1
|
He S, Huang Y, Liu J, Liu H, Chen Y, Zou T, Sun J, Wang W, Wei H, Yu CY. A Metformin-Based Multifunctional Nanoplatform as a DNA Damage Amplifier for Maximized Radio-Immunotherapy to Overcome Radiotherapy Resistance. ACS NANO 2025; 19:14848-14864. [PMID: 40207668 DOI: 10.1021/acsnano.4c18627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/11/2025]
Abstract
Radiotherapy (RT) has been highlighted to be an effective strategy for antitumor immunity activation by causing direct DNA damages, but it generally suffers from low response rates due to the compromised cytosolic DNA (cDNA) recognition by cyclic GMP-AMP synthase (cGAS). Simultaneous DNA repair and clearance system regulation for enhanced cDNA accumulation is a useful approach to improve immune response rates, which remains seldom reported to our knowledge. Here, we report the construction of a metformin (MET)-based multifunctional nanocomplex, CS-MET/siTREX1 (CSMT), consisting of biguanide-decorated CS (CS-MET) as the vector and 3'-5' DNA exonuclease TREX1 siRNA (siTREX1) as the therapeutic gene for RT-induced antitumor immunity enhancement by amplifying the initial DNA damage signals. The uniqueness of this study is the development of CSMT as a specific DNA damage amplifier to promote cDNA accumulation for maximizing radio-immunotherapy and circumventing RT resistance. Specifically, the CSMT nanocomplexes show not only enhanced gene transfection efficiency by MET modification but also synergistic therapeutic effects including MET's inhibition on DNA repair and siTREX1's attenuation on cDNA clearance, which leads to the greatest inhibitory effect in a Hepa1-6 proximal/distal tumor model with a high tumor growth inhibition (TGI) value of 99.1% for the primary tumor and significantly compromised distal tumor growth by inducing immunogenic cell death (ICD), promoting tumor-associated neutrophil (TAN) polarization, and stimulating tumor-specific memory T-cell generation. Overall, the CSMT nanocomplexes developed herein hold great translatable promises for overcoming RT resistance in clinics.
Collapse
Affiliation(s)
- Shuangyan He
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Yun Huang
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Jia Liu
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Hongdu Liu
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Yalan Chen
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Ting Zou
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Jian Sun
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - WuZhou Wang
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Hua Wei
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Cui-Yun Yu
- Affiliated Hospital of Hunan Academy of Chinese Medicine Hunan Academy of Chinese Medicine, Changsha 410013, China
| |
Collapse
|
2
|
Zhang B, Xu P, Ablasser A. Regulation of the cGAS-STING Pathway. Annu Rev Immunol 2025; 43:667-692. [PMID: 40085836 DOI: 10.1146/annurev-immunol-101721-032910] [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] [Indexed: 03/16/2025]
Abstract
The cGAS-cGAMP-STING pathway is essential for immune defense against pathogens. Upon binding DNA, cGAS synthesizes cGAMP, which activates STING, leading to potent innate immune effector responses. However, lacking specific features to distinguish between self and nonself DNA, cGAS-STING immunity requires precise regulation to prevent aberrant activation. Several safeguard mechanisms acting on different levels have evolved to maintain tolerance to self DNA and ensure immune homeostasis under normal conditions. Disruption of these safeguards can lead to erroneous activation by self DNA, resulting in inflammatory conditions but also favorable antitumor immunity. Insights into structural and cellular checkpoints that control and terminate cGAS-STING signaling are essential for comprehending and manipulating DNA-triggered innate immunity in health and disease.
Collapse
Affiliation(s)
- Bing Zhang
- Global Health Institute, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland;
| | - Pengbiao Xu
- Global Health Institute, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland;
| | - Andrea Ablasser
- Global Health Institute, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland;
- Institute for Cancer Research (ISREC), Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
3
|
Mu W, Xu G, Li L, Wen J, Xiu Y, Zhao J, Liu T, Wei Z, Luo W, Yang H, Wu Z, Zhan X, Xiao X, Bai Z. Carnosic Acid Directly Targets STING C-Terminal Tail to Improve STING-Mediated Inflammatory Diseases. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2417686. [PMID: 39965124 PMCID: PMC11984877 DOI: 10.1002/advs.202417686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 01/30/2025] [Indexed: 02/20/2025]
Abstract
cGAS (cyclic GMP-AMP synthase)-STING (stimulator of interferon genes) signaling plays a vital role in innate immunity, while its deregulation may lead to a wide variety of autoinflammatory and autoimmune diseases. It is essential to identify specifically effective lead compounds to inhibit the signaling. Herein, it is shown that carnosic acid (CA), an active ingredient of medicinal plant Rosmarinus officinalis L., specifically suppressed cGAS-STING pathway activation and the subsequent inflammatory responses. Mechanistically, CA directly bound to STING C-terminal tail (CTT), impeded the recruitment of TANK-binding kinase 1 (TBK1) onto STING signalosome, thereby blocking the phosphorylation of STING and interferon regulatory factor 3 (IRF3) nuclear translocation. Importantly, CA dramatically attenuated STING-mediated inflammatory responses in vivo. Consistently, CA has a salient ameliorative effect on autoinflammatory disease model mediated by Trex1 deficiency, via inhibition of the cGAS-STING signaling. Notably, the study further indicates that phenolic hydroxyl groups are essential for CA-mediated STING inhibitory activity. Collectively, the results thus identify STING as one of the crucial targets of CA for mediating CA's anti-inflammatory activity, and further reveal that STING CTT may be a novel promising target for drug development.
Collapse
Affiliation(s)
- Wenqing Mu
- Department of Hepatologythe Fifth Medical Center of PLA General HospitalBeijing100039China
- School of Traditional Chinese MedicineCapital Medical UniversityBeijing100069China
- State Key Laboratory of Radiation Medicine and ProtectionInstitutes for Translational MedicineSoochow UniversityJiangsu215123China
| | - Guang Xu
- School of Traditional Chinese MedicineCapital Medical UniversityBeijing100069China
| | - Ling Li
- Beijing Institute of BiotechnologyBeijing100071China
| | - Jincai Wen
- Department of Hepatologythe Fifth Medical Center of PLA General HospitalBeijing100039China
- Military Institute of Chinese MateriaFifth Medical Center of Chinese PLA General HospitalBeijing100039China
| | - Ye Xiu
- Department of Hepatologythe Fifth Medical Center of PLA General HospitalBeijing100039China
- Military Institute of Chinese MateriaFifth Medical Center of Chinese PLA General HospitalBeijing100039China
| | - Jia Zhao
- Department of Hepatologythe Fifth Medical Center of PLA General HospitalBeijing100039China
- Military Institute of Chinese MateriaFifth Medical Center of Chinese PLA General HospitalBeijing100039China
| | - Tingting Liu
- Department of Hepatologythe Fifth Medical Center of PLA General HospitalBeijing100039China
- Military Institute of Chinese MateriaFifth Medical Center of Chinese PLA General HospitalBeijing100039China
| | - Ziying Wei
- Department of Hepatologythe Fifth Medical Center of PLA General HospitalBeijing100039China
- Military Institute of Chinese MateriaFifth Medical Center of Chinese PLA General HospitalBeijing100039China
| | - Wei Luo
- Department of Hepatologythe Fifth Medical Center of PLA General HospitalBeijing100039China
- Military Institute of Chinese MateriaFifth Medical Center of Chinese PLA General HospitalBeijing100039China
| | - Huijie Yang
- Department of Hepatologythe Fifth Medical Center of PLA General HospitalBeijing100039China
- Military Institute of Chinese MateriaFifth Medical Center of Chinese PLA General HospitalBeijing100039China
| | - Zhixin Wu
- Department of Hepatologythe Fifth Medical Center of PLA General HospitalBeijing100039China
- Military Institute of Chinese MateriaFifth Medical Center of Chinese PLA General HospitalBeijing100039China
| | - Xiaoyan Zhan
- Department of Hepatologythe Fifth Medical Center of PLA General HospitalBeijing100039China
- Military Institute of Chinese MateriaFifth Medical Center of Chinese PLA General HospitalBeijing100039China
| | - Xiaohe Xiao
- Department of Hepatologythe Fifth Medical Center of PLA General HospitalBeijing100039China
- Military Institute of Chinese MateriaFifth Medical Center of Chinese PLA General HospitalBeijing100039China
| | - Zhaofang Bai
- Department of Hepatologythe Fifth Medical Center of PLA General HospitalBeijing100039China
- Military Institute of Chinese MateriaFifth Medical Center of Chinese PLA General HospitalBeijing100039China
| |
Collapse
|
4
|
Qiu GH, Fu M, Zheng X, Huang C. Protection of the genome and the central exome by peripheral non-coding DNA against DNA damage in health, ageing and age-related diseases. Biol Rev Camb Philos Soc 2025; 100:508-529. [PMID: 39327815 DOI: 10.1111/brv.13151] [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: 09/26/2023] [Revised: 09/15/2024] [Accepted: 09/18/2024] [Indexed: 09/28/2024]
Abstract
DNA in eukaryotic genomes is under constant assault from both exogenous and endogenous sources, leading to DNA damage, which is considered a major molecular driver of ageing. Fortunately, the genome and the central exome are safeguarded against these attacks by abundant peripheral non-coding DNA. Non-coding DNA codes for small non-coding RNAs that inactivate foreign nucleic acids in the cytoplasm and physically blocks these attacks in the nucleus. Damage to non-coding DNA produced during such blockage is removed in the form of extrachromosomal circular DNA (eccDNA) through nucleic pore complexes. Consequently, non-coding DNA serves as a line of defence for the exome against DNA damage. The total amount of non-coding DNA/heterochromatin declines with age, resulting in a decrease in both physical blockage and eccDNA exclusion, and thus an increase in the accumulation of DNA damage in the nucleus during ageing and in age-related diseases. Here, we summarize recent evidence supporting a protective role of non-coding DNA in healthy and pathological states and argue that DNA damage is the proximate cause of ageing and age-related genetic diseases. Strategies aimed at strengthening the protective role of non-coding DNA/heterochromatin could potentially offer better systematic protection for the dynamic genome and the exome against diverse assaults, reduce the burden of DNA damage to the exome, and thus slow ageing, counteract age-related genetic diseases and promote a healthier life for individuals.
