary: The transcription factor NF-κB, together with downstream Wnt and Sox9, is required for Paneth and goblet cell fate decisions and for maintenance of the small intestinal stem cell niche.
Collapse
Affiliation(s)
- Cristina Brischetto
- Signal Transduction in Tumor Cells, Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Str. 10, 13092 Berlin, Germany
| | - Karsten Krieger
- Signal Transduction in Tumor Cells, Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Str. 10, 13092 Berlin, Germany
| | - Christian Klotz
- Unit for Mycotic and Parasitic Agents and Mycobacteria, Robert Koch-Institute (RKI), 13353 Berlin, Germany
| | - Inge Krahn
- Signal Transduction in Tumor Cells, Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Str. 10, 13092 Berlin, Germany
| | - Séverine Kunz
- CF Electron Microscopy, Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Str. 10, 13092 Berlin, Germany
| | - Marina Kolesnichenko
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Charité-Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Patrick Mucka
- Signal Transduction in Tumor Cells, Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Str. 10, 13092 Berlin, Germany
| | - Julian Heuberger
- Signal Transduction in Development and Cancer, Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Str. 10, 13092 Berlin, Germany.,Medical Department, Division of Gastroenterology and Hepatology, Charité University Medicine, 13353 Berlin, Germany
| | - Claus Scheidereit
- Signal Transduction in Tumor Cells, Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Str. 10, 13092 Berlin, Germany
| | - Ruth Schmidt-Ullrich
- Signal Transduction in Tumor Cells, Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Str. 10, 13092 Berlin, Germany
| |
Collapse
|
42
|
Meier-Soelch J, Mayr-Buro C, Juli J, Leib L, Linne U, Dreute J, Papantonis A, Schmitz ML, Kracht M. Monitoring the Levels of Cellular NF-κB Activation States. Cancers (Basel) 2021; 13:5351. [PMID: 34771516 PMCID: PMC8582385 DOI: 10.3390/cancers13215351] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 10/18/2021] [Indexed: 12/12/2022] Open
Abstract
The NF-κB signaling system plays an important regulatory role in the control of many biological processes. The activities of NF-κB signaling networks and the expression of their target genes are frequently elevated in pathophysiological situations including inflammation, infection, and cancer. In these conditions, the outcome of NF-κB activity can vary according to (i) differential activation states, (ii) the pattern of genomic recruitment of the NF-κB subunits, and (iii) cellular heterogeneity. Additionally, the cytosolic NF-κB activation steps leading to the liberation of DNA-binding dimers need to be distinguished from the less understood nuclear pathways that are ultimately responsible for NF-κB target gene specificity. This raises the need to more precisely determine the NF-κB activation status not only for the purpose of basic research, but also in (future) clinical applications. Here we review a compendium of different methods that have been developed to assess the NF-κB activation status in vitro and in vivo. We also discuss recent advances that allow the assessment of several NF-κB features simultaneously at the single cell level.
Collapse
Affiliation(s)
- Johanna Meier-Soelch
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University, 35392 Giessen, Germany; (J.M.-S.); (C.M.-B.); (J.J.); (L.L.)
| | - Christin Mayr-Buro
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University, 35392 Giessen, Germany; (J.M.-S.); (C.M.-B.); (J.J.); (L.L.)
| | - Jana Juli
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University, 35392 Giessen, Germany; (J.M.-S.); (C.M.-B.); (J.J.); (L.L.)
| | - Lisa Leib
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University, 35392 Giessen, Germany; (J.M.-S.); (C.M.-B.); (J.J.); (L.L.)
| | - Uwe Linne
- Mass Spectrometry Facility of the Department of Chemistry, Philipps University, 35032 Marburg, Germany;
| | - Jan Dreute
- Institute of Biochemistry, Justus Liebig University, 35392 Giessen, Germany;
| | - Argyris Papantonis
- Institute of Pathology, University Medical Center Göttingen, 37075 Göttingen, Germany;
| | - M. Lienhard Schmitz
- Institute of Biochemistry, Justus Liebig University, 35392 Giessen, Germany;
| | - Michael Kracht
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University, 35392 Giessen, Germany; (J.M.-S.); (C.M.-B.); (J.J.); (L.L.)
