Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Mol Cell ; 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-39032489

ABSTRACT

The modular Integrator complex is a transcription regulator that is essential for embryonic development. It attenuates coding gene expression via premature transcription termination and performs 3'-processing of non-coding RNAs. For both activities, Integrator requires endonuclease activity that is harbored by an RNA cleavage module consisting of INTS4-9-11. How correct assembly of Integrator modules is achieved remains unknown. Here, we show that BRAT1 and WDR73 are critical biogenesis factors for the human cleavage module. They maintain INTS9-11 inactive during maturation by physically blocking the endonuclease active site and prevent premature INTS4 association. Furthermore, BRAT1 facilitates import of INTS9-11 into the nucleus, where it is joined by INTS4. Final BRAT1 release requires locking of the mature cleavage module conformation by inositol hexaphosphate (IP6). Our data explain several neurodevelopmental disorders caused by BRAT1, WDR73, and INTS11 mutations as Integrator assembly defects and reveal that IP6 is an essential co-factor for cleavage module maturation.

2.
Plant Cell ; 35(9): 3303-3324, 2023 09 01.
Article in English | MEDLINE | ID: mdl-37220754

ABSTRACT

Flowering is the transition from vegetative to reproductive growth and is critical for plant adaptation and reproduction. FLOWERING LOCUS C (FLC) plays a central role in flowering time control, and dissecting its regulation mechanism provides essential information for crop improvement. Here, we report that DECAPPING5 (DCP5), a component of processing bodies (P-bodies), regulates FLC transcription and flowering time in Arabidopsis (Arabidopsis thaliana). DCP5 and its interacting partner SISTER OF FCA (SSF) undergo liquid-liquid phase separation (LLPS) that is mediated by their prion-like domains (PrDs). Enhancing or attenuating the LLPS of both proteins using transgenic methods greatly affects their ability to regulate FLC and flowering time. DCP5 regulates FLC transcription by modulating RNA polymerase II enrichment at the FLC locus. DCP5 requires SSF for FLC regulation, and loss of SSF or its PrD disrupts DCP5 function. Our results reveal that DCP5 interacts with SSF, and the nuclear DCP5-SSF complex regulates FLC expression at the transcriptional level.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Arabidopsis/metabolism , Arabidopsis Proteins/metabolism , Co-Repressor Proteins/genetics , Co-Repressor Proteins/metabolism , Flowers/physiology , Gene Expression Regulation, Plant/genetics , MADS Domain Proteins/genetics , MADS Domain Proteins/metabolism , Mutation , Processing Bodies , Reproduction
3.
BMC Plant Biol ; 24(1): 188, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38486139

ABSTRACT

BACKGROUND: Proper flowering time is important for the growth and development of plants, and both too early and too late flowering impose strong negative influences on plant adaptation and seed yield. Thus, it is vitally important to study the mechanism underlying flowering time control in plants. In a previous study by the authors, genome-wide association analysis was used to screen the candidate gene SISTER OF FCA (SSF) that regulates FLOWERING LOCUS C (FLC), a central gene encoding a flowering suppressor in Arabidopsis thaliana. RESULTS: SSF physically interacts with Protein arginine methyltransferase 5 (PRMT5, SKB1). Subcellular co-localization analysis showed that SSF and SKB1 interact in the nucleus. Genetically, SSF and SKB1 exist in the same regulatory pathway that controls FLC expression. Furthermore, RNA-sequencing analysis showed that both SSF and SKB1 regulate certain common pathways. CONCLUSIONS: This study shows that PRMT5 interacts with SSF, thus controlling FLC expression and facilitating flowering time control.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Arabidopsis/metabolism , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Flowers/metabolism , Gene Expression Regulation, Plant , Genome-Wide Association Study , MADS Domain Proteins/genetics , MADS Domain Proteins/metabolism
4.
BMC Plant Biol ; 22(1): 328, 2022 Jul 08.
Article in English | MEDLINE | ID: mdl-35799118

