Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 110
Filter
2.
Cell Rep ; 43(9): 114691, 2024 Sep 24.
Article in English | MEDLINE | ID: mdl-39235944

ABSTRACT

The strategy of lowering cholesterol levels by promoting cholesterol excretion is still lacking, and few molecular targets act on multiple cholesterol metabolic processes. In this study, we find that Nogo-B deficiency/inhibition simultaneously promotes hepatic uptake of cholesterol and cholesterol excretion. Nogo-B deficiency decreases cholesterol levels by activating ATP-binding cassette transporters (ABCs), apolipoprotein E (ApoE), and low-density lipoprotein receptor (LDLR) expression. We discover that Nogo-B interacts with liver X receptor α (LXRα), and Nogo-B deficiency inhibits ubiquitination degradation of LXRα, thereby enhancing its function on cholesterol excretion. Decreased cellular cholesterol levels further activate SREBP2 and LDLR expression, thereby promoting hepatic uptake of cholesterol. Nogo-B inhibition decreases atherosclerotic plaques and cholesterol levels in mice, and Nogo-B levels are correlated to cholesterol levels in human plasma. In this study, Nogo-B deficiency/inhibition not only promotes hepatic uptake of blood cholesterol but also facilitates cholesterol excretion. This study reports a strategy to lower cholesterol levels by inhibiting Nogo-B expression to promote hepatic cholesterol uptake and cholesterol excretion.


Subject(s)
Cholesterol , Hypercholesterolemia , Nogo Proteins , Receptors, LDL , Animals , Humans , Male , Mice , Apolipoproteins E/metabolism , Cholesterol/metabolism , Hypercholesterolemia/metabolism , Hypercholesterolemia/pathology , Liver/metabolism , Liver X Receptors/metabolism , Mice, Inbred C57BL , Mice, Knockout , Nogo Proteins/antagonists & inhibitors , Nogo Proteins/metabolism , Receptors, LDL/metabolism , Sterol Regulatory Element Binding Protein 2/metabolism , Ubiquitination
3.
Nat Commun ; 15(1): 6845, 2024 Aug 10.
Article in English | MEDLINE | ID: mdl-39122737

ABSTRACT

Glucagon-like peptide 1 (GLP1), which is mainly processed and cleaved from proglucagon in enteroendocrine cells (EECs) of the intestinal tract, acts on the GLP1 receptor in pancreatic cells to stimulate insulin secretion and to inhibit glucagon secretion. However, GLP1 processing is not fully understood. Here, we show that reticulon 4B (Nogo-B), an endoplasmic reticulum (ER)-resident protein, interacts with the major proglucagon fragment of proglucagon to retain proglucagon on the ER, thereby inhibiting PCSK1-mediated cleavage of proglucagon in the Golgi. Intestinal Nogo-B knockout in male type 2 diabetes mellitus (T2DM) mice increases GLP1 and insulin levels and decreases glucagon levels, thereby alleviating pancreatic injury and insulin resistance. Finally, we identify aberrantly elevated Nogo-B expression and inhibited proglucagon cleavage in EECs from diabetic patients. Our study reveals the subcellular regulatory processes involving Nogo-B during GLP1 production and suggests intestinal Nogo-B as a potential therapeutic target for T2DM.


Subject(s)
Diabetes Mellitus, Type 2 , Endoplasmic Reticulum , Glucagon-Like Peptide 1 , Nogo Proteins , Proglucagon , Proprotein Convertase 1 , Animals , Humans , Male , Mice , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/genetics , Endoplasmic Reticulum/metabolism , Enteroendocrine Cells/metabolism , Glucagon-Like Peptide 1/metabolism , Golgi Apparatus/metabolism , HEK293 Cells , Insulin/metabolism , Insulin Resistance , Intestines/pathology , Mice, Inbred C57BL , Mice, Knockout , Nogo Proteins/metabolism , Nogo Proteins/genetics , Proglucagon/metabolism , Proglucagon/genetics , Proprotein Convertase 1/metabolism , Proprotein Convertase 1/genetics , Protein Binding , Proteolysis
4.
Nat Commun ; 15(1): 6843, 2024 Aug 10.
Article in English | MEDLINE | ID: mdl-39122671

