Waiting
Login processing...

Trial ends in Request Full Access Tell Your Colleague About Jove

Immunology and Infection

Adjuvant Activity of Mycobacterium paratuberculosis in Enhancing the Immunogenicity of Autoantigens During Experimental Autoimmune Encephalomyelitis

Published: May 12, 2023 doi: 10.3791/65422

Summary

Here, we present an alternative protocol to actively induce experimental autoimmune encephalomyelitis in C57BL/6 mice, using the immunogenic epitope myelin oligodendrocyte glycoprotein (MOG)35-55 suspended in incomplete Freund's adjuvant containing the heat-killed Mycobacterium avium subspecies paratuberculosis.

Abstract

Experimental autoimmune encephalomyelitis (EAE) induced by myelin oligodendrocyte glycoprotein (MOG) requires immunization by a MOG peptide emulsified in complete Freund's adjuvant (CFA) containing inactivated Mycobacterium tuberculosis. The antigenic components of the mycobacterium activate dendritic cells to stimulate T-cells to produce cytokines that promote the Th1 response via toll-like receptors. Therefore, the amount and species of mycobacteria present during the antigenic challenge are directly related to the development of EAE. This methods paper presents an alternative protocol to induce EAE in C57BL/6 mice using a modified incomplete Freund's adjuvant containing the heat-killed Mycobacterium avium subspecies paratuberculosis strain K-10.

M. paratuberculosis, a member of the Mycobacterium avium complex, is the causative agent of Johne's disease in ruminants and has been identified as a risk factor for several human T-cell-mediated disorders, including multiple sclerosis. Overall, mice immunized with Mycobacterium paratuberculosis showed earlier onset and greater disease severity than mice immunized with CFA containing the strain of M. tuberculosis H37Ra at the same doses of 4 mg/mL. The antigenic determinants of Mycobacterium avium subspecies paratuberculosis (MAP) strain K-10 were able to induce a strong Th1 cellular response during the effector phase, characterized by significantly higher numbers of T-lymphocytes (CD4+ CD27+), dendritic cells (CD11c+ I-A/I-E+), and monocytes (CD11b+ CD115+) in the spleen compared to mice immunized with CFA. Furthermore, the proliferative T-cell response to the MOG peptide appeared to be highest in M. paratuberculosis-immunized mice. The use of an encephalitogen (e.g., MOG35-55) emulsified in an adjuvant containing M. paratuberculosis in the formulation may be an alternative and validated method to activate dendritic cells for priming myelin epitope-specific CD4+ T-cells during the induction phase of EAE.

Introduction

Experimental autoimmune encephalomyelitis (EAE) is a common model for the study of human demyelinating disorders1. There are several models of EAE: active immunization using different myelin peptides in combination with potent adjuvants, passive immunization by in vitro transfer of myelin-specific CD4+ lymphocytes, and transgenic models of spontaneous EAE2. Each of these models has specific features that allow different aspects of EAE to be studied, such as the onset, effector phase, or chronic phase. The myelin oligodendrocyte glycoprotein (MOG) model of EAE is a good model to study the immune-mediated mechanisms of chronic neuroinflammation and demyelination, as it is characterized by mononuclear inflammatory infiltration, demyelination in peripheral white matter, and reduced recovery after the disease peak1.

MOG-EAE is induced by immunization of susceptible mice with the peptide MOG35-55 in complete Freund's adjuvant (CFA), followed by an intraperitoneal injection of pertussis toxin. This increases the permeability of the blood-brain barrier and allows myelin-specific T-cells activated in the periphery to reach the central nervous system (CNS), where they will be reactivated3. CFA plays a key role in the induction of EAE by enhancing the antigen uptake by antigen-presenting cells and the expression of cytokines related to humoral- and cell-mediated responses4. This mechanism is mainly due to the presence of killed Mycobacterium tuberculosis emulsified in oil, the components of which provide a strong stimulus for the immune system5. In fact, the induction of EAE is directly related to the amount of mycobacterium present during the antigenic challenge6.

The addition of other killed mycobacteria, such as Mycobacterium butyricum, to incomplete Freund's adjuvant7, as well as the effect of adjuvant combinations8, can modulate the clinical course of EAE and consequently influence the reproducibility of the results. Mycobacterium avium subspecies paratuberculosis (MAP), the etiologic agent of Johne's disease in ruminants, has been associated with inflammatory disorders of the human CNS9, as its antigenic components are capable of eliciting a strong humoral- and cell-mediated response in patients with multiple sclerosis and neuromyelitis optica spectrum disorder9. Therefore, in this protocol, we show an alternative and reproducible method to induce MOG-EAE by replacing M. tuberculosis in CFA with M. paratuberculosis.