ABSTRACT

BACKGROUND: Flowering time is an important agronomic trait of crops and significantly affects plant adaptation and seed production. Flowering time varies greatly among maize (Zea mays) inbred lines, but the genetic basis of this variation is not well understood. Here, we report the comprehensive genetic architecture of six flowering time-related traits using a recombinant inbred line (RIL) population obtained from a cross between two maize genotypes, B73 and Abe2, and combined with genome-wide association studies to identify candidate genes that affect flowering time. RESULTS: Our results indicate that these six traits showed extensive phenotypic variation and high heritability in the RIL population. The flowering time of this RIL population showed little correlation with the leaf number under different environmental conditions. A genetic linkage map was constructed by 10,114 polymorphic markers covering the whole maize genome, which was applied to QTL mapping for these traits, and identified a total of 82 QTLs that contain 13 flowering genes. Furthermore, a combined genome-wide association study and linkage mapping analysis revealed 17 new candidate genes associated with flowering time. CONCLUSIONS: In the present study, by using genetic mapping and GWAS approaches with the RIL population, we revealed a list of genomic regions and candidate genes that were significantly associated with flowering time. This work provides an important resource for the breeding of flowering time traits in maize.


Subject(s)
Genome-Wide Association Study , Zea mays , Chromosome Mapping/methods , Genetic Linkage , Genome-Wide Association Study/methods , Phenotype , Plant Breeding , Polymorphism, Single Nucleotide/genetics , Quantitative Trait Loci/genetics , Zea mays/genetics
5.
Theor Appl Genet ; 133(10): 2797-2810, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32535640

ABSTRACT

KEY MESSAGE: A novel genomic region controlling thermotolerance at flowering was identified by the combination of whole genomic re-sequencing and bulked segregant analysis in maize. The increasing frequency of extreme high temperature has brought a great threat to the development of maize throughout its life cycle, especially during the flowering phase. However, the genetic basis of thermotolerance at flowering in maize remains poorly understood. Here, we characterized a thermotolerant maize ecotype Abe2 and dissected its genetic basis using a F2:8 recombinant inbred line (RIL) population generated from a cross between Abe2 and B73. After continuous high temperature stress above 35 °C for 17 days, Abe2 and B73 show distinct leaf scorching phenotype under field conditions. To identify the genomic regions associated with the phenotypic variation, we applied a combination of whole genomic re-sequencing and bulked segregant analysis, and revealed 10,316,744 SNPs and 1,488,302 InDels between the two parental lines, and 2,693,054 SNPs and 313,757 InDels between the two DNA pools generated from the thermos-tolerant and the sensitive individuals of the RIL, of which, 108,655 and 17,853 SNPs may cause nonsynonymous variations. Finally, a 7.41 Mb genomic region on chromosome 1 was identified, and 7 candidate genes were annotated to participate in high temperature-related stress response. A candidate gene Zm00001d033339 encoding a serine/threonine protein kinase was proposed to be the most likely causative gene contributing to the thermotolerance at flowering by involving in stomatal movement (GO: 0010119) via Abscisic acid (ABA) pathway (KO04075). This work could provide an opportunity for gene cloning and pyramiding breeding to improve thermotolerance at flowering in maize.


Subject(s)
Flowers/physiology , Genome, Plant , Thermotolerance , Zea mays/genetics , INDEL Mutation , Phenotype , Polymorphism, Single Nucleotide , Quantitative Trait Loci , Whole Genome Sequencing , Zea mays/physiology
6.
Immunity ; 34(1): 85-95, 2011 Jan 28.
Article in English | MEDLINE | ID: mdl-21194983

