Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 273
Filter
1.
Genes Dev ; 35(9-10): 602-618, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33888558

ABSTRACT

The DNA damage response (DDR) fulfils essential roles to preserve genome integrity. Targeting the DDR in tumors has had remarkable success over the last decade, exemplified by the licensing of PARP inhibitors for cancer therapy. Recent studies suggest that the application of DDR inhibitors impacts on cellular innate and adaptive immune responses, wherein key DNA repair factors have roles in limiting chronic inflammatory signaling. Antitumor immunity plays an emerging part in cancer therapy, and extensive efforts have led to the development of immune checkpoint inhibitors overcoming immune suppressive signals in tumors. Here, we review the current understanding of the molecular mechanisms underlying DNA damage-triggered immune responses, including cytosolic DNA sensing via the cGAS/STING pathway. We highlight the implications of DDR components for therapeutic outcomes of immune checkpoint inhibitors or their use as biomarkers. Finally, we discuss the rationale for novel combinations of DDR inhibitors with antagonists of immune checkpoints and current hindrances limiting their broader therapeutic applications.


Subject(s)
DNA Repair/physiology , Immunity, Cellular/genetics , Immunotherapy , Neoplasms/therapy , Adaptive Immunity/genetics , DNA Damage/immunology , Discoidin Domain Receptors/antagonists & inhibitors , Humans , Immune Checkpoint Inhibitors/therapeutic use , Neoplasms/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Protein Kinase Inhibitors/therapeutic use
2.
Nat Immunol ; 17(3): 230-40, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26882261

ABSTRACT

Inflammation is emerging as one of the hallmarks of cancer, yet its role in most tumors remains unclear. Whereas a minority of solid tumors are associated with overt inflammation, long-term treatment with non-steroidal anti-inflammatory drugs is remarkably effective in reducing cancer rate and death. This indicates that inflammation might have many as-yet-unrecognized facets, among which an indolent course might be far more prevalent than previously appreciated. In this Review, we explore the various inflammatory processes underlying the development and progression of colorectal cancer and discuss anti-inflammatory means for its prevention and treatment.


Subject(s)
Adenocarcinoma/immunology , Adenoma/immunology , Carcinogenesis/immunology , Colorectal Neoplasms/immunology , Dysbiosis/immunology , Gastrointestinal Microbiome/immunology , Inflammation , Inflammatory Bowel Diseases/immunology , Intestinal Mucosa/immunology , Adenocarcinoma/drug therapy , Adenocarcinoma/prevention & control , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/prevention & control , DNA Damage/immunology , Disease Progression , Humans , Immunity, Innate/immunology , Immunity, Mucosal/immunology , Interleukin-1beta/antagonists & inhibitors , Janus Kinases/antagonists & inhibitors , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Receptors, Interleukin-6/antagonists & inhibitors , STAT Transcription Factors/antagonists & inhibitors , Tumor Necrosis Factor-alpha/antagonists & inhibitors
3.
Nature ; 594(7861): 100-105, 2021 06.
Article in English | MEDLINE | ID: mdl-33981041

ABSTRACT

Ageing of the immune system, or immunosenescence, contributes to the morbidity and mortality of the elderly1,2. To define the contribution of immune system ageing to organism ageing, here we selectively deleted Ercc1, which encodes a crucial DNA repair protein3,4, in mouse haematopoietic cells to increase the burden of endogenous DNA damage and thereby senescence5-7 in the immune system only. We show that Vav-iCre+/-;Ercc1-/fl mice were healthy into adulthood, then displayed premature onset of immunosenescence characterized by attrition and senescence of specific immune cell populations and impaired immune function, similar to changes that occur during ageing in wild-type mice8-10. Notably, non-lymphoid organs also showed increased senescence and damage, which suggests that senescent, aged immune cells can promote systemic ageing. The transplantation of splenocytes from Vav-iCre+/-;Ercc1-/fl or aged wild-type mice into young mice induced senescence in trans, whereas the transplantation of young immune cells attenuated senescence. The treatment of Vav-iCre+/-;Ercc1-/fl mice with rapamycin reduced markers of senescence in immune cells and improved immune function11,12. These data demonstrate that an aged, senescent immune system has a causal role in driving systemic ageing and therefore represents a key therapeutic target to extend healthy ageing.