Collapse
Affiliation(s)
- Guo-Hua Qiu
- College of Life Sciences, Longyan University, Longyan, 364012, People's Republic of China
- Fujian Provincial Key Laboratory of Preventive Veterinary Medicine and Biotechnology, Engineering Research Center for the Prevention and Control of Animal-Origin Zoonosis, Key Laboratory for the Prevention and Control of Animal Infectious Diseases and Biotechnology, Fujian Province Universities, Longyan, People's Republic of China
| | - Mingjun Fu
- College of Life Sciences, Longyan University, Longyan, 364012, People's Republic of China
- Fujian Provincial Key Laboratory of Preventive Veterinary Medicine and Biotechnology, Engineering Research Center for the Prevention and Control of Animal-Origin Zoonosis, Key Laboratory for the Prevention and Control of Animal Infectious Diseases and Biotechnology, Fujian Province Universities, Longyan, People's Republic of China
| | - Xintian Zheng
- College of Life Sciences, Longyan University, Longyan, 364012, People's Republic of China
- Fujian Provincial Key Laboratory of Preventive Veterinary Medicine and Biotechnology, Engineering Research Center for the Prevention and Control of Animal-Origin Zoonosis, Key Laboratory for the Prevention and Control of Animal Infectious Diseases and Biotechnology, Fujian Province Universities, Longyan, People's Republic of China
| | - Cuiqin Huang
- College of Life Sciences, Longyan University, Longyan, 364012, People's Republic of China
- Fujian Provincial Key Laboratory of Preventive Veterinary Medicine and Biotechnology, Engineering Research Center for the Prevention and Control of Animal-Origin Zoonosis, Key Laboratory for the Prevention and Control of Animal Infectious Diseases and Biotechnology, Fujian Province Universities, Longyan, People's Republic of China
| |
Collapse
|
5
|
Andarawi S, Vodickova L, Uttarilli A, Hanak P, Vodicka P. Defective DNA repair: a putative nexus linking immunological diseases, neurodegenerative disorders, and cancer. Mutagenesis 2025; 40:4-19. [PMID: 39937585 DOI: 10.1093/mutage/geae029] [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: 02/16/2024] [Accepted: 01/30/2025] [Indexed: 02/13/2025] Open
Abstract
DNA damage is a common event in cells, resulting from both internal and external factors. The maintenance of genomic integrity is vital for cellular function and physiological processes. The inadequate repair of DNA damage results in the genomic instability, which has been associated with the development and progression of various human diseases. Accumulation of DNA damage can lead to multiple diseases, such as neurodegenerative disorders, cancers, immune deficiencies, infertility, and ageing. This comprehensive review delves the impact of alterations in DNA damage response genes (DDR) and tries to elucidate how and to what extent the same traits modulate diverse major human diseases, such as cancer, neurodegenerative diseases, and immunological disorders. DDR is apparently the trait connecting important complex disorders in humans. However, the pathogenesis of the above disorders and diseases are different and lead to divergent consequences. It is important to discover the switch(es) that direct further the pathogenic process either to proliferative, or degenerative diseases. Our understanding of the influence of DNA damage on diverse human disorders may enable the development of the strategies to prevent, diagnose, and treat these diseases. In our article, we analysed publicly available GWAS summary statistics from the NHGRI-EBI GWAS Catalog and identified 12 009 single-nucleotide polymorphisms (SNPs) associated with cancer. Among these, 119 SNPs were found in DDR pathways, exhibiting significant P-values. Additionally, we identified 44 SNPs linked to various cancer types and neurodegenerative diseases (NDDs), including four located in DDR-related genes: ATM, CUX2, and WNT3. Furthermore, 402 SNPs were associated with both cancer and immunological disorders, with two found in the DDR gene RAD51B. This highlights the versatility of the DDR pathway in multifactorial diseases. However, the specific mechanisms that regulate DDR to initiate distinct pathogenic processes remain to be elucidated.
Collapse
Affiliation(s)
- Safaa Andarawi
- Department of the Molecular Biology of Cancer, Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 142 00 Prague, Czech Republic
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Alej Svobody 1655/77, 32300 Pilsen, Czech Republic
| | - Ludmila Vodickova
- Department of the Molecular Biology of Cancer, Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 142 00 Prague, Czech Republic
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Alej Svobody 1655/77, 32300 Pilsen, Czech Republic
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Albertov 4, 128 00 Prague, Czech Republic
| | - Anusha Uttarilli
- Department of the Molecular Biology of Cancer, Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 142 00 Prague, Czech Republic
| | - Petr Hanak
- Department of the Molecular Biology of Cancer, Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 142 00 Prague, Czech Republic
| | - Pavel Vodicka
- Department of the Molecular Biology of Cancer, Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 142 00 Prague, Czech Republic
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Alej Svobody 1655/77, 32300 Pilsen, Czech Republic
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Albertov 4, 128 00 Prague, Czech Republic
| |
Collapse
|
6
|
Hernandez FJ. Nucleases: From Primitive Immune Defenders to Modern Biotechnology Tools. Immunology 2025; 174:279-286. [PMID: 39686519 PMCID: PMC11799398 DOI: 10.1111/imm.13884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 10/22/2024] [Accepted: 11/29/2024] [Indexed: 12/18/2024] Open
Abstract
The story of nucleases begins on the ancient battlefields of early Earth, where simple bacteria fought to survive against viral invaders. Nucleases are enzymes that degrade nucleic acids, with restriction endonucleases emerging as some of the earliest defenders, cutting foreign DNA to protect their bacteria hosts. However, bacteria sought more than just defence. They evolved the CRISPR-Cas system, an adaptive immune mechanism capable of remembering past invaders. The now-famous Cas9 nuclease, a key player in this system, has been harnessed for genome editing, revolutionising biotechnology. Over time, nucleases evolved from basic viral defence tools into complex regulators of immune function in higher organisms. In humans, DNases and RNases maintain immune balance by clearing cellular debris, preventing autoimmunity, and defending against pathogens. These enzymes have transformed from simple bacterial defenders to critical players in both human immunity and biotechnology. This review explores the evolutionary history of nucleases and their vital roles as protectors in the story of life's defence mechanisms.
Collapse
Affiliation(s)
- Frank J. Hernandez
- Department of Physics, Chemistry and BiologyLinköping UniversityLinköpingSweden
- Department of Bioengineering and Biosciences, TECNUNNavarra UniversityDonostiaSpain
- IKERBASQUE, Basque Foundation for ScienceBilbaoSpain
| |
Collapse
|
7
|
Khedr S, Dissanayake LV, Alsheikh AJ, Zietara A, Spires DR, Kerketta R, Mathison AJ, Urrutia R, Palygin O, Staruschenko A. Role of cGAS/STING pathway in aging and sexual dimorphism in diabetic kidney disease. JCI Insight 2024; 10:e174126. [PMID: 39589791 PMCID: PMC11721291 DOI: 10.1172/jci.insight.174126] [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: 07/20/2023] [Accepted: 11/19/2024] [Indexed: 11/27/2024] Open
Abstract
Diabetic kidney disease (DKD) is the leading cause of chronic renal pathology. Understanding the molecular underpinnings of DKD is critical to designing tailored therapeutic approaches. Here, we focused on sex differences and the contribution of aging toward the progression of DKD. To explore these questions, we utilized young (12 weeks old) and aged (approximately 50 weeks old) type 2 diabetic nephropathy (T2DN) rats. We revealed that the cyclic GMP-AMP synthase (cGAS)/stimulator of interferon genes (STING) pathway was upregulated in T2DN rats compared with nondiabetic Wistar rats and in type 2 diabetic human kidneys. The activation of the cGAS/STING signaling pathway exhibited distinct protein expression profiles between male and female T2DN rats, with these differences becoming more pronounced with aging. RNA-Seq analysis of the kidney cortex in both male and female T2DN rats, at both younger and older ages, revealed several key molecules, highlighting crucial genes within the cGAS/STING pathway. Thus, our study delved deep into understanding the intricate sexual differences in the development and progression of DKD and we propose the cGAS/STING pathway as an essential contributor to disease development.
Collapse
Affiliation(s)
- Sherif Khedr
- Department of Physiology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Lashodya V. Dissanayake
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida, USA
| | - Ammar J. Alsheikh
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Adrian Zietara
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida, USA
| | - Denisha R. Spires
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Romica Kerketta
- Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Angela J. Mathison
- Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Raul Urrutia
- Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Oleg Palygin
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Alexander Staruschenko
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida, USA
- Hypertension and Kidney Research Center, University of South Florida, Tampa, Florida, USA
- James A. Haley Veterans’ Hospital, Tampa, Florida, USA
| |
Collapse
|
8
|
Sun H, Luo M, Zhou M, Zheng L, Li H, Esworthy RS, Shen B. Structure-specific nucleases in genome dynamics and strategies for targeting cancers. J Mol Cell Biol 2024; 16:mjae019. [PMID: 38714348 PMCID: PMC11574390 DOI: 10.1093/jmcb/mjae019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 04/21/2024] [Accepted: 05/06/2024] [Indexed: 05/09/2024] Open
Abstract
Nucleases are a super family of enzymes that hydrolyze phosphodiester bonds present in genomes. They widely vary in substrates, causing differentiation in cleavage patterns and having a diversified role in maintaining genetic material. Through cellular evolution of prokaryotic to eukaryotic, nucleases become structure-specific in recognizing its own or foreign genomic DNA/RNA configurations as its substrates, including flaps, bubbles, and Holliday junctions. These special structural configurations are commonly found as intermediates in processes like DNA replication, repair, and recombination. The structure-specific nature and diversified functions make them essential to maintaining genome integrity and evolution in normal and cancer cells. In this article, we review their roles in various pathways, including Okazaki fragment maturation during DNA replication, end resection in homology-directed recombination repair of DNA double-strand breaks, DNA excision repair and apoptosis DNA fragmentation in response to exogenous DNA damage, and HIV life cycle. As the nucleases serve as key points for the DNA dynamics, cellular apoptosis, and cancer cell survival pathways, we discuss the efforts in the field in developing the therapeutic regimens, taking advantage of recently available knowledge of their diversified structures and functions.
Collapse
Affiliation(s)
- Haitao Sun
- Medicinal Plant Resources and Protection Research Center, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, China
- Department of Cancer Genetics and Epigenetics, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Megan Luo
- Department of Cancer Genetics and Epigenetics, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Mian Zhou
- Department of Cancer Genetics and Epigenetics, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Li Zheng
- Department of Cancer Genetics and Epigenetics, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Hongzhi Li
- Department of Cancer Genetics and Epigenetics, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - R Steven Esworthy
- Department of Cancer Genetics and Epigenetics, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Binghui Shen
- Department of Cancer Genetics and Epigenetics, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| |
Collapse
|
9
|
Liang X, Chen J, Yan P, Chen Z, Gao C, Bai R, Tang J. The highly conserved region within exonuclease III-like in PML-I regulates the cytoplasmic localization of PML-NBs. J Biol Chem 2024; 300:107872. [PMID: 39395810 PMCID: PMC11602975 DOI: 10.1016/j.jbc.2024.107872] [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: 04/13/2024] [Revised: 09/12/2024] [Accepted: 09/25/2024] [Indexed: 10/14/2024] Open
Abstract
The sub-nuclear protein structure PML-NB regulates a wide range of important cellular functions, while its abnormal cytoplasmic localization may have pathological consequences. However, the nature of this aberrant localization remains poorly understood. In this study, we unveil that PML-I, the most conserved and abundant structural protein of PML-NB, possesses potent cytoplasmic targeting ability within the N-terminal half of the exonuclease III-like domain encoded by its unique exon 9, independent of the known nuclear localization signal. Fusion of this region to PML-VI can relocate PML-VI from the nucleus to the cytosol. Structural and deletion analysis revealed that the cytoplasmic targeting ability of this domain was restrained by the sequences encoded by exon 8a and the 3' portion of exon 9 in PML-I. Deletion of either of these regions relocates PML-I to the cytosol. Furthermore, we observed a potential interaction between the ER-localized TREX1 and the cytoplasmic-located PML-I mutants. Our results suggest that perturbation of the EXO-like domain of PML-I may represent an important mode to translocate PMLs from the nucleus to the cytosol, thereby interfering with the normal nuclear functions of PML-NBs.