Subscription Required. Please recommend JoVE to your librarian.

Protocol

All mouse experiments were approved by the Institutional Animal Care and Use Committee of the Juntendo University School of Medicine (Approval Number 290238) and were conducted in accordance with the National Institutes of Health Guidelines for Animal Experimentation.

1. General comments on the experiment

  1. House the mice in individual cages in the animal facility under controlled, pathogen-free conditions at 23 °C ± 2 °C with 50% ± 10% humidity, and a 12 h light/dark cycle with ad libitum access to food and water.
  2. Inject the mice with phosphate-buffered saline (PBS) without antigen and CFA, and use them as negative and positive controls, respectively.

2. Preparation of mycobacterial antigens

  1. Grow the M. paratuberculosis K-10 strain in Middlebrook liquid medium 7H9 enriched with 10% OADC (oleic acid, albumin, dextrose, catalase), 0.005% Tween 80, and 2 mg/L of mycobactin J for 2 weeks in T25 tissue culture flasks at 37 °C. Assess the colony growth regularly by visual inspection.
    CAUTION: Handle M. paratuberculosis in a Biosafety Level 2 (BSL-2) facility.
  2. Grow the enrichment cultureina 500 mL Erlenmeyer flask with a suspension of 1.7 × 105 colony-forming units (CFU)/mL in a final volume of 200 mL (same medium as step 2.1) in a shaking incubator for 1 week.
    NOTE: As contaminating organisms may affect the results, the bacteria must be inoculated onto Middlebrook 7H10 solid media to determine colony morphology and to verify purity by conventional polymerase chain reaction (PCR) detection kits for M. paratuberculosis.
  3. Inactivate the bacterial suspensions for 5-10 min at 70 °C and centrifuge at 3,000 × g for 10 min. Washed the weighed pellet in PBS twice, disrupt by sonication, and store at -20 °C.
  4. Transfer the pellet to a sterile container and freeze it with dry ice or liquid nitrogen. Transfer immediately to a freeze-drying chamber.
  5. Freeze-dry (4 h at -50 °C under vacuum) M. paratuberculosis according to the machine manual.
  6. At the end of lyophilization, remove the stainless-steel container from the freeze-dryer and transfer it to a bio-cleaning bench. Use a stainless-steel spatula to dislodge the dried MAP lumps adhering to the inner wall of the stainless-steel container and pulverize them as finely as possible.
  7. Weigh the pulverized cells using an electronic balance and manually place them in an aseptic 10 mL vial at a concentration of 10 mg/mL.
    NOTE: The cell density was quantified as grams of dry weight per liter of sample. After 3 weeks, 1 mg (wet weight) of bacterial cell pellet contains approximately 2.5 × 108 CFU.

3. Preparation of MOG 35-55 emulsion

  1. Grind the contents of one vial of dried M. paratuberculosis (10 mg) to a fine powder with a mortar and pestle and add 10 mL to incomplete Freund's adjuvant to obtain a 10 mg/mL stock solution. Store at -4 °C.
  2. Prior to immunization, dilute the stock solution to a final concentration of 4 mg/mL.
  3. Dilute the peptide MOG35-55 in ddH2O to a final concentration of 2 mg/mL and store at -20 °C.
  4. Using a homogenizer, mix the MOG35-55 solution (2 mg/mL) with an equal volume of adjuvant (4 mg/mL) from step 3.2 in a 5 mL tube until a thick emulsion is formed. After every 10 s of mixing, place the solution on ice for 20 s, and spin the tube to recover all the solution.
  5. Transfer the emulsion into a 1 mL syringe, remove all air, and add a 27 G needle. The emulsion is now ready for injection.
    NOTE. Since some loss of the viscous emulsion of MOG35-55 occurs during preparation, it is best to prepare 1.5 times the amount needed.

4. Animal immunization

  1. Anesthetize the animals with inhalation of isoflurane to minimize stress on the animals.
  2. Inject the emulsion (200 µL containing 200 µg/mouse of MOG35-55) subcutaneously into the lower back.
  3. Administer a dose of pertussis toxin (100 µL containing 200 ng/mouse) intraperitoneally on days 0 and 2 after immunization.
    ​CAUTION: Pertussis toxin has a multiple suppressive effect on the immune system; avoid ingestion and contact with eyes and skin.