ABSTRACT

The generation of tumor-directed cytotoxic T lymphocytes is considered crucial for the induction of antitumor immunity. To activate these CD8(+) T cells, antigen-presenting cells (APCs) must initially acquire tumor cell-associated antigens. The major source of tumor antigens is dead tumor cells, but little is known about how APCs in draining lymph nodes acquire and crosspresent these antigens. Here we show that CD169(+) macrophages phagocytose dead tumor cells transported via lymphatic flow and subsequently crosspresent tumor antigens to CD8(+) T cells. Subcutaneous immunization with irradiated tumor cells protects mice from syngenic tumor. However, tumor antigen-specific CD8(+) T cell activation and subsequent antitumor immunity are severely impaired in mice depleted with CD169(+) macrophages. Neither migratory dendritic cells (DCs) nor lymph node-resident conventional DCs are essential for the crosspresentation of tumor antigens. Thus, we have identified CD169(+) macrophages as lymph node-resident APCs dominating early activation of tumor antigen-specific CD8(+) T cells.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Lymph Nodes/pathology , Lymphoma, T-Cell/immunology , Macrophages/metabolism , Membrane Glycoproteins/biosynthesis , Receptors, Immunologic/biosynthesis , Animals , Antigens, Neoplasm/immunology , CD11c Antigen/biosynthesis , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cell Movement/immunology , Cross-Priming , Immunization , Lymphocyte Activation , Lymphoma, T-Cell/pathology , Macrophages/immunology , Macrophages/pathology , Mice , Mice, Transgenic , Phagocytosis/immunology , Sialic Acid Binding Ig-like Lectin 1
7.
Cell Physiol Biochem ; 47(5): 2159-2169, 2018.
Article in English | MEDLINE | ID: mdl-29975939

ABSTRACT

BACKGROUND/AIMS: Histone acetylation has been demonstrated to be associated with inflammation response. Histone acetyltransferase (HAT) Mof, specifically acetylating lysine 16 of histone H4 (H4K16), has been reported to regulate T cell differentiation. In addition, it has been suggested that acetylation of H4K16 is associated with the inflammatory response. We evaluated the role and potential mechanism of Mof in the development of experimental colitis. METHODS: We used Mof conditional knockout mice to study the role of Mof in dextran sulfate sodium (DSS)-induced colitis and detected the differential expression of genes due to Mof deficiency involved in the inflammatory response, particularly the Th17 signaling pathway, by western blotting, quantitative PCR and RNA sequencing (RNA-seq). RESULTS: A significant elevation of Mof was observed in colonic tissues of mice with DSS-induced colitis. Mof deficiency alleviated the severity of DSS- induced colitis in mice. We found that Th17 signaling pathway associated genes, including Il17a, Il22, RORγt, RORα, Stat3, TGF-ß 1, and Il6, were downregulated in colon tissues with Mof deficiency. RNA-seq data analysis suggested that 68 genes were related to inflammatory response processing and 47 genes were downregulated in Mof defective colon tissues. CONCLUSION: Our study demonstrated that HAT Mof is involved in the development of colitis, and the lack of Mof ameliorates DSS-induced colitis in mice.


Subject(s)
Colitis/enzymology , Dextran Sulfate/toxicity , Histone Acetyltransferases/metabolism , Signal Transduction , Th17 Cells/metabolism , Animals , Colitis/chemically induced , Colitis/genetics , Colitis/pathology , Histone Acetyltransferases/genetics , Mice , Mice, Knockout , Th17 Cells/pathology
8.
Cell Immunol ; 331: 168-177, 2018 09.
Article in English | MEDLINE | ID: mdl-30103869

ABSTRACT

Macrophages are represented in all tissues by phenotypically distinct resident populations that show great functional diversity. Macrophages generally play a protumoral role, and they are attractive targets for cancer therapy. In this study, we found that CD169+ macrophages depletion inhibited the growth of established Lewis lung carcinoma tumors in mice. Benefits must be weighed against potential adverse effects in cancer therapy. Here, we investigated the adverse effects of CD169+ macrophages depletion on bone and bone marrow in mice bearing Lewis lung carcinoma tumors. Our studies showed that depletion of CD169+ macrophages in LLC tumor-bearing mice disrupted bone homeostasis, including bone weight loss and bone mineral density decrease. Further studies revealed that bone marrow erythropoiesis was severely impaired after depletion of CD169+ macrophages in LLC tumor-bearing mice. Our findings suggest that depletion of macrophages for cancer therapy may be associated with potential adverse effects that need to be recognized, prevented, and optimally managed.