ABSTRACT

Despite the potential of small molecules and recombinant proteins to enhance the efficiency of homology-directed repair (HDR), single-stranded DNA (ssDNA) donors, as currently designed and chemically modified, remain suboptimal for precise gene editing. Here, we screen the biased ssDNA binding sequences of DNA repair-related proteins and engineer RAD51-preferred sequences into HDR-boosting modules for ssDNA donors. Donors with these modules exhibit an augmented affinity for RAD51, thereby enhancing HDR efficiency across various genomic loci and cell types when cooperated with Cas9, nCas9, and Cas12a. By combining with an inhibitor of non-homologous end joining (NHEJ) or the HDRobust strategy, these modular ssDNA donors achieve up to 90.03% (median 74.81%) HDR efficiency. The HDR-boosting modules targeting an endogenous protein enable a chemical modification-free strategy to improve the efficacy of ssDNA donors for precise gene editing.


Subject(s)
DNA, Single-Stranded , Gene Editing , Rad51 Recombinase , Recombinational DNA Repair , DNA, Single-Stranded/metabolism , DNA, Single-Stranded/genetics , Humans , Gene Editing/methods , Rad51 Recombinase/metabolism , Rad51 Recombinase/genetics , CRISPR-Cas Systems , HEK293 Cells , CRISPR-Associated Proteins/metabolism , CRISPR-Associated Proteins/genetics , CRISPR-Associated Protein 9/metabolism , CRISPR-Associated Protein 9/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Endodeoxyribonucleases/metabolism , Endodeoxyribonucleases/genetics , DNA End-Joining Repair
5.
Sci Immunol ; 9(96): eadj5465, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38875319

ABSTRACT

Nucleic acids are major structures detected by the innate immune system. Although intracellular single-stranded DNA (ssDNA) accumulates during pathogen infection or disease, it remains unclear whether and how intracellular ssDNA stimulates the innate immune system. Here, we report that intracellular ssDNA triggers cytokine expression and cell death in a CGT motif-dependent manner. We identified Schlafen 11 (SLFN11) as an ssDNA-activated RNase, which is essential for the innate immune responses induced by intracellular ssDNA and adeno-associated virus infection. We found that SLFN11 directly binds ssDNA containing CGT motifs through its carboxyl-terminal domain, translocates to the cytoplasm upon ssDNA recognition, and triggers innate immune responses through its amino-terminal ribonuclease activity that cleaves transfer RNA (tRNA). Mice deficient in Slfn9, a mouse homolog of SLFN11, exhibited resistance to CGT ssDNA-induced inflammation, acute hepatitis, and septic shock. This study identifies CGT ssDNA and SLFN11/9 as a class of immunostimulatory nucleic acids and pattern recognition receptors, respectively, and conceptually couples DNA immune sensing to controlled RNase activation and tRNA cleavage.


Subject(s)
DNA, Single-Stranded , Immunity, Innate , Mice, Inbred C57BL , Animals , Female , Humans , Male , Mice , DNA, Single-Stranded/immunology , HEK293 Cells , Immunity, Innate/immunology , Mice, Knockout , Nuclear Proteins/immunology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Ribonucleases/immunology , Ribonucleases/metabolism
6.
Chin Med J (Engl) ; 137(8): 921-935, 2024 Apr 20.
Article in English | MEDLINE | ID: mdl-38527930

ABSTRACT

ABSTRACT: Caloric restriction (CR) is a well-established dietary intervention known to extend healthy lifespan and exert positive effects on aging-related diseases, including cardiovascular conditions. Sirtuins, a family of nicotinamide adenine dinucleotide (NAD + )-dependent histone deacetylases, have emerged as key regulators of cellular metabolism, stress responses, and the aging process, serving as energy status sensors in response to CR. However, the mechanism through which CR regulates Sirtuin function to ameliorate cardiovascular disease remains unclear. This review not only provided an overview of recent research investigating the interplay between Sirtuins and CR, specifically focusing on their potential implications for cardiovascular health, but also provided a comprehensive summary of the benefits of CR for the cardiovascular system mediated directly via Sirtuins. CR has also been shown to have considerable impact on specific metabolic organs, leading to the production of small molecules that enter systemic circulation and subsequently regulate Sirtuin activity within the cardiovascular system. The direct and indirect effects of CR offer a potential mechanism for Sirtuin modulation and subsequent cardiovascular protection. Understanding the interplay between CR and Sirtuins will provide new insights for the development of interventions to prevent and treat cardiovascular diseases.