5. Clinical assessment

  1. Monitor the mice daily for weight and clinical signs. Use the following scoring system: 0 = no clinical signs; 1 = flaccid tail; 2 = impairment of the righting reflex and weakness of the hind limbs; 3 = complete paralysis of the hind limbs; 4 = complete paralysis of the hind limbs with partial paralysis of the forelimbs; 5 = moribund.
    NOTE: Upon the initial observation of clinical signs, animals are provided with increased access to water and food. Rodent chow is softened and moistened, then placed on the cage floor from the food hopper. In the case of dehydrated animals, subcutaneous fluid supplementation is administered, or a rodent chow slurry is given via oral gavage. If a mouse necessitates this type of assistance for over 3 days, euthanasia will be conducted.
  2. To avoid or minimize animal pain and distress, use the following human endpoints: body weight loss greater than 20%, clinical score ≥4.0, absence of the righting reflex at score 3, and when the animal is unable to access feed or water for 24 h.
    NOTE: The mice were euthanized on day 30 after EAE induction by intraperitoneal injections of pentobarbital (≥150 mg/kg).

Subscription Required. Please recommend JoVE to your librarian.

Representative Results

Groups of C57BL/6 mice (total n = 15/group) were immunized with MOG35-55 in an emulsion containing M. paratuberculosis or by the common method with CFA. All groups of mice manifested an acute monophasic disease characterized by a single peak of disability observed at 14-17 days, followed by a partial recovery of symptoms over the next 10 days (Figure 1A). Mice immunized with the adjuvant containing M. paratuberculosis, irrespective of sex, showed an earlier onset from 8 to 9 days after immunization and greater severity in the acute phase than mice immunized with CFA (Figure 1B).There was no significant difference in body weight among the groups (Figure 1C). Cytofluorimetric analysis of spleen cells from EAE mice and proliferative activity of T-lymphocytes assessed by 3H-thymidine incorporation were performed, as previously published11. All spleen cells of EAE mice, regardless of the adjuvant used, showed a strong proliferative response to the peptide MOG35-55, whereas they did not proliferate in response to the control peptide ovalbumin (Figure 1D). M. paratuberculosis-immunized EAE mice showed an increased proportion of T-lymphocytes (CD4+ CD27+), dendritic cells (CD11c+I-A/I-E+), and monocytes (CD11b+CD115+) in the spleen compared to CFA-immunized mice during the acute phase of EAE (Figure 1E). Histological analysis by hematoxylin-eosin revealed a typical perivascular and meningeal mononuclear inflammatory infiltration in the brain and spinal cord (Figure 1F).

Figure 1
Figure 1: Representative EAE results. (A) Clinical scores of mice evaluated daily; (B) differences in disease course; (C) body weight; (D) T-cell proliferative response to the MOG35-55 peptide; (E) immune cell populations in splenocytes during the acute phase; (F) histological images of spinal cord sections (4x magnification) stained with hematoxylin-eosin(scale bar = 200 µm) during the acute phase. Arrows indicate inflammatory infiltrates. The data show combined results of three independent experiments (n = 5 mice/group/experiment). Data are expressed as mean ± standard deviation calculated using a one-way ANOVA followed by Bonferroni's post hoc test or Mann-Whitney U test. Abbreviations: EAE = experimental autoimmune encephalomyelitis; MOG = myelin oligodendrocyte glycoprotein; PBS = phosphate-buffered saline; CFA = complete Freund's adjuvant. Please click here to view a larger version of this figure.

Subscription Required. Please recommend JoVE to your librarian.

Discussion

We demonstrated a robust alternative protocol to actively induce severe EAE in C57BL/6J mice using the peptide MOG35-55 emulsified in an adjuvant containing M. paratuberculosis10. The induction of EAE by this method resulted in a more severe disease than that induced by the common protocol with CFA. This difference could be due to the different lipidic components in the cell wall of the mycobacteria11. In fact, unlike other mycobacteria, M. paratuberculosis produces a lipopentapeptide antigen on the cell wall surface instead of glycopeptidolipids11. This lipopentapeptide did not cross-react with other closely related species and has been shown to be a target for a strong specific humoral response in patients with autoimmune disorders12. Mycobacteria possess pathogen-associated molecular patterns that play different roles in immune responses9. For instance, we have recently shown that oral immunization with M. paratuberculosis increases the severity of MOG-EAE13, whereas vaccination with bacillus Calmette-Guerin (BCG)-Tokyo-172 appears to have a protective role in the progression of active and spontaneous EAE models14.