Collapse
Affiliation(s)
- Xinxin Liang
- National Key Laboratory of Veterinary Public Health Security, Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jinwen Chen
- National Key Laboratory of Veterinary Public Health Security, Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Peijie Yan
- National Key Laboratory of Veterinary Public Health Security, Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Zhongzhou Chen
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Chao Gao
- National Key Laboratory of Veterinary Public Health Security, Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Rulan Bai
- National Key Laboratory of Veterinary Public Health Security, Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jun Tang
- National Key Laboratory of Veterinary Public Health Security, Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China.
| |
Collapse
|
10
|
Mistriotis P, Wisniewski EO, Si BR, Kalab P, Konstantopoulos K. Coordinated in confined migration: crosstalk between the nucleus and ion channel-mediated mechanosensation. Trends Cell Biol 2024; 34:809-825. [PMID: 38290913 PMCID: PMC11284253 DOI: 10.1016/j.tcb.2024.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 12/22/2023] [Accepted: 01/05/2024] [Indexed: 02/01/2024]
Abstract
Cell surface and intracellular mechanosensors enable cells to perceive different geometric, topographical, and physical cues. Mechanosensitive ion channels (MICs) localized at the cell surface and on the nuclear envelope (NE) are among the first to sense and transduce these signals. Beyond compartmentalizing the genome of the cell and its transcription, the nucleus also serves as a mechanical gauge of different physical and topographical features of the tissue microenvironment. In this review, we delve into the intricate mechanisms by which the nucleus and different ion channels regulate cell migration in confinement. We review evidence suggesting an interplay between macromolecular nuclear-cytoplasmic transport (NCT) and ionic transport across the cell membrane during confined migration. We also discuss the roles of the nucleus and ion channel-mediated mechanosensation, whether acting independently or in tandem, in orchestrating migratory mechanoresponses. Understanding nuclear and ion channel sensing, and their crosstalk, is critical to advancing our knowledge of cell migration in health and disease.
Collapse
Affiliation(s)
| | - Emily O Wisniewski
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA; Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Bishwa R Si
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA; Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Petr Kalab
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA.
| | - Konstantinos Konstantopoulos
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA; Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD 21218, USA; Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA; Department of Oncology, The Johns Hopkins University, Baltimore, MD 21205, USA.
| |
Collapse
|
11
|
Shim A, Luan X, Zhou W, Crow YJ, Maciejowski J. Mutations in the non-catalytic polyproline motif destabilize TREX1 and amplify cGAS-STING signaling. Hum Mol Genet 2024; 33:1555-1566. [PMID: 38796715 PMCID: PMC11373327 DOI: 10.1093/hmg/ddae089] [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: 02/16/2024] [Revised: 04/25/2024] [Indexed: 05/28/2024] Open
Abstract
The cGAS-STING pathway detects cytosolic DNA and activates a signaling cascade that results in a type I interferon (IFN) response. The endoplasmic reticulum (ER)-associated exonuclease TREX1 suppresses cGAS-STING by eliminating DNA from the cytosol. Mutations that compromise TREX1 function are linked to autoinflammatory disorders, including systemic lupus erythematosus (SLE) and Aicardi-Goutières syndrome (AGS). Despite key roles in regulating cGAS-STING and suppressing excessive inflammation, the impact of many disease-associated TREX1 mutations-particularly those outside of the core catalytic domains-remains poorly understood. Here, we characterize a recessive AGS-linked TREX1 P61Q mutation occurring within the poorly characterized polyproline helix (PPII) motif. In keeping with its position outside of the catalytic core or ER targeting motifs, neither the P61Q mutation, nor aggregate proline-to-alanine PPII mutation, disrupts TREX1 exonuclease activity, subcellular localization, or cGAS-STING regulation in overexpression systems. Introducing targeted mutations into the endogenous TREX1 locus revealed that PPII mutations destabilize the protein, resulting in impaired exonuclease activity and unrestrained cGAS-STING activation. Overall, these results demonstrate that TREX1 PPII mutations, including P61Q, impair proper immune regulation and lead to autoimmune disease through TREX1 destabilization.
Collapse
Affiliation(s)
- Abraham Shim
- Molecular Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY10065, USA
| | - Xiaohan Luan
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong518055, China
| | - Wen Zhou
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong518055, China
| | - Yanick J Crow
- MRC Human Genetics Unit, Institute of Genetics and Cancer, The University of Edinburgh, Edinburgh, UK
| | - John Maciejowski
- Molecular Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY10065, USA
| |
Collapse
|
12
|
Ruan K, Bai G, Fang Y, Li D, Li T, Liu X, Lu B, Lu Q, Songyang Z, Sun S, Wang Z, Zhang X, Zhou W, Zhang H. Biomolecular condensates and disease pathogenesis. SCIENCE CHINA. LIFE SCIENCES 2024; 67:1792-1832. [PMID: 39037698 DOI: 10.1007/s11427-024-2661-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 06/21/2024] [Indexed: 07/23/2024]
Abstract
Biomolecular condensates or membraneless organelles (MLOs) formed by liquid-liquid phase separation (LLPS) divide intracellular spaces into discrete compartments for specific functions. Dysregulation of LLPS or aberrant phase transition that disturbs the formation or material states of MLOs is closely correlated with neurodegeneration, tumorigenesis, and many other pathological processes. Herein, we summarize the recent progress in development of methods to monitor phase separation and we discuss the biogenesis and function of MLOs formed through phase separation. We then present emerging proof-of-concept examples regarding the disruption of phase separation homeostasis in a diverse array of clinical conditions including neurodegenerative disorders, hearing loss, cancers, and immunological diseases. Finally, we describe the emerging discovery of chemical modulators of phase separation.
Collapse
Affiliation(s)
- Ke Ruan
- The First Affiliated Hospital & School of Life Sciences, Ministry of Education Key Laboratory for Membrane-less Organelles & Cellular Dynamics, Hefei National Research Center for Interdisciplinary Sciences at the Microscale, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.
| | - Ge Bai
- Nanhu Brain-computer Interface Institute, Hangzhou, 311100, China.
- Department of Neurology of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| | - Yanshan Fang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 201210, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Dan Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, 200030, China.
| | - Tingting Li
- Department of Biomedical Informatics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China.
| | - Xingguo Liu
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
- Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 510000, China.
| | - Boxun Lu
- Neurology Department at Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, School of Life Sciences, Fudan University, Shanghai, 200433, China.
| | - Qing Lu
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, 200030, China.
| | - Zhou Songyang
- State Key Laboratory of Biocontrol, MOE Key Laboratory of Gene Function and Regulation and Guangzhou Key Laboratory of Healthy Aging Research, School of Life Sciences, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510275, China.
| | - Shuguo Sun
- Department of Human Anatomy, Histology and Embryology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Zheng Wang
- The Second Affiliated Hospital, School of Basic Medical Sciences, Institute of Biomedical Innovation, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China.
| | - Xin Zhang
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, China.
| | - Wen Zhou
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China.
| | - Hong Zhang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
13
|
Wang L, Zhou W. Phase separation as a new form of regulation in innate immunity. Mol Cell 2024; 84:2410-2422. [PMID: 38936362 DOI: 10.1016/j.molcel.2024.06.004] [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: 04/08/2024] [Revised: 05/22/2024] [Accepted: 06/05/2024] [Indexed: 06/29/2024]
Abstract
Innate immunity is essential for the host against pathogens, cancer, and autoimmunity. The innate immune system encodes many sensor, adaptor, and effector proteins and relies on the assembly of higher-order signaling complexes to activate immune defense. Recent evidence demonstrates that many of the core complexes involved in innate immunity are organized as liquid-like condensates through a mechanism known as phase separation. Here, we discuss phase-separated condensates and their diverse functions. We compare the biochemical, structural, and mechanistic details of solid and liquid-like assemblies to explore the role of phase separation in innate immunity. We summarize the emerging evidence for the hypothesis that phase separation is a conserved mechanism that controls immune responses across the tree of life. The discovery of phase separation in innate immunity provides a new foundation to explain the rules that govern immune system activation and will enable the development of therapeutics to treat immune-related diseases properly.
Collapse
Affiliation(s)
- Lei Wang
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Wen Zhou
- Shenzhen Key Laboratory of Biomolecular Assembling and Regulation, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China; Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China.
| |
Collapse
|
14
|
Prakash P, Khodke P, Balasubramaniam M, Davids BO, Hollis T, Davis J, Kumbhar B, Dash C. Three prime repair exonuclease 1 preferentially degrades the integration-incompetent HIV-1 DNA through favorable kinetics, thermodynamic, structural, and conformational properties. J Biol Chem 2024; 300:107438. [PMID: 38838778 PMCID: PMC11259700 DOI: 10.1016/j.jbc.2024.107438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 05/17/2024] [Accepted: 05/24/2024] [Indexed: 06/07/2024] Open
Abstract
HIV-1 integration into the human genome is dependent on 3'-processing of the viral DNA. Recently, we reported that the cellular Three Prime Repair Exonuclease 1 (TREX1) enhances HIV-1 integration by degrading the unprocessed viral DNA, while the integration-competent 3'-processed DNA remained resistant. Here, we describe the mechanism by which the 3'-processed HIV-1 DNA resists TREX1-mediated degradation. Our kinetic studies revealed that the rate of cleavage (kcat) of the 3'-processed DNA was significantly lower (approximately 2-2.5-fold) than the unprocessed HIV-1 DNA by TREX1. The kcat values of human TREX1 for the processed U5 and U3 DNA substrates were 3.8 s-1 and 4.5 s-1, respectively. In contrast, the unprocessed U5 and U3 substrates were cleaved at 10.2 s-1 and 9.8 s-1, respectively. The efficiency of degradation (kcat/Km) of the 3'-processed DNA (U5-70.2 and U3-28.05 pM-1s-1) was also significantly lower than the unprocessed DNA (U5-103.1 and U3-65.3 pM-1s-1). Furthermore, the binding affinity (Kd) of TREX1 was markedly lower (∼2-fold) for the 3'-processed DNA than the unprocessed DNA. Molecular docking and dynamics studies revealed distinct conformational binding modes of TREX1 with the 3'-processed and unprocessed HIV-1 DNA. Particularly, the unprocessed DNA was favorably positioned in the active site with polar interactions with the catalytic residues of TREX1. Additionally, a stable complex was formed between TREX1 and the unprocessed DNA compared the 3'-processed DNA. These results pinpoint the mechanism by which TREX1 preferentially degrades the integration-incompetent HIV-1 DNA and reveal the unique structural and conformational properties of the integration-competent 3'-processed HIV-1 DNA.