| |
Collapse
|
43
|
Jochems F, Thijssen B, De Conti G, Jansen R, Pogacar Z, Groot K, Wang L, Schepers A, Wang C, Jin H, Beijersbergen RL, Leite de Oliveira R, Wessels LFA, Bernards R. The Cancer SENESCopedia: A delineation of cancer cell senescence. Cell Rep 2021; 36:109441. [PMID: 34320349 PMCID: PMC8333195 DOI: 10.1016/j.celrep.2021.109441] [Citation(s) in RCA: 117] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 05/29/2021] [Accepted: 07/02/2021] [Indexed: 12/17/2022] Open
Abstract
Cellular senescence is characterized as a stable proliferation arrest that can be triggered by multiple stresses. Most knowledge about senescent cells is obtained from studies in primary cells. However, senescence features may be different in cancer cells, since the pathways that are involved in senescence induction are often deregulated in cancer. We report here a comprehensive analysis of the transcriptome and senolytic responses in a panel of 13 cancer cell lines rendered senescent by two distinct compounds. We show that in cancer cells, the response to senolytic agents and the composition of the senescence-associated secretory phenotype are more influenced by the cell of origin than by the senescence trigger. Using machine learning, we establish the SENCAN gene expression classifier for the detection of senescence in cancer cell samples. The expression profiles and senescence classifier are available as an interactive online Cancer SENESCopedia.
Collapse
Affiliation(s)
- Fleur Jochems
- Division of Molecular Carcinogenesis, Oncode Institute, Netherlands Cancer Institute, Plesmanlaan 121, 1066 Amsterdam, the Netherlands
| | - Bram Thijssen
- Division of Molecular Carcinogenesis, Oncode Institute, Netherlands Cancer Institute, Plesmanlaan 121, 1066 Amsterdam, the Netherlands
| | - Giulia De Conti
- Division of Molecular Carcinogenesis, Oncode Institute, Netherlands Cancer Institute, Plesmanlaan 121, 1066 Amsterdam, the Netherlands
| | - Robin Jansen
- Division of Molecular Carcinogenesis, Oncode Institute, Netherlands Cancer Institute, Plesmanlaan 121, 1066 Amsterdam, the Netherlands
| | - Ziva Pogacar
- Division of Molecular Carcinogenesis, Oncode Institute, Netherlands Cancer Institute, Plesmanlaan 121, 1066 Amsterdam, the Netherlands
| | - Kelvin Groot
- Division of Molecular Carcinogenesis, Oncode Institute, Netherlands Cancer Institute, Plesmanlaan 121, 1066 Amsterdam, the Netherlands
| | - Liqin Wang
- Division of Molecular Carcinogenesis, Oncode Institute, Netherlands Cancer Institute, Plesmanlaan 121, 1066 Amsterdam, the Netherlands
| | - Arnout Schepers
- Division of Molecular Carcinogenesis, Oncode Institute, Netherlands Cancer Institute, Plesmanlaan 121, 1066 Amsterdam, the Netherlands
| | - Cun Wang
- Division of Molecular Carcinogenesis, Oncode Institute, Netherlands Cancer Institute, Plesmanlaan 121, 1066 Amsterdam, the Netherlands
| | - Haojie Jin
- Division of Molecular Carcinogenesis, Oncode Institute, Netherlands Cancer Institute, Plesmanlaan 121, 1066 Amsterdam, the Netherlands
| | - Roderick L Beijersbergen
- Division of Molecular Carcinogenesis, The NKI Robotics and Screening Center, Netherlands Cancer Institute, Plesmanlaan 121, 1066 Amsterdam, the Netherlands
| | - Rodrigo Leite de Oliveira
- Division of Molecular Carcinogenesis, Oncode Institute, Netherlands Cancer Institute, Plesmanlaan 121, 1066 Amsterdam, the Netherlands
| | - Lodewyk F A Wessels
- Division of Molecular Carcinogenesis, Oncode Institute, Netherlands Cancer Institute, Plesmanlaan 121, 1066 Amsterdam, the Netherlands; Faculty of EEMCS, Delft University of Technology, Delft, the Netherlands.