Subject(s)
Aging/immunology , Aging/physiology , Immune System/immunology , Immune System/physiology , Immunosenescence/immunology , Immunosenescence/physiology , Organ Specificity/immunology , Organ Specificity/physiology , Aging/drug effects , Aging/pathology , Animals , DNA Damage/immunology , DNA Damage/physiology , DNA Repair/immunology , DNA Repair/physiology , DNA-Binding Proteins/genetics , Endonucleases/genetics , Female , Healthy Aging/immunology , Healthy Aging/physiology , Homeostasis/immunology , Homeostasis/physiology , Immune System/drug effects , Immunosenescence/drug effects , Male , Mice , Organ Specificity/drug effects , Rejuvenation , Sirolimus/pharmacology , Spleen/cytology , Spleen/transplantation
4.
Immunity ; 45(4): 903-916, 2016 10 18.
Article in English | MEDLINE | ID: mdl-27742546

ABSTRACT

Immune aging manifests with a combination of failing adaptive immunity and insufficiently restrained inflammation. In patients with rheumatoid arthritis (RA), T cell aging occurs prematurely, but the mechanisms involved and their contribution to tissue-destructive inflammation remain unclear. We found that RA CD4+ T cells showed signs of aging during their primary immune responses and differentiated into tissue-invasive, proinflammatory effector cells. RA T cells had low expression of the double-strand-break repair nuclease MRE11A, leading to telomeric damage, juxtacentromeric heterochromatin unraveling, and senescence marker upregulation. Inhibition of MRE11A activity in healthy T cells induced the aging phenotype, whereas MRE11A overexpression in RA T cells reversed it. In human-synovium chimeric mice, MRE11Alow T cells were tissue-invasive and pro-arthritogenic, and MRE11A reconstitution mitigated synovitis. Our findings link premature T cell aging and tissue-invasiveness to telomere deprotection and heterochromatin unpacking, identifying MRE11A as a therapeutic target to combat immune aging and suppress dysregulated tissue inflammation.


Subject(s)
Arthritis, Rheumatoid/immunology , Cellular Senescence/immunology , DNA-Binding Proteins/immunology , Deoxyribonucleases/immunology , T-Lymphocytes/immunology , Animals , Cells, Cultured , DNA Damage/immunology , DNA Repair/immunology , Female , Humans , Inflammation/immunology , Leukocytes, Mononuclear/immunology , Mice , Synovitis/immunology , Telomere/immunology , Up-Regulation/immunology
5.
BMC Cancer ; 24(1): 561, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38711034

ABSTRACT

Modulation of DNA damage repair in lung squamous cell carcinoma (LUSC) can result in the generation of neoantigens and heightened immunogenicity. Therefore, understanding DNA damage repair mechanisms holds significant clinical relevance for identifying targets for immunotherapy and devising therapeutic strategies. Our research has unveiled that the tumor suppressor zinc finger protein 750 (ZNF750) in LUSC binds to the promoter region of tenascin C (TNC), leading to reduced TNC expression. This modulation may impact the malignant behavior of tumor cells and is associated with patient prognosis. Additionally, single-cell RNA sequencing (scRNA-seq) of LUSC tissues has demonstrated an inverse correlation between ZNF750/TNC expression levels and immunogenicity. Manipulation of the ZNF750-TNC axis in vitro within LUSC cells has shown differential sensitivity to CD8+ cells, underscoring its pivotal role in regulating cellular immunogenicity. Further transcriptome sequencing analysis, DNA damage repair assay, and single-strand break analyses have revealed the involvement of the ZNF750-TNC axis in determining the preference for homologous recombination (HR) repair or non-homologous end joining (NHEJ) repair of DNA damage. with involvement of the Hippo/ERK signaling pathway. In summary, this study sheds light on the ZNF750-TNC axis's role in DNA damage repair regulation in LUSC, laying a groundwork for future translational research in immune cell therapy for LUSC.


Subject(s)
Carcinoma, Squamous Cell , DNA Damage , Lung Neoplasms , Tenascin , Humans , Lung Neoplasms/immunology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Tenascin/genetics , Tenascin/metabolism , DNA Damage/immunology , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Transcription Factors/metabolism , Transcription Factors/genetics , Promoter Regions, Genetic , Prognosis , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism
6.
Nat Immunol ; 12(8): 778-85, 2011 Jul 03.
Article in English | MEDLINE | ID: mdl-21725321

ABSTRACT

The transcription factors that regulate differentiation into the monocyte subset in bone marrow have not yet been identified. Here we found that the orphan nuclear receptor NR4A1 controlled the differentiation of Ly6C- monocytes. Ly6C- monocytes, which function in a surveillance role in circulation, were absent from Nr4a1-/- mice. Normal numbers of myeloid progenitor cells were present in Nr4a1-/- mice, which indicated that the defect occurred during later stages of monocyte development. The defect was cell intrinsic, as wild-type mice that received bone marrow from Nr4a1-/- mice developed fewer patrolling monocytes than did recipients of wild-type bone marrow. The Ly6C- monocytes remaining in the bone marrow of Nr4a1-/- mice were arrested in S phase of the cell cycle and underwent apoptosis. Thus, NR4A1 functions as a master regulator of the differentiation and survival of 'patrolling' Ly6C- monocytes.