Subject(s)
Bone Marrow/immunology , Bone and Bones/immunology , Carcinoma, Lewis Lung/immunology , Erythropoiesis/immunology , Homeostasis/immunology , Macrophages/immunology , Animals , Bone Density/drug effects , Bone Density/immunology , Bone Marrow/metabolism , Bone and Bones/drug effects , Bone and Bones/metabolism , Carcinoma, Lewis Lung/drug therapy , Carcinoma, Lewis Lung/genetics , Cell Line, Tumor , Cells, Cultured , Diphtheria Toxin/administration & dosage , Diphtheria Toxin/pharmacology , Erythropoiesis/genetics , Heparin-binding EGF-like Growth Factor/genetics , Heparin-binding EGF-like Growth Factor/immunology , Heparin-binding EGF-like Growth Factor/metabolism , Homeostasis/drug effects , Homeostasis/genetics , Macrophages/drug effects , Macrophages/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Sialic Acid Binding Ig-like Lectin 1/genetics , Sialic Acid Binding Ig-like Lectin 1/immunology , Sialic Acid Binding Ig-like Lectin 1/metabolism
9.
Immunology ; 148(2): 174-86, 2016 06.
Article in English | MEDLINE | ID: mdl-26868141

ABSTRACT

Macrophages and dendritic cells (DCs) in murine spleen are essential for the maintenance of immune homeostasis by elimination of blood-borne foreign particles and organisms. It has been reported that splenic DCs, especially CD8α(+) CD103(+) DCs, are responsible for tolerance to apoptosis-associated antigens. However, the molecular mechanism by which these DCs maintain immune homeostasis by blood-borne apoptotic cell clearance remains elusive. Here, we found that the CCL22/CCR4 axis played a critical role in the maintenance of immune homeostasis during apoptotic cell clearance by splenic CD8α(+) CD103(+) DCs. The present results revealed that systemic administration of apoptotic cells rapidly induced a large number of CCL22 and CCR4(+) regulatory T (Treg) cells in the spleen of C57BL/6J mice. Further study demonstrated that CD8α(+) CD103(+) DCs dominantly produce much higher CCL22 than CD8α(+) CD103(-) DCs. Moreover, the transient deletion of CD8α(+) CD103(+) DCs caused a decrease in CCL22 levels together with CCR4(+) Treg cell percentage. Subsequently, the levels of some pro-inflammatory cytokines, such as interleukin-17 and interferon-γ in the spleen with the absence of CD8α(+) CD103(+) DCs increased in response to the administration of apoptotic cells. Hence, intravenous injection of apoptotic cells induced a subsequent increase in CCL22 expression and CCR4(+) Treg cells, which contribute to the maintenance of immune homeostasis at least partially by splenic CD8α(+) CD103(+) DCs.


Subject(s)
Apoptosis/immunology , Chemokine CCL22/metabolism , Dendritic Cells/immunology , Receptors, CCR4/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , Antigens, CD/metabolism , CD8 Antigens/metabolism , Cells, Cultured , Homeostasis/immunology , Integrin alpha Chains/metabolism , Interferon-gamma/metabolism , Interleukin-17/metabolism , Mice , Mice, Inbred C57BL , Spleen/pathology
10.
EMBO J ; 29(14): 2421-32, 2010 07 21.
Article in English | MEDLINE | ID: mdl-20551902

ABSTRACT

Despite extensive investigations of Cbl-interacting protein of 85 kDa (CIN85) in receptor trafficking and cytoskeletal dynamics, little is known about its functions in vivo. Here, we report the study of a mouse deficient of the two CIN85 isoforms expressed in the central nervous system, exposing a function of CIN85 in dopamine receptor endocytosis. Mice lacking CIN85 exon 2 (CIN85(Deltaex2)) show hyperactivity phenotypes, characterized by increased physical activity and exploratory behaviour. Interestingly, CIN85(Deltaex2) animals display abnormally high levels of dopamine and D2 dopamine receptors (D2DRs) in the striatum, an important centre for the coordination of animal behaviour. Importantly, CIN85 localizes to the post-synaptic compartment of striatal neurons in which it co-clusters with D2DRs. Moreover, it interacts with endocytic regulators such as dynamin and endophilins in the striatum. Absence of striatal CIN85 causes insufficient complex formation of endophilins with D2DRs in the striatum and ultimately decreased D2DR endocytosis in striatal neurons in response to dopamine stimulation. These findings indicate an important function of CIN85 in the regulation of dopamine receptor functions and provide a molecular explanation for the hyperactive behaviour of CIN85(Deltaex2) mice.


