Indexed In : Science Citation Index Expanded(SCIE), MEDLINE,
Pubmed/Pubmed Central, Elsevier Bibliographic, Google Scholar,
Databases(Scopus & Embase), KCI, KoreaMed, DOAJ
Gut and Liver is an international journal of gastroenterology, focusing on the gastrointestinal tract, liver, biliary tree, pancreas, motility, and neurogastroenterology. Gut atnd Liver delivers up-to-date, authoritative papers on both clinical and research-based topics in gastroenterology. The Journal publishes original articles, case reports, brief communications, letters to the editor and invited review articles in the field of gastroenterology. The Journal is operated by internationally renowned editorial boards and designed to provide a global opportunity to promote academic developments in the field of gastroenterology and hepatology. +MORE
Yong Chan Lee |
Professor of Medicine Director, Gastrointestinal Research Laboratory Veterans Affairs Medical Center, Univ. California San Francisco San Francisco, USA |
Jong Pil Im | Seoul National University College of Medicine, Seoul, Korea |
Robert S. Bresalier | University of Texas M. D. Anderson Cancer Center, Houston, USA |
Steven H. Itzkowitz | Mount Sinai Medical Center, NY, USA |
All papers submitted to Gut and Liver are reviewed by the editorial team before being sent out for an external peer review to rule out papers that have low priority, insufficient originality, scientific flaws, or the absence of a message of importance to the readers of the Journal. A decision about these papers will usually be made within two or three weeks.
The remaining articles are usually sent to two reviewers. It would be very helpful if you could suggest a selection of reviewers and include their contact details. We may not always use the reviewers you recommend, but suggesting reviewers will make our reviewer database much richer; in the end, everyone will benefit. We reserve the right to return manuscripts in which no reviewers are suggested.
The final responsibility for the decision to accept or reject lies with the editors. In many cases, papers may be rejected despite favorable reviews because of editorial policy or a lack of space. The editor retains the right to determine publication priorities, the style of the paper, and to request, if necessary, that the material submitted be shortened for publication.
Jae-Young Lee1 , Hyun Woo Ma2 , Ji Hyung Kim2 , I Seul Park2 , Mijeong Son2 , Keun Ho Ryu3 , Jieun Shin3 , Seung Won Kim2,4 , Jae Hee Cheon2,4
Correspondence to: Jae Hee Cheon
ORCID https://orcid.org/0000-0002-2282-8904
E-mail GENIUSHEE@yuhs.ac
Seung Won Kim
ORCID https://orcid.org/0000-0002-1692-1192
E-mail swk21c@hanmail.net
Jae-Young Lee and Hyun Woo Ma contributed equally to this work as first authors.
This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.
Gut Liver 2023;17(5):766-776. https://doi.org/10.5009/gnl220159
Published online September 27, 2022, Published date September 15, 2023
Copyright © Gut and Liver.
Background/Aims: The purpose of the current study was to examine the anti-inflammatory effects of CKD-506, a novel histone deacetylase 6 inhibitor, on human peripheral blood mononuclear cells (PBMCs) and CD4+ T cells and to explore the relationship between CKD-506 and gut epithelial barrier function.
Methods: Lipopolysaccharide-stimulated human PBMCs from inflammatory bowel disease (IBD) patients were treated with CKD-506, and tumor necrosis factor (TNF)-α expression was measured using an enzyme-linked immunosorbent assay. The proliferation of CD4+ T cells from IBD patients was evaluated using flow cytometric analysis. The effects of CKD-506 on gut barrier function in a cell line and colon organoids, based on examinations of mRNA production, goblet cell differentiation, and E-cadherin recovery, were investigated using quantitative reverse transcription polymerase chain reaction, immunofluorescence, and a fluorescein isothiocyanate-dextran permeability assay.
Results: Secretion of TNF-α, a pivotal pro-inflammatory mediator in IBD, by lipopolysaccharide-triggered PBMCs was markedly decreased by CKD-506 treatment in a dose-dependent manner and to a greater extent than by tofacitinib or tubastatin A treatment. E-cadherin mRNA expression and goblet cell differentiation increased significantly and dose-dependently in HT-29 cells in response to CKD-506, and inhibition of E-cadherin loss after TNF-α stimulation was significantly reduced both in HT-29 cells and gut organoids. Caco-2 cells treated with CKD-506 showed a significant reduction in barrier permeability in a dose-dependent manner.
Conclusions: The present study demonstrated that CKD-506 has anti-inflammatory effects on PBMCs and CD4 T cells and improves gut barrier function, suggesting its potential as a small-molecule therapeutic option for IBD.