Subject(s)
Antigens, Ly/immunology , Apoptosis/immunology , Bone Marrow/immunology , Cell Differentiation/immunology , Monocytes/immunology , Nuclear Receptor Subfamily 4, Group A, Member 1/immunology , Animals , Cell Cycle/immunology , DNA Damage/immunology , Flow Cytometry , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , RNA, Messenger/chemistry , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Specific Pathogen-Free Organisms
7.
J Neurosci ; 41(45): 9286-9307, 2021 11 10.
Article in English | MEDLINE | ID: mdl-34593604

ABSTRACT

Adult-onset neurodegenerative diseases are often accompanied by evidence of a chronic inflammation that includes activation of microglial cells and altered levels of brain cytokines. Aspects of this response are likely secondary reactions to neurodegeneration, but for many illnesses the inflammation may itself be an early and even causative disease event. In such cases, the inflammation is referred to as "sterile" as it occurs in the absence of an actual bacterial or viral pathogen. A potent trigger of sterile inflammation in CNS microglia has been shown to be the presence of DNA in the cytoplasm (cytoDNA) induced either by direct DNA damage or by inhibited DNA repair. We have shown that cytoDNA comes from the cell nucleus as a result of insufficient DNA damage repair. Using wild-type and Atm-/- mouse microglia, we extend these observations here by showing that its genomic origins are not random, but rather are heavily biased toward transcriptionally inactive, intergenic regions, in particular repetitive elements and AT-rich sequences. Once released from the genome, in both males and females, we show that cytoDNA is actively exported to the cytoplasm by a CRM1-dependent mechanism. In the cytoplasm, it is degraded either by a cytosolic exonuclease, Trex1, or an autophagy pathway that ends with degradation in the lysosome. Blocking the accumulation of cytoDNA prevents the emergence of the sterile inflammation reaction. These findings offer new insights into the emergence of sterile inflammation and offer novel approaches that may be of use in combatting a wide range of neurodegenerative conditions.SIGNIFICANCE STATEMENT Sterile inflammation describes a state where the defenses of the immune system are activated in the absence of a true pathogen. A potent trigger of this unorthodox response is the presence of DNA in the cytoplasm, which immune cells interpret as an invading virus or pathogen. We show that when DNA damage increases, fragments of the cell's own genome are actively exported to the cytoplasm where they are normally degraded. If this degradation is incomplete an immune reaction is triggered. Both age and stress increase DNA damage, and as age-related neurodegenerative diseases are frequently accompanied by a chronic low-level inflammation, strategies that reduce the induction of cytoplasmic DNA or speed its clearance become attractive therapeutic targets.


Subject(s)
Cytoplasm/immunology , DNA Damage/immunology , DNA/immunology , Inflammation/immunology , Repetitive Sequences, Nucleic Acid/immunology , Animals , Cytoplasm/metabolism , DNA/metabolism , DNA Repair , Female , Inflammation/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/immunology , Microglia/metabolism
8.
J Virol ; 95(16): e0076021, 2021 07 26.
Article in English | MEDLINE | ID: mdl-34037418

ABSTRACT

Pseudorabies virus (PRV) is the causative pathogen of Aujeszky's disease in pigs. Although vaccination is currently applied to prevent the morbidity of PRV infection, new applications are urgently needed to control this infectious disease. Poly(ADP-ribose) polymerase 1 (PARP1) functions in DNA damage repair. We report here that pharmacological and genetic inhibition of PARP1 significantly influenced PRV replication. Moreover, we demonstrate that inhibition of PARP1 induced DNA damage response and antiviral innate immunity. Mechanistically, PARP1 inhibition-induced DNA damage response resulted in the release of double-stranded DNA (dsDNA) into the cytosol, where dsDNA interacted with cyclic GMP-AMP (cGAMP) synthase (cGAS). cGAS subsequently catalyzed cGAMP production to activate the STING/TBK1/IRF3 innate immune signaling pathway. Furthermore, challenge of mice with PARP1 inhibitor stimulated antiviral innate immunity and protected mice from PRV infection in vivo. Our results demonstrate that PARP1 inhibitors may be used as a new strategy to prevent Aujeszky's disease in pigs. IMPORTANCE Aujeszky's disease is a notifiable infectious disease of pigs and causes economic losses worldwide in the pig industry. The causative pathogen is PRV, which is a member of the subfamily Alphaherpesvirinae of the family Herpesviridae. PRV has a wide range of hosts, such as ruminants, carnivores, and rodents. More seriously, recent reports suggest that PRV can cause human endophthalmitis and encephalitis, which indicates that PRV may be a potential zoonotic pathogen. Although vaccination is currently the major strategy used to control the disease, new applications are also urgently needed for the pig industry and public health. We report here that inhibition of PARP1 induces DNA damage-induced antiviral innate immunity through the cGAS-STING signaling pathway. Therefore, PARP1 is a therapeutic target for PRV infection as well as alphaherpesvirus infection.