Subject(s)
Behavior, Animal/physiology , Endocytosis/physiology , Neoplasm Proteins/metabolism , Nerve Tissue Proteins/metabolism , Protein Isoforms/metabolism , Receptors, Dopamine D2/metabolism , Adaptor Proteins, Signal Transducing , Animals , Brain/anatomy & histology , Brain/metabolism , Dopamine Agonists/metabolism , Dopamine Antagonists/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Knockout , Motor Activity/physiology , Neoplasm Proteins/genetics , Nerve Tissue Proteins/genetics , Neurons/cytology , Neurons/metabolism , Protein Isoforms/genetics , Receptors, Dopamine D2/genetics
11.
Transl Oncol ; 15(1): 101306, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34883446

ABSTRACT

Radiofrequency ablation (RFA) is a widely used and effective treatment for primary or metastatic liver cancer with small-size lesions. However, the therapeutic effectiveness of RFA in controlling metastatic lesion or recurrence is still limited. As the major cell population in tumor microenvironment (TME), macrophages have been reported to be recruited to RFA-treated lesion, but their roles are still unclear. Herein, we successfully established the mouse model mimicking RFA-induced abscopal effect, in which RFA eliminated the local orthotopic liver tumor but failed to control growth of distant tumor. Correspondently, RFA suppressed protumoral activation of local tumor-associated macrophages (TAMs), but failed to reprogram TAMs in distance. Importantly, although RFA led to reduced proportion of hepatic CD169+ macrophages in local and decreased expression of immune inhibitory molecules Tim-3 and PD-L1, these alterations were not observed for CD169+ macrophages in distant TME. Further RNA-seq and flow cytometry analysis showed that hepatic CD169+ macrophages contributed to reprograming TME through recruiting CD8+ T/NK cells and suppressing accumulation of MDSCs/Tregs. Consistently, depletion of CD169+ macrophages in CD169-DTR mouse greatly promoted liver tumor progression and largely dampened RFA-induced tumor suppression. Notably, transfer of CD169+ macrophages synergistically enhanced RFA-induced inhibition of distant tumor. To our knowledge, this is the first study which demonstrates hepatic CD169+ macrophages as a key factor responsible for RFA-induced abscopal effect. Our data suggest RFA with transfer of CD169+ macrophages as a promising combination therapy to lessen metastasis or recurrence of liver cancer in patients.

12.
Front Neuroanat ; 16: 978641, 2022.
Article in English | MEDLINE | ID: mdl-36059431

ABSTRACT

The adenosine A2A receptor (A2AR), a G protein-coupled receptor, is involved in numerous and varied physiological and pathological processes, including inflammation, immune responses, blood flow, and neurotransmission. Accordingly, it has become an important drug target for the treatment of neuropsychiatric disorders. However, the exact brain distribution of A2AR in regions outside the striatum that display relatively low levels of endogenous A2AR expression has hampered the exploration of A2AR functions under both physiological and pathological conditions. To further study the detailed distribution of the A2AR in low-expression regions, we have generated A2AR knock-in mice in which the 3xHA-2xMyc epitope tag sequence was fused to the C-terminus of A2AR (A2AR-tag mice) via CRISPR/Cas9 technology. Here, using CRISPR/Cas9 technology, we have generated A2AR knock-in mice in which the 3xHA-2xMyc epitope tag sequence was fused to the C-terminus of A2AR (A2AR-tag mice). The A2AR-tag mice exhibited normal locomotor activity and emotional state. Consistent with previous studies, A2AR fluorescence was widely detected in the striatum, nucleus accumbens, and olfactory tubercles, with numerous labeled cells being evident in these regions in the A2AR-tag mouse. Importantly, we also identified the presence of a few but clearly labeled cells in heterogeneous brain regions where A2AR expression has not previously been unambiguously detected, including the lateral septum, hippocampus, amygdala, cerebral cortex, and gigantocellular reticular nucleus. The A2AR-tag mouse represents a novel useful genetic tool for monitoring the expression of A2AR and dissecting its functions in brain regions other than the striatum.