| | - René Bernards
- Division of Molecular Carcinogenesis, Oncode Institute, Netherlands Cancer Institute, Plesmanlaan 121, 1066 Amsterdam, the Netherlands.
| |
Collapse
|
44
|
Fafián‐Labora JA, O’Loghlen A. NF-κB/IKK activation by small extracellular vesicles within the SASP. Aging Cell 2021; 20:e13426. [PMID: 34187082 PMCID: PMC8282244 DOI: 10.1111/acel.13426] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 05/26/2021] [Accepted: 06/05/2021] [Indexed: 12/18/2022] Open
Abstract
Cellular senescence plays an important role in different biological and pathological conditions. Senescent cells communicate with their microenvironment through a plethora of soluble factors, metalloproteases and extracellular vesicles (EV). Although much is known about the role that soluble factors play in senescence, the downstream signalling pathways activated by EV in senescence is unknown. To address this, we performed a small molecule inhibitor screen and have identified the IκB kinases IKKε, IKKα and IKKβ as essential for senescence mediated by EV (evSASP). By using pharmacological inhibitors of IKKε, IKKα and IKKβ, in addition to CRISPR/Cas9 targeting their respective genes, we find these pathways are important in mediating senescence. In addition, we find that senescence activation is dependent on canonical NF‐κB transcription factors where siRNA targeting p65 prevent senescence. Importantly, these IKK pathways are also relevant to ageing as knockout of IKKA, IKKB and IKKE avoid the activation of senescence. Altogether, these findings open a new potential line of investigation in the field of senescence by targeting the negative effects of the evSASP independent of particular EV contents.
Collapse
Affiliation(s)
- Juan Antonio Fafián‐Labora
- Epigenetics & Cellular Senescence Group Blizard Institute Barts and The London School of Medicine and Dentistry Queen Mary University of London London UK
| | - Ana O’Loghlen
- Epigenetics & Cellular Senescence Group Blizard Institute Barts and The London School of Medicine and Dentistry Queen Mary University of London London UK
| |
Collapse
|
45
|
Yu R, Liang J, Liu Q, Niu XZ, Lopez DH, Hou S. The relationship of CCL4, BCL2A1, and NFKBIA genes with premature aging in women of Yin deficiency constitution. Exp Gerontol 2021; 149:111316. [PMID: 33766622 DOI: 10.1016/j.exger.2021.111316] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 03/10/2021] [Accepted: 03/11/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Traditional Chinese Medicine (TCM) defined constitution as a health statue or physical fitness that determines individual susceptibility to diseases. Yin deficiency constitution (YinDC) is a type of constitution closely related to aging. Previous studies found that the characteristic genes of YinDC are part of the inflammatory aging signaling pathways (e.g., NF-kappa B). Therefore, the aim of the study was to further reveal the dysregulation of genes associated with inflammatory aging in YinDC women. METHODS This study adopted the industrial standard of constitutional judgment, and screened YinDC (n = 30) and Balanced constitution (BC) (n = 30) from women between the ages of 35 to 49, a range categorized as the degenerating period by TCM. Five genes CCL4, BCL2A1, NFKBIA, TAK1, and IL-8 were analyzed by qRT-PCR. RESULTS Logistical regression revealed the correlation between body constitution and the expression of the five genes: the expression of NFKBIA and CCL4 mRNA was significantly up-regulated, whereas the expression of BCL2A1 mRNA was significantly down-regulated in YinDC (P < 0.05). Age or weight, when included in the model, did not affected the correlations. CONCLUSION Increased mRNA expression of CCL4 and NFKBIA and decreased mRNA expression of BCL2A1 may be the molecular basis of premature aging of YinDC women. These results provide a mechanistic basis for early conditioning of YinDC, anti-aging, and the prevention of aging-related diseases.