Subject(s)
Antiviral Agents/immunology , DNA Damage/immunology , Immunity, Innate/drug effects , Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors , Pseudorabies/drug therapy , Animals , Antiviral Agents/pharmacology , Cell Line , Herpesvirus 1, Suid/drug effects , Herpesvirus 1, Suid/physiology , Humans , Membrane Proteins/metabolism , Mice , Nucleotidyltransferases/metabolism , Poly (ADP-Ribose) Polymerase-1/genetics , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Pseudorabies/immunology , Signal Transduction/drug effects , Signal Transduction/immunology , Swine , Virus Replication/drug effects
9.
PLoS Pathog ; 16(3): e1008429, 2020 03.
Article in English | MEDLINE | ID: mdl-32208449

ABSTRACT

Chromatin dynamics regulated by epigenetic modification is crucial in genome stability and gene expression. Various epigenetic mechanisms have been identified in the pathogenesis of human diseases. Here, we examined the effects of ten epigenetic agents on pseudorabies virus (PRV) infection by using GFP-reporter assays. Inhibitors of bromodomain protein 4 (BRD4), which receives much more attention in cancer than viral infection, was found to exhibit substantial anti-viral activity against PRV as well as a range of DNA and RNA viruses. We further demonstrated that BRD4 inhibition boosted a robust innate immune response. BRD4 inhibition also de-compacted chromatin structure and induced the DNA damage response, thereby triggering the activation of cGAS-mediated innate immunity and increasing host resistance to viral infection both in vitro and in vivo. Mechanistically, the inhibitory effect of BRD4 inhibition on viral infection was mainly attributed to the attenuation of viral attachment. Our findings reveal a unique mechanism through which BRD4 inhibition restrains viral infection and points to its potent therapeutic value for viral infectious diseases.


Subject(s)
Cell Cycle Proteins/immunology , DNA Damage/immunology , DNA Viruses/immunology , Immunity, Innate , Nuclear Proteins/immunology , RNA Viruses/immunology , Transcription Factors/immunology , A549 Cells , Animals , Chlorocebus aethiops , DNA Virus Infections/immunology , Dogs , Female , HEK293 Cells , HeLa Cells , Humans , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred BALB C , NIH 3T3 Cells , RAW 264.7 Cells , RNA Virus Infections/immunology , Swine , Vero Cells
10.
Hepatology ; 74(5): 2380-2394, 2021 11.
Article in English | MEDLINE | ID: mdl-34110660

ABSTRACT

BACKGROUND AND AIMS: Hepatitis C virus (HCV) leads to a high rate of chronic infection and T cell dysfunction. Although it is well known that chronic antigenic stimulation is a driving force for impaired T cell functions, the precise mechanisms underlying immune activation-induced T cell dysfunctions during HCV infection remain elusive. APPROACH AND RESULTS: Here, we demonstrated that circulating CD4+ T cells from patients who are chronically HCV-infected exhibit an immune activation status, as evidenced by the overexpression of cell activation markers human leukocyte antigen-antigen D-related, glucose transporter 1, granzyme B, and the short-lived effector marker CD127- killer cell lectin-like receptor G1+ . In contrast, the expression of stem cell-like transcription factor T cell factor 1 and telomeric repeat-binding factor 2 (TRF2) are significantly reduced in CD4+ T cells from patients who are chronically HCV-infected compared with healthy participants (HP). Mechanistic studies revealed that CD4+ T cells from participants with HCV exhibit phosphoinositide 3-kinase/Akt/mammalian target of rapamycin signaling hyperactivation on T cell receptor stimulation, promoting proinflammatory effector cell differentiation, telomeric DNA damage, and cellular apoptosis. Inhibition of Akt signaling during T cell activation preserved the precursor memory cell population and prevented inflammatory effector cell expansion, DNA damage, and apoptotic death. Moreover, knockdown of TRF2 reduced HP T cell stemness and triggered telomeric DNA damage and cellular apoptosis, whereas overexpression of TRF2 in CD4 T cells prevented telomeric DNA damage. CONCLUSIONS: These results suggest that modulation of immune activation through inhibiting Akt signaling and protecting telomeres through enhancing TRF2 expression may open therapeutic strategies to fine tune the adaptive immune responses in the setting of persistent immune activation and inflammation during chronic HCV infection.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , DNA Damage/immunology , Hepacivirus/genetics , Hepatitis C, Chronic/genetics , Hepatitis C, Chronic/immunology , Telomere/genetics , Adult , Aged , Apoptosis/genetics , Apoptosis/immunology , Cells, Cultured , DNA Damage/genetics , Female , Gene Knockdown Techniques/methods , Hepatitis C, Chronic/virology , Humans , Lymphocyte Activation , Male , Middle Aged , Persistent Infection/genetics , Persistent Infection/immunology , Persistent Infection/virology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Viral/genetics , Signal Transduction/genetics , Signal Transduction/immunology , TOR Serine-Threonine Kinases/metabolism , Telomeric Repeat Binding Protein 2/genetics , Telomeric Repeat Binding Protein 2/metabolism , Transduction, Genetic/methods , Young Adult
11.
J Immunol ; 205(8): 2222-2230, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32887747