Subject(s)
Caloric Restriction , Cardiovascular Diseases , Sirtuins , Humans , Sirtuins/metabolism , Sirtuins/physiology , Cardiovascular Diseases/metabolism , Animals
7.
EBioMedicine ; 101: 104995, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38350330

ABSTRACT

RNA splicing is an important RNA processing step required by multiexon protein-coding mRNAs and some noncoding RNAs. Precise RNA splicing is required for maintaining gene and cell function; however, mis-spliced RNA transcripts can lead to loss- or gain-of-function effects in human diseases. Mis-spliced RNAs induced by gene mutations or the dysregulation of splicing regulators may result in frameshifts, nonsense-mediated decay (NMD), or inclusion/exclusion of exons. Genetic animal models have characterised multiple splicing factors required for cardiac development or function. Moreover, sarcomeric and ion channel genes, which are closely associated with cardiovascular function and disease, are hotspots for AS. Here, we summarise splicing factors and their targets that are associated with cardiovascular diseases, introduce some therapies potentially related to pathological AS targets, and raise outstanding questions and future directions in this field.


Subject(s)
Alternative Splicing , Cardiovascular Diseases , Animals , Humans , Cardiovascular Diseases/genetics , Cardiovascular Diseases/therapy , Mutation , Nonsense Mediated mRNA Decay , RNA Splicing Factors/genetics
8.
Cell Metab ; 36(1): 7-9, 2024 01 02.
Article in English | MEDLINE | ID: mdl-38171339

ABSTRACT

Spinal cord-associated disorders are common in the elderly population; however, the mechanisms underlying spinal aging remain elusive. In a recent Nature paper, Sun et al. systemically analyzed aged spines in nonhuman primates and identified a new cluster of CHIT1-positive microglia that drives motor neuron senescence and subsequent spine aging.


Subject(s)
Motor Neurons , Spinal Cord , Animals , Humans , Aged , Aging/physiology , Microglia
9.
Sheng Li Xue Bao ; 75(6): 946-952, 2023 Dec 25.
Article in Chinese | MEDLINE | ID: mdl-38151356

ABSTRACT

Our previous study has shown that p66Shc plays an important role in the process of myocardial regeneration in newborn mice, and p66Shc deficiency leads to weakened myocardial regeneration in newborn mice. This study aims to explore the role of p66Shc protein in myocardial injury repair after myocardial infarction in adult mice, in order to provide a new target for the treatment of myocardial injury after myocardial infarction. Mouse myocardial infarction models of adult wild-type (WT) and p66Shc knockout (KO) were constructed by anterior descending branch ligation. The survival rate and heart-to-body weight ratio of two models were compared and analyzed. Masson's staining was used to identify scar area of injured myocardial tissue, and myocyte area was determined by wheat germ agglutinin (WGA) staining. TUNEL staining was used to detect the cardiomyocyte apoptosis. The protein expression of brain natriuretic peptide (BNP), a common marker of myocardial hypertrophy, was detected by Western blotting. The results showed that there was no significant difference in survival rate, myocardial scar area, myocyte apoptosis, and heart weight to body weight ratio between the WT and p66ShcKO mice after myocardial infarction surgery. Whereas the protein expression level of BNP in the p66ShcKO mice was significantly down-regulated compared with that in the WT mice. These results suggest that, unlike in neonatal mice, the deletion of p66Shc has no significant effect on myocardial injury repair after myocardial infarction in adult mice.


Subject(s)
Myocardial Infarction , Oxidative Stress , Animals , Mice , Body Weight , Cicatrix/metabolism , Mice, Knockout , Myocardial Infarction/genetics , Shc Signaling Adaptor Proteins/metabolism , Src Homology 2 Domain-Containing, Transforming Protein 1/genetics , Src Homology 2 Domain-Containing, Transforming Protein 1/metabolism
12.
Signal Transduct Target Ther ; 8(1): 255, 2023 07 03.
Article in English | MEDLINE | ID: mdl-37394473