Collapse
Affiliation(s)
- Ruoxi Yu
- National Institute of Traditional Chinese Medicine Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Jinfeng Liang
- School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Qi Liu
- School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xi-Zhi Niu
- Department of Chemical and Environmental Engineering, Department of Environmental Sciences, University of Arizona, Tucson, AZ 85719, USA
| | - David Humberto Lopez
- Department of Chemical and Environmental Engineering, Department of Environmental Sciences, University of Arizona, Tucson, AZ 85719, USA
| | - Shujuan Hou
- National Institute of Traditional Chinese Medicine Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| |
Collapse
|
46
|
Kolesnichenko M, Mikuda N, Höpken UE, Kärgel E, Uyar B, Tufan AB, Milanovic M, Sun W, Krahn I, Schleich K, von Hoff L, Hinz M, Willenbrock M, Jungmann S, Akalin A, Lee S, Schmidt-Ullrich R, Schmitt CA, Scheidereit C. Transcriptional repression of NFKBIA triggers constitutive IKK- and proteasome-independent p65/RelA activation in senescence. EMBO J 2021; 40:e104296. [PMID: 33459422 PMCID: PMC7957429 DOI: 10.15252/embj.2019104296] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 12/07/2020] [Accepted: 12/09/2020] [Indexed: 12/11/2022] Open
Abstract
The IκB kinase (IKK)‐NF‐κB pathway is activated as part of the DNA damage response and controls both inflammation and resistance to apoptosis. How these distinct functions are achieved remained unknown. We demonstrate here that DNA double‐strand breaks elicit two subsequent phases of NF‐κB activation in vivo and in vitro, which are mechanistically and functionally distinct. RNA‐sequencing reveals that the first‐phase controls anti‐apoptotic gene expression, while the second drives expression of senescence‐associated secretory phenotype (SASP) genes. The rapidly activated first phase is driven by the ATM‐PARP1‐TRAF6‐IKK cascade, which triggers proteasomal destruction of inhibitory IκBα, and is terminated through IκBα re‐expression from the NFKBIA gene. The second phase, which is activated days later in senescent cells, is on the other hand independent of IKK and the proteasome. An altered phosphorylation status of NF‐κB family member p65/RelA, in part mediated by GSK3β, results in transcriptional silencing of NFKBIA and IKK‐independent, constitutive activation of NF‐κB in senescence. Collectively, our study reveals a novel physiological mechanism of NF‐κB activation with important implications for genotoxic cancer treatment.
Collapse
Affiliation(s)
- Marina Kolesnichenko
- Signal Transduction in Tumor Cells, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Nadine Mikuda
- Signal Transduction in Tumor Cells, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Uta E Höpken
- Microenvironmental Regulation in Autoimmunity and Cancer, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Eva Kärgel
- Signal Transduction in Tumor Cells, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Bora Uyar
- Bioinformatics/Mathematical Modeling Platform, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Ahmet Bugra Tufan
- Signal Transduction in Tumor Cells, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Maja Milanovic
- Department of Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin, Berlin, Germany
| | - Wei Sun
- Laboratory for Functional Genomics and Systems Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Inge Krahn
- Signal Transduction in Tumor Cells, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Kolja Schleich
- Department of Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin, Berlin, Germany
| | - Linda von Hoff
- Signal Transduction in Tumor Cells, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Michael Hinz
- Signal Transduction in Tumor Cells, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Michael Willenbrock
- Signal Transduction in Tumor Cells, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Sabine Jungmann
- Signal Transduction in Tumor Cells, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Altuna Akalin
- Bioinformatics/Mathematical Modeling Platform, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Soyoung Lee
- Department of Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin, Berlin, Germany
| | - Ruth Schmidt-Ullrich
- Signal Transduction in Tumor Cells, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Clemens A Schmitt
- Department of Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin, Berlin, Germany
| | - Claus Scheidereit
- Signal Transduction in Tumor Cells, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| |
Collapse
|