Potential limitations of the protocol include the self-limited monophasic course of EAE, which may became chronic if the concentrations of mycobacterium in the adjuvant and pertussis toxin are doubled. This aspect is very important to consider, especially when studying the neurodegenerative aspect of EAE using knockout mice, as the absence of recovery phase may be caused by the high doses of the reagents used and not by the lack of function of certain genes. Furthermore, although the protocol described above can be applied to other EAE protocols by varying the strains and age of mice or the type and amount of protein, to ensure that EAE experiments are performed in a methodologically correct manner, experimental groups must be matched for age and sex.

Since the incidence of the disease may vary, recommendations for troubleshooting are: the optimal M. paratuberculosis concentration can range from 2 to 4 mg/mL, and a three-way connector can be used for mixing small volumes of the antigen in the aqueous phase. We have described a method for preparing the emulsion by using a homogenizer; however, an alternative and simple method for mixing small volumes of antigen in the aqueous phase, in the oil phase, involves the use of a three-way connector fitting, to which two glass syringes are connected. In conclusion, we identified M. paratuberculosis as a potent adjuvant candidate in the induction of active EAE. Further studies on the mode of action and the type of immunity induced in different animals species will increase confidence in the possibility of adopting this alternative method to understand the mechanism of neuroinflammation.

Subscription Required. Please recommend JoVE to your librarian.

Disclosures

The authors have no conflicts of interest to disclose.

Acknowledgments

This work received support from a grant from the Japanese Society for the promotion of Science (grant no. JP 23K14675).

Materials

Name Company Catalog Number Comments
anti-mouse CD115 antibody Biolegend, USA 135505 for cytofluorimetry 1:1,000
anti-mouse CD11b antibody Biolegend, USA 101215 for cytofluorimetry 1:1,000
anti-mouse CD11c antibody Biolegend, USA 117313 for cytofluorimetry 1:1,000
anti-mouse CD16/32  antibody Biolegend, USA 101302 for cytofluorimetry 1:1,000
anti-mouse CD4  antibody Biolegend, USA 116004 for cytofluorimetry 1:1,000
anti-mouse CD8a  antibody Biolegend, USA 100753 for cytofluorimetry 1:1,000
anti-mouse I-A/I-E antibody Biolegend, USA 107635 for cytofluorimetry 1:1,000
anti-mouse Ly-6C  antibody Biolegend, USA 128023 for cytofluorimetry 1:1,000
BBL Middlebrook OADC Enrichment Thermo Fisher Scientific, USA BD 211886 for isolation and cultivation of mycobacteria
C57BL/6J mice Charles River Laboratory, Japan 3 weeks old, male and female
FBS 10279-106 Gibco Life Techologies, USA 42F9155K for cell culture, warm at 37 °C before use
Freeze Dryer machine Eyela, Tokyo, Japan FDU-1200 for bacteria lyophilization
incomplete e Freund’s adjuvant Difco Laboratories, MD, USA 263810 for use in adjuvant
Middlebrook 7H9 Broth Difco Laboratories, MD, USA 90003-876 help in the growth of Mycobacteria
Mycobacterium avium subsp. paratuberculosis K-10 ATCC, USA BAA-968 bacteria from bovine origin
Mycobacterium tuberculosis H37 Ra, Desiccated BD Biosciences, USA 743-26880-EA for use in adjuvant
Mycobactin J Allied Laboratory, MO, USA growth promoter
Myelin Oligodendrocyte Glycolipid (MOG) 35-55 AnaSpec, USA AS-60130-10 encephalotigenic peptide
Ovalbumin (257-264) Sigma-Aldrich, USA S7951-1MG negative control antigen  for proliferative assay
pertussis toxin solution Fujifilm Wako, Osaka Japan 168-22471 From gram-negative bacteria Bordetella pertussi, increases blood-brain barrier permeability
Polytron homogenizer PT 3100 Kinematica for mixing the antigen with the adjuvant
RPMI 1640 with L-glutamine Gibco Life Techologies, USA 11875093 For cell culture
Thymidine, [Methyl-3H], in 2% ethanol, 1 mCi PerkinElmer, Waltham, MA, USA NET027W001MC for proliferation assay, use (1 μCi/well)
Zombie NIR Fixable Viability Kit Biolegend, USA 423105  cytofluorimetry, for cell viability