ABSTRACT

Thoracic aortic aneurysms (TAAs) develop asymptomatically and are characterized by dilatation of the aorta. This is considered a life-threating vascular disease due to the risk of aortic rupture and without effective treatments. The current understanding of the pathogenesis of TAA is still limited, especially for sporadic TAAs without known genetic mutation. Sirtuin 6 (SIRT6) expression was significantly decreased in the tunica media of sporadic human TAA tissues. Genetic knockout of Sirt6 in mouse vascular smooth muscle cells accelerated TAA formation and rupture, reduced survival, and increased vascular inflammation and senescence after angiotensin II infusion. Transcriptome analysis identified interleukin (IL)-1ß as a pivotal target of SIRT6, and increased IL-1ß levels correlated with vascular inflammation and senescence in human and mouse TAA samples. Chromatin immunoprecipitation revealed that SIRT6 bound to the Il1b promoter to repress expression partly by reducing the H3K9 and H3K56 acetylation. Genetic knockout of Il1b or pharmacological inhibition of IL-1ß signaling with the receptor antagonist anakinra rescued Sirt6 deficiency mediated aggravation of vascular inflammation, senescence, TAA formation and survival in mice. The findings reveal that SIRT6 protects against TAA by epigenetically inhibiting vascular inflammation and senescence, providing insight into potential epigenetic strategies for TAA treatment.


Subject(s)
Aortic Aneurysm, Thoracic , Sirtuins , Humans , Mice , Animals , Aortic Aneurysm, Thoracic/genetics , Aortic Aneurysm, Thoracic/metabolism , Aortic Aneurysm, Thoracic/pathology , Inflammation/genetics , Angiotensin II/genetics , Angiotensin II/pharmacology , Epigenesis, Genetic/genetics , Sirtuins/genetics
13.
Eur Heart J ; 44(29): 2746-2759, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37377116

ABSTRACT

AIMS: The mechanisms underlying ageing-induced vascular remodelling remain unclear. This study investigates the role and underlying mechanisms of the cytoplasmic deacetylase sirtuin 2 (SIRT2) in ageing-induced vascular remodelling. METHODS AND RESULTS: Transcriptome and quantitative real-time PCR data were used to analyse sirtuin expression. Young and old wild-type and Sirt2 knockout mice were used to explore vascular function and pathological remodelling. RNA-seq, histochemical staining, and biochemical assays were used to evaluate the effects of Sirt2 knockout on the vascular transcriptome and pathological remodelling and explore the underlying biochemical mechanisms. Among the sirtuins, SIRT2 had the highest levels in human and mouse aortas. Sirtuin 2 activity was reduced in aged aortas, and loss of SIRT2 accelerated vascular ageing. In old mice, SIRT2 deficiency aggravated ageing-induced arterial stiffness and constriction-relaxation dysfunction, accompanied by aortic remodelling (thickened vascular medial layers, breakage of elastin fibres, collagen deposition, and inflammation). Transcriptome and biochemical analyses revealed that the ageing-controlling protein p66Shc and metabolism of mitochondrial reactive oxygen species (mROS) contributed to SIRT2 function in vascular ageing. Sirtuin 2 repressed p66Shc activation and mROS production by deacetylating p66Shc at lysine 81. Elimination of reactive oxygen species by MnTBAP repressed the SIRT2 deficiency-mediated aggravation of vascular remodelling and dysfunction in angiotensin II-challenged and aged mice. The SIRT2 coexpression module in aortas was reduced with ageing across species and was a significant predictor of age-related aortic diseases in humans. CONCLUSION: The deacetylase SIRT2 is a response to ageing that delays vascular ageing, and the cytoplasm-mitochondria axis (SIRT2-p66Shc-mROS) is important for vascular ageing. Therefore, SIRT2 may serve as a potential therapeutic target for vascular rejuvenation.


Subject(s)
Sirtuin 2 , Vascular Remodeling , Mice , Humans , Animals , Aged , Sirtuin 2/metabolism , Src Homology 2 Domain-Containing, Transforming Protein 1 , Reactive Oxygen Species/metabolism , Aging , Mice, Knockout
14.
Int J Biol Macromol ; 242(Pt 4): 125151, 2023 Jul 01.
Article in English | MEDLINE | ID: mdl-37270127