13.
J Immunol ; 182(7): 4127-36, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19299710

ABSTRACT

Apoptotic cell clearance by dendritic cells (DCs) plays a crucial role in the maintenance of self-tolerance. In spleen, CD8alpha(+) DCs are thought to be responsible for this phenomenon by phagocytosing circulating apoptotic cells. However, as CD8alpha(+) DCs are believed to be predominantly localized in the T cell zone, it remains unclear how these DCs phagocytose blood-borne apoptotic cells accumulated in the marginal zone (MZ). In this study, we identified a subpopulation of CD8alpha(+) DCs responsible for tolerance induction to cell-associated Ags. Among splenic CD8alpha(+) DCs, the CD103(+),CD207(+) subset was preferentially localized in the MZ and dominantly phagocytosed blood-borne apoptotic cells. After phagocytosis of apoptotic cells, this DC subset migrated into the T cell zone for cross-presentation of cell-associated Ags. Stimulation of TLRs induced the disappearance of this DC subset. Consequently, CD8alpha(+) DCs neither phagocytosed injected apoptotic cells nor presented cell-associated Ags in mice treated with TLR ligands. Transient ablation of this DC subset by cytochrome c injection resulted in a failure of tolerance induction to cell-associated Ags, indicating that this DC subset is essential for tolerance induction by apoptotic cell clearance.


Subject(s)
CD8 Antigens/immunology , Dendritic Cells/cytology , Dendritic Cells/immunology , Immune Tolerance , Spleen/cytology , Animals , Antigen Presentation/immunology , Antigens, CD/immunology , Antigens, CD/metabolism , Antigens, Surface/immunology , Antigens, Surface/metabolism , Apoptosis/immunology , CD8 Antigens/metabolism , Cell Movement/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Gene Expression Profiling , Immunohistochemistry , Integrin alpha Chains/immunology , Integrin alpha Chains/metabolism , Lectins, C-Type/immunology , Lectins, C-Type/metabolism , Mannose-Binding Lectins/immunology , Mannose-Binding Lectins/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Phagocytosis/immunology , Spleen/immunology
14.
Biomed Rep ; 14(2): 26, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33408860

ABSTRACT

CD169+ macrophages are a unique type of macrophage subset that differ from M1 and M2 macrophages. CD169+ macrophages are present in multiple tissues and organs throughout the body and are primarily expressed in secondary lymphoid organs. These cells are primarily divided across three locations in secondary lymphoid organs: The metallophilic marginal zone of the spleen, the subcapsular sinus and the medulla of the lymph nodes. Due to their unique location distribution in vivo and the presence of the CD169 molecule on their surfaces, CD169+ macrophages are reported to serve important roles in several processes, such as phagocytosis, antigen presentation, immune tolerance, viral infection and inflammatory responses. At the same time, it has been reported that CD169+ macrophages may also serve an important role in anti-tumour immunity. The present review focuses on the research progress surrounding the function of CD169+ macrophages in a variety of diseases, such as viral infection, autoimmune diseases and tumours.

15.
Sci Adv ; 7(34)2021 08.
Article in English | MEDLINE | ID: mdl-34417177

ABSTRACT

Tuberculosis-causing mycobacteria have thick cell-wall and capsule layers that are formed from complex structures. Protein secretion across these barriers depends on a specialized protein secretion system, but none has been reported. We show that Mycobacterium tuberculosis Rv3705c and its homologous MSMEG_6251 in Mycobacterium smegmatis are tube-forming proteins in the mycobacterial envelope (TiME). Crystallographic and cryo-EM structures of these two proteins show that both proteins form rotationally symmetric rings. Two layers of TiME rings pack together in a tail-to-tail manner into a ring-shaped complex, which, in turn, stacks together to form tubes. M. smegmatis TiME was detected mainly in the cell wall and capsule. Knocking out the TiME gene markedly decreased the amount of secreted protein in the M. smegmatis culture medium, and expression of this gene in knocked-out strain partially restored the level of secreted protein. Our structure and functional data thus suggest that TiME forms a protein transport tube across the mycobacterial outer envelope.