ABSTRACT

CRISPR/Cas9 technology has revolutionized rapid and reliable gene editing in cells. Although many cell types have been subjected to CRISPR/Cas9-mediated gene editing, there is no evidence of success in genetic alteration of Ag-experienced memory CD8 T cells. In this study, we show that CRISPR/Cas9-mediated gene editing in memory CD8 T cells precludes their proliferation after Ag re-encounter in vivo. This defect is mediated by the proapoptotic transcription factor p53, a sensor of DNA damage. Temporarily inhibiting p53 function offers a window of opportunity for the memory CD8 T cells to repair the DNA damage, facilitating robust recall responses on Ag re-encounter. We demonstrate this by functionally altering memory CD8 T cells using CRISPR/Cas9-mediated targeted gene disruption under the aegis of p53siRNA in the mouse model. Our approach thus adapts the CRISPR/Cas9 technology for memory CD8 T cells to undertake gene editing in vivo, for the first time, to our knowledge.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CRISPR-Cas Systems , Cell Proliferation/genetics , Immunologic Memory/genetics , Tumor Suppressor Protein p53 , Animals , Antigens/immunology , DNA Damage/genetics , DNA Damage/immunology , Mice , Mice, Transgenic , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/immunology
12.
J Immunol ; 204(6): 1437-1447, 2020 03 15.
Article in English | MEDLINE | ID: mdl-32034061

ABSTRACT

DNA damage could lead to the accumulation of cytosolic DNA, and the cytosolic DNA-sensing pathway has been implicated in multiple inflammatory diseases. However, the role of cytosolic DNA-sensing pathway in asthma pathogenesis is still unclear. This article explored the role of airway epithelial cyclic GMP-AMP synthase (cGAS), the major sensor of cytosolic dsDNA, in asthma pathogenesis. Cytosolic dsDNA accumulation in airway epithelial cells (ECs) was detected in the setting of allergic inflammation both in vitro and in vivo. Mice with cGAS deletion in airway ECs were used for OVA- or house dust mite (HDM)-induced allergic airway inflammation. Additionally, the effects of cGAS knockdown on IL-33-induced GM-CSF production and the mechanisms by which IL-33 induced cytosolic dsDNA accumulation in human bronchial epithelial (HBE) cells were explored. Increased accumulation of cytosolic dsDNA was observed in airway epithelium of OVA- or HDM-challenged mice and in HBE cells treated with IL-33. Deletion of cGAS in the airway ECs of mice significantly attenuated the allergic airway inflammation induced by OVA or HDM. Mechanistically, cGAS participates in promoting TH2 immunity likely via regulating the production of airway epithelial GM-CSF. Furthermore, Mito-TEMPO could reduce IL-33-induced cytoplasmic dsDNA accumulation in HBE cells possibly through suppressing the release of mitochondrial DNA into the cytosol. In conclusion, airway epithelial cGAS plays an important role via sensing the cytosolic dsDNA in asthma pathogenesis and could serve as a promising therapeutic target against allergic airway inflammation.


Subject(s)
Airway Remodeling/immunology , Asthma/immunology , Epithelial Cells/immunology , Nucleotidyltransferases/metabolism , Respiratory Mucosa/immunology , Allergens/administration & dosage , Allergens/immunology , Animals , Antigens, Dermatophagoides/administration & dosage , Antigens, Dermatophagoides/immunology , Asthma/pathology , Cytosol/immunology , Cytosol/metabolism , DNA Damage/immunology , DNA, Mitochondrial/immunology , DNA, Mitochondrial/metabolism , Dermatophagoides pteronyssinus/immunology , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gene Knockdown Techniques , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Interleukin-33/immunology , Interleukin-33/metabolism , Mice , Mice, Transgenic , Mitochondria/metabolism , Nucleotidyltransferases/genetics , Ovalbumin/administration & dosage , Ovalbumin/immunology , Respiratory Mucosa/cytology , Respiratory Mucosa/pathology
13.
Nature ; 536(7614): 91-5, 2016 08 04.
Article in English | MEDLINE | ID: mdl-27350335

ABSTRACT

Recognition of neoantigens that are formed as a consequence of DNA damage is likely to form a major driving force behind the clinical activity of cancer immunotherapies such as T-cell checkpoint blockade and adoptive T-cell therapy. Therefore, strategies to selectively enhance T-cell reactivity against genetically defined neoantigens are currently under development. In mouse models, T-cell pressure can sculpt the antigenicity of tumours, resulting in the emergence of tumours that lack defined mutant antigens. However, whether the T-cell-recognized neoantigen repertoire in human cancers is constant over time is unclear. Here we analyse the stability of neoantigen-specific T-cell responses and the antigens they recognize in two patients with stage IV melanoma treated by adoptive T-cell transfer. The T-cell-recognized neoantigens can be selectively lost from the tumour cell population, either by overall reduced expression of the genes or loss of the mutant alleles. Notably, loss of expression of T-cell-recognized neoantigens was accompanied by development of neoantigen-specific T-cell reactivity in tumour-infiltrating lymphocytes. These data demonstrate the dynamic interactions between cancer cells and T cells, which suggest that T cells mediate neoantigen immunoediting, and indicate that the therapeutic induction of broad neoantigen-specific T-cell responses should be used to avoid tumour resistance.