ABSTRACT

Protein post-translational modifications (PTMs) are important regulators of protein functions and produce proteome complexity. SIRT1 has NAD+-dependent deacylation of acyl-lysine residues. The present study aimed to explore the correlation between lysine crotonylation (Kcr) on cardiac function and rhythm in Sirt1 cardiac-specific knockout (ScKO) mice and related mechanism. Quantitative proteomics and bioinformatics analysis of Kcr were performed in the heart tissue of ScKO mice established with a tamoxifen-inducible Cre-loxP system. The expression and enzyme activity of crotonylated protein were assessed by western blot, co-immunoprecipitation, and cell biology experiment. Echocardiography and electrophysiology were performed to investigate the influence of decrotonylation on cardiac function and rhythm in ScKO mice. The Kcr of SERCA2a was significantly increased on Lys120 (1.973 folds). The activity of SERCA2a decreased due to lower binding energy of crotonylated SERCA2a and ATP. Changes in expression of PPAR-related proteins suggest abnormal energy metabolism in the heart. ScKO mice had cardiac hypertrophy, impaired cardiac function, and abnormal ultrastructure and electrophysiological activities. We conclude that knockout of SIRT1 alters the ultrastructure of cardiac myocytes, induces cardiac hypertrophy and dysfunction, causes arrhythmia, and changes energy metabolism by regulating Kcr of SERCA2a. These findings provide new insight into the role of PTMs in heart diseases.


Subject(s)
Heart Diseases , Lysine , Sarcoplasmic Reticulum Calcium-Transporting ATPases , Animals , Mice , Arrhythmias, Cardiac , Cardiomegaly/genetics , Lysine/chemistry , Mice, Knockout , Protein Processing, Post-Translational , Sirtuin 1/genetics , Sirtuin 1/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/chemistry , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
15.
Life Sci Alliance ; 6(6)2023 06.
Article in English | MEDLINE | ID: mdl-37037595

ABSTRACT

Enhancer of zeste homolog 2 (EZH2) is an important transcriptional regulator in development that catalyzes H3K27me3. The role of EZH2 in epicardial development is still unknown. In this study, we show that EZH2 is expressed in epicardial cells during both human and mouse heart development. Ezh2 epicardial deletion resulted in impaired epicardial cell migration, myocardial hypoplasia, and defective coronary plexus development, leading to embryonic lethality. By using RNA sequencing, we identified that EZH2 controls the transcription of tissue inhibitor of metalloproteinase 3 (TIMP3) in epicardial cells during heart development. Loss-of-function studies revealed that EZH2 promotes epicardial cell migration by suppressing TIMP3 expression. We also found that epicardial Ezh2 deficiency-induced TIMP3 up-regulation leads to extracellular matrix reconstruction in the embryonic myocardium by mass spectrometry. In conclusion, our results demonstrate that EZH2 is required for epicardial cell migration because it blocks Timp3 transcription, which is vital for heart development. Our study provides new insight into the function of EZH2 in cell migration and epicardial development.


Subject(s)
Cell Movement , Enhancer of Zeste Homolog 2 Protein , Heart , Animals , Humans , Mice , Cell Movement/genetics , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Heart/growth & development
16.
Brain Pathol ; 33(4): e13157, 2023 07.
Article in English | MEDLINE | ID: mdl-36974636

ABSTRACT

Mitochondrial encephalomyopathies (ME) are frequently associated with mutations of mitochondrial DNA, but the pathogenesis of a subset of ME (sME) remains elusive. Here we report that haploinsufficiency of a mitochondrial inner membrane protein, Mic60, causes progressive neurological abnormalities with insulted mitochondrial structure and neuronal loss in mice. In addition, haploinsufficiency of Mic60 reduces mitochondrial membrane potential and cellular ATP production, increases reactive oxygen species, and alters mitochondrial oxidative phosphorylation complexes in neurons in an age-dependent manner. Moreover, haploinsufficiency of Mic60 compromises brain glucose intake and oxygen consumption in mice, resembling human ME syndrome. We further discover that MIC60 protein expression declined significantly in human sME, implying that insufficient MIC60 may contribute for pathogenesis of human ME. Notably, systemic administration of antioxidant N-acetylcysteine largely reverses mitochondrial dysfunctions and metabolic disorders in haplo-insufficient Mic60 mice, also restores neurological abnormal symptom. These results reveal Mic60 is required in the maintenance of mitochondrial integrity and function, and likely a potential therapeutics target for mitochondrial encephalomyopathies.