Subject(s)
Bacterial Proteins , Mycobacterium tuberculosis , Bacterial Proteins/metabolism , Cell Wall/genetics , Mycobacterium smegmatis/genetics , Mycobacterium smegmatis/metabolism , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/metabolism
16.
Int Immunopharmacol ; 78: 106012, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31865052

ABSTRACT

Macrophages are recognized as one of the major cell types in tumor microenvironment, and macrophage infiltration has been predominantly associated with poor prognosis among patients with breast cancer. Using the murine models of triple-negative breast cancer in CD169-DTR mice, we found that CD169+ macrophages support tumor growth and metastasis. CD169+ macrophage depletion resulted in increased accumulation of CD8+ T cells within tumor, and produced significant expansion of CD8+ T cells in circulation and spleen. In addition, we observed that CD169+ macrophage depletion alleviated tumor-induced splenomegaly in mice, but had no improvement in bone loss and repression of bone marrow erythropoiesis in tumor-bearing mice. Cancer cells and tumor associated macrophages exploit the upregulation of the immunosuppressive protein PD-L1 to subvert T cell-mediated immune surveillance. Within the tumor microenvironment, our understanding of the regulation of PD-L1 protein expression is limited. We showed that there was a 5-fold higher relative expression of PD-L1 on macrophages as compared with 4T1 tumor cells; coculture of macrophages with 4T1 cells augmented PD-L1 levels on macrophages, but did not upregulate the expression of PD-L1 on 4T1 cells. JAK2/STAT3 signaling pathway was activated in macrophages after coculture, and we further identified the JAK2 as a critical regulator of PD-L1 expression in macrophages during coculture with 4T1 cells. Collectively, our data reveal that breast cancer cells and CD169+ macrophages exhibit bidirectional interactions that play a critical role in tumor progression, and inhibition of JAK2 signaling pathway in CD169+ macrophages may be potential strategy to block tumor microenvironment-derived immune escape.


Subject(s)
B7-H1 Antigen/metabolism , Janus Kinase 2/metabolism , Macrophages/immunology , Triple Negative Breast Neoplasms/immunology , Tumor Escape/immunology , Tumor Microenvironment/immunology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , B7-H1 Antigen/immunology , Cell Communication/immunology , Cell Culture Techniques , Cell Line, Tumor/transplantation , Coculture Techniques , Diphtheria Toxin/pharmacology , Disease Models, Animal , Female , Humans , Janus Kinase 2/antagonists & inhibitors , Macrophages/drug effects , Macrophages/metabolism , Mice , Primary Cell Culture , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Sialic Acid Binding Ig-like Lectin 1/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology , Triple Negative Breast Neoplasms/pathology , Tumor Escape/drug effects , Up-Regulation
17.
Nat Commun ; 11(1): 5830, 2020 11 17.
Article in English | MEDLINE | ID: mdl-33203912

ABSTRACT

The identification and functional characterization of natural variants in plants are essential for understanding phenotypic adaptation. Here we identify a molecular variation in At2g47310 that contributes to the natural variation in flowering time in Arabidopsis thaliana accessions. This gene, which we term SISTER of FCA (SSF), functions in an antagonistic manner to its close homolog FCA. Genome-wide association analysis screens two major haplotypes of SSF associated with the natural variation in FLC expression, and a single polymorphism, SSF-N414D, is identified as a main contributor. The SSF414N protein variant interacts more strongly with CUL1, a component of the E3 ubiquitination complex, than the SSF414D form, mediating differences in SSF protein degradation and FLC expression. FCA and SSF appear to have arisen through gene duplication after dicot-monocot divergence, with the SSF-N414D polymorphism emerging relatively recently within A. thaliana. This work provides a good example for deciphering the functional importance of natural polymorphisms in different organisms.


Subject(s)
Arabidopsis/physiology , Flowers/physiology , Polymorphism, Genetic , Adaptation, Physiological/genetics , Arabidopsis/genetics , Arabidopsis Proteins/genetics , Chromatin/genetics , Chromatin/metabolism , Cullin Proteins/genetics , Cullin Proteins/metabolism , Flowers/genetics , Gene Duplication , Gene Expression Regulation, Plant , Genome-Wide Association Study , MADS Domain Proteins/genetics , Phylogeography , Plants, Genetically Modified , Protein Stability , RNA-Binding Proteins/genetics
18.
Inflamm Bowel Dis ; 25(9): 1510-1521, 2019 08 20.
Article in English | MEDLINE | ID: mdl-31107535