Collapse
Affiliation(s)
- Prem Prakash
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College, Nashville, Tennessee, USA
| | - Purva Khodke
- Department of Biological Sciences, Sunandan Divatia School of Science, SVKM's NMIMS (Deemed-to-be-) University, Mumbai, Maharashtra, India
| | - Muthukumar Balasubramaniam
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College, Nashville, Tennessee, USA
| | - Benem-Orom Davids
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York City, New York, USA
| | - Thomas Hollis
- Department of Biochemistry and Center for Structural Biology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Jamaine Davis
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College, Nashville, Tennessee, USA
| | - Bajarang Kumbhar
- Department of Biological Sciences, Sunandan Divatia School of Science, SVKM's NMIMS (Deemed-to-be-) University, Mumbai, Maharashtra, India
| | - Chandravanu Dash
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College, Nashville, Tennessee, USA; Center for AIDS Health Disparities Research, Meharry Medical College, Nashville, Tennessee, USA; Department of Microbiology, Immunology, and Physiology, Meharry Medical College, Nashville, Tennessee, USA.
| |
Collapse
|
15
|
Manthei KA, Munson LM, Nandakumar J, Simmons LA. Structural and biochemical characterization of the mitomycin C repair exonuclease MrfB. Nucleic Acids Res 2024; 52:6347-6359. [PMID: 38661211 PMCID: PMC11194089 DOI: 10.1093/nar/gkae308] [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: 02/18/2024] [Revised: 04/03/2024] [Accepted: 04/10/2024] [Indexed: 04/26/2024] Open
Abstract
Mitomycin C (MMC) repair factor A (mrfA) and factor B (mrfB), encode a conserved helicase and exonuclease that repair DNA damage in the soil-dwelling bacterium Bacillus subtilis. Here we have focused on the characterization of MrfB, a DEDDh exonuclease in the DnaQ superfamily. We solved the structure of the exonuclease core of MrfB to a resolution of 2.1 Å, in what appears to be an inactive state. In this conformation, a predicted α-helix containing the catalytic DEDDh residue Asp172 adopts a random coil, which moves Asp172 away from the active site and results in the occupancy of only one of the two catalytic Mg2+ ions. We propose that MrfB resides in this inactive state until it interacts with DNA to become activated. By comparing our structure to an AlphaFold prediction as well as other DnaQ-family structures, we located residues hypothesized to be important for exonuclease function. Using exonuclease assays we show that MrfB is a Mg2+-dependent 3'-5' DNA exonuclease. We show that Leu113 aids in coordinating the 3' end of the DNA substrate, and that a basic loop is important for substrate binding. This work provides insight into the function of a recently discovered bacterial exonuclease important for the repair of MMC-induced DNA adducts.
Collapse
Affiliation(s)
- Kelly A Manthei
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Lia M Munson
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Jayakrishnan Nandakumar
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Lyle A Simmons
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
16
|
Tasharrofi B, Karimzadeh P, Asadollahi M, Hasani S, Heidari M, Keramatipour M. Aicardi-Goutières Syndrome Type 1: A Novel Missense Variant and Review of the Mutational Spectrum. IRANIAN JOURNAL OF CHILD NEUROLOGY 2024; 18:117-129. [PMID: 38988838 PMCID: PMC11231675 DOI: 10.22037/ijcn.v18i3.43274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 12/20/2023] [Indexed: 07/12/2024]
Abstract
Objectives Mutations in the TREX1 gene cause Aicardi-Goutières syndrome (AGS) 1, associated with a spectrum of autoimmune and neurodegenerative manifestations. AGS 1, the most severe neonatal type of AGS, is characterized by abnormal neurologic findings, visual inattention, hepatosplenomegaly, thrombocytopenia, skin rash, restlessness, and fever. Materials & Methods The present study described two affected siblings from an Iranian family whose phenotypes overlap with intrauterine infections. They had almost similar presentations, including developmental delay, microcephaly, no fix and follow epileptic seizures and the same pattern of brain CT scan involvements. Following clinical and paraclinical assessments, whole-exome sequencing was employed to determine the disease-causing variant, and subsequently, PCR-Sanger sequencing was performed to indicate the segregation pattern of the candidate variant in family members. Results Genetic analysis revealed a novel homozygous missense variant (c.461A>C; p.D154A) in the TREX1 gene in affected family members. Sanger sequencing of other family members showed the expected zygosities. Conclusion This study identifies a novel mutation in the TREX1 gene in this family and highlights the efficiency of next-generation sequencing-based techniques for obtaining a definite diagnosis in patients with early-onset encephalopathy.
Collapse
Affiliation(s)
- Behnoosh Tasharrofi
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Parvaneh Karimzadeh
- Pediatric Neurology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Pediatric Neurology Department, Mofid Children's Hospital, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mostafa Asadollahi
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sepideh Hasani
- Watson Genetic Laboratory, North Kargar Street, Tehran, Iran & Department of Medical Genetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Morteza Heidari
- Department of Pediatrics, Division of Pediatric Neurology, Children›s Medical Center, Pediatrics Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Keramatipour
- Watson Genetic Laboratory, North Kargar Street, Tehran, Iran & Department of Medical Genetics, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
17
|
Gu X, Chen Y, Cao K, Tu M, Liu W, Ju J. Therapeutic landscape in systemic lupus erythematosus: mtDNA activation of the cGAS-STING pathway. Int Immunopharmacol 2024; 133:112114. [PMID: 38652968 DOI: 10.1016/j.intimp.2024.112114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 04/16/2024] [Accepted: 04/16/2024] [Indexed: 04/25/2024]
Abstract
Mitochondrial DNA (mtDNA) serves as a pivotal immune stimulus in the immune response. During stress, mitochondria release mtDNA into the cytoplasm, where it is recognized by the cytoplasmic DNA receptor cGAS. This activation initiates the cGAS-STING-IRF3 pathway, culminating in an inflammatory response. The cGAS-STING pathway has emerged as a critical mediator of inflammatory responses in microbial infections, stress, autoimmune diseases, chronic illnesses, and tissue injuries. Systemic lupus erythematosus (SLE) is a chronic autoimmune disease characterized by connective tissue involvement across various bodily systems. Its hallmark is the production of numerous autoantibodies, which prompt the immune system to target and damage the body's own tissues, resulting in organ and tissue damage. Increasing evidence implicates the cGAS-STING pathway as a significant contributor to SLE pathogenesis. This article aims to explore the role of the mtDNA-triggered cGAS-STING pathway and its mechanisms in SLE, with the goal of providing novel insights for clinical interventions.
Collapse
Affiliation(s)
- Xiaotian Gu
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang 261053, China
| | - Yong Chen
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang 261053, China
| | - Kunyu Cao
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang 261053, China
| | - Miao Tu
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang 261053, China
| | - Wan Liu
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang 261053, China.
| | - Jiyu Ju
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang 261053, China.
| |
Collapse
|
18
|
Tani T, Mathsyaraja H, Campisi M, Li ZH, Haratani K, Fahey CG, Ota K, Mahadevan NR, Shi Y, Saito S, Mizuno K, Thai TC, Sasaki N, Homme M, Yusuf CFB, Kashishian A, Panchal J, Wang M, Wolf BJ, Barbie TU, Paweletz CP, Gokhale PC, Liu D, Uppaluri R, Kitajima S, Cain J, Barbie DA. TREX1 Inactivation Unleashes Cancer Cell STING-Interferon Signaling and Promotes Antitumor Immunity. Cancer Discov 2024; 14:752-765. [PMID: 38227896 PMCID: PMC11062818 DOI: 10.1158/2159-8290.cd-23-0700] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 11/28/2023] [Accepted: 01/08/2024] [Indexed: 01/18/2024]
Abstract
A substantial fraction of cancers evade immune detection by silencing Stimulator of Interferon Genes (STING)-Interferon (IFN) signaling. Therapeutic reactivation of this program via STING agonists, epigenetic, or DNA-damaging therapies can restore antitumor immunity in multiple preclinical models. Here we show that adaptive induction of three prime exonuclease 1 (TREX1) restrains STING-dependent nucleic acid sensing in cancer cells via its catalytic function in degrading cytosolic DNA. Cancer cell TREX1 expression is coordinately induced with STING by autocrine IFN and downstream STAT1, preventing signal amplification. TREX1 inactivation in cancer cells thus unleashes STING-IFN signaling, recruiting T and natural killer (NK) cells, sensitizing to NK cell-derived IFNγ, and cooperating with programmed cell death protein 1 blockade in multiple mouse tumor models to enhance immunogenicity. Targeting TREX1 may represent a complementary strategy to induce cytosolic DNA and amplify cancer cell STING-IFN signaling as a means to sensitize tumors to immune checkpoint blockade (ICB) and/or cell therapies. SIGNIFICANCE STING-IFN signaling in cancer cells promotes tumor cell immunogenicity. Inactivation of the DNA exonuclease TREX1, which is adaptively upregulated to limit pathway activation in cancer cells, recruits immune effector cells and primes NK cell-mediated killing. Targeting TREX1 has substantial therapeutic potential to amplify cancer cell immunogenicity and overcome ICB resistance. This article is featured in Selected Articles from This Issue, p. 695.
Collapse
Affiliation(s)
- Tetsuo Tani
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Contributed equally
| | | | - Marco Campisi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Ze-Hua Li
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Koji Haratani
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Caroline G. Fahey
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Keiichi Ota
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Navin R. Mahadevan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Yingxiao Shi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Shin Saito
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Kei Mizuno
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Tran C. Thai
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Nobunari Sasaki
- Department of Cell Biology, Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Mizuki Homme
- Department of Cell Biology, Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Choudhury Fabliha B. Yusuf
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | | | - Min Wang
- Gilead Sciences, Foster City, CA, USA
| | | | - Thanh U. Barbie
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Brigham and Women's Hospital, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Cloud P. Paweletz
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Prafulla C Gokhale
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - David Liu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Ravindra Uppaluri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Shunsuke Kitajima
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Cell Biology, Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | | | - David A. Barbie
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
19
|
Wenzl SJ, de Oliveira Mann CC. How enzyme-centered approaches are advancing research on cyclic oligo-nucleotides. FEBS Lett 2024; 598:839-863. [PMID: 38453162 DOI: 10.1002/1873-3468.14838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 03/09/2024]
Abstract
Cyclic nucleotides are the most diversified category of second messengers and are found in all organisms modulating diverse pathways. While cAMP and cGMP have been studied over 50 years, cyclic di-nucleotide signaling in eukaryotes emerged only recently with the anti-viral molecule 2´3´cGAMP. Recent breakthrough discoveries have revealed not only the astonishing chemical diversity of cyclic nucleotides but also surprisingly deep-rooted evolutionary origins of cyclic oligo-nucleotide signaling pathways and structural conservation of the proteins involved in their synthesis and signaling. Here we discuss how enzyme-centered approaches have paved the way for the identification of several cyclic nucleotide signals, focusing on the advantages and challenges associated with deciphering the activation mechanisms of such enzymes.