Subject(s)
Mitochondrial Encephalomyopathies , Animals , Mice , Humans , Mitochondrial Encephalomyopathies/genetics , Mitochondrial Encephalomyopathies/metabolism , Mitochondrial Proteins/metabolism , Mitochondria/metabolism , DNA, Mitochondrial , Antioxidants
17.
J Mol Cell Cardiol ; 177: 21-27, 2023 04.
Article in English | MEDLINE | ID: mdl-36827872

ABSTRACT

The longevity protein p66Shc is essential for the senescence signaling that is involved in heart regeneration and remodeling. However, the exact role of p66Shc in heart regeneration is unknown. In this study, we found that p66Shc deficiency decreased neonatal mouse cardiomyocyte (CM) proliferation and impeded neonatal heart regeneration after apical resection injury. RNA sequencing and functional verification demonstrated that p66Shc regulated CM proliferation by activating ß-catenin signaling. These findings reveal the critical role of p66Shc in neonatal heart regeneration and provide new insights into senescence signaling in heart regeneration.


Subject(s)
Signal Transduction , Animals , Mice , Phosphorylation , Shc Signaling Adaptor Proteins/genetics , Shc Signaling Adaptor Proteins/metabolism , Src Homology 2 Domain-Containing, Transforming Protein 1/genetics , Src Homology 2 Domain-Containing, Transforming Protein 1/metabolism
18.
Adv Healthc Mater ; 12(5): e2202010, 2023 02.
Article in English | MEDLINE | ID: mdl-36416442

ABSTRACT

Reactive oxygen species (ROS) have been implicated in multiple cellular processes, and an imbalance in redox homeostasis gives rise to diseases, therefore, reestablishing redox homeostasis is a way to cure. Here, copper-based metal-organic networks (Cu-MON) are generated by one-step reaction using anti-inflammatory and antioxidant baicalein as organic ligand and pro-angiogenic copper as metal ions. Phosphate buffered saline is required for triggering Cu-MON formation, and baicalein regulates the morphology and particle size of Cu-MON. Cu-MON are composed of Cu-baicalein complexes (82.08 wt%) and Cu3 (PO4 )2 ·3H2 O (17.92 wt%), thus exhibit a variable catalase-like activity against different H2 O2 levels due to the reversible change between Cu2+ /Cu1+ /Cu0 species. Intramuscular injection of Cu-MON significantly increases blood flow of ischemic limb in diabetic mice, enhances the relative activities of redox-related enzymes in ischemic muscle, thus effectively ameliorating the oxidative damage. Taken together, through moderate and dynamic "precise homeostasis regulation of cells," Cu-MON can be an efficient therapeutic strategy for peripheral arterial disease with diabetic complications.


Subject(s)
Copper , Diabetes Mellitus, Experimental , Mice , Animals , Oxidation-Reduction , Reactive Oxygen Species
19.
Proc Natl Acad Sci U S A ; 119(26): e2204289119, 2022 06 28.
Article in English | MEDLINE | ID: mdl-35727985

ABSTRACT

Behçet's disease (BD) is a chronic vasculitis characterized by systemic immune aberrations. However, a comprehensive understanding of immune disturbances in BD and how they contribute to BD pathogenesis is lacking. Here, we performed single-cell and bulk RNA sequencing to profile peripheral blood mononuclear cells (PBMCs) and isolated monocytes from BD patients and healthy donors. We observed prominent expansion and transcriptional changes in monocytes in PBMCs from BD patients. Deciphering the monocyte heterogeneity revealed the accumulation of C1q-high (C1qhi) monocytes in BD. Pseudotime inference indicated that BD monocytes markedly shifted their differentiation toward inflammation-accompanied and C1qhi monocyte-ended trajectory. Further experiments showed that C1qhi monocytes enhanced phagocytosis and proinflammatory cytokine secretion, and multiplatform analyses revealed the significant clinical relevance of this subtype. Mechanistically, C1qhi monocytes were induced by activated interferon-γ (IFN-γ) signaling in BD patients and were decreased by tofacitinib treatment. Our study illustrates the BD immune landscape and the unrecognized contribution of C1qhi monocytes to BD hyperinflammation, showing their potential as therapeutic targets and clinical assessment indexes.


Subject(s)
Behcet Syndrome , Complement C1q , Monocytes , Behcet Syndrome/genetics , Behcet Syndrome/immunology , Complement C1q/genetics , Complement C1q/immunology , Humans , Monocytes/immunology , RNA-Seq , Single-Cell Analysis
SELECTION OF CITATIONS
SEARCH DETAIL