Keywords: HDAC6 inhibitor, Inflammatory bowel diseases, Barrier function, T-cell
Inflammatory bowel disease (IBD) represents a group of chronic immune-mediated diseases of the gastrointestinal tract, characterized by recurrent inflammation and consequential damage of the gastrointestinal tract.1,2 IBD includes Crohn’s disease and ulcerative colitis (UC),3 which show both overlapping and distinct clinical and pathological characteristics.4 IBD is associated with lifelong relapsing inflammatory symptoms and disabling complications. However, the pathophysiology of IBD has not been clearly elucidated.5 Various therapeutic options are available,6 but the current medical treatments including aminosalicylates, corticosteroids, immunomodulators, and biologics such as anti-tumor necrosis factor (TNF)-α have shown limited clinical efficacy as well as a high incidence of side effects, such as increased infectious complications and drug resistance.7-9 Recently, tofacitinib, a Janus kinase inhibitor, has emerged as a small-molecule drug used for treating UC and has been found to be effective in inducing and maintaining remission.10 Tofacitinib decreases the proliferation of CD4+ T cells, inhibits the production of cytokines, such as interleukin (IL)-17 and interferon-γ,11-14 and increases the gut barrier by suppressing CLAUDIN2.15 Notwithstanding, the use of tofacitinib in IBD has several limitations including an increased risk of infection and long-term safety concerns.16 Thus, more efficacious and safer small-molecule medicines for IBD are necessary.17,18
The influence of epigenetics on the pathogenesis of IBD has been an area of intense interest for the past few decades.19 Histone deacetylases (HDACs) are a family of mostly ubiquitous enzymes that remove acetyl groups from lysines on histone proteins to regulate gene transcription.20 Various types of HDACs have been found to be closely associated with intestinal inflammation, and the specific functions of individual HDACs have been investigated through studies on HDAC inhibitors.21 Pan-HDAC inhibitors, or non-selective HDAC inhibitors, such as givinostat and suberoylanilide hydroxamic acid (also known as vorinostat), were the first to be studied.22 Although non-selective HDAC inhibitors show anti-inflammatory potential
CKD-506 is a HDAC6 inhibitor that shows highly specific anti-HDAC6 activity and is expected to show better efficacy than previously developed HDAC6 inhibitors in managing immune-mediated inflammatory diseases.31 CKD-506 was previously reported to decrease the production of numerous pro-inflammatory cytokines in the serum and kidneys in a murine systemic lupus erythematosus model.31 A phase I clinical trial of CKD-506 did not reveal notable safety issues (EudraCT number: 2016-002816-42). Previous studies have investigated the efficacy of CKD-506 in a murine colitis model and murine cell lines.1 However, the effects of CKD-506 on immune cells including T cells and on intestinal barrier function, which prevents unwanted excess immune response by protecting from contact of luminal antigens, have not been examined. Moreover, to the best of our knowledge, there has not yet been a study on the anti-inflammatory effect of CKD-506 using samples of IBD patients, although there are clear differences between mouse and human models in terms of drug responses.32 Therefore, we conducted an
Based on the necessity of exploring the mechanism of action of HDAC6 inhibitors in IBD treatment, the purpose of this study was to examine the effects of CKD-506 on immune cells including T cells and on gut barrier function
Samples were collected from disease-involved proximal colon specimens from patients with active UC at Severance Hospital, Seoul, Korea. Colon biopsy samples that were obtained from UC patients by colonoscopy (n=3) were used for establishment of colon organoids. Blood samples were obtained from UC (n=3) or Crohn’s disease (n=2) patients who had been newly diagnosed with IBD and had never been treated by immunosuppressants or anti-TNF drugs. Healthy control blood samples (n=4) were obtained from patients during medical checkups. All patients were free of other diseases including arthritis. The study was approved by the Institutional Review Board of Severance Hospital (IRB number: 4-2012-0302). All patients and controls provided written informed consent, and all methods were performed in accordance with relevant guidelines and regulations. Patient characteristics are described in Table 1.
Table 1 Clinical and Demographic Characteristics of Inflammatory Bowel Disease Patients and Healthy Controls
Characteristic | UC | CD | Healthy control |
---|---|---|---|
No. of patients | 6 | 2 | 4 |
Male/female | 4/2 | 1/1 | 2/2 |
Age, mean±SD, yr | 31.2±11.2 | 30.2±12.6 | 31.3±8.5 |
UC, ulcerative colitis; CD, Crohn’s disease.
Blood samples were centrifuged with Ficoll Paque Plus (GE Healthcare Life Science, Uppsala, Sweden) and Leucosep (Greiner Bio-One, Kremsmünster, Austria). Red blood cells were removed with RBC Lysis Buffer (BioLegend, San Diego, CA, USA). Separated PBMCs were cultured in 12-well plates (Corning, NY, USA) at a density of 1×105 cells per well in Roswell Park Memorial Institute medium (RPMI) supplemented with 10% heat-inactivated fetal bovine serum (FBS) and 1% penicillin/streptomycin (Thermo Fisher Scientific, Pittsburgh, PA, USA) in a humidified 37℃ incubator in 5% CO2 and stimulated with lipopolysaccharide (LPS). All drugs (tofacitinib, Janus kinase inhibitor, and tubastatin A) were kindly provided by Chong Kun Dang Pharmaceuticals (Seoul, Korea).
To evaluate cytokine levels, PBMC cultured media was collected and processed using the human TNF-α enzyme-linked immunosorbent assay kit (BioLegend). All procedures were performed according to the manufacturer’s instructions. Human colon carcinoma cell lines HT-29 (HTB-38TM; Korea Cell Line Bank, Seoul, Korea), HT29-LuciaTM AhR cells (InvivoGen, San Diego, CA, USA), and Caco-2 (HTB-37TM; American Type Culture Collection, Manassas, VA, USA), which are widely used intestinal epithelial cell lines
CD4+ T cells were enriched with CD4 MicroBeads, Human (Miltenyi Biotec, Bergisch Gladbach, Germany) and stained with CellTrace Violet (Thermo Fisher Scientific). After staining, CD4+ T cells were activated by pre-coated anti-human CD3 antibody (clone: OKT3) (Thermo Fisher Scientific) for 5 days in RPMI medium supplemented with 10% heat-inactivated FBS and 1% penicillin/streptomycin in a humidified 37℃ incubator of 5% CO2. Cells stained with anti-CD3 APC-Cy7 antibody and anti-CD4 fluorescein isothiocyanate (FITC) antibody (Thermo Fisher Scientific) were analyzed by FACSVerse (BD Bioscience, San Jose, NJ, USA).
Total RNA was extracted using TRIzol Reagent (Thermo Fisher Scientific) and reverse-transcribed using a High-Capacity cDNA Reverse Transcription Kit (Thermo Fisher Scientific) according to the provided protocol. Amplification was performed using SYBR Green Master Mix (Thermo Fisher Scientific) for 45 cycles using the following thermocycling steps: 95℃ for 30 seconds, 60℃ to 63℃ for 30 seconds, and 72℃ for 40 seconds. All real-time polymerase chain reactions were performed using the relative standard curve. Results are presented as fold change compared with the control sample after normalization to the level of β-actin mRNA. Primers are listed in Table 2.
Table 2 Human Primers Used for qRT-PCR
Gene | Sequence (5'-3') |
---|---|
β-Actin | F: CTCTTCCAGCCTTCCTTCCTG |
R: CAGCACTGTGTTGGCGTACAG | |
KLF4 | F: CGGACATCAACGACGTGAG |
R: GACGCCTTCAGCACGAACT | |
E-cadherin | F: AGCCATGTACGTTGCTATCC |
R: CGTAGCACAGCTTCTCCTTAAT | |
LL37 | F: AGGATTGTGACTTCAAGAAGGACG |
R: GTTTATTTCTCAGAGCCCAGAAGC |
qRT-PCR, quantitative reverse transcription polymerase chain reaction; F, forward primer; R, reverse primer.
After at least six biopsy samples were collected, washed with ice-cold phosphate-buffered saline, and stripped of the underlying muscle layers with surgical scissors, tissues were chopped into approximately 5-mm pieces and further washed with ice-cold phosphate-buffered saline. Organoid culture was performed as previous study.33,34 Briefly, crypt isolation was performed using Gentle Cell Dissociation Reagent (Stemcell Technologies, Vancouver, BC, Canada). Isolated crypts were embedded in Matrigel (Corning) at a density of 1,000 per 50 µL Matrigel and seeded in 12-well plates. Matrigel was incubated at 37℃. After the Matrigel hardened, crypts were cultured with IntestiCult Organoid Growth Medium (Human) (Stemcell Technologies) supplemented with Y-27632 dihydrochloride (Sigma-Aldrich, St. Louis, MO, USA).
Organoids and HT-29 cells were stained with Alexa Fluor 488-conjugated anti-E-cadherin antibody (24E10) (Cell Signaling Technology, Berkeley, CA, USA) and Alexa Fluor 488-conjugated Zo-1 antibody, and nuclei were counterstained with 4',6-diamidino-2-phenylindole (DAPI; Thermo Fisher Scientific). Images were obtained at Yonsei Advanced Imaging Center in cooperation with Carl Zeiss Microscopy, Yonsei University College of Medicine, using an LSM780 confocal microscope (Carl Zeiss, Oberkochen, Germany).
Alcian blue staining was performed using an Alcian Blue Stain Kit (ab150662; Abcam, Cambridge, MA, USA) according to the manufacturer’s instructions.
Caco-2 cell monolayers cultured on transwell chambers (0.4 µm pore, 3460, Corning) were stimulated with recombinant human TNF-α (40 ng/mL) (R&D Systems, Minneapolis, MN, USA) and treated with CKD-506, tofacitinib, or tubastatin A. After 48 hours of exposure, FITC-dextran (4 kDa) was applied to the transwell chambers. The basolateral media was collected after 2 hours and transferred to 96-well microplates (Berthold Technologies, Bad Wildbad, Germany). The fluorescence intensity of basolateral media was measured with Thermo Fisher Scientific Varioskan® Flash.
HT29-LuciaTM AhR cells were plated into 96-well plate at a density of 1×105 cells and pre-treated with CKD-506 (0.1, 1.0, or 3.0 µM), tofacitinib (1.0 µM), or tubastatin A (1.0 µM) for 18 hours. Luciferase assay was measured according to the manufacturer’s protocol, and the results are expressed in terms of relative luciferase activity.
Experimental results were expressed as mean value and standard error of the mean. GraphPad software (La Jolla, CA, USA) was used for statistical analysis. We tested normality of data and the significance of the differences between the test conditions was assessed using Mann-Whitney test or Tukey's multiple comparisons post-test. The p-values <0.05 were considered statistically significant.
As CD4+ T cells, which are involved in the pathophysiology of IBD, are a major source of TNF-α production, we evaluated the relationship between CD4+ T cells and CKD-506. To determine the effects of CKD-506 on CD4+ T cell proliferation, cells were evaluated by flow cytometeric analysis after treatment with CKD-506 (0.1, 1.0, or 3.0 µM) or positive controls (tofacitinib, Janus kinase inhibitor, and tubastatin A, HDAC6 inhibitor) (Fig. 1A). A dose-dependent inhibition of T cell proliferation by CKD-506 was shown in the IBD patient group as well as the healthy control group. T cell proliferation was drastically inhibited when treated with tofacitinib at 1.0 µM but not when treated with tubastatin A.
Defects in barrier function of the intestinal epithelium allow penetration of pathogenic bacteria through the epithelium, which eventually invade the bloodstream and cause systemic inflammation.35 LPS, which is an endotoxin found in the outer membrane of Gram-negative bacteria, induces TNF-α production by stimulating PBMCs in blood.36 To examine the influence of CKD-506 on LPS-triggered inflammatory cytokine production in PBMCs of IBD patients, TNF-α level was measured by enzyme-linked immunosorbent assay after LPS stimulation and CKD-506 treatment. TNF-α secretion was reduced by CKD-506 in LPS-triggered PBMCs in a dose-dependent manner (Fig. 1B).
IECs, which are involved in the initial pathogenesis of IBD-associated inflammation,2,37 were identified as a main target of HDAC6 inhibitor in previous studies.1,2 To evaluate the effects of CKD-506 on IECs, mRNA expression of genes associated with gut barrier function in HT-29 cells (an IEC cell line) was measured (Fig. 2A-C). The mRNA expressions of E-cadherin, OCCLUDIN, and CLAUDIN1 increased in a dose-dependent manner in cells treated with CKD-506 (Fig. 2A). Consistently, immunofluorescence stain showed that Zo-1 was increased in CKD-506-treated groups, compared to control or tofacitinib groups (Fig. 2D and E). mRNA expression of aryl hydrocarbon receptor (AHR) and Krüppel-like factor 4 (KLF4), transcription factors that induces IL-22 and goblet cell differentiation, respectively, was also increased, in a dose-dependent manner, in HT-29 cells treated with CKD-506 but not those treated with tofacitinib or tubastatin (Fig. 2B). We noted no significant changes in gene expression of peroxisome proliferator-activated receptor γ (PPARγ) in the normal state of HT-29 cells (data not shown). The mRNA of cathelicidin (LL-37), an antimicrobial peptide, was expressed at higher level after CKD-506 treatment compared with tofacitinib or tubastatin A treatment (Fig. 2C). These results suggest that CKD-506 induces expression of genes related to intestinal epithelial barrier function.
To further investigate the relationship between CKD-506 and differentiation of goblet cells, mucin production was analyzed after Alcian blue staining. The integrated density of mucin significantly increased in a dose-dependent manner in cells treated with different doses of CKD-506 (Fig. 2F and G). HT-29 cells treated with 1.0 µM CKD-506 and 1.0 µM tubastatin A produced more mucin than cells treated with 1.0 µM tofacitinib, although the difference was not statistically significant.
AHR, an important transcription factor for barrier function, blocks epithelial-mesenchymal transition and E-cadherin reduction in epithelial cells.38 To confirm the regulation by AHR, we performed AHR-promoter assay using HT29-AHR-reporter cells. We found that CKD-506 significantly induced the gene expression and promoter activity of AHR in HT-29 cells (Fig. 2H). Taken together, these results indicate that CKD-506 enhances intestinal epithelial barrier function and improves the function of goblet cells.
E-cadherin is a key component of the adherens junction that is essential to maintaining normal gut epithelial barrier function.39 The increase in mRNA expression of E-cadherin in HT-29 cells treated with CKD-506, as shown in Fig. 2, led us to investigate the association between CKD-506 and gut barrier function. For a closer examination of the influence of CKD-506 on E-cadherin, the expression of E-cadherin in HT-29 after TNF-α stimulation followed by CKD-506 treatment was analyzed using immunofluorescence. Under the same condition of TNF-α stimulation, HT-29 cells treated with a high dose (3.0 µM) of CKD-506 showed significantly higher E-cadherin expression than HT-29 cells without small-molecule treatment (Fig. 3A). The representative immunofluorescence results from each group are demonstrated in Fig. 3B. Treatment with CKD-506 (1.0 µM) showed better efficacy of barrier function restoration than did tubastatin A of the same concentration.
Intestinal organoids are three-dimensional tissues that have epithelial regenerative capacity and exhibit the same
To identify the intestinal epithelial barrier function in another cell line, we stimulated Caco-2 cells with TNF-α and investigated related gene expression. Interestingly, HDAC6 inhibitors CKD-506 and tubastatin both significantly increased AHR expression, whereas tofacitinib did not. Meanwhile, only CKD-506 significantly increased CLAUDIN1 and PPARγ expression, while tofacitinib and tubastatin did not (Fig. 3C and D).
To investigate the direct impact of CKD-506 on cell permeability, a FITC-dextran permeability assay was performed on Caco-2 cells, in which permeability was triggered by TNF-α, followed by treatment with CKD-506, tofacitinib, or tubastatin A. All groups treated with different concentrations of CKD-506 showed restoration of barrier function (Fig. 4C). A dose-response relationship regarding the restoration of epithelial barrier function was demonstrated with high statistical significance. Taken together, these results suggest that CKD-506 has the potential to restore gut barrier function.
A previous study suggested that HDAC6 affects CD4+ T cell activation, while another study suggested that HDAC6 deficiency had no obvious effect on CD4+ T cell development in murine models.41,42 To investigate the influence of HDAC6 on CD4+ T cells, we conducted an
Pro-inflammatory cytokines such as TNF-α and interferon-γ affect the expression of tight-junction proteins in IECs and disrupt intestinal barrier function.54,55 The mRNA expression of E-cadherin was highly increased in cells treated with CKD-506. E-cadherin is a key component of the adherens junction, making it essential in normal gut barrier function.35,39 Our
Many previous studies have demonstrated that HDAC6 inhibitors suppress pro-inflammatory cytokine production
In conclusion, we showed that CKD-506 suppresses TNF-α production in immune cells and CD4+ T cell proliferation and has potential in restoring intestinal epithelial function to a more effective level than other HDAC6 inhibitors. Our findings indicate that CKD-506 is a promising and competent candidate for small-molecule medicine for IBD treatment.
This work was supported by a National Research Foundation of Korea (NRF) grant funded by the Korean government (MSIT) (2020R1A2C2003638).
J.H.C. is an editorial board member of the journal but was not involved in the peer reviewer selection, evaluation, or decision process of this article. No other potential conflicts of interest relevant to this article were reported.
Study concept and design: H.W.M., J.H.C., S.W.K. Data acquisition: J.Y.L., H.W.M. Data analysis and interpretation: J.Y.L., H.W.M., S.W.K. Drafting of the manuscript: J.Y.L., S.W.K. Critical revision of the manuscript for important intellectual content: J.H.C. Statistical analysis: J.Y.L., H.W.M., S.W.K. Obtained funding: J.H.C. Technical or material support: I.S.P., M.S., J.H.K., K.H.R., J.S. Study supervision: S.W.K., J.H.C. Approval of final manuscript: all authors.
Gut and Liver 2023; 17(5): 766-776
Published online September 15, 2023 https://doi.org/10.5009/gnl220159
Copyright © Gut and Liver.
Jae-Young Lee1 , Hyun Woo Ma2 , Ji Hyung Kim2 , I Seul Park2 , Mijeong Son2 , Keun Ho Ryu3 , Jieun Shin3 , Seung Won Kim2,4 , Jae Hee Cheon2,4
1Department of Medicine, 2Department of Internal Medicine and Institute of Gastroenterology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 3Department of Non-Clinical Study, CKD Research Institute, CKD Pharmaceutical Co., Yongin, and 4Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
Correspondence to:Jae Hee Cheon
ORCID https://orcid.org/0000-0002-2282-8904
E-mail GENIUSHEE@yuhs.ac
Seung Won Kim
ORCID https://orcid.org/0000-0002-1692-1192
E-mail swk21c@hanmail.net
Jae-Young Lee and Hyun Woo Ma contributed equally to this work as first authors.
This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.
Background/Aims: The purpose of the current study was to examine the anti-inflammatory effects of CKD-506, a novel histone deacetylase 6 inhibitor, on human peripheral blood mononuclear cells (PBMCs) and CD4+ T cells and to explore the relationship between CKD-506 and gut epithelial barrier function.
Methods: Lipopolysaccharide-stimulated human PBMCs from inflammatory bowel disease (IBD) patients were treated with CKD-506, and tumor necrosis factor (TNF)-α expression was measured using an enzyme-linked immunosorbent assay. The proliferation of CD4+ T cells from IBD patients was evaluated using flow cytometric analysis. The effects of CKD-506 on gut barrier function in a cell line and colon organoids, based on examinations of mRNA production, goblet cell differentiation, and E-cadherin recovery, were investigated using quantitative reverse transcription polymerase chain reaction, immunofluorescence, and a fluorescein isothiocyanate-dextran permeability assay.
Results: Secretion of TNF-α, a pivotal pro-inflammatory mediator in IBD, by lipopolysaccharide-triggered PBMCs was markedly decreased by CKD-506 treatment in a dose-dependent manner and to a greater extent than by tofacitinib or tubastatin A treatment. E-cadherin mRNA expression and goblet cell differentiation increased significantly and dose-dependently in HT-29 cells in response to CKD-506, and inhibition of E-cadherin loss after TNF-α stimulation was significantly reduced both in HT-29 cells and gut organoids. Caco-2 cells treated with CKD-506 showed a significant reduction in barrier permeability in a dose-dependent manner.
Conclusions: The present study demonstrated that CKD-506 has anti-inflammatory effects on PBMCs and CD4 T cells and improves gut barrier function, suggesting its potential as a small-molecule therapeutic option for IBD.
Keywords: HDAC6 inhibitor, Inflammatory bowel diseases, Barrier function, T-cell
Inflammatory bowel disease (IBD) represents a group of chronic immune-mediated diseases of the gastrointestinal tract, characterized by recurrent inflammation and consequential damage of the gastrointestinal tract.1,2 IBD includes Crohn’s disease and ulcerative colitis (UC),3 which show both overlapping and distinct clinical and pathological characteristics.4 IBD is associated with lifelong relapsing inflammatory symptoms and disabling complications. However, the pathophysiology of IBD has not been clearly elucidated.5 Various therapeutic options are available,6 but the current medical treatments including aminosalicylates, corticosteroids, immunomodulators, and biologics such as anti-tumor necrosis factor (TNF)-α have shown limited clinical efficacy as well as a high incidence of side effects, such as increased infectious complications and drug resistance.7-9 Recently, tofacitinib, a Janus kinase inhibitor, has emerged as a small-molecule drug used for treating UC and has been found to be effective in inducing and maintaining remission.10 Tofacitinib decreases the proliferation of CD4+ T cells, inhibits the production of cytokines, such as interleukin (IL)-17 and interferon-γ,11-14 and increases the gut barrier by suppressing CLAUDIN2.15 Notwithstanding, the use of tofacitinib in IBD has several limitations including an increased risk of infection and long-term safety concerns.16 Thus, more efficacious and safer small-molecule medicines for IBD are necessary.17,18
The influence of epigenetics on the pathogenesis of IBD has been an area of intense interest for the past few decades.19 Histone deacetylases (HDACs) are a family of mostly ubiquitous enzymes that remove acetyl groups from lysines on histone proteins to regulate gene transcription.20 Various types of HDACs have been found to be closely associated with intestinal inflammation, and the specific functions of individual HDACs have been investigated through studies on HDAC inhibitors.21 Pan-HDAC inhibitors, or non-selective HDAC inhibitors, such as givinostat and suberoylanilide hydroxamic acid (also known as vorinostat), were the first to be studied.22 Although non-selective HDAC inhibitors show anti-inflammatory potential
CKD-506 is a HDAC6 inhibitor that shows highly specific anti-HDAC6 activity and is expected to show better efficacy than previously developed HDAC6 inhibitors in managing immune-mediated inflammatory diseases.31 CKD-506 was previously reported to decrease the production of numerous pro-inflammatory cytokines in the serum and kidneys in a murine systemic lupus erythematosus model.31 A phase I clinical trial of CKD-506 did not reveal notable safety issues (EudraCT number: 2016-002816-42). Previous studies have investigated the efficacy of CKD-506 in a murine colitis model and murine cell lines.1 However, the effects of CKD-506 on immune cells including T cells and on intestinal barrier function, which prevents unwanted excess immune response by protecting from contact of luminal antigens, have not been examined. Moreover, to the best of our knowledge, there has not yet been a study on the anti-inflammatory effect of CKD-506 using samples of IBD patients, although there are clear differences between mouse and human models in terms of drug responses.32 Therefore, we conducted an
Based on the necessity of exploring the mechanism of action of HDAC6 inhibitors in IBD treatment, the purpose of this study was to examine the effects of CKD-506 on immune cells including T cells and on gut barrier function
Samples were collected from disease-involved proximal colon specimens from patients with active UC at Severance Hospital, Seoul, Korea. Colon biopsy samples that were obtained from UC patients by colonoscopy (n=3) were used for establishment of colon organoids. Blood samples were obtained from UC (n=3) or Crohn’s disease (n=2) patients who had been newly diagnosed with IBD and had never been treated by immunosuppressants or anti-TNF drugs. Healthy control blood samples (n=4) were obtained from patients during medical checkups. All patients were free of other diseases including arthritis. The study was approved by the Institutional Review Board of Severance Hospital (IRB number: 4-2012-0302). All patients and controls provided written informed consent, and all methods were performed in accordance with relevant guidelines and regulations. Patient characteristics are described in Table 1.
Table 1 . Clinical and Demographic Characteristics of Inflammatory Bowel Disease Patients and Healthy Controls.
Characteristic | UC | CD | Healthy control |
---|---|---|---|
No. of patients | 6 | 2 | 4 |
Male/female | 4/2 | 1/1 | 2/2 |
Age, mean±SD, yr | 31.2±11.2 | 30.2±12.6 | 31.3±8.5 |
UC, ulcerative colitis; CD, Crohn’s disease..
Blood samples were centrifuged with Ficoll Paque Plus (GE Healthcare Life Science, Uppsala, Sweden) and Leucosep (Greiner Bio-One, Kremsmünster, Austria). Red blood cells were removed with RBC Lysis Buffer (BioLegend, San Diego, CA, USA). Separated PBMCs were cultured in 12-well plates (Corning, NY, USA) at a density of 1×105 cells per well in Roswell Park Memorial Institute medium (RPMI) supplemented with 10% heat-inactivated fetal bovine serum (FBS) and 1% penicillin/streptomycin (Thermo Fisher Scientific, Pittsburgh, PA, USA) in a humidified 37℃ incubator in 5% CO2 and stimulated with lipopolysaccharide (LPS). All drugs (tofacitinib, Janus kinase inhibitor, and tubastatin A) were kindly provided by Chong Kun Dang Pharmaceuticals (Seoul, Korea).
To evaluate cytokine levels, PBMC cultured media was collected and processed using the human TNF-α enzyme-linked immunosorbent assay kit (BioLegend). All procedures were performed according to the manufacturer’s instructions. Human colon carcinoma cell lines HT-29 (HTB-38TM; Korea Cell Line Bank, Seoul, Korea), HT29-LuciaTM AhR cells (InvivoGen, San Diego, CA, USA), and Caco-2 (HTB-37TM; American Type Culture Collection, Manassas, VA, USA), which are widely used intestinal epithelial cell lines
CD4+ T cells were enriched with CD4 MicroBeads, Human (Miltenyi Biotec, Bergisch Gladbach, Germany) and stained with CellTrace Violet (Thermo Fisher Scientific). After staining, CD4+ T cells were activated by pre-coated anti-human CD3 antibody (clone: OKT3) (Thermo Fisher Scientific) for 5 days in RPMI medium supplemented with 10% heat-inactivated FBS and 1% penicillin/streptomycin in a humidified 37℃ incubator of 5% CO2. Cells stained with anti-CD3 APC-Cy7 antibody and anti-CD4 fluorescein isothiocyanate (FITC) antibody (Thermo Fisher Scientific) were analyzed by FACSVerse (BD Bioscience, San Jose, NJ, USA).
Total RNA was extracted using TRIzol Reagent (Thermo Fisher Scientific) and reverse-transcribed using a High-Capacity cDNA Reverse Transcription Kit (Thermo Fisher Scientific) according to the provided protocol. Amplification was performed using SYBR Green Master Mix (Thermo Fisher Scientific) for 45 cycles using the following thermocycling steps: 95℃ for 30 seconds, 60℃ to 63℃ for 30 seconds, and 72℃ for 40 seconds. All real-time polymerase chain reactions were performed using the relative standard curve. Results are presented as fold change compared with the control sample after normalization to the level of β-actin mRNA. Primers are listed in Table 2.
Table 2 . Human Primers Used for qRT-PCR.
Gene | Sequence (5'-3') |
---|---|
β-Actin | F: CTCTTCCAGCCTTCCTTCCTG |
R: CAGCACTGTGTTGGCGTACAG | |
KLF4 | F: CGGACATCAACGACGTGAG |
R: GACGCCTTCAGCACGAACT | |
E-cadherin | F: AGCCATGTACGTTGCTATCC |
R: CGTAGCACAGCTTCTCCTTAAT | |
LL37 | F: AGGATTGTGACTTCAAGAAGGACG |
R: GTTTATTTCTCAGAGCCCAGAAGC |
qRT-PCR, quantitative reverse transcription polymerase chain reaction; F, forward primer; R, reverse primer..
After at least six biopsy samples were collected, washed with ice-cold phosphate-buffered saline, and stripped of the underlying muscle layers with surgical scissors, tissues were chopped into approximately 5-mm pieces and further washed with ice-cold phosphate-buffered saline. Organoid culture was performed as previous study.33,34 Briefly, crypt isolation was performed using Gentle Cell Dissociation Reagent (Stemcell Technologies, Vancouver, BC, Canada). Isolated crypts were embedded in Matrigel (Corning) at a density of 1,000 per 50 µL Matrigel and seeded in 12-well plates. Matrigel was incubated at 37℃. After the Matrigel hardened, crypts were cultured with IntestiCult Organoid Growth Medium (Human) (Stemcell Technologies) supplemented with Y-27632 dihydrochloride (Sigma-Aldrich, St. Louis, MO, USA).
Organoids and HT-29 cells were stained with Alexa Fluor 488-conjugated anti-E-cadherin antibody (24E10) (Cell Signaling Technology, Berkeley, CA, USA) and Alexa Fluor 488-conjugated Zo-1 antibody, and nuclei were counterstained with 4',6-diamidino-2-phenylindole (DAPI; Thermo Fisher Scientific). Images were obtained at Yonsei Advanced Imaging Center in cooperation with Carl Zeiss Microscopy, Yonsei University College of Medicine, using an LSM780 confocal microscope (Carl Zeiss, Oberkochen, Germany).
Alcian blue staining was performed using an Alcian Blue Stain Kit (ab150662; Abcam, Cambridge, MA, USA) according to the manufacturer’s instructions.
Caco-2 cell monolayers cultured on transwell chambers (0.4 µm pore, 3460, Corning) were stimulated with recombinant human TNF-α (40 ng/mL) (R&D Systems, Minneapolis, MN, USA) and treated with CKD-506, tofacitinib, or tubastatin A. After 48 hours of exposure, FITC-dextran (4 kDa) was applied to the transwell chambers. The basolateral media was collected after 2 hours and transferred to 96-well microplates (Berthold Technologies, Bad Wildbad, Germany). The fluorescence intensity of basolateral media was measured with Thermo Fisher Scientific Varioskan® Flash.
HT29-LuciaTM AhR cells were plated into 96-well plate at a density of 1×105 cells and pre-treated with CKD-506 (0.1, 1.0, or 3.0 µM), tofacitinib (1.0 µM), or tubastatin A (1.0 µM) for 18 hours. Luciferase assay was measured according to the manufacturer’s protocol, and the results are expressed in terms of relative luciferase activity.
Experimental results were expressed as mean value and standard error of the mean. GraphPad software (La Jolla, CA, USA) was used for statistical analysis. We tested normality of data and the significance of the differences between the test conditions was assessed using Mann-Whitney test or Tukey's multiple comparisons post-test. The p-values <0.05 were considered statistically significant.
As CD4+ T cells, which are involved in the pathophysiology of IBD, are a major source of TNF-α production, we evaluated the relationship between CD4+ T cells and CKD-506. To determine the effects of CKD-506 on CD4+ T cell proliferation, cells were evaluated by flow cytometeric analysis after treatment with CKD-506 (0.1, 1.0, or 3.0 µM) or positive controls (tofacitinib, Janus kinase inhibitor, and tubastatin A, HDAC6 inhibitor) (Fig. 1A). A dose-dependent inhibition of T cell proliferation by CKD-506 was shown in the IBD patient group as well as the healthy control group. T cell proliferation was drastically inhibited when treated with tofacitinib at 1.0 µM but not when treated with tubastatin A.
Defects in barrier function of the intestinal epithelium allow penetration of pathogenic bacteria through the epithelium, which eventually invade the bloodstream and cause systemic inflammation.35 LPS, which is an endotoxin found in the outer membrane of Gram-negative bacteria, induces TNF-α production by stimulating PBMCs in blood.36 To examine the influence of CKD-506 on LPS-triggered inflammatory cytokine production in PBMCs of IBD patients, TNF-α level was measured by enzyme-linked immunosorbent assay after LPS stimulation and CKD-506 treatment. TNF-α secretion was reduced by CKD-506 in LPS-triggered PBMCs in a dose-dependent manner (Fig. 1B).
IECs, which are involved in the initial pathogenesis of IBD-associated inflammation,2,37 were identified as a main target of HDAC6 inhibitor in previous studies.1,2 To evaluate the effects of CKD-506 on IECs, mRNA expression of genes associated with gut barrier function in HT-29 cells (an IEC cell line) was measured (Fig. 2A-C). The mRNA expressions of E-cadherin, OCCLUDIN, and CLAUDIN1 increased in a dose-dependent manner in cells treated with CKD-506 (Fig. 2A). Consistently, immunofluorescence stain showed that Zo-1 was increased in CKD-506-treated groups, compared to control or tofacitinib groups (Fig. 2D and E). mRNA expression of aryl hydrocarbon receptor (AHR) and Krüppel-like factor 4 (KLF4), transcription factors that induces IL-22 and goblet cell differentiation, respectively, was also increased, in a dose-dependent manner, in HT-29 cells treated with CKD-506 but not those treated with tofacitinib or tubastatin (Fig. 2B). We noted no significant changes in gene expression of peroxisome proliferator-activated receptor γ (PPARγ) in the normal state of HT-29 cells (data not shown). The mRNA of cathelicidin (LL-37), an antimicrobial peptide, was expressed at higher level after CKD-506 treatment compared with tofacitinib or tubastatin A treatment (Fig. 2C). These results suggest that CKD-506 induces expression of genes related to intestinal epithelial barrier function.
To further investigate the relationship between CKD-506 and differentiation of goblet cells, mucin production was analyzed after Alcian blue staining. The integrated density of mucin significantly increased in a dose-dependent manner in cells treated with different doses of CKD-506 (Fig. 2F and G). HT-29 cells treated with 1.0 µM CKD-506 and 1.0 µM tubastatin A produced more mucin than cells treated with 1.0 µM tofacitinib, although the difference was not statistically significant.
AHR, an important transcription factor for barrier function, blocks epithelial-mesenchymal transition and E-cadherin reduction in epithelial cells.38 To confirm the regulation by AHR, we performed AHR-promoter assay using HT29-AHR-reporter cells. We found that CKD-506 significantly induced the gene expression and promoter activity of AHR in HT-29 cells (Fig. 2H). Taken together, these results indicate that CKD-506 enhances intestinal epithelial barrier function and improves the function of goblet cells.
E-cadherin is a key component of the adherens junction that is essential to maintaining normal gut epithelial barrier function.39 The increase in mRNA expression of E-cadherin in HT-29 cells treated with CKD-506, as shown in Fig. 2, led us to investigate the association between CKD-506 and gut barrier function. For a closer examination of the influence of CKD-506 on E-cadherin, the expression of E-cadherin in HT-29 after TNF-α stimulation followed by CKD-506 treatment was analyzed using immunofluorescence. Under the same condition of TNF-α stimulation, HT-29 cells treated with a high dose (3.0 µM) of CKD-506 showed significantly higher E-cadherin expression than HT-29 cells without small-molecule treatment (Fig. 3A). The representative immunofluorescence results from each group are demonstrated in Fig. 3B. Treatment with CKD-506 (1.0 µM) showed better efficacy of barrier function restoration than did tubastatin A of the same concentration.
Intestinal organoids are three-dimensional tissues that have epithelial regenerative capacity and exhibit the same
To identify the intestinal epithelial barrier function in another cell line, we stimulated Caco-2 cells with TNF-α and investigated related gene expression. Interestingly, HDAC6 inhibitors CKD-506 and tubastatin both significantly increased AHR expression, whereas tofacitinib did not. Meanwhile, only CKD-506 significantly increased CLAUDIN1 and PPARγ expression, while tofacitinib and tubastatin did not (Fig. 3C and D).
To investigate the direct impact of CKD-506 on cell permeability, a FITC-dextran permeability assay was performed on Caco-2 cells, in which permeability was triggered by TNF-α, followed by treatment with CKD-506, tofacitinib, or tubastatin A. All groups treated with different concentrations of CKD-506 showed restoration of barrier function (Fig. 4C). A dose-response relationship regarding the restoration of epithelial barrier function was demonstrated with high statistical significance. Taken together, these results suggest that CKD-506 has the potential to restore gut barrier function.
A previous study suggested that HDAC6 affects CD4+ T cell activation, while another study suggested that HDAC6 deficiency had no obvious effect on CD4+ T cell development in murine models.41,42 To investigate the influence of HDAC6 on CD4+ T cells, we conducted an
Pro-inflammatory cytokines such as TNF-α and interferon-γ affect the expression of tight-junction proteins in IECs and disrupt intestinal barrier function.54,55 The mRNA expression of E-cadherin was highly increased in cells treated with CKD-506. E-cadherin is a key component of the adherens junction, making it essential in normal gut barrier function.35,39 Our
Many previous studies have demonstrated that HDAC6 inhibitors suppress pro-inflammatory cytokine production
In conclusion, we showed that CKD-506 suppresses TNF-α production in immune cells and CD4+ T cell proliferation and has potential in restoring intestinal epithelial function to a more effective level than other HDAC6 inhibitors. Our findings indicate that CKD-506 is a promising and competent candidate for small-molecule medicine for IBD treatment.
This work was supported by a National Research Foundation of Korea (NRF) grant funded by the Korean government (MSIT) (2020R1A2C2003638).
J.H.C. is an editorial board member of the journal but was not involved in the peer reviewer selection, evaluation, or decision process of this article. No other potential conflicts of interest relevant to this article were reported.
Study concept and design: H.W.M., J.H.C., S.W.K. Data acquisition: J.Y.L., H.W.M. Data analysis and interpretation: J.Y.L., H.W.M., S.W.K. Drafting of the manuscript: J.Y.L., S.W.K. Critical revision of the manuscript for important intellectual content: J.H.C. Statistical analysis: J.Y.L., H.W.M., S.W.K. Obtained funding: J.H.C. Technical or material support: I.S.P., M.S., J.H.K., K.H.R., J.S. Study supervision: S.W.K., J.H.C. Approval of final manuscript: all authors.
Table 1 Clinical and Demographic Characteristics of Inflammatory Bowel Disease Patients and Healthy Controls
Characteristic | UC | CD | Healthy control |
---|---|---|---|
No. of patients | 6 | 2 | 4 |
Male/female | 4/2 | 1/1 | 2/2 |
Age, mean±SD, yr | 31.2±11.2 | 30.2±12.6 | 31.3±8.5 |
UC, ulcerative colitis; CD, Crohn’s disease.
Table 2 Human Primers Used for qRT-PCR
Gene | Sequence (5'-3') |
---|---|
β-Actin | F: CTCTTCCAGCCTTCCTTCCTG |
R: CAGCACTGTGTTGGCGTACAG | |
KLF4 | F: CGGACATCAACGACGTGAG |
R: GACGCCTTCAGCACGAACT | |
E-cadherin | F: AGCCATGTACGTTGCTATCC |
R: CGTAGCACAGCTTCTCCTTAAT | |
LL37 | F: AGGATTGTGACTTCAAGAAGGACG |
R: GTTTATTTCTCAGAGCCCAGAAGC |
qRT-PCR, quantitative reverse transcription polymerase chain reaction; F, forward primer; R, reverse primer.