Collapse
Affiliation(s)
- Simon J Wenzl
- Department of Bioscience, TUM School of Natural Sciences, Technical University of Munich, Garching, Germany
| | - Carina C de Oliveira Mann
- Department of Bioscience, TUM School of Natural Sciences, Technical University of Munich, Garching, Germany
| |
Collapse
|
20
|
Queiroz MAF, de Oliveira AQT, Moura TCF, Brito WRDS, Santana EGM, de Lima LLP, Lopes FT, Bichara CDA, Amoras EDSG, Ishak R, Vallinoto IMVC, Vallinoto ACR. The Expression Levels of TREX1 and IFN-α Are Associated with Immune Reconstitution in HIV-1-Infected Individuals. Viruses 2024; 16:499. [PMID: 38675842 PMCID: PMC11054413 DOI: 10.3390/v16040499] [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: 02/14/2024] [Revised: 03/21/2024] [Accepted: 03/22/2024] [Indexed: 04/28/2024] Open
Abstract
TREX1 acts in the initial prevention of an autoimmune response, but it may contribute to the permissiveness of retrovirus infections. This study investigated the association between the levels of TREX1 gene expression with the polymorphisms TREX1 rs3135941 (T/C) and TREX1 rs3135945 (G/A), and the presence of antinuclear antibodies (ANA) in antiretroviral therapy (ART)-naïve individuals and after 1 year of treatment. Blood samples from 119 individuals with HIV-1 were subjected to genotyping of polymorphisms and quantification of TREX1 gene expression and HIV-1 viral load by qPCR. The concentration of IFN-α and the number of CD4+/CD8+ T lymphocytes were determined by ELISA and flow cytometry, respectively; ANA was investigated by immunofluorescence. A control group of 167 seronegative individuals was used for the comparison of genotypic frequencies. The frequency of the polymorphisms were not associated with HIV infection or with variations in the expression of TREX1 and IFN-α (p > 0.05). ART-naïve individuals exhibited higher TREX1 expression and lower IFN-α expression. After 1 year of ART, TREX1 levels were reduced, while IFN-α and CD4+ T lymphocytes were elevated (p < 0.05). Some individuals on ART presented ANA. These results suggest that ART-mediated restoration of immune competence is associated with a reduction in TREX1 expression, which may induce the development of ANA, regardless of the polymorphism investigated.
Collapse
Affiliation(s)
- Maria Alice Freitas Queiroz
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66.075-110, PA, Brazil; (A.Q.T.d.O.); (T.C.F.M.); (W.R.d.S.B.); (E.G.M.S.); (L.L.P.d.L.); (F.T.L.); (C.D.A.B.); (E.d.S.G.A.); (R.I.); (I.M.V.C.V.); (A.C.R.V.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66.075-110, PA, Brazil
| | - Allysson Quintino Tenório de Oliveira
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66.075-110, PA, Brazil; (A.Q.T.d.O.); (T.C.F.M.); (W.R.d.S.B.); (E.G.M.S.); (L.L.P.d.L.); (F.T.L.); (C.D.A.B.); (E.d.S.G.A.); (R.I.); (I.M.V.C.V.); (A.C.R.V.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66.075-110, PA, Brazil
| | - Tuane Carolina Ferreira Moura
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66.075-110, PA, Brazil; (A.Q.T.d.O.); (T.C.F.M.); (W.R.d.S.B.); (E.G.M.S.); (L.L.P.d.L.); (F.T.L.); (C.D.A.B.); (E.d.S.G.A.); (R.I.); (I.M.V.C.V.); (A.C.R.V.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66.075-110, PA, Brazil
| | - Wandrey Roberto dos Santos Brito
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66.075-110, PA, Brazil; (A.Q.T.d.O.); (T.C.F.M.); (W.R.d.S.B.); (E.G.M.S.); (L.L.P.d.L.); (F.T.L.); (C.D.A.B.); (E.d.S.G.A.); (R.I.); (I.M.V.C.V.); (A.C.R.V.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66.075-110, PA, Brazil
| | - Emmanuelle Giuliana Mendes Santana
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66.075-110, PA, Brazil; (A.Q.T.d.O.); (T.C.F.M.); (W.R.d.S.B.); (E.G.M.S.); (L.L.P.d.L.); (F.T.L.); (C.D.A.B.); (E.d.S.G.A.); (R.I.); (I.M.V.C.V.); (A.C.R.V.)
| | - Lorena Leticia Peixoto de Lima
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66.075-110, PA, Brazil; (A.Q.T.d.O.); (T.C.F.M.); (W.R.d.S.B.); (E.G.M.S.); (L.L.P.d.L.); (F.T.L.); (C.D.A.B.); (E.d.S.G.A.); (R.I.); (I.M.V.C.V.); (A.C.R.V.)
| | - Felipe Teixeira Lopes
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66.075-110, PA, Brazil; (A.Q.T.d.O.); (T.C.F.M.); (W.R.d.S.B.); (E.G.M.S.); (L.L.P.d.L.); (F.T.L.); (C.D.A.B.); (E.d.S.G.A.); (R.I.); (I.M.V.C.V.); (A.C.R.V.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66.075-110, PA, Brazil
| | - Carlos David Araújo Bichara
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66.075-110, PA, Brazil; (A.Q.T.d.O.); (T.C.F.M.); (W.R.d.S.B.); (E.G.M.S.); (L.L.P.d.L.); (F.T.L.); (C.D.A.B.); (E.d.S.G.A.); (R.I.); (I.M.V.C.V.); (A.C.R.V.)
| | - Ednelza da Silva Graça Amoras
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66.075-110, PA, Brazil; (A.Q.T.d.O.); (T.C.F.M.); (W.R.d.S.B.); (E.G.M.S.); (L.L.P.d.L.); (F.T.L.); (C.D.A.B.); (E.d.S.G.A.); (R.I.); (I.M.V.C.V.); (A.C.R.V.)
| | - Ricardo Ishak
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66.075-110, PA, Brazil; (A.Q.T.d.O.); (T.C.F.M.); (W.R.d.S.B.); (E.G.M.S.); (L.L.P.d.L.); (F.T.L.); (C.D.A.B.); (E.d.S.G.A.); (R.I.); (I.M.V.C.V.); (A.C.R.V.)
| | - Izaura Maria Vieira Cayres Vallinoto
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66.075-110, PA, Brazil; (A.Q.T.d.O.); (T.C.F.M.); (W.R.d.S.B.); (E.G.M.S.); (L.L.P.d.L.); (F.T.L.); (C.D.A.B.); (E.d.S.G.A.); (R.I.); (I.M.V.C.V.); (A.C.R.V.)
| | - Antonio Carlos Rosário Vallinoto
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66.075-110, PA, Brazil; (A.Q.T.d.O.); (T.C.F.M.); (W.R.d.S.B.); (E.G.M.S.); (L.L.P.d.L.); (F.T.L.); (C.D.A.B.); (E.d.S.G.A.); (R.I.); (I.M.V.C.V.); (A.C.R.V.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66.075-110, PA, Brazil
| |
Collapse
|
21
|
Prakash P, Khodke P, Balasubramaniam M, Davids BO, Hollis T, Davis J, Pandhare J, Kumbhar B, Dash C. Three Prime Repair Exonuclease 1 preferentially degrades the integration-incompetent HIV-1 DNA through favorable kinetics, thermodynamic, structural and conformational properties. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.19.585766. [PMID: 38562877 PMCID: PMC10983988 DOI: 10.1101/2024.03.19.585766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
HIV-1 integration into the human genome is dependent on 3'-processing of the reverse transcribed viral DNA. Recently, we reported that the cellular Three Prime Repair Exonuclease 1 (TREX1) enhances HIV-1 integration by degrading the unprocessed viral DNA, while the integration-competent 3'-processed DNA remained resistant. Here, we describe the mechanism by which the 3'-processed HIV-1 DNA resists TREX1-mediated degradation. Our kinetic studies revealed that the rate of cleavage (kcat) of the 3'-processed DNA was significantly lower than the unprocessed HIV-1 DNA by TREX1. The efficiency of degradation (kcat/KM) of the 3'-processed DNA was also significantly lower than the unprocessed DNA. Furthermore, the binding affinity (Kd) of TREX1 was markedly lower to the 3'-processed DNA compared to the unprocessed DNA. Molecular docking and dynamics studies revealed distinct conformational binding modes of TREX1 with the 3'-processed and unprocessed HIV-1 DNA. Particularly, the unprocessed DNA was favorably positioned in the active site with polar interactions with the catalytic residues of TREX1. Additionally, a stable complex was formed between TREX1 and the unprocessed DNA compared the 3'-processed DNA. These results pinpoint the biochemical mechanism by which TREX1 preferentially degrades the integration-incompetent HIV-1 DNA and reveal the unique structural and conformational properties of the integration-competent 3'-processed HIV-1 DNA.
Collapse
Affiliation(s)
- Prem Prakash
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College, Nashville, Tennessee, 37208, USA
| | - Purva Khodke
- Sunandan Divatia School of Science, NMIMS University, Mumbai, 400056, India
| | - Muthukumar Balasubramaniam
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College, Nashville, Tennessee, 37208, USA
| | - Benem-Orom Davids
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York City, New York, 10032, USA
| | - Thomas Hollis
- Department of Biochemistry and Center for Structural Biology, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - Jamaine Davis
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College, Nashville, Tennessee, 37208, USA
| | - Jui Pandhare
- Center for AIDS Health Disparities Research, Meharry Medical College, Nashville, Tennessee, 37208, USA
| | - Bajarang Kumbhar
- Sunandan Divatia School of Science, NMIMS University, Mumbai, 400056, India
| | - Chandravanu Dash
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College, Nashville, Tennessee, 37208, USA
- Center for AIDS Health Disparities Research, Meharry Medical College, Nashville, Tennessee, 37208, USA
- Department of Microbiology, Immunology, and Physiology, Meharry Medical College, Nashville, Tennessee, 37208, USA
| |
Collapse
|
22
|
Guo X, Yang L, Wang J, Wu Y, Li Y, Du L, Li L, Fang Z, Zhang X. The cytosolic DNA-sensing cGAS-STING pathway in neurodegenerative diseases. CNS Neurosci Ther 2024; 30:e14671. [PMID: 38459658 PMCID: PMC10924111 DOI: 10.1111/cns.14671] [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: 08/31/2023] [Revised: 02/10/2024] [Accepted: 02/27/2024] [Indexed: 03/10/2024] Open
Abstract
BACKGROUND With the widespread prevalence of neurodegenerative diseases (NDs) and high rates of mortality and disability, it is imminent to find accurate targets for intervention. There is growing evidence that neuroimmunity is pivotal in the pathology of NDs and that interventions targeting neuroimmunity hold great promise. Exogenous or dislocated nucleic acids activate the cytosolic DNA sensor cyclic GMP-AMP synthase (cGAS), activating the stimulator of interferon genes (STING). The activated STING triggers innate immune responses and then the cGAS-STING signaling pathway links abnormal nucleic acid sensing to the immune response. Recently, numerous studies have shown that neuroinflammation regulated by cGAS-STING signaling plays an essential role in NDs. AIMS In this review, we summarized the mechanism of cGAS-STING signaling in NDs and focused on inhibitors targeting cGAS-STING. CONCLUSION The cGAS-STING signaling plays an important role in the pathogenesis of NDs. Inhibiting the cGAS-STING signaling may provide new measures in the treatment of NDs.
