Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
FASEB J ; 35(11): e22009, 2021 11.
Article in English | MEDLINE | ID: mdl-34694026

ABSTRACT

Tuberculosis (TB), which is caused by Mycobacterium tuberculosis (Mtb), remains a major cause of morbidity and mortality worldwide. Increasing lines of evidence indicate that certain individuals, which are termed resisters, are naturally resistant to TB infection. The resister phenotype has been linked to host efficient innate immune responses, but the underlying mechanisms and the key immune factors remain unclear. Here, we find that upon Mtb infection, monocyte-derived macrophages (MDMs) from TB resisters exhibited distinctly higher production of TNF-α, IL-1ß and IL-6, higher ratio of bacteria in acidic vacuoles, and lower intracellular bacterial loads, as compared to that from the healthy controls, individuals with latent TB infection, and TB patients. Such enhanced anti-Mtb immune capacity of macrophages from resisters largely depends on histone deacetylase 6 (HDAC6), whose expression is specifically maintained in MDMs from TB resisters during Mtb infection. Furthermore, we demonstrate that HDAC6 is required for acidification of Mtb-containing phagosomes in macrophages, thus controlling the intracellular survival of Mtb. Taken together, these findings unravel an indispensable role of HDAC6 in human innate resistance against Mtb infection, suggesting that HDAC6 may serve as a marker for individual TB risk as well as a novel host-directed anti-TB therapeutic target.


Subject(s)
Disease Resistance , Histone Deacetylase 6/immunology , Immunity, Innate , Macrophages/immunology , Tuberculosis/immunology , Adult , Biomarkers/metabolism , Case-Control Studies , Female , Humans , Macrophages/cytology , Male , Middle Aged
2.
STAR Protoc ; 2(3): 100674, 2021 09 17.
Article in English | MEDLINE | ID: mdl-34337443

ABSTRACT

Ubiquitin stress-induced NEDDylation leads to the formation of aggresome-like bodies (ALBs) in the perinuclear region of cells. Therefore, imaging analysis is essential for characterizing the biological phenotypes of ALBs. Here, we describe a protocol to monitor ALBs induced by ubiquitin stress using immunocytochemistry and to quantify cells containing ALBs. This optimized protocol details the use of readily available materials and reagents and can be applied to explore diverse molecules involved in stress-induced ALBs. For complete details on the use and execution of this protocol, please refer to Kim et al. (2021).


Subject(s)
Cytosol , Microscopy, Confocal/methods , Molecular Biology/methods , Fluorescent Antibody Technique , HeLa Cells , Histone Deacetylase 6/immunology , Histone Deacetylase 6/metabolism , Humans , Immunohistochemistry/methods , Inclusion Bodies/metabolism , NEDD8 Protein/immunology , NEDD8 Protein/metabolism , Ubiquitin/metabolism
3.
Bioorg Chem ; 109: 104754, 2021 04.
Article in English | MEDLINE | ID: mdl-33677416

ABSTRACT

Tumor immunotherapy is currently subject of intense scientific and clinical developments. In previous decade, therapists used natural immune system from the human body to treat several diseases. Although tumor immune disease is a big challenge, combinatorial therapeutic strategy has been succeeded to show the clinical significance. In this context, we discuss the HDAC6 and tumor immune diseases relationship. Also, we summarized the current state of knowledge that based on the combination treatments of the HDAC6 inhibitors (HDAC6is) with antitumor immunomodulatory agents. We observed that, the combination therapies slow down the tumor immune diseases by blocking the aggresome and proteasome pathway. The combination therapy was able to reduce M2 macrophage and increasing PD-L1 blockade sensitivity. Most importantly, multiple combinations of HDAC6is with other agents may consider as potential strategies to treat tumor immune diseases, by reducing the side effects and improve efficacy for the future clinical development.


Subject(s)
Antineoplastic Agents/pharmacology , Histone Deacetylase 6/antagonists & inhibitors , Histone Deacetylase Inhibitors/pharmacology , Immunologic Factors/pharmacology , Immunotherapy , Neoplasms/therapy , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Histone Deacetylase 6/chemistry , Histone Deacetylase 6/immunology , Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors/chemistry , Humans , Immunologic Factors/chemical synthesis , Immunologic Factors/chemistry , Molecular Structure , Neoplasms/immunology , Neoplasms/pathology
4.
Front Immunol ; 11: 1776, 2020.
Article in English | MEDLINE | ID: mdl-32849638