Subject(s)
Antigens, Neoplasm/immunology , DNA Damage/immunology , Melanoma/immunology , T-Lymphocytes/immunology , Adoptive Transfer , Alleles , Animals , Antigens, Neoplasm/biosynthesis , Antigens, Neoplasm/genetics , Cell Line, Tumor , DNA Damage/genetics , Disease Models, Animal , Down-Regulation , Epitopes, T-Lymphocyte/biosynthesis , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Humans , L Cells , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma/genetics , Melanoma/pathology , Melanoma/therapy , Mice , Mutation , T-Lymphocytes/cytology , Tumor Escape/immunology
14.
Immunol Rev ; 281(1): 99-114, 2018 01.
Article in English | MEDLINE | ID: mdl-29247998

ABSTRACT

Recognition of pathogens and altered self must be efficient and highly specific to orchestrate appropriate responses while limiting excessive inflammation and autoimmune reaction to normal self. AIM2 is a member of innate immune sensors that detects the presence of DNA, arguably the most conserved molecules in living organisms. However, AIM2 achieves specificity by detecting altered or mislocalized DNA molecules. It can detect damaged DNA, and the aberrant presence of DNA within the cytosolic compartment such as genomic DNA released into the cytosol upon loss of nuclear envelope integrity. AIM2 is also a key sensor of pathogens that detects the presence of foreign DNA accumulating in the cytosol during the life cycle of intracellular pathogens including viruses, bacteria, and parasites. AIM2 activation initiates the assembly of the inflammasome, an innate immune complex that leads to the activation of inflammatory caspases. This triggers the maturation and secretion of the cytokines IL-1ß and IL-18. It can also initiate pyroptosis, a proinflammatory form of cell death. The AIM2 inflammasome contributes to physiological responses and diseases. It is a key player in host defenses, but its deregulation can contribute immune-linked diseases, such as autoinflammatory and autoimmune pathologies. Moreover, AIM2 may play a role in cancer development. Recent studies have shown that the detection of self-DNA species by AIM2 is an important factor that contributes to diseases associated with perturbation of cellular homeostasis. Thus, in addition of being a sensor of pathogen associated molecular patterns (PAMPs), the AIM2 inflammasome is emerging as a key guardian of cellular integrity.


Subject(s)
DNA Damage/immunology , DNA-Binding Proteins/metabolism , Inflammasomes/metabolism , Animals , Caspases/metabolism , Cell Death , DNA/immunology , Homeostasis , Host-Pathogen Interactions , Humans , Immunity, Innate , Pathogen-Associated Molecular Pattern Molecules/immunology
15.
Nat Immunol ; 10(8): 817-21, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19621044

ABSTRACT

During B cell and T cell development, the lymphoid-specific proteins RAG-1 and RAG-2 act together to initiate the assembly of antigen receptor genes through a series of site-specific somatic DNA rearrangements that are collectively called variable-diversity-joining (V(D)J) recombination. In the past 20 years, a great deal has been learned about the enzymatic activities of the RAG-1-RAG-2 complex. Recent studies have identified several new and exciting regulatory functions of the RAG-1-RAG-2 complex. Here we discuss some of these functions and suggest that the RAG-1-RAG-2 complex nucleates a specialized subnuclear compartment that we call the 'V(D)J recombination factory'.


Subject(s)
DNA-Binding Proteins/immunology , Gene Rearrangement/immunology , Homeodomain Proteins/immunology , Models, Biological , Nuclear Proteins/immunology , Recombination, Genetic , VDJ Recombinases/immunology , Animals , B-Lymphocytes/immunology , Chromatin/metabolism , DNA Damage/immunology , DNA Repair/immunology , Histones/metabolism , Humans , Protein Binding
16.
FASEB J ; 34(6): 8574-8595, 2020 06.
Article in English | MEDLINE | ID: mdl-32369236

ABSTRACT

Although effective immunological diagnostic systems for autoimmune bullous skin diseases (AIBD) have been established, there are still unidentified cutaneous autoantigens. The purpose of this study is to investigative whether anti-human serum albumin (HSA) autoantibodies exist in AIBD sera and their potential pathogenesis. By immunoprecipitation-immunoblotting, immunofluorescence assay, anti-HSA autoantibodies could be detected in AIBD sera; by ELISAs, positive rates of AIBD sera for IgG and IgA anti-HSA autoantibodies were 29% and 34%, respectively. The IgG anti-HSA autoantibodies in ABID sera recognized a number of HSA antigen epitopes and therefore a polyclonal antibody against HSA were next employed to study its pathogenesis. In vitro cell and tissue culture models, anti-HSA antibody could influence DNA damage-related signaling proteins, via activation of phospho-p38 signaling pathway. This is the first report that an autoantibody may influence DNA damage-related signaling proteins. Statistical analyses also proved that anti-HSA autoantibodies were positively correlated with various known autoantibodies and clinical features of ABID patients. In summary, IgG and IgA autoantibodies to HSA may have diagnosis values for AIBD. DNA damage-related signaling proteins might be involved in the pathogenic role of anti-HSA autoantibodies in AIBD. Phospho-p38 signaling pathway is a potential target for treatment of AIBD positive for serum anti-HSA autoantibodies.