Collapse
Affiliation(s)
- Xiaofeng Guo
- Department of Critical Care Medicine, Xijing HospitalThe Fourth Military Medical UniversityChina
- Department of Intensive Care UnitJoint Logistics Force No. 988 HospitalZhengzhouChina
| | - Lin Yang
- Department of Critical Care Medicine, Xijing HospitalThe Fourth Military Medical UniversityChina
| | - Jiawei Wang
- Department of Critical Care Medicine, Xijing HospitalThe Fourth Military Medical UniversityChina
| | - You Wu
- Department of Critical Care Medicine, Xijing HospitalThe Fourth Military Medical UniversityChina
| | - Yi Li
- Department of Critical Care Medicine, Xijing HospitalThe Fourth Military Medical UniversityChina
| | - Lixia Du
- Department of Critical Care Medicine, Xijing HospitalThe Fourth Military Medical UniversityChina
| | - Ling Li
- Department of Critical Care Medicine, Xijing HospitalThe Fourth Military Medical UniversityChina
| | - Zongping Fang
- Department of Critical Care Medicine, Xijing HospitalThe Fourth Military Medical UniversityChina
- Department of Anesthesiology, Xijing HospitalFourth Military Medical UniversityShaanxiChina
- Translational Research Institute of Brain and Brain‐Like Intelligence, Shanghai Fourth People's Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Xijing Zhang
- Department of Critical Care Medicine, Xijing HospitalThe Fourth Military Medical UniversityChina
| |
Collapse
|
23
|
Manthei KA, Munson LM, Nandakumar J, Simmons LA. Structural and biochemical characterization of the mitomycin C repair exonuclease MrfB. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.15.580553. [PMID: 38405983 PMCID: PMC10889028 DOI: 10.1101/2024.02.15.580553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Mitomycin C (MMC) repair factor A (mrfA) and factor B (mrfB), encode a conserved helicase and exonuclease that repair DNA damage in the soil-dwelling bacterium Bacillus subtilis. Here we have focused on the characterization of MrfB, a DEDDh exonuclease in the DnaQ superfamily. We solved the structure of the exonuclease core of MrfB to a resolution of 2.1 Å, in what appears to be an inactive state. In this conformation, a predicted α-helix containing the catalytic DEDDh residue Asp172 adopts a random coil, which moves Asp172 away from the active site and results in the occupancy of only one of the two catalytic Mg2+ ions. We propose that MrfB resides in this inactive state until it interacts with DNA to become activated. By comparing our structure to an AlphaFold prediction as well as other DnaQ-family structures, we located residues hypothesized to be important for exonuclease function. Using exonuclease assays we show that MrfB is a Mg2+-dependent 3'-5' DNA exonuclease. We show that Leu113 aids in coordinating the 3' end of the DNA substrate, and that a basic loop is important for substrate binding. This work provides insight into the function of a recently discovered bacterial exonuclease important for the repair of MMC-induced DNA adducts.
Collapse
Affiliation(s)
- Kelly A. Manthei
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Lia M. Munson
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Jayakrishnan Nandakumar
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Lyle A. Simmons
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
24
|
Wei X, Jing J, Huang R, Zhou T, Wu L, Ou G, Wu Y, Hu J, Zhu W, Wu Y, Li Y, Zhang S, You Z. QFAE-nB alleviates pulmonary fibrosis by inhibiting the STING pathway in mice. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117295. [PMID: 37806536 DOI: 10.1016/j.jep.2023.117295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 09/29/2023] [Accepted: 10/06/2023] [Indexed: 10/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Pulmonary fibrosis (PF) is an irreversible lung disease that severely affects human respiratory function. Traditionally, the natural plant Quzhou Fructus Arantii (QFA) has therapeutic effects on respiratory diseases. However, the effects and the mechanism of anti-fibrotic have not been elucidated. AIM OF THE STUDY In this study, QFAE-nB was extracted from QFA, the aims of this study include understanding the correlation between Bleomycin (BLM)-induced PF and STING pathway in mice, as well as exploring the role and mechanisms of QFAE-nB in the treatment of PF. MATERIALS AND METHODS QFAE-nB was extracted from QFA, six main chemical components in QFAE-nB were identified by HPLC-QTOF-MS/MS, and quantitative analysis was conducted by HPLC. qPCR and Western blot were used to verify the molecular mechanism of QFAE-nB, and the anti-fibrotic effect of QFAE-nB was determined by hematoxylin-eosin (HE) staining and Masson staining as well as immunohistochemistry. TREX1-KO and STING-KO mice were used to verify the relationship between STING and PF and the important target action of QFAE-nB. RESULTS Six main flavonoids in QFAE-nB were identified as eriocitrin (0.76%), neoeriocitrin (2.79%), narirutin (4.31%), naringin (35.41%), hesperidin (1.74%), and neohesperidin (27.18%). The results showed that BLM-induced PF was associated with its exacerbated release of proinflammatory factors and chemokines in lung tissues. In addition, QFAE-nB alleviated BLM-induced lung fibrosis in mice by inhibiting the activation of the STING signaling pathway and reducing the signal transduction of TBK1-IRF3 and TBK1-NF-κB pathways. Notably, knockout of the TREX1 gene caused massive inflammation and even induced PF in the lung tissues, whereas QFAE-nB effectively alleviated inflammation and reduced PF. The deletion of the STING gene suppressed BLM-induced PF and inflammation, but STING-KO mice treated with QFAE-nB showed even lower expression levels of proinflammatory factors and chemokine. CONCLUSIONS The STING pathway plays an important role in PF, and QFAE-nB alleviates PF by mainly targeting the inhibition of the STING pathway to reduce inflammation. Together, the study paves the way for targeting the STING pathway in PF treatment.
Collapse
Affiliation(s)
- Xueping Wei
- School of Public Health, Hangzhou Medical College, Hangzhou, China; Key Discipline of Zhejiang Province in Public Health and Preventive Medicine (First Class, Category A), Hangzhou Medical College, China
| | - Junsong Jing
- School of Public Health, Hangzhou Medical College, Hangzhou, China; Key Discipline of Zhejiang Province in Public Health and Preventive Medicine (First Class, Category A), Hangzhou Medical College, China
| | - Rongrong Huang
- School of Public Health, Hangzhou Medical College, Hangzhou, China; Key Discipline of Zhejiang Province in Public Health and Preventive Medicine (First Class, Category A), Hangzhou Medical College, China
| | - Ting Zhou
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, China
| | - Lianhao Wu
- School of Public Health, Hangzhou Medical College, Hangzhou, China; Key Discipline of Zhejiang Province in Public Health and Preventive Medicine (First Class, Category A), Hangzhou Medical College, China
| | - Guoteng Ou
- School of Public Health, Hangzhou Medical College, Hangzhou, China; Key Discipline of Zhejiang Province in Public Health and Preventive Medicine (First Class, Category A), Hangzhou Medical College, China
| | - Youping Wu
- The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Jingjin Hu
- School of Public Health, Hangzhou Medical College, Hangzhou, China; Key Discipline of Zhejiang Province in Public Health and Preventive Medicine (First Class, Category A), Hangzhou Medical College, China
| | - Wenwen Zhu
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, China
| | - Yueguo Wu
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, China
| | - Yuanyuan Li
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, China.
| | - Sheng Zhang
- Center for Safety Evaluation and Research, Hangzhou Medical College, Hangzhou, China.
| | - Zhenqiang You
- School of Public Health, Hangzhou Medical College, Hangzhou, China; School of Environmental and Chemical Engineering, Shanghai University, Shanghai, China; Key Discipline of Zhejiang Province in Public Health and Preventive Medicine (First Class, Category A), Hangzhou Medical College, China.
| |
Collapse
|
25
|
Luan X, Wang L, Song G, Zhou W. Innate immune responses to RNA: sensing and signaling. Front Immunol 2024; 15:1287940. [PMID: 38343534 PMCID: PMC10854198 DOI: 10.3389/fimmu.2024.1287940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 01/11/2024] [Indexed: 02/15/2024] Open
Abstract
Nucleic acids are among the most essential PAMPs (pathogen-associated molecular patterns). Animals have evolved numerous sensors to recognize nucleic acids and trigger immune signaling against pathogen replication, cellular stress and cancer. Many sensor proteins (e.g., cGAS, AIM2, and TLR9) recognize the molecular signature of infection or stress and are responsible for the innate immune response to DNA. Remarkably, recent evidence demonstrates that cGAS-like receptors acquire the ability to sense RNA in some forms of life. Compared with the nucleic-acid sensing by cGAS, innate immune responses to RNA are based on various RNA sensors, including RIG-I, MDA5, ADAR1, TLR3/7/8, OAS1, PKR, NLRP1/6, and ZBP1, via a broad-spectrum signaling axis. Importantly, new advances have brought to light the potential clinical application of targeting these signaling pathways. Here, we highlight the latest discoveries in the field. We also summarize the activation and regulatory mechanisms of RNA-sensing signaling. In addition, we discuss how RNA sensing is tightly controlled in cells and why the disruption of immune homeostasis is linked to disease.
Collapse
Affiliation(s)
- Xiaohan Luan
- Shenzhen Key Laboratory of Biomolecular Assembling and Regulation, Southern University of Science and Technology, Shenzhen, Guangdong, China
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Lei Wang
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Guangji Song
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Wen Zhou
- Shenzhen Key Laboratory of Biomolecular Assembling and Regulation, Southern University of Science and Technology, Shenzhen, Guangdong, China
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
| |
Collapse
|
26
|
Shim A, Luan X, Zhou W, Crow Y, Maciejowski J. Mutations in the non-catalytic polyproline motif destabilize TREX1 and amplify cGAS-STING signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.04.574136. [PMID: 38260344 PMCID: PMC10802300 DOI: 10.1101/2024.01.04.574136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
The cGAS-STING pathway detects cytosolic DNA and activates a signaling cascade that results in a type I interferon (IFN) response. The endoplasmic reticulum (ER)-associated exonuclease TREX1 suppresses cGAS-STING by eliminating DNA from the cytosol. Mutations that compromise TREX1 function are linked to autoinflammatory disorders, including systemic lupus erythematosus (SLE) and Aicardi-Goutières syndrome (AGS). Despite key roles in regulating cGAS-STING and suppressing excessive inflammation, the impact of many disease-associated TREX1 mutations - particularly those outside of the core catalytic domains - remains poorly understood. Here, we characterize a recessive AGS-linked TREX1 P61Q mutation occurring within the poorly characterized polyproline helix (PPII) motif. In keeping with its position outside of the catalytic core or ER targeting motifs, neither the P61Q mutation, nor aggregate proline-to-alanine PPII mutation, disrupt TREX1 exonuclease activity, subcellular localization, or cGAS-STING regulation in overexpression systems. Introducing targeted mutations into the endogenous TREX1 locus revealed that PPII mutations destabilize the protein, resulting in impaired exonuclease activity and unrestrained cGAS-STING activation. Overall, these results demonstrate that TREX1 PPII mutations, including P61Q, impair proper immune regulation and lead to autoimmune disease through TREX1 destabilization.