ABSTRACT

Macrophages are derived from monocytes in the bone marrow and play an important role in anti-viral innate immune responses. Macrophages produce cytokines such as interferons and IL-10 upon viral infection to modulate anti-viral immune responses. Type I interferons (IFNs) promote anti-viral defense. IL-10 is a suppressor cytokine that down-regulates anti-viral immune responses. HDAC6 is a tubulin deacetylase that can modulate microtubule dynamics and microtubule-mediated cell signaling pathways. In the present study, we investigated the potential role of HDAC6 in macrophage anti-viral responses by examining poly (I:C)-induced IFN-ß and IL-10 production in mouse bone marrow-derived macrophages (BMDMs). We also investigated the role of HDAC6 in poly (I:C)-induced anti-viral signaling such as TBK1, GSK-3ß, and Akt activation in mouse BMDMs. Our data showed that HDAC6 deletion enhanced poly (I:C)-induced INF-ß expression in macrophages by up-regulating TBK1 activity and eliminating the inhibitory regulation of GSK-3ß. Furthermore, HDAC6 deletion inhibited poly (I:C)-induced suppressor cytokine IL-10 production in the BMDMs, which was associated with the inhibition of Akt activation. Our results suggest that HDAC6 modulates IFN-ß and IL-10 production in macrophages through its regulation of TBK1, GSK-3ß, and Akt signaling. HDAC6 could act as a suppressor of anti-viral innate immune responses in macrophages.


Subject(s)
Gene Expression Regulation/immunology , Histone Deacetylase 6/immunology , Macrophages/immunology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Immunity, Innate/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Poly I-C/immunology , Poly I-C/pharmacology , Virus Diseases/immunology
5.
Viruses ; 12(7)2020 07 06.
Article in English | MEDLINE | ID: mdl-32640546

ABSTRACT

The host innate defence against influenza virus infection is an intricate system with a plethora of antiviral factors involved. We have identified host histone deacetylase 6 (HDAC6) as an anti-influenza virus factor in cultured cells. Consistent with this, we report herein that HDAC6 knockout (KO) mice are more susceptible to influenza virus A/PR/8/1934 (H1N1) infection than their wild type (WT) counterparts. The KO mice lost weight faster than the WT mice and, unlike WT mice, could not recover their original body weight. Consequently, more KO mice succumbed to infection, which corresponded with higher lung viral loads. Conversely, the expression of the critical innate antiviral response genes interferon alpha/beta, CD80, CXCL10 and IL15 was significantly downregulated in KO mouse lungs compared to WT mouse lungs. These data are consistent with the known function of HDAC6 of de-acetylating the retinoic acid inducible gene-I (RIG-I) and activating the host innate antiviral response cascade. Loss of HDAC6 thus leads to a blunted innate response and increased susceptibility of mice to influenza A virus infection.


Subject(s)
Disease Susceptibility , Histone Deacetylase 6/genetics , Immunity, Innate/genetics , Influenza A Virus, H1N1 Subtype/physiology , Orthomyxoviridae Infections/genetics , Animals , Cell Line , DEAD Box Protein 58/genetics , Female , Histone Deacetylase 6/immunology , Lung/virology , Male , Mice , Mice, Knockout , Orthomyxoviridae Infections/immunology , Viral Load , Virus Replication
6.
Hepatology ; 71(6): 1967-1987, 2020 06.
Article in English | MEDLINE | ID: mdl-31539182

ABSTRACT

BACKGROUND AND AIMS: Hepatocellular carcinoma (HCC) is often accompanied by resistance to immunotherapies despite the presence of tumor-infiltrating lymphocytes. We report that histone deacetylase 6 (HDAC6) represses interleukin-17 (IL-17)-producing helper T (TH 17) cell pathogenicity and the antitumor immune response, dependent on its deacetylase activity. APPROACH AND RESULTS: Adoptive transfer of HDAC6-deficient TH 17 cells impedes HCC growth, dependent on elevated IL-17A, by enhancing the production of antitumor cytokine and cluster of differentiation 8-positive (CD8+) T cell-mediated antitumor responses. Intriguingly, HDAC6-depleted T cells trigger programmed cell death protein 1 (PD-1)-PD-1 ligand 1 expression to achieve a strong synergistic effect to sensitize advanced HCC to an immune checkpoint blocker, while blockade of IL-17A partially suppresses it. Mechanistically, HDAC6 limits TH 17 pathogenicity and the antitumor effect through regulating forkhead box protein O1 (FoxO1). HDAC6 binds and deacetylates cytosolic FoxO1 at K242, which is required for its nuclear translocation and stabilization to repress retinoic acid-related orphan receptor gamma (RoRγt), the transcription factor of TH 17 cell. This regulation of HDAC6 for murine and human TH 17 cell is highly conserved. CONCLUSIONS: These results demonstrate that targeting the cytosolic HDAC6-FoxO1 axis reprograms the pathogenicity and antitumor response of TH 17 cells in HCC, with a pathogenicity-driven responsiveness to facilitate immunotherapies.