Subject(s)
Autoantibodies/immunology , Autoimmune Diseases/immunology , Serum Albumin, Human/immunology , Skin Diseases, Vesiculobullous/immunology , Adult , Aged , Aged, 80 and over , Autoantigens/immunology , Cell Line , Cell Line, Tumor , Child , DNA Damage/immunology , Epitopes/immunology , Female , Humans , Immunoblotting/methods , Immunoglobulin A/immunology , Immunoglobulin G/immunology , Male , Middle Aged , Signal Transduction/immunology , p38 Mitogen-Activated Protein Kinases/immunology
17.
J Immunol ; 202(10): 2924-2944, 2019 05 15.
Article in English | MEDLINE | ID: mdl-30988120

ABSTRACT

Clonal expansion of B cell chronic lymphocytic leukemia (B-CLL) occurs within lymphoid tissue pseudofollicles. IL-15, a stromal cell-associated cytokine found within spleens and lymph nodes of B-CLL patients, significantly boosts in vitro cycling of blood-derived B-CLL cells following CpG DNA priming. Both IL-15 and CpG DNA are elevated in microbe-draining lymphatic tissues, and unraveling the basis for IL-15-driven B-CLL growth could illuminate new therapeutic targets. Using CpG DNA-primed human B-CLL clones and approaches involving both immunofluorescent staining and pharmacologic inhibitors, we show that both PI3K/AKT and JAK/STAT5 pathways are activated and functionally important for IL-15→CD122/ɣc signaling in ODN-primed cells expressing activated pSTAT3. Furthermore, STAT5 activity must be sustained for continued cycling of CFSE-labeled B-CLL cells. Quantitative RT-PCR experiments with inhibitors of PI3K and STAT5 show that both contribute to IL-15-driven upregulation of mRNA for cyclin D2 and suppression of mRNA for DNA damage response mediators ATM, 53BP1, and MDC1. Furthermore, protein levels of these DNA damage response molecules are reduced by IL-15, as indicated by Western blotting and immunofluorescent staining. Bioinformatics analysis of ENCODE chromatin immunoprecipitation sequencing data from cell lines provides insight into possible mechanisms for STAT5-mediated repression. Finally, pharmacologic inhibitors of JAKs and STAT5 significantly curtailed B-CLL cycling when added either early or late in a growth response. We discuss how the IL-15-induced changes in gene expression lead to rapid cycling and possibly enhanced mutagenesis. STAT5 inhibitors might be an effective modality for blocking B-CLL growth in patients.


Subject(s)
Cyclin D2/immunology , DNA Damage/immunology , Interleukin-15/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Proto-Oncogene Proteins c-akt/immunology , STAT5 Transcription Factor/immunology , Signal Transduction/immunology , Adaptor Proteins, Signal Transducing/immunology , Adult , Aged , Aged, 80 and over , Ataxia Telangiectasia Mutated Proteins/immunology , Cell Cycle Proteins/immunology , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic/immunology , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Male , Middle Aged , Tumor Suppressor p53-Binding Protein 1/immunology , Up-Regulation/immunology
18.
Future Oncol ; 17(13): 1683-1694, 2021 May.
Article in English | MEDLINE | ID: mdl-33726502

ABSTRACT

Radiation therapy (RT) in some cases results in a systemic anticancer response known as the abscopal effect. Multiple hypotheses support the role of immune activation initiated by RT-induced DNA damage. Optimal radiation dose is necessary to promote the cGAS-STING pathway in response to radiation and initiate an IFN-1 signaling cascade that promotes the maturation and migration of dendritic cells to facilitate antigen presentation and stimulation of cytotoxic T cells. T cells then exert a targeted response throughout the body at areas not subjected to RT. These effects are further augmented through the use of immunotherapeutic drugs resulting in increased T-cell activity. Tumor-infiltrating lymphocyte presence and TREX1, KPNA2 and p53 signal expression are being explored as prognostic biomarkers.