ABSTRACT

BACKGROUND: Inflammatory bowel disease (IBD) is a chronic disease of the intestinal tract in which excessive activation of inflammatory response is correlated. Cyanidin-3-O-glucoside (C3G) is a powerful anti-inflammatory agent, widely existing in fruits and vegetables. However, the role of C3G has rarely been investigated in dextran sulfate sodium (DSS)-induced colitis. METHODS: In an attempt to elucidate the possible mechanism of IBD and develop new efficient therapeutic methods for colitis, we evaluated the effects of C3G on DSS-induced colitis. DSS-induced colitic C57BL/6 mice were intraperitoneal injected with 1ug C3G or phosphate buffer every 2 days, a total of 3 times; the changes in macrophages and regular T cells were analyzed by flow cytometry and immunofluorescence. Cytokines and chemokines were measured by real-time quantitative polymerase chain reaction. RESULTS: The results showed that C3G treatment did not cause changes in body weight and colon length as much as those of DSS-treated mice only. Cytokine expression levels such as interleukin (IL)- 6, IL-1ß, IL-18, tumor necrosis factor α, interferon γ (IFN γ) in colons and mesenteric lymph nodes (mLNs) from C3G-treated mice were lower than those from colitic mice. Meanwhile, C3G injection inhibited the decrease in CCL22 levels and Tregs induction in colitic mice. Furthermore, the activation of macrophages by LPS and increase of CD169+ cells induced by type I IFN could be inhibited by C3G directly in vitro. CONCLUSIONS: The study is the first to demonstrate strong effects of C3G to alleviate DSS-induced colonic damage in mice. The effect of C3G on DSS-induced colitis clearly showed a decrease of CD169+ macrophages in both the colon and mLNs. An increase of CD169+ cells induced by type I IFN could be inhibited by C3G. All these data suggest that the role of C3G in colitic inflammation was mediated at least partially by CD169+ cells and the type I IFN pathway.


Subject(s)
Anthocyanins/pharmacology , Colitis/prevention & control , Dextran Sulfate/toxicity , Glucosides/pharmacology , Macrophages, Peritoneal/drug effects , Sialic Acid Binding Ig-like Lectin 1/metabolism , T-Lymphocytes, Regulatory/drug effects , Animals , Cells, Cultured , Chemokine CCL22/genetics , Chemokine CCL22/metabolism , Colitis/chemically induced , Colitis/immunology , Colitis/pathology , Female , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/pathology , Male , Mice , Mice, Inbred C57BL , Sialic Acid Binding Ig-like Lectin 1/genetics , T-Lymphocytes, Regulatory/immunology
19.
Int Immunopharmacol ; 63: 1-8, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30056257

ABSTRACT

Acute lung injury (ALI) is a serious complication among patients with acute kidney injury (AKI) that is a systemic inflammatory disease with high morbidity and mortality. The pathophysiology of AKI-associated ALI is poorly understood. G-CSF regulates the production and function of neutrophils that mediate lung injury via elastase and other mediators. Here, we used a mouse model of adenine-induced AKI to determine the roles of G-CSF and neutrophil elastase in AKI-associated ALI. We confirmed that ALI was associated with high serum G-CSF levels, and elevated neutrophil elastase activity in the lungs and serum of mice with adenine-induced AKI. Systemic administration of G-CSF-specific neutralizing antibody normalized granulopoiesis, pulmonary neutrophil infiltration, and neutrophil elastase activity, conferring improved lung architecture in mice with adenine-induced AKI. Further studies revealed that macrophages secreted G-CSF upon urea stimulation. Consequently, G-CSF could be a target for new anti-lung injury strategy in patients with AKI.


Subject(s)
Acute Kidney Injury/immunology , Acute Lung Injury/immunology , Granulocyte Colony-Stimulating Factor/immunology , Acute Kidney Injury/chemically induced , Acute Kidney Injury/complications , Acute Lung Injury/etiology , Adenine , Animals , Bronchoalveolar Lavage Fluid/immunology , Female , Leukocyte Elastase/immunology , Macrophages, Peritoneal/immunology , Mice, Inbred C57BL , Urea/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL