Waiting
Login processing...

Trial ends in Request Full Access Tell Your Colleague About Jove

Immunology and Infection

In Vivo Augmentation of Gut-Homing Regulatory T Cell Induction

Published: January 22, 2020 doi: 10.3791/60585

Summary

Here we present a protocol for in vivo augmentation of gut-homing regulatory T cell induction. In this protocol, dendritic cells are engineered to locally produce high concentrations of the active vitamin D (1,25-dihydroxyvitamin D or 1,25[OH]2D) and the active vitamin A (retinoic acid or RA) de novo.

Abstract

Inflammatory bowel disease (IBD) is an inflammatory chronic disease in the gastrointestinal tract (GUT). In the United States, there are approximately 1.4 million IBD patients. It is generally accepted that a dysregulated immune response to gut bacteria initiates the disease and disrupts the mucosal epithelial barrier. We recently show that gut-homing regulatory T (Treg) cells are a promising therapy for IBD. Accordingly, this article presents a protocol for in vivo augmentation of gut-homing Treg cell induction. In this protocol, dendritic cells are engineered to produce locally high concentrations of two molecules de novo, active vitamin D (1,25-dihydroxyvitamin D or 1,25[OH]2D) and active vitamin A (retinoic acid or RA). We chose 1,25(OH)2D and RA based on previous findings showing that 1,25(OH)2D can induce the expression of regulatory molecules (e.g., forkhead box P3 and interleukin-10) and that RA can stimulate the expression of gut-homing receptors in T cells. To generate such engineered dendritic cells, we use a lentiviral vector to transduce dendritic cells to overexpress two genes. One gene is the cytochrome P450 family 27 subfamily B member 1 that encodes 25-hydroxyvitamin D 1α-hydroxylase, which physiologically catalyzes the synthesis of 1,25(OH)2D. The other gene is the aldehyde dehydrogenase 1 family member A2 that encodes retinaldehyde dehydrogenase 2, which physiologically catalyzes the synthesis of RA. This protocol can be used for future investigation of gut-homing Treg cells in vivo.

Introduction

Inflammatory bowel disease (IBD) is an inflammatory chronic disease in the gastrointestinal tract (GUT). In the United States, there are approximately 1.4 million IBD patients. It is generally accepted that a dysregulated immune response to gut bacteria initiates the disease and disrupts the mucosal epithelial barrier1,2. For this reason, currently available U.S. Food and Drug Administration (FDA)-approved drugs inhibit the functions of inflammatory mediators or block the homing of immune cells into the gut. However, the inflammatory mediators and immune cells that are targeted are also necessary for immune defenses. As a result, the inflammatory mediator inhibitors compromise systemic immune defense and the immune cell homing blockers weaken gut immune defense, both of which can lead to severe consequences3,4. In addition, the immune cell homing blockers can also block the homing of regulatory T (Treg) cells into the gut and hence can worsen the already compromised gut immune tolerance in IBD patients. Furthermore, blocking of Treg cell homing into the gut may also lead to systemic immune suppression due to the accumulation of Treg cells in the blood5. Finally, inhibitors and blockers function transiently and, thereby, require frequent administrations. Frequent administration of these inhibitors and blockers may further exacerbate the untoward side effects.

Recently, we proposed a novel strategy that can potentially mitigate or even eliminate the side effects associated with current drugs for IBD treatment6. This strategy augments the induction of gut-homing Treg cells in peripheral lymphoid tissues6. The rationale of this strategy is that gut-homing Treg cells specifically home to the gut and hence will not compromise systemic immune defenses. In addition, since Treg cells can potentially form memory7,8, gut-homing Treg cells can potentially provide a stable control of the chronic gut inflammation in IBD patients and, thereby, treatment should not need to be administered as frequently. Furthermore, since this strategy augments the induction of gut-homing Treg cells in vivo, it does not have the concern of in vivo instability in a highly proinflammatory environment that is associated with adoptive transfer of in vitro generated Treg cells9,10. In this regard, in vitro generated Treg cells are one of the proposed strategies for the treatment of autoimmune diseases11,12,13 and transplant rejection14,15. Finally, in this strategy, dendritic cells (DCs) are engineered to produce locally high concentrations of two molecules de novo: active vitamin D (1,25-dihydroxyvitamin D or 1,25[OH]2D) and active vitamin A (retinoic acid or RA). We chose 1,25(OH)2D and RA because 1,25(OH)2D can induce the expression of regulatory molecules (e.g., forkhead box P3 [foxp3] and interleukin-10 [IL-10])16,17 and that RA can stimulate the expression of gut-homing receptors in T cells18. Because both 1,25(OH)2D and RA can also tolerize DCs28,29, we reason that the engineered DCs will be stably maintained in a tolerogenic status in vivo and hence circumvent the in vivo instability concerns that are associated with in vitro generated tolerogenic DCs (TolDCs)19,20,21. In this respect, TolDCs are also one of the proposed strategies for in vivo augmentation of Treg cell functions19,20,21. To support our reasoning, we have shown that the engineered DCs, upon in vivo delivery, can augment the induction of gut-homing Treg cells in peripheral lymphoid tissues6.

An additional advantage of our proposed strategy is that 1,25(OH)2D also has other functions that can potentially benefit IBD patients. These other functions include the ability of 1,25(OH)2D to stimulate the secretion of antimicrobials22 and to suppresses carcinogenesis23. Infections and cancers are frequently associated with IBD24,25.

To generate the DCs that can produce locally high concentrations of both 1,25(OH)2D and RA de novo, we use a lentiviral vector to engineer DCs to overexpress two genes. One gene is the cytochrome P450 family 27 subfamily B member 1 (CYP27B1) that encodes 25-hydroxyvitamin D 1α-hydroxylase (1α-hydroxylase), which physiologically catalyzes the synthesis of 1,25(OH)2D. The other gene is the aldehyde dehydrogenase 1 family member A2 (ALDH1a2) that encodes retinaldehyde dehydrogenase 2 (RALDH2), which physiologically catalyzes the synthesis of RA6.

Because in vivo augmentation of gut-homing Treg cell induction is potentially important in the treatment of IBD, in the following protocol we will detail the procedures for the generation of the 1α-hydroxylase-RALDH2-overexpressing DCs (DC-CYP-ALDH cells) that can be used for the future investigation of gut-homing Treg cells in vivo.

Subscription Required. Please recommend JoVE to your librarian.

Protocol

All in vivo animal study protocols were reviewed and approved by the Loma Linda University Institutional Animal Care and Use Committee (IACUC) as well as the Animal Care and Use Review Office (ACURO) of the US Army Medical Research and Materiel Command (USAMRMC) of the Department of Defense.

1. Preparation of the Lentivirus that Expresses both 1α-hydroxylase and RALDH2 (lenti-CYP-ALDH Virus)

  1. Day 0: In the early morning, prepare 5 x 105 cells/mL of 293T cells in the CM-10-D cell culture medium.
  2. Seed 20 mL/plate in 150 mm x 25 mm culture dishes. Culture the cells at 37 °C and 5% CO2 for 24 h. Confluency will reach ~80–90% after 24 h.
  3. Day 1: Make 2x HBS (50 mM HEPES, 280 mM NaCl, and 1.5 mM Na2HPO4, pH 7.1). Aliquot and store at -20 °C.
  4. Make 2 M CaCl2 and store at room temperature.
  5. For each 150 mm x 25 mm culture dish, prepare a DNA precipitation mixture in a sterile 50 mL culture tube. First, add 1,620 µL of 2x HBS, 9.5 µg of PMD2G (VSVG), 17.5 µg of pCMVR8.74 (Capsid), 27 µg of Lenti-CYP-ALDH plasmid (a lentiviral vector that carries both CYP27B1 and ALDH1a2). Second, add H2O to a final volume of 3,037.5 µL.
  6. In the last, add 202.5 µL of 2 M CaCl2 dropwise while mixing the solution to final concentrations of 1x HBS and 125 mM CaCl2. CaCl2 must be the last to be added. Leave the transfection mixtures at room temperature for 20 min with occasional mixing. For multiple 150 mm x 25 mm culture dishes, scale up accordingly.
  7. Add the 3,240 µL of DNA precipitation mixture dropwise to one 150 mm x 25 mm culture dish containing the 293T cells. Gently swirl the culture dish side to side while adding the DNA precipitation mixture. Incubate the dish at 37 °C and 5% CO2 for 24 h.
  8. Day 2: Remove the calcium phosphate transfection solution, wash gently with 1x PBS, and refeed the cell culture with CM-4-D cell culture medium. Incubate the cells at 37 °C and 5% CO2 for 24 h.
  9. Day 3: Harvest the supernatants in sterile storage bottles and store at 4–8 °C. Replenish the cell culture with fresh CM-4-D cell culture medium. Culture the cells at 37 °C and 5% CO2 for another 24 h.
  10. Day 4: Harvest the supernatants into sterile storage bottles. Filter the supernatants from Day 3 and Day 4 through a 0.45 µm filter. To concentrate the VSVG pseudotype virus, transfer the supernatants into centrifuge tubes and spin at 4,780 x g for 24 h at 4 °C.
  11. Day 5: Pour the supernatants off the pellets (the pellet is often visible) and allow the tubes to drain on a paper towel in a sterile biosafety cabinet for several minutes.
  12. Resuspend the pellets by pipetting with sterile PBS containing 5% glycerol. The volume for resuspension will be 33.3 µL per 150 mm x 25 mm culture dish.
  13. Aliquot 200 µL/tube and store at -80 °C. Make a separate aliquot of 50 µL/vial for titration purposes. Titers should be within the range of 108–109 transducing units (TUs)/mL.
  14. Add bleach into the culture dishes and discard them.
    NOTE: These transfected cells are the biggest safety concern in the protocol since all lentiviral elements are expressed in the cells.

2. Generation of Bone Marrow Derived DCs (BMDCs)

  1. Harvest tibias and femurs from 4–5 Balb/c mice into a 50 mL sterile polypropylene tube containing the culture medium6.
  2. Transfer the tibias and femurs into a 100 mm x 20 mm culture dish containing the culture medium. Remove tissues such as muscles from the tibias and femurs using dissecting scissors and forceps.
  3. Cut both ends of the tibias and femurs to expose the bone marrow cavity.
  4. Draw 10 mL culture medium in a 10 mL syringe attached to a 30 G needle.
  5. Carefully insert the needle into the bone marrow cavity and flush the bone marrow into a clean 50 mL sterile polypropylene tube. Repeat this procedure if necessary, to ensure that the bone marrow has been completely flushed out.
  6. Pellet the bone marrow cells by centrifugation (400 x g) at 2–8 °C for 5 min. Aspirate the supernatant.
  7. Resuspend the pellet with 5 mL of 1x red blood cell lysis buffer.
  8. Incubate the cells at room temperature for 4–5 min with occasional shaking.
  9. Stop the reaction by adding 30 mL of culture medium.
  10. Pellet the cells by centrifugation (400 x g) at 2–8 °C for 5 min. Resuspend the cells in 10 mL of CM-10-R cell culture medium. If needed, the cells can be passed through a 40 µm cell strainer to remove debris.
  11. Perform a cell count and adjust the cells to 1 x 106 cells/mL using CM-10-R cell culture medium.
  12. Add recombinant murine granulocyte/macrophage colony-stimulating factor (GM-CSF, final concentration = 100 U/mL) and murine interleukin-4 (IL-4, final concentration = 10 U/mL).
    NOTE: The stock solutions of GM-CSF and IL-4 are stored at -80 °C at 100,000 U/mL (1,000x) and 10,000 U/mL (1,000x), respectively.
  13. Distribute the cells into 6 well culture plates at 4 mL/well and culture the cells at 37 °C and 5% CO2 for 48 h.
  14. Remove nonadherent cells with gentle pipetting.
  15. Add fresh CM-10-R cell culture medium containing the GM-CSF (100 U/mL) and IL-4 (10 U/mL). Culture the cells for another 48 h.
  16. Harvest nonadherent cells (BMDCs) for transduction by the lenti-CYP-ALDH virus.

3. Transduction of DCs with lenti-CYP-ALDH Virus to Generate DC-CYP-ALDH Cells

  1. Prepare 1 x 106 DCs/well in a total volume of 0.5 mL of CM-10-R cell culture medium containing 50 µL of virus and 8 µg/mL protamine in a 6 well culture plate.
  2. Culture the cells at 37 °C and 5% CO2 for 24 h.
  3. Replace the medium with fresh CM-10-R cell culture medium and culture the cells at 37 °C and 5% CO2 for another 24 h. At this time point, the enzymatic activities of 1α-hydroxylase and RALDH2 can be evaluated (see step 4). If necessary, repeat steps 3.1–3.3 for a second transduction.
  4. Add lipopolysaccharide (100 ng/mL) and culture the cells for 24 h.
  5. Harvest the cells (DC-CYP-ALDH cells) for experiments.

4. Evaluation of the Overexpressed 1α-hydroxylase and RALDH2 in DC-CYP-ALDH Cells

  1. Evaluation of the overexpressed 1α-hydroxylase in DC-CYP-ALDH cells
    1. Seed 1.0 x 106 DC-CYP-ALDH cells/well in 2 mL of CM-10-R cell culture medium into 12 well culture plates.
    2. Add 25(OH)D to the final concentration of 2.5 μM. Incubate the DC-CYP-ALDH cells for 24 h.
    3. Harvest the supernatants and store at -20 °C.
    4. Assay for 1,25(OH)2D concentrations in the supernatants using a commercially available radioimmunoassay (RIA) (see Table of Materials).
    5. Determine the expression of 1α-hydroxylase in DC-CYP-ALDH cells by fluorescence activated cell sorting (FACS) using an anti-CYP27B1 antibody.
  2. Evaluation of the overexpressed RALDH2 in DC-CYP-ALDH cells
    NOTE: The expression and activity of the overexpressed RALDH2 are determined using a commercial aldehyde dehydrogenase (ALDH) detection kit (see Table of Materials).
    1. Seed 1 mL of DC-CYP-ALDH or control cells (1 x 105 cells/mL in assay buffer) in each of two 5 mL tubes (one labeled as "Control" and the other labeled as "Test").
    2. Add 10 µL of diethylaminobenzaldehyde (DEAB) solution (1.5 mM in 95% ethanol) to each of the control tubes and mix immediately. DEAB is a RALDH2 inhibitor.
    3. Add 5 µL of activated RALDH fluorescence substrate (300 µM) to the Control and Test tubes and mix immediately.
    4. Incubate the tubes for 45 min.
    5. Pellet the cells by centrifugation at 400 x g at 2–8 °C for 5 min. Aspirate the supernatants.
    6. Reconstitute the cells in 200 µL of assay buffer.
    7. Keep the cells on ice and proceed for analysis by FACS.

Subscription Required. Please recommend JoVE to your librarian.

Representative Results

DC-CYP-ALDH cells expressed significantly increased amount of 1α-hydroxylase. To determine whether DC-CYP-ALDH cells generated from BMDCs expressed a significantly increased amount of 1α-hydroxylase, BMDCs were transduced with the lenti-CYP-ALDH virus to produce bone-marrow-derived DC-CYP-ALDH cells (BMDC-CYP-ALDH cells). Subsequently, the BMDC-CYP-ALDH cells were examined for the expression of 1α-hydroxylase by FACS. Our data showed that the BMDC-CYP-ALDH cells, when compared to the parental BMDCs, displayed enhanced expression of the 1α-hydroxylase (Figure 1A). We also determined the enzymatic activity of the 1α-hydroxylase in the BMDC-CYP-ALDH cells. To do so, 1.0 x 106 BMDC-CYP-ALDH cells in 2 mL of CM-10-R cell culture medium were added into 12 well culture plates. 25(OH)D was then added to the cell culture at the final concentration of 2.5 μM. The cells were cultured at 37 ˚C and 5% CO2 for 24 h and the supernatants were harvested for the measurement of 1,25(OH)2D using a radioimmunoassay (RIA). Our data showed that 1,25(OH)2D concentrations in the culture supernatants of the BMDC-CYP cells (BMDCs transduced with lenti-CYP-GFP virus) and the BMDC-CYP-ALDH cells were each approximately 20x higher than those of the parental BMDCs and the BMDC-ALDH cells (BMDCs transduced with lenti-ALDH virus) (Figure 1B).

DC-CYP-ALDH cells expressed significantly increased amount of RALDH2. To determine whether BMDC-CYP-ALDH cells expressed a significantly increased amount of RALDH2, a RALDH2 substrate was added into the cell cultures in the presence or absence of the RALDH2 inhibitor diethylaminobenzaldehyde (DEAB) (15 µM). The fluorescent product retained inside the cells was analyzed by FACS. Our data showed that mean fluorescence intensities (MFIs) of the BMDC-CYP-ALDH cells were approximately 6x higher than those of the parental BMDCs, suggesting that the BMDC-CYP-ALDH cells, when compared to the parental BMDCs, expressed significantly enhanced RALDH2 enzymatic activity (Figure 2A,B).

DC-CYP-ALDH cells augmented the induction of foxp3+CCR9+ gut-homing Treg cells in vitro. To investigate whether DC-CYP-ALDH cells were able to augment the induction of gut-homing Treg cells in vitro, we transduced DC2.4 cells (a bone marrow-derived DC line26,27,28,29), with lenti-CYP-ALDH virus and generated DC2.4-CYP-ALDH cells. Subsequently, we determined whether the DC2.4-CYP-ALDH cells were able to augment the induction of gut-homing Treg cells in vitro. Accordingly, naive CD4+ T cells were purified from C57BL/6 mice. Purified naive CD4+ T cells at 5 x 105 cells/well were then cocultured with either the parental DC2.4 cells (1 x 105 cells/well) or the DC2.4-CYP-ALDH cells (1 x 105 cells/well) in 24 well culture plates in a serum-free medium in the presence of an anti-CD3 monoclonal antibody (5 µg/mL) and recombinant human IL-2 (50 U/mL). In addition, 25(OH)D and retinol at various concentrations were also added into the cultures. The cells were incubated at 37 ˚C and 5% CO2. Five days later, the cells were analyzed by FACS for the expressions of foxp3 and c-c chemokine receptor type 9 (CCR9). Our data showed that in the presence of the substrates, the DC2.4 cells did not significantly change the abundance of foxp3+CCR9+ cells in the CD4+ T cell populations (Figure 3A,B). In contrast, the DC2.4-CYP-ALDH cells significantly enhanced the abundance of foxp3+CCR9+ cells among CD4+ T cells. In addition, the more 25(OH)D added, the greater the ability of the DC2.4-CYP-ALDH cells to increase the abundance of foxp3+CCR9+ cells among CD4+ T cells. Therefore, our data support that the DC-CYP-ALDH cells can augment the induction of gut-homing Treg cells in vitro.

DC-CYP-ALDH cells augmented the induction of foxp3+CCR9+ gut-homing Treg cells in vivo. To determine whether DC-CYP-ALDH cells could augment the induction of gut-homing Treg cells in vivo, we intraperitoneally administered one of the following cells into Balb/c mice: the parental DC2.4 cells, the DC2.4-CYP cells (DC2.4 cells transduced with lenti-CYP-GFP virus), and the DC-2.4-CYP-ALDH cells. Four days after the cell administration, mesenteric lymph nodes were examined by FACS (Figure 4A). Our data showed that the DC2.4-CYP-ALDH cells, when compared to the controls, significantly increased the abundance of foxp3+CCR9+ cells among CD3+ T cells (Figure 4B,C). Based on these results, we conclude that the DC-CYP-ALDH cell administration significantly augments the induction of foxp3+CCR9+ T cells in peripheral lymphoid tissues.

Figure 1
Figure 1: DC-CYP-ALDH cells expressed significantly increased amount of 1α-hydroxylase. (A) BMDC-CYP-ALDH cells were generated and analyzed by FACS. A representative FACS plot shows the expression of 1α-hydroxylase in the parental BMDCs and the BMDC-CYP-ALDH cells (gated on live cells). (B) 1α-hydroxylase substrate (i.e., 25(OH)D) was added into the DC cultures. 24 h later, the supernatants were collected and 1,25(OH)2D concentrations were measured. The data show concentrations of 1,25(OH)2D in the cultures of the parental BMDCs, the BMDC-CYP cells, the BMDC-ALDH cells, and the BMDC-CYP-ALDH cells. **p < 0.01. ANOVA test. n = 4. This figure is adapted from Xu et al.6. Copyright 2019. The American Association of Immunologist, Inc. Please click here to view a larger version of this figure.

Figure 2
Figure 2: DC-CYP-ALDH cells expressed significantly increased amount of RALDH2. (A) BMDC-CYP-ALDH cells were generated and analyzed as described in the protocol. Representative overlaid FACS plots show the BODIPY aminoacetate fluorescence in the BMDCs and the BMDC-CYP-ALDH cells in the presence (+DEAB) or absence (-DEAB) of the RALHD2 inhibitor diethylaminobenzaldehyde (DEAB). (B) Mean fluorescence intensities (MFIs) of BODIPY aminoacetate in the BMDCs and the BMDC-CYP-ALDH cells in the absence of DEAB. *p < 0.05; t-test; n = 4. This figure is adapted from Xu et al.6. Copyright 2019. The American Association of Immunologist, Inc. Please click here to view a larger version of this figure.

Figure 3
Figure 3: DC-CYP-ALDH cells augmented the induction of foxp3+CCR9+ gut-homing Treg cells in vitro. (A) CD4+ naive T cells were isolated from C57BL/6 mouse spleens. The CD4+ T cells were then activated in cultures by an anti-CD3 mAb (5 µg/mL) in the presence of either the parental DC2.4 cells or the DC2.4-CYP-ALDH cells. Additionally, the cultures were added with the indicated concentrations of 25(OH)D and retinol. Five days later, the cells were collected and analyzed by FACS for the expressions of foxp3 and CCR9 in CD3+CD4+ T cell population. Representative FACS plots show the expressions of foxp3 and CCR9 in the CD3+CD4+ T cell populations. (B) Cumulative data from (A). *p < 0.05; ANOVA test; n = 4. This figure is adapted from Xu et al.6. Copyright 2019. The American Association of Immunologist, Inc. Please click here to view a larger version of this figure.

Figure 4
Figure 4: DC-CYP-ALDH cells augmented the induction of foxp3+CCR9+ gut-homing Treg cells in vivo. (A) Balb/c mice intraperitoneally (i.p.) received one of the following DC transfers (Transfer): no DC transfer (No transfer), parental DC2.4 cells, DC2.4-CYP cells, and DC2.4-CYP-ALDH cells. Four days later, mesenteric lymph nodes (MLNs) were analyzed by FACS. (B) Representative FACS plots show the expressions of foxp3 and CCR9 in CD3+ T cell population. (C) Cumulative data from (B) show the percentage of foxp3+CCR9+ cells in the CD3+ T cell population. Cells were gated on CD3+ T cells for all the analyses. Where applicable, the data presented are means ± SEM. *p < 0.05; ANOVA test; n = 4–6. This figure is adapted from Xu et al.6. Copyright 2019. The American Association of Immunologist, Inc. Please click here to view a larger version of this figure.

Subscription Required. Please recommend JoVE to your librarian.

Discussion

In this article we describe the use of DC-CYP-ALDH cells, for augmenting the induction of gut-homing Treg cells in peripheral lymphoid tissues. Our data have shown that the DC-CYP-ALDH cells can synthesize locally high concentrations de novo of both 1,25(OH)2D and RA in vitro in the presence of corresponding substrates (i.e., 25[OH]D and retinol, respectively). Because sufficient blood concentrations of 25(OH)D and retinol can be easily achieved through vitamin D and A supplementations respectively in patients who have deficiencies30,31, we reason that the DC-CYP-ALDH cells can augment the induction of gut-homing Treg cells in peripheral lymphoid tissues when normal blood concentrations of 25(OH)D and retinol are present. To support this reasoning, our data demonstrates that in normal healthy animals that do not have vitamin D and vitamin A deficiencies, the DC-CYP-ALDH cells augment the induction of Treg cells that express both regulatory molecules (i.e., foxp3 and IL-10) and a gut-homing receptor (i.e., CCR9). Therefore, this technology can be used for further investigation of gut-homing Treg cells for the treatment of IBD.

One critical step of this protocol is the production of lenti-CYP-ALDH virus with high titers. The preferred virus titers should be 108–109 TUs/mL. A high titer of the lenti-CYP-ALDH virus is necessary for a high transduction efficiency in DCs.

Another critical step of this protocol is the transduction efficiency in DCs. Because DC-CYP-ALDH cells are not tolerized in vitro in this technology, it is essential that the transduction rate be more than 90% to ensure that the DC-CYP-ALDH cells can efficiently augment the induction of gut-homing Treg cells in vivo. In addition, the DC-CYP-ALDH cells can be further purified by FACS before in vivo administration32.

A unique advantage of this protocol is that the DC-CYP-ALDH cells do not need to be tolerized in vitro before in vivo administration. It is expected that the DC-CYP-ALDH cells, as a result of the combined actions of 1,25(OH)2D and RA, will be maintained in a tolerogenic status in vivo because both 1,25(OH)2D and RA have been shown to tolerize DCs33,34. Therefore, we anticipate that the DC-CYP-ALDH cells will not have instability concerns in an in vivo proinflammatory environment.

Currently, we have only demonstrated that DC-CYP-ALDH cells can increase the frequency (number) of gut-homing Treg cells in peripheral lymphoid tissues and intestines6. Consequently, regulatory function in the intestines as a whole is enhanced because the percent of Treg cells in the intestines is increased. Figure 4 shows that when compared to those with control treatments, intraperitoneal treatment with DC-CYP-ALDH cells significantly increased the percentage of CCR9+foxp3+ Treg cells in the mesenteric lymph nodes, meaning that more Treg cells in the mesenteric lymph nodes are able to specifically home into the intestinal tissues. Figure 4 further shows that most foxp3+ T cells in mesenteric lymph nodes are negative for CCR9 and therefore do not have gut-homing capacity. However, whether DC-CYP-ALDH cells can also enhance the regulatory function of each Treg cell per se (such as enhanced expression levels of foxp3 and/or IL-10) requires further investigation.

The reagents and materials described here are for animal studies only. However, the protocol is applicable for human studies by using corresponding human reagents and materials, except that DCs will be generated from peripheral blood monocytes in humans. The ultimate goal of this protocol is the generation of clinical grade DC-CYP-ALDH cells for the treatment of IBD.

Subscription Required. Please recommend JoVE to your librarian.

Disclosures

Drs. Xiaolei Tang and David J. Baylink are inventors of a pending patent related to this study.

Acknowledgments

This work was supported by the Office of the Assistant Secretary of Defense for Health Affairs through the Peer Reviewed Medical Research Program under Award No. W81XWH-15-1-0240 (XT). Opinions, interpretations, conclusions, and recommendations are those of the author and are not necessarily endorsed by the Department of Defense. This work was also partially supported by Research Innovation Grants from the Department of Medicine at Loma Linda University (681207-2967 [XT and GG], 681205-2967 [XT], and 325491 [DJB]).

Materials

Name Company Catalog Number Comments
10 mL syringes ThermoFisher Scientific Cat# 03-377-23
100 mm x 20 mm culture dishes Sigma-Aldrich Cat# CLS430167
12-well culture plates ThermoFisher Scientific Cat# 07-200-82
150 mm x 25 mm culture dishes Sigma-Aldrich Cat# CLS430559
25-hydroxycholecalciferol (25[OH]D) Sigma-Aldrich Cat# H4014
293T cells ATCC CRL-3216
2-mercaptoethanol ThermoFisher Scientific Cat#: 21985023
6-well culture plates ThermoFisher Scientific Cat# 07-200-83
ALDEFLUOR kit Stemcell Technologies Cat# 01700
Anti-CYP27B1 Abcam Cat# ab95047
BD FACSAria II BD Biosciences N/A
CaCl2 Sigma-Aldrich Cat# C1016
CM-10-D cell culture medium DMEM medium containing 10% fetal bovine serum (FBS), 100 U/ml penicillin/streptomycin, 0.055 mM 2-mercaptoethanol (2-ME), 1 mM sodium pyruvate, 0.1 mM nonessential amino acid, and 2 mM L-glutamine.
CM-10-R cell culture medium RPMI 1640 medium (no glutamine) containing 10% fetal bovine serum (FBS), 100 U/ml penicillin/streptomycin, 0.055 mM 2-mercaptoethanol (2-ME), 1 mM sodium pyruvate, 0.1 mM nonessential amino acid, and 2 mM L-glutamine.
CM-4-D cell culture medium DMEM medium containing 4% fetal bovine serum (FBS), 100 U/ml penicillin/streptomycin, 0.055 mM 2-mercaptoethanol (2-ME), 1 mM sodium pyruvate, 0.1 mM nonessential amino acid, and 2 mM L-glutamine.
Corning bottle-top vacuum filters, 0.22 mM, 500 mL Sigma-Aldrich Cat# CLS430513
Corning bottle-top vacuum filters, 0.45 mM, 500 mL Sigma-Aldrich Cat# CLS430514
Dissecting scissor ThermoFisher Scientific Cat# 08-940
DMEM medium ThermoFisher Scientific Cat# 11960044
Fetal bovine serum ThermoFisher Scientific Cat# 16000044
Forceps ThermoFisher Scientific Cat# 22-327379
Gibco ACK lysing buffer ThermoFisher Scientific Cat# A1049201
Glycerol Sigma-Aldrich Cat# G5516
Goat anti-rabbit IgG Abcam Cat# ab205718
HEPES Millipore Cat# 391340
Lenti-CYP-ALDH Custom-made 1.6-kb mouse CYP27B1 and ALDH1a2 cDNAs were amplified by PCR using a plasmid containing the CYP27B1 cDNA and a plasmid containing the ALDH1a2 cDNA respectively (GeneCopoeia). The amplified CYP27B1 cDNA fragment with a 5' KOZAK ribosome entry sequence was cloned into the pRRL-SIN.cPPt.PGKGFP.WPRE lentiviral vector (Addgene). The resulting construct was designated as lenti-CYP-GFP. The amplified ALDH1a2 cDNA fragment was cloned into the lenti-CYP-GFP to replace the GFP and was designated as lenti-CYP-ALDH. This bicistronic plasmid expresses CYP27B1 controlled by SFFV promoter and ALDH1a2 controlled by PGK promoter.
L-glutamine ThermoFisher Scientific Cat#25030081
Lipopolysaccharide Sigma-Aldrich Cat# L3755
Murine GM-CSF Peprotech Cat# 315-03
Murine IL-4 Peprotech Cat# 214-14
Na2HPO4 Sigma-Aldrich Cat# NIST2186II
NaCl Sigma-Aldrich Cat# S9888
Needles ThermoFisher Scientific Cat# 14-841-02
Nonessential Amino Acids ThermoFisher Scientific Cat#: 11140076
pCMVR8.74 Addgene Plasmid# 22036
Penicillin/Streptomycin ThermoFisher Scientific Cat#15140148
Phoshate Balanced Solution (PBS) ThermoFisher Scientific Cat#: 20012027
PMD2G Addgene Plasmid# 12259
Polypropylene tube, 15 mL ThermoFisher Scientific Cat# AM12500
Polypropylene tube, 50 mL ThermoFisher Scientific Cat# AM12502
Protamine sulfate Sigma-Aldrich Cat# P3369
Rabbit polycloncal IgG isotype control Abcam Cat# ab171870
Radioimmunoassay for 1,25(OH)2D measurement Heartland Assays
RPMI 1640 medium, no glutamine ThermoFisher Scientific Cat# 21870076
Sodium pyruvat ThermoFisher Scientific Cat#: 11360070
Sorvall Legend XTR Centrifuge ThermoFisher Scientific Cat# 75004521
Sterile Cell strainers, 40 mm ThermoFisher Scientific Cat# 07-201-430
Sterile storage bottles, 500 mL ThermoFisher Scientific Cat# CLS431432

DOWNLOAD MATERIALS LIST

References

  1. Abraham, C., Cho, J. H. Inflammatory bowel disease. New England Journal of Medicine. 361 (21), 2066-2078 (2009).
  2. Kaser, A., Zeissig, S., Blumberg, R. S. Inflammatory bowel disease. Annual Reviews in Immunology. 28, 573-621 (2010).
  3. Clifford, D. B., et al. Natalizumab-associated progressive multifocal leukoencephalopathy in patients with multiple sclerosis: lessons from 28 cases. Lancet Neurology. 9 (4), 438-446 (2010).
  4. Linda, H., et al. Progressive multifocal leukoencephalopathy after natalizumab monotherapy. New England Journal of Medicine. 361 (11), 1081-1087 (2009).
  5. Fischer, A., et al. Differential effects of alpha4beta7 and GPR15 on homing of effector and regulatory T cells from patients with UC to the inflamed gut in vivo. Gut. 65 (10), 1642-1664 (2016).
  6. Xu, Y., et al. In Vivo Generation of Gut-Homing Regulatory T Cells for the Suppression of Colitis. Journal of Immunology. 202 (12), 3447-3457 (2019).
  7. Rosenblum, M. D., Way, S. S., Abbas, A. K. Regulatory T cell memory. Nature Reviews Immunology. 16 (2), 90-101 (2016).
  8. Grimm, A. J., Kontos, S., Diaceri, G., Quaglia-Thermes, X., Hubbell, J. A. Memory of tolerance and induction of regulatory T cells by erythrocyte-targeted antigens. Science Report. 5, 15907 (2015).
  9. Kim, H. J., et al. Stable inhibitory activity of regulatory T cells requires the transcription factor Helios. Science. 350 (6258), 334-339 (2015).
  10. Bhela, S., et al. The Plasticity and Stability of Regulatory T Cells during Viral-Induced Inflammatory Lesions. Journal of Immunology. 199 (4), 1342-1352 (2017).
  11. Bluestone, J. A., et al. Type 1 diabetes immunotherapy using polyclonal regulatory T cells. Science Translational Medicine. 7 (315), (2015).
  12. Marek-Trzonkowska, N., et al. Therapy of type 1 diabetes with CD4(+)CD25(high)CD127-regulatory T cells prolongs survival of pancreatic islets - results of one year follow-up. Clinical Immunology. 153 (1), 23-30 (2014).
  13. Desreumaux, P., et al. Safety and efficacy of antigen-specific regulatory T-cell therapy for patients with refractory Crohn's disease. Gastroenterology. 143 (5), 1201-1202 (2012).
  14. Di Ianni, M., et al. Tregs prevent GVHD and promote immune reconstitution in HLA-haploidentical transplantation. Blood. 117 (14), 3921-3928 (2011).
  15. Brunstein, C. G., et al. Infusion of ex vivo expanded T regulatory cells in adults transplanted with umbilical cord blood: safety profile and detection kinetics. Blood. 117 (3), 1061-1070 (2011).
  16. Kang, S. W., et al. 1,25-Dihyroxyvitamin D3 promotes FOXP3 expression via binding to vitamin D response elements in its conserved noncoding sequence region. Journal of Immunology. 188 (11), 5276-5282 (2012).
  17. Correale, J., Ysrraelit, M. C., Gaitan, M. I. Immunomodulatory effects of Vitamin D in multiple sclerosis. Brain. 132, Pt 5 1146-1160 (2009).
  18. Iwata, M., et al. Retinoic acid imprints gut-homing specificity on T cells. Immunity. 21 (4), 527-538 (2004).
  19. Steinman, R. M., Banchereau, J. Taking dendritic cells into medicine. Nature. 449 (7161), 419-426 (2007).
  20. Vicente-Suarez, I., Brayer, J., Villagra, A., Cheng, F., Sotomayor, E. M. TLR5 ligation by flagellin converts tolerogenic dendritic cells into activating antigen-presenting cells that preferentially induce T-helper 1 responses. Immunology Letters. 125 (2), 114-118 (2009).
  21. Danova, K., et al. NF-kappaB, p38 MAPK, ERK1/2, mTOR, STAT3 and increased glycolysis regulate stability of paricalcitol/dexamethasone-generated tolerogenic dendritic cells in the inflammatory environment. Oncotarget. 6 (16), 14123-14138 (2015).