Subject(s)
Chemoradiotherapy/methods , Dendritic Cells/immunology , Immune Checkpoint Inhibitors/therapeutic use , Neoplasms/radiotherapy , Biomarkers, Tumor/analysis , Biomarkers, Tumor/metabolism , Cell Movement/radiation effects , Clinical Trials as Topic , DNA Damage/immunology , DNA Damage/radiation effects , Dendritic Cells/radiation effects , Exodeoxyribonucleases/analysis , Exodeoxyribonucleases/metabolism , Humans , Immune Checkpoint Inhibitors/pharmacology , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/radiation effects , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/mortality , Phosphoproteins/analysis , Phosphoproteins/metabolism , Prognosis , Progression-Free Survival , Radiotherapy Dosage , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/radiation effects , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Tumor Microenvironment/radiation effects , Tumor Suppressor Protein p53/analysis , Tumor Suppressor Protein p53/metabolism , alpha Karyopherins/analysis , alpha Karyopherins/metabolism
19.
J Allergy Clin Immunol ; 145(1): 324-334, 2020 01.
Article in English | MEDLINE | ID: mdl-31604087

ABSTRACT

BACKGROUND: Wiskott-Aldrich syndrome (WAS) is an X-linked primary immune deficiency disorder resulting from Wiskott-Aldrich syndrome protein (WASp) deficiency. Lymphocytes from patients with WAS manifest increased DNA damage and lymphopenia from cell death, yet how WASp influences DNA damage-linked cell survival is unknown. A recently described mechanism promoting cell survival after ionizing radiation (IR)-induced DNA damage involves fragmentation and dispersal of the Golgi apparatus, known as the Golgi-dispersal response (GDR), which uses the Golgi phosphoprotein 3 (GOLPH3)-DNA-dependent protein kinase (DNA-PK)-myosin XVIIIA-F-actin signaling pathway. OBJECTIVE: We sought to define WASp's role in the DNA damage-induced GDR and its disruption as a contributor to the development of radiosensitivity-linked immunodeficiency in patients with WAS. METHODS: In human TH and B-cell culture systems, DNA damage-induced GDR elicited by IR or radiomimetic chemotherapy was monitored in the presence or absence of WASp or GOLPH3 alone or both together. RESULTS: WASp deficiency completely prevents the development of IR-induced GDR in human TH and B cells, despite the high DNA damage load. Loss of WASp impedes nuclear translocation of GOLPH3 and its colocalization with the DNA-dependent protein kinase catalytic subunit (DNA-PKcs). Surprisingly, however, depletion of GOLPH3 alone or depolymerization of F-actin in WASp-sufficient TH cells still allows development of robust GDR, suggesting that WASp, but not GOLPH3, is essential for GDR and cell survival after IR-induced DNA-damage in human lymphocytes. CONCLUSION: The study identifies WASp as a novel effector of the nucleus-to-Golgi cell-survival pathway triggered by IR-induced DNA damage in cells of the hematolymphoid lineage and proposes an impaired GDR as a new cause for development of a "radiosensitive" form of immune dysregulation in patients with WAS.


Subject(s)
B-Lymphocytes/immunology , DNA Damage/immunology , Golgi Apparatus/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , Wiskott-Aldrich Syndrome Protein Family/immunology , DNA Damage/genetics , DNA-Activated Protein Kinase/genetics , DNA-Activated Protein Kinase/immunology , Golgi Apparatus/genetics , Humans , Membrane Proteins/genetics , Membrane Proteins/immunology , Wiskott-Aldrich Syndrome/genetics , Wiskott-Aldrich Syndrome/immunology , Wiskott-Aldrich Syndrome Protein Family/genetics
20.
Immunol Rev ; 280(1): 74-82, 2017 11.
Article in English | MEDLINE | ID: mdl-29027228

ABSTRACT

A single protein, HMGB1, directs the triggering of inflammation, innate and adaptive immune responses, and tissue healing after damage. HMGB1 is the best characterized damage-associated molecular pattern (DAMP), proteins that are normally inside the cell but are released after cell death, and allow the immune system to distinguish between antigens that are dangerous or not. Notably, cells undergoing severe stress actively secrete HMGB1 via a dedicated secretion pathway: HMGB1 is relocated from the nucleus to the cytoplasm and then to secretory lysosomes or directly to the extracellular space. Extracellular HMGB1 (either released or secreted) triggers inflammation and adaptive immunological responses by switching among multiple oxidation states, which direct the mutually exclusive choices of different binding partners and receptors. Immune cells are first recruited to the damaged tissue and then activated; thereafter, HMGB1 supports tissue repair and healing, by coordinating the switch of macrophages to a tissue-healing phenotype, activation and proliferation of stem cells, and neoangiogenesis. Inevitably, HMGB1 also orchestrates the support of stressed but illegitimate tissues: tumors. Concomitantly, HMGB1 enhances the immunogenicity of mutated proteins in the tumor (neoantigens), promoting anti-tumor responses and immunological memory. Tweaking the activities of HMGB1 in inflammation, immune responses and tissue repair could bring large rewards in the therapy of multiple medical conditions, including cancer.


Subject(s)
Adaptive Immunity , Cell Death , DNA Damage/immunology , HMGB1 Protein/immunology , Immunity, Innate , Inflammation/immunology , Animals , Humans , Oxidation-Reduction , Wound Healing
SELECTION OF CITATIONS
SEARCH DETAIL