Collapse
Affiliation(s)
- Abraham Shim
- Molecular Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Xiaohan Luan
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Wen Zhou
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Yanick Crow
- MRC Human Genetics Unit, Institute of Genetics and Cancer, The University of Edinburgh, Edinburgh, UK
| | - John Maciejowski
- Molecular Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
27
|
Huang KW, Wu CY, Toh SI, Liu TC, Tu CI, Lin YH, Cheng AJ, Kao YT, Chu JW, Hsiao YY. Molecular insight into the specific enzymatic properties of TREX1 revealing the diverse functions in processing RNA and DNA/RNA hybrids. Nucleic Acids Res 2023; 51:11927-11940. [PMID: 37870446 PMCID: PMC10681709 DOI: 10.1093/nar/gkad910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 09/28/2023] [Accepted: 10/06/2023] [Indexed: 10/24/2023] Open
Abstract
In various autoimmune diseases, dysfunctional TREX1 (Three prime Repair Exonuclease 1) leads to accumulation of endogenous single-stranded DNA (ssDNA), double-stranded DNA (dsDNA) and DNA/RNA hybrids in the cytoplasm and triggers immune activation through the cGAS-STING pathway. Although inhibition of TREX1 could be a useful strategy for cancer immunotherapy, profiling cellular functions in terms of its potential substrates is a key step. Particularly important is the functionality of processing DNA/RNA hybrids and RNA substrates. The exonuclease activity measurements conducted here establish that TREX1 can digest both ssRNA and DNA/RNA hybrids but not dsRNA. The newly solved structures of TREX1-RNA product and TREX1-nucleotide complexes show that 2'-OH does not impose steric hindrance or specific interactions for the recognition of RNA. Through all-atom molecular dynamics simulations, we illustrate that the 2'-OH-mediated intra-chain hydrogen bonding in RNA would affect the binding with TREX1 and thereby reduce the exonuclease activity. This notion of higher conformational rigidity in RNA leading TREX1 to exhibit weaker catalytic cleavage is further validated by the binding affinity measurements with various synthetic DNA-RNA junctions. The results of this work thus provide new insights into the mechanism by which TREX1 processes RNA and DNA/RNA hybrids and contribute to the molecular-level understanding of the complex cellular functions of TREX1 as an exonuclease.
Collapse
Affiliation(s)
- Kuan-Wei Huang
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
- Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
| | - Chia-Yun Wu
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
- Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
| | - Shu-Ing Toh
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
- Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
| | - Tung-Chang Liu
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
- Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
| | - Chun-I Tu
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
- Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
| | - Yin-Hsin Lin
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
| | - An-Ju Cheng
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
| | - Ya-Ting Kao
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
- Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
- Institute of Bioinformatics and Systems Biology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
- Center for Intelligent Drug Systems and Smart Bio-devices (IDSB), National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
| | - Jhih-Wei Chu
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
- Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
- Institute of Bioinformatics and Systems Biology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
- Center for Intelligent Drug Systems and Smart Bio-devices (IDSB), National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
| | - Yu-Yuan Hsiao
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
- Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
- Institute of Bioinformatics and Systems Biology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
- Center for Intelligent Drug Systems and Smart Bio-devices (IDSB), National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
- Drug Development and Value Creation Research Center, Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
| |
Collapse
|
28
|
García-Giménez J, Córdoba-David G, Rayego-Mateos S, Cannata-Ortiz P, Carrasco S, Ruiz-Ortega M, Fernandez-Fernandez B, Ortiz A, Ramos AM. STING1 deficiency ameliorates immune-mediated crescentic glomerulonephritis in mice. J Pathol 2023; 261:309-322. [PMID: 37650295 DOI: 10.1002/path.6177] [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: 04/25/2023] [Revised: 07/10/2023] [Accepted: 07/20/2023] [Indexed: 09/01/2023]
Abstract
Rapidly progressive/crescentic glomerulonephritis (RPGN/CGN) involves the formation of glomerular crescents by maladaptive differentiation of parietal epithelial cells that leads to rapid loss of renal function. The molecular mechanisms of crescent formation are poorly understood. Therefore, new insights into molecular mechanisms could identify alternative therapeutic targets for RPGN/CGN. Analysis of kidney biopsies from patients with RPGN revealed increased interstitial, glomerular, and tubular expression of STING1, an accessory protein of the c-GAS-dependent DNA-sensing pathway, which was also observed in murine nephrotoxic nephritis induced by an anti-GBM antibody. STING1 was expressed by key cell types involved in RPGN and crescent formation such as glomerular parietal epithelial cells, and tubular cells as well as by inflammation accessory cells. In functional in vivo studies, Sting1-/- mice with nephrotoxic nephritis had lower kidney cytokine expression, milder kidney infiltration by innate and adaptive immune cells, and decreased disease severity. Pharmacological STING1 inhibition mirrored these findings. Direct STING1 agonism in parietal and tubular cells activated the NF-κB-dependent cytokine response and the interferon-induced genes (ISGs) program. These responses were also triggered in a STING1-dependent manner by the pro-inflammatory cytokine TWEAK. These results identify STING1 activation as a pathological mechanism in RPGN/CGN and TWEAK as an activator of STING1. Pharmacological strategies targeting STING1, or upstream regulators may therefore be potential alternatives to treat RPGN. © 2023 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Jorge García-Giménez
- Department of Nephrology and Hypertension, IIS-Fundación Jiménez Díaz-Universidad Autónoma de Madrid, Madrid, Spain
| | - Gina Córdoba-David
- Department of Nephrology and Hypertension, IIS-Fundación Jiménez Díaz-Universidad Autónoma de Madrid, Madrid, Spain
| | - Sandra Rayego-Mateos
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz-Universidad Autónoma de Madrid, Madrid, Spain
| | - Pablo Cannata-Ortiz
- Department of Pathology, IIS-Fundación Jiménez Díaz-Universidad Autónoma de Madrid, Madrid, Spain
| | - Susana Carrasco
- Department of Nephrology and Hypertension, IIS-Fundación Jiménez Díaz-Universidad Autónoma de Madrid, Madrid, Spain
| | - Marta Ruiz-Ortega
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz-Universidad Autónoma de Madrid, Madrid, Spain
- RICORS2040, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Beatriz Fernandez-Fernandez
- Department of Nephrology and Hypertension, IIS-Fundación Jiménez Díaz-Universidad Autónoma de Madrid, Madrid, Spain
- RICORS2040, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Medicine, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Alberto Ortiz
- Department of Nephrology and Hypertension, IIS-Fundación Jiménez Díaz-Universidad Autónoma de Madrid, Madrid, Spain
- RICORS2040, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Pharmacology, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Adrián M Ramos
- Department of Nephrology and Hypertension, IIS-Fundación Jiménez Díaz-Universidad Autónoma de Madrid, Madrid, Spain
- RICORS2040, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
29
|
Dudka D, Akins RB, Lampson MA. FREEDA: An automated computational pipeline guides experimental testing of protein innovation. J Cell Biol 2023; 222:e202212084. [PMID: 37358475 PMCID: PMC10292211 DOI: 10.1083/jcb.202212084] [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: 12/17/2022] [Revised: 04/22/2023] [Accepted: 06/07/2023] [Indexed: 06/27/2023] Open
Abstract
Cell biologists typically focus on conserved regions of a protein, overlooking innovations that can shape its function over evolutionary time. Computational analyses can reveal potential innovations by detecting statistical signatures of positive selection that lead to rapid accumulation of beneficial mutations. However, these approaches are not easily accessible to non-specialists, limiting their use in cell biology. Here, we present an automated computational pipeline FREEDA that provides a simple graphical user interface requiring only a gene name; integrates widely used molecular evolution tools to detect positive selection in rodents, primates, carnivores, birds, and flies; and maps results onto protein structures predicted by AlphaFold. Applying FREEDA to >100 centromere proteins, we find statistical evidence of positive selection within loops and turns of ancient domains, suggesting innovation of essential functions. As a proof-of-principle experiment, we show innovation in centromere binding of mouse CENP-O. Overall, we provide an accessible computational tool to guide cell biology research and apply it to experimentally demonstrate functional innovation.
Collapse
Affiliation(s)
- Damian Dudka
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - R. Brian Akins
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael A. Lampson
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
30
|
Mathavarajah S, Vergunst KL, Habib EB, Williams SK, He R, Maliougina M, Park M, Salsman J, Roy S, Braasch I, Roger A, Langelaan D, Dellaire G. PML and PML-like exonucleases restrict retrotransposons in jawed vertebrates. Nucleic Acids Res 2023; 51:3185-3204. [PMID: 36912092 PMCID: PMC10123124 DOI: 10.1093/nar/gkad152] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 02/16/2023] [Accepted: 02/21/2023] [Indexed: 03/14/2023] Open
Abstract
We have uncovered a role for the promyelocytic leukemia (PML) gene and novel PML-like DEDDh exonucleases in the maintenance of genome stability through the restriction of LINE-1 (L1) retrotransposition in jawed vertebrates. Although the mammalian PML protein forms nuclear bodies, we found that the spotted gar PML ortholog and related proteins in fish function as cytoplasmic DEDDh exonucleases. In contrast, PML proteins from amniote species localized both to the cytoplasm and formed nuclear bodies. We also identified the PML-like exon 9 (Plex9) genes in teleost fishes that encode exonucleases. Plex9 proteins resemble TREX1 but are unique from the TREX family and share homology to gar PML. We also characterized the molecular evolution of TREX1 and the first non-mammalian TREX1 homologs in axolotl. In an example of convergent evolution and akin to TREX1, gar PML and zebrafish Plex9 proteins suppressed L1 retrotransposition and could complement TREX1 knockout in mammalian cells. Following export to the cytoplasm, the human PML-I isoform also restricted L1 through its conserved C-terminus by enhancing ORF1p degradation through the ubiquitin-proteasome system. Thus, PML first emerged as a cytoplasmic suppressor of retroelements, and this function is retained in amniotes despite its new role in the assembly of nuclear bodies.