DOWNLOAD MATERIALS LIST

References

  1. Bittner, S., Afzali, A. M., Wiendl, H., Meuth, S. G. Myelin oligodendrocyte glycoprotein (MOG35-55) induced experimental autoimmune encephalomyelitis (EAE) in C57BL/6 mice. Journal of Visualized Experiments. (86), e51275 (2014).
  2. Constantinescu, C. S., Farooqi, N., O'Brien, K., Gran, B. Experimental autoimmune encephalomyelitis (EAE) as a model for multiple sclerosis (MS). British Journal of Pharmacology. 164 (4), 1079-1106 (2011).
  3. Lu, C., et al. Pertussis toxin induces angiogenesis in brain microvascular endothelial cells. Journal of Neuroscience Research. 86 (12), 2624-2640 (2008).
  4. Awate, S., Babiuk, L. A., Mutwiri, G. Mechanisms of action of adjuvants. Frontiers in Immunology. 4, 114 (2013).
  5. Kubota, M., et al. Adjuvant activity of Mycobacteria-derived mycolic acids. Heliyon. 6 (5), e04064 (2020).
  6. Nicolo, C., et al. Mycobacterium tuberculosis in the adjuvant modulates the balance of Th immune response to self-antigen of the CNS without influencing a "core" repertoire of specific T cells. International Immunology. 18 (2), 363-374 (2006).
  7. O'Connor, R. A., et al. Adjuvant immunotherapy of experimental autoimmune encephalomyelitis: immature myeloid cells expressing CXCL10 and CXCL16 attract CXCR3+CXCR6+ and myelin-specific T cells to the draining lymph nodes rather than the central nervous system. Journal of Immunology. 188 (5), 2093-2101 (2012).
  8. Libbey, J. E., Fujinami, R. S. Experimental autoimmune encephalomyelitis as a testing paradigm for adjuvants and vaccines. Vaccine. 29 (17), 3356-3362 (2011).
  9. Cossu, D., Yokoyama, K., Hattori, N. Conflicting role of Mycobacterium species in multiple sclerosis. Frontiers in Neurology. 8, 216 (2017).
  10. Cossu, D., Yokoyama, K., Sakanishi, T., Momotani, E., Hattori, N. Adjuvant and antigenic properties of Mycobacterium avium subsp. paratuberculosis on experimental autoimmune encephalomyelitis. Journal of Neuroimmunology. 330, 174-177 (2019).
  11. Biet, F., et al. Lipopentapeptide induces a strong host humoral response and distinguishes Mycobacterium avium subsp. paratuberculosis from M. avium subsp. avium. Vaccine. 26 (2), 257-268 (2008).
  12. Cossu, D., Yokoyama, K., Tomizawa, Y., Momotani, E., Hattori, N. Altered humoral immunity to mycobacterial antigens in Japanese patients affected by inflammatory demyelinating diseases of the central nervous system. Scientific Reports. 7 (1), 3179 (2017).
  13. Cossu, D., et al. A mucosal immune response induced by oral administration of heat-killed Mycobacterium avium subsp. paratuberculosis exacerbates EAE. Journal of Neuroimmunology. 352, 577477 (2021).
  14. Cossu, D., Yokoyama, K., Sakanishi, T., Sechi, L. A., Hattori, N. Bacillus Calmette-Guerin Tokyo-172 vaccine provides age-related neuroprotection in actively induced and spontaneous experimental autoimmune encephalomyelitis models. Clinical and Experimental Immunology. 6, (2023).

Tags

Adjuvant Activity Mycobacterium Paratuberculosis Immunogenicity Autoantigens Experimental Autoimmune Encephalomyelitis Neurological Diseases Treatment Movement Disorders Interactive Diseases Pathogens Etiology Inflammatory Diseases Multiple Sclerosis NMOSD Genetic Factors Environmental Factors Humoral Response Cell-mediated Response Gut Immune Brain Axis EAE Model Mycobacterium Tuberculosis Lipo Pentapeptide Antigen Glycopeptide Lipids Immune Response Peripheral Nervous System Central Nervous System
Adjuvant Activity of <em>Mycobacterium paratuberculosis</em> in Enhancing the Immunogenicity of Autoantigens During Experimental Autoimmune Encephalomyelitis
Play Video
PDF DOI DOWNLOAD MATERIALS LIST

Cite this Article

Cossu, D., Tomizawa, Y., Momotani,More

Cossu, D., Tomizawa, Y., Momotani, E., Yokoyama, K., Hattori, N. Adjuvant Activity of Mycobacterium paratuberculosis in Enhancing the Immunogenicity of Autoantigens During Experimental Autoimmune Encephalomyelitis. J. Vis. Exp. (195), e65422, doi:10.3791/65422 (2023).

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