Subject(s)
Carcinoma, Hepatocellular , Histone Deacetylase 6/immunology , Interleukin-17/immunology , Liver Neoplasms , Animals , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/therapy , Cell Line , Cellular Reprogramming/drug effects , Cellular Reprogramming/immunology , Forkhead Box Protein O1/pharmacology , Humans , Immune Checkpoint Inhibitors/pharmacology , Immunotherapy/methods , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Liver Neoplasms/therapy , Mice , Programmed Cell Death 1 Receptor/immunology , Receptors, Retinoic Acid/immunology , Signal Transduction/drug effects , Signal Transduction/immunology , T-Lymphocytes, Helper-Inducer/immunology , Retinoic Acid Receptor gamma
7.
Sci Rep ; 9(1): 6136, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30992475

ABSTRACT

Histone deacetylases (HDACs) are involved in diverse cellular regulatory mechanisms including non-canonical functions outside the chromatin environment. Several publications have demonstrated that selective HDAC inhibitors (HDACi) can influence tumor immunogenicity and the functional activity of specific immune cells. In particular, the selective inhibition of HDAC6 has been reported to decrease tumor growth in several malignancies. However, there is still no clarity about the cellular components mediating this effect. In this study, we evaluated the HDAC6i Nexturastat A as a priming agent to facilitate the transition of the tumor microenvironment from "cold" to "hot", and potentially augment immune check-point blockade therapies. This combination modality demonstrated to significantly reduce tumor growth in syngeneic melanoma tumor models. Additionally, we observed a complete neutralization of the up-regulation of PD-L1 and other immunosuppressive pathways induced by the treatment with anti-PD-1 blockade. This combination also showed profound changes in the tumor microenvironment such as enhanced infiltration of immune cells, increased central and effector T cell memory, and a significant reduction of pro-tumorigenic M2 macrophages. The evaluation of individual components of the tumor microenvironment suggested that the in vivo anti-tumor activity of HDAC6i is mediated by its effect on tumor cells and tumor-associated macrophages, and not directly over T cells. Overall, our results indicate that selective HDAC6i could be used as immunological priming agents to sensitize immunologically "cold" tumors and subsequently improve ongoing immune check-point blockade therapies.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Histone Deacetylase 6/antagonists & inhibitors , Histone Deacetylase Inhibitors/therapeutic use , Macrophages/drug effects , Neoplasms/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Animals , Anti-Inflammatory Agents/therapeutic use , B7-H1 Antigen/immunology , Histone Deacetylase 6/immunology , Immune Tolerance/drug effects , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Neoplasms/immunology , Programmed Cell Death 1 Receptor/immunology , Tumor Microenvironment/drug effects
8.
PLoS Pathog ; 13(12): e1006799, 2017 12.
Article in English | MEDLINE | ID: mdl-29281743

ABSTRACT

Recent evidence on HDAC6 function underlines its role as a key protein in the innate immune response to viral infection. However, whether HDAC6 regulates innate immunity during bacterial infection remains unexplored. To assess the role of HDAC6 in the regulation of defence mechanisms against intracellular bacteria, we used the Listeria monocytogenes (Lm) infection model. Our data show that Hdac6-/- bone marrow-derived dendritic cells (BMDCs) have a higher bacterial load than Hdac6+/+ cells, correlating with weaker induction of IFN-related genes, pro-inflammatory cytokines and nitrite production after bacterial infection. Hdac6-/- BMDCs have a weakened phosphorylation of MAPK signalling in response to Lm infection, suggesting altered Toll-like receptor signalling (TLR). Compared with Hdac6+/+ counterparts, Hdac6-/- GM-CSF-derived and FLT3L-derived dendritic cells show weaker pro-inflammatory cytokine secretion in response to various TLR agonists. Moreover, HDAC6 associates with the TLR-adaptor molecule Myeloid differentiation primary response gene 88 (MyD88), and the absence of HDAC6 seems to diminish the NF-κB induction after TLR stimuli. Hdac6-/- mice display low serum levels of inflammatory cytokine IL-6 and correspondingly an increased survival to a systemic infection with Lm. The impaired bacterial clearance in the absence of HDAC6 appears to be caused by a defect in autophagy. Hence, Hdac6-/- BMDCs accumulate higher levels of the autophagy marker p62 and show defective phagosome-lysosome fusion. These data underline the important function of HDAC6 in dendritic cells not only in bacterial autophagy, but also in the proper activation of TLR signalling. These results thus demonstrate an important regulatory role for HDAC6 in the innate immune response to intracellular bacterial infection.


Subject(s)
Autophagy/immunology , Histone Deacetylase 6/immunology , Immunity, Innate , Listeria monocytogenes/immunology , Listeria monocytogenes/pathogenicity , Toll-Like Receptors/immunology , Animals , Dendritic Cells/immunology , Female , Histone Deacetylase 6/deficiency , Histone Deacetylase 6/genetics , Host-Pathogen Interactions/immunology , Humans , Interleukin-6/blood , Listeriosis/enzymology , Listeriosis/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/immunology , Signal Transduction/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...