Collapse
Affiliation(s)
| | - Kathleen L Vergunst
- Department of Biochemistry & Molecular Biology, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Elias B Habib
- Department of Pathology, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Shelby K Williams
- Department of Biochemistry & Molecular Biology, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Raymond He
- Department of Biochemistry & Molecular Biology, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Maria Maliougina
- Department of Biochemistry & Molecular Biology, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Mika Park
- Department of Pathology, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Jayme Salsman
- Department of Pathology, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Stéphane Roy
- Department of Stomatology, Faculty of Dentistry, Université de Montréal, Montréal, QB, Canada
| | - Ingo Braasch
- Michigan State University, Department of Integrative Biology and Ecology, Evolution, and Behavior Program, East Lansing, MI, USA
| | - Andrew J Roger
- Department of Biochemistry & Molecular Biology, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - David N Langelaan
- Department of Biochemistry & Molecular Biology, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Graham Dellaire
- Department of Pathology, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
- Department of Biochemistry & Molecular Biology, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
31
|
Fang L, Ying S, Xu X, Wu D. TREX1 cytosolic DNA degradation correlates with autoimmune disease and cancer immunity. Clin Exp Immunol 2023; 211:193-207. [PMID: 36745566 PMCID: PMC10038326 DOI: 10.1093/cei/uxad017] [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: 10/24/2022] [Revised: 01/22/2023] [Accepted: 02/03/2023] [Indexed: 02/07/2023] Open
Abstract
The N-terminal domain of Three Prime Repair Exonuclease 1 (TREX1) is catalytically active and can degrade dsDNA or ssDNA in the cytosol, whereas the C-terminal domain is primarily involved in protein localization. TREX1 deficiency induces cytosolic DNA accumulation as well as activation of the cGAS-STING-IFN signaling pathway, which results in tissue inflammation and autoimmune diseases. Furthermore, TREX1 expression in cancer immunity can be adaptively regulated to promote tumor proliferation, making it a promising therapeutic target.
Collapse
Affiliation(s)
- Liwei Fang
- Pediatric Neurorehabilitation Center, Pediatric Department, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Songcheng Ying
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Xi Xu
- Department of Plastic Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - De Wu
- Pediatric Neurorehabilitation Center, Pediatric Department, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
32
|
Molecular Mechanisms of Neutrophil Extracellular Trap (NETs) Degradation. Int J Mol Sci 2023; 24:ijms24054896. [PMID: 36902325 PMCID: PMC10002918 DOI: 10.3390/ijms24054896] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/24/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023] Open
Abstract
Although many studies have been exploring the mechanisms driving NETs formation, much less attention has been paid to the degradation and elimination of these structures. The NETs clearance and the effective removal of extracellular DNA, enzymatic proteins (neutrophil elastase, proteinase 3, myeloperoxidase) or histones are necessary to maintain tissue homeostasis, to prevent inflammation and to avoid the presentation of self-antigens. The persistence and overabundance of DNA fibers in the circulation and tissues may have dramatic consequences for a host leading to the development of various systemic and local damage. NETs are cleaved by a concerted action of extracellular and secreted deoxyribonucleases (DNases) followed by intracellular degradation by macrophages. NETs accumulation depends on the ability of DNase I and DNAse II to hydrolyze DNA. Furthermore, the macrophages actively engulf NETs and this event is facilitated by the preprocessing of NETs by DNase I. The purpose of this review is to present and discuss the current knowledge about the mechanisms of NETs degradation and its role in the pathogenesis of thrombosis, autoimmune diseases, cancer and severe infections, as well as to discuss the possibilities for potential therapeutic interventions. Several anti-NETs approaches had therapeutic effects in animal models of cancer and autoimmune diseases; nevertheless, the development of new drugs for patients needs further study for an effective development of clinical compounds that are able to target NETs.
Collapse
|
33
|
Dudka D, Akins RB, Lampson MA. FREEDA: an automated computational pipeline guides experimental testing of protein innovation by detecting positive selection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.27.530329. [PMID: 36909479 PMCID: PMC10002610 DOI: 10.1101/2023.02.27.530329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
Abstract
Cell biologists typically focus on conserved regions of a protein, overlooking innovations that can shape its function over evolutionary time. Computational analyses can reveal potential innovations by detecting statistical signatures of positive selection that leads to rapid accumulation of beneficial mutations. However, these approaches are not easily accessible to non-specialists, limiting their use in cell biology. Here, we present an automated computational pipeline FREEDA (Finder of Rapidly Evolving Exons in De novo Assemblies) that provides a simple graphical user interface requiring only a gene name, integrates widely used molecular evolution tools to detect positive selection, and maps results onto protein structures predicted by AlphaFold. Applying FREEDA to >100 mouse centromere proteins, we find evidence of positive selection in intrinsically disordered regions of ancient domains, suggesting innovation of essential functions. As a proof-of-principle experiment, we show innovation in centromere binding of CENP-O. Overall, we provide an accessible computational tool to guide cell biology research and apply it to experimentally demonstrate functional innovation.
Collapse
|
34
|
Berger MB, Cisneros GA. Distal Mutations in the β-Clamp of DNA Polymerase III* Disrupt DNA Orientation and Affect Exonuclease Activity. J Am Chem Soc 2023; 145:3478-3490. [PMID: 36745735 PMCID: PMC10237177 DOI: 10.1021/jacs.2c11713] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
DNA polymerases are responsible for the replication and repair of DNA found in all DNA-based organisms. DNA Polymerase III is the main replicative polymerase of E. coli and is composed of over 10 proteins. A subset of these proteins (Pol III*) includes the polymerase (α), exonuclease (ϵ), clamp (β), and accessory protein (θ). Mutations of residues in, or around the active site of the catalytic subunits (α and ϵ), can have a significant impact on catalysis. However, the effects of distal mutations in noncatalytic subunits on the activity of catalytic subunits are less well-characterized. Here, we investigate the effects of two Pol III* variants, β-L82E/L82'E and β-L82D/L82'D, on the proofreading reaction catalyzed by ϵ. MD simulations reveal major changes in the dynamics of Pol III*, which extend throughout the complex. These changes are mostly induced by a shift in the position of the DNA substrate inside the β-clamp, although no major structural changes are observed in the protein complex. Quantum mechanics/molecular mechanics (QM/MM) calculations indicate that the β-L82D/L82'D variant has reduced catalytic proficiency due to highly endoergic reaction energies resulting from structural changes in the active site and differences in the electric field at the active site arising from the protein and substrate. Conversely, the β-L82E/L82'E variant is predicted to maintain proofreading activity, exhibiting a similar reaction barrier for nucleotide excision compared with the WT system. However, significant differences in the reaction mechanism are obtained due to the changes induced by the mutations on the β-clamp.
Collapse
Affiliation(s)
- Madison B Berger
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, Texas 75080, United States
| | - G Andrés Cisneros
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, Texas 75080, United States
- Department of Physics, University of Texas at Dallas, Richardson, Texas 75080, United States
| |
Collapse
|
35
|
Bin Khathlan Y, Almutairi S, Albadr FB, Alangari AA, Alsultan A. Case report: Durable response to ruxolitinib in a child with TREX1-related disorder. Front Pediatr 2023; 11:1178919. [PMID: 37187582 PMCID: PMC10175768 DOI: 10.3389/fped.2023.1178919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 04/10/2023] [Indexed: 05/17/2023] Open
Abstract
Background JAK inhibitors are useful in treating interferonopathies, presumably because they downregulate the JAK/STAT signaling. There are limited studies about the safety and effectiveness of using JAK inhibitors in children with TREX1-related disorders. Case presentation We report an 8-year-old female who presented at five years of age with features suggestive of hemophagocytic lymphohistiocytosis (HLH)-like disorder. The infectious disease workup was negative. Neurological assessment was normal. A brain CT scan was performed because of headache. It showed a faint subcortical calcification at right frontal lobe and almost symmetrical calcification within the basal ganglia. Brain MRI showed bilateral symmetrical globus pallidus, high T1 signal intensities, and a few scattered nonspecific FLAIR hyperintensities in subcortical and deep white matter. IVIG as an immune modulating agent was administered initially which led to the resolution of fever, improvement of blood count parameters, inflammatory markers, and normalization of liver enzymes. The child remained afebrile with no significant events for several months, then had disease flare up. The patient was started on pulse methylprednisolone 30 mg/kg for three days, then continued on 2 mg/kg. Whole exome sequencing revealed a novel heterozygous missense TREX1 mutation NM_016381.3:c.223G > A p.(Glu75Lys). The child was started on ruxolitinib, 5 mg orally twice daily. The child has prolonged, durable remission after initiating ruxolitinib with no adverse effects. Steroids were tapered off and the patient is no longer on IVIG. The patient is still on ruxolitinib for more than two years. Conclusion This case highlights the potential role of ruxolitinib in the treatment of TREX1-related disorders. A longer follow-up period is required to evaluate the long-term outcome.
Collapse
Affiliation(s)
- Yasir Bin Khathlan
- Department of Pediatrics, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Sajdi Almutairi
- Department of Pediatrics, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Fahad B. Albadr
- Department of Radiology and Medical Imaging, King Saud University Medical City and College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Abdullah A. Alangari
- Department of Pediatrics, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Abdulrahman Alsultan
- Department of Pediatrics, College of Medicine, King Saud University, Riyadh, Saudi Arabia
- Oncology Center, King Saud University Medical City, Riyadh, Saudi Arabia
- Correspondence: Abdulrahman Alsultan
| |
Collapse
|
36
|
Kimura S, Srisuknimit V, McCarty KL, Dedon PC, Kranzusch PJ, Waldor MK. Sequential action of a tRNA base editor in conversion of cytidine to pseudouridine. Nat Commun 2022; 13:5994. [PMID: 36220828 PMCID: PMC9553926 DOI: 10.1038/s41467-022-33714-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 09/28/2022] [Indexed: 11/08/2022] Open
Abstract
Post-transcriptional RNA editing modulates gene expression in a condition-dependent fashion. We recently discovered C-to-Ψ editing in Vibrio cholerae tRNA. Here, we characterize the biogenesis, regulation, and functions of this previously undescribed RNA editing process. We show that an enzyme, TrcP, mediates the editing of C-to-U followed by the conversion of U to Ψ, consecutively. AlphaFold-2 predicts that TrcP consists of two globular domains (cytidine deaminase and pseudouridylase) and a long helical domain. The latter domain tethers tRNA substrates during both the C-to-U editing and pseudouridylation, likely enabling a substrate channeling mechanism for efficient catalysis all the way to the terminal product. C-to-Ψ editing both requires and suppresses other modifications, creating an interdependent network of modifications in the tRNA anticodon loop that facilitates coupling of tRNA modification states to iron availability. Our findings provide mechanistic insights into an RNA editing process that likely promotes environmental adaptation.
Collapse
Affiliation(s)
- Satoshi Kimura
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA.
- Department of Microbiology, Harvard Medical School, Boston, MA, USA.
- Howard Hughes Medical Institute, Boston, MA, USA.
| | - Veerasak Srisuknimit
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
| | - Kacie L McCarty
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Peter C Dedon
- Department of Biological Engineering, Massachusetts Institution of Technology, Cambridge, MA, USA
- Singapore-MIT Alliance for Research and Technology Antimicrobial Resistance Interdisciplinary Research Group, Singapore, Singapore
| | - Philip J Kranzusch
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Matthew K Waldor
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA.
- Department of Microbiology, Harvard Medical School, Boston, MA, USA.
- Howard Hughes Medical Institute, Boston, MA, USA.
| |
Collapse
|