  22. Liu, P. T., et al. Toll-like receptor triggering of a vitamin D-mediated human antimicrobial response. Science. 311 (5768), 1770-1773 (2006).
  23. Cao, H., et al. Application of vitamin D and vitamin D analogs in acute myelogenous leukemia. Experimental Hematology. 50, 1-12 (2017).
  24. Anderson, A., et al. Lasting Impact of Clostridium difficile Infection in Inflammatory Bowel Disease: A Propensity Score Matched Analysis. Inflammatory Bowel Disease. 23 (12), 2180-2188 (2017).
  25. Tsai, J. H., et al. Association of Aneuploidy and Flat Dysplasia With Development of High-Grade Dysplasia or Colorectal Cancer in Patients With Inflammatory Bowel Disease. Gastroenterology. 153 (6), 1492-1495 (2017).
  26. Lee, H. W., et al. Tracking of dendritic cell migration into lymph nodes using molecular imaging with sodium iodide symporter and enhanced firefly luciferase genes. Science Reports. 5, 9865 (2015).
  27. Shen, Z., Reznikoff, G., Dranoff, G., Rock, K. L. Cloned dendritic cells can present exogenous antigens on both MHC class I and class II molecules. Journal of Immunology. 158 (6), 2723-2730 (1997).
  28. Okada, N., et al. Administration route-dependent vaccine efficiency of murine dendritic cells pulsed with antigens. British Journal of Cancer. 84 (11), 1564-1570 (2001).
  29. Li, C. H., et al. Dendritic cells, engineered to overexpress 25-hydroxyvitamin D 1alpha-hydroxylase and pulsed with a myelin antigen, provide myelin-specific suppression of ongoing experimental allergic encephalomyelitis. FASEB J. , (2017).
  30. Narula, N., et al. Impact of High-Dose Vitamin D3 Supplementation in Patients with Crohn's Disease in Remission: A Pilot Randomized Double-Blind Controlled Study. Digestive Disease Science. 62 (2), 448-455 (2017).
  31. Ahmad, S. M., et al. Vitamin A Supplementation during Pregnancy Enhances Pandemic H1N1 Vaccine Response in Mothers, but Enhancement of Transplacental Antibody Transfer May Depend on When Mothers Are Vaccinated during Pregnancy. Journal of Nutrition. 148 (12), 1968-1975 (2018).
  32. Noronha, S. M. R., et al. Aldefluor protocol to sort keratinocytes stem cells from skin. Acta Cirurgica Brasileira. 32 (11), 984-994 (2017).
  33. Ferreira, G. B., et al. Vitamin D3 Induces Tolerance in Human Dendritic Cells by Activation of Intracellular Metabolic Pathways. Cell Reports. 10 (5), 711-725 (2015).
  34. Bakdash, G., Vogelpoel, L. T., van Capel, T. M., Kapsenberg, M. L., de Jong, E. C. Retinoic acid primes human dendritic cells to induce gut-homing, IL-10-producing regulatory T cells. Mucosal Immunology. 8 (2), 265-278 (2015).

Tags

In Vivo Augmentation Gut-homing Regulatory T Cell Induction Therapeutic Strategy Human Applications Peripheral Lymphoid Tissues Pro-inflammatory Environments Active Vitamin D Vitamin A Engineered Dendritic Cells CM-10-D Cell Culture Medium 293T Cells HBS Solution Envelope Plasmid Packaging Plasmid LENTI-CYP-ALDH Plasmid Calcium Chloride
In Vivo Augmentation of Gut-Homing Regulatory T Cell Induction
Play Video
PDF DOI DOWNLOAD MATERIALS LIST

Cite this Article

Bi, H., Wasnik, S., Baylink, D. J.,More

Bi, H., Wasnik, S., Baylink, D. J., Liu, C., Tang, X. In Vivo Augmentation of Gut-Homing Regulatory T Cell Induction. J. Vis. Exp. (155), e60585, doi:10.3791/60585 (2020).

Less
Copy Citation Download Citation Reprints and Permissions
View Video

Get cutting-edge science videos from JoVE sent straight to your inbox every month.

Waiting X
Simple Hit Counter