Open access peer-reviewed chapter

Mycobacterium ulcerans Disease and Host Immune Responses

Written By

Michael S. Avumegah

Submitted: 14 January 2022 Reviewed: 21 February 2022 Published: 14 May 2022

DOI: 10.5772/intechopen.103843

From the Edited Volume

New Advances in Neglected Tropical Diseases

Edited by Márcia Aparecida Sperança

Chapter metrics overview

170 Chapter Downloads

View Full Metrics

Abstract

Mycobacterium ulcerans is the causative agent of the subcutaneous necrotic condition known as Buruli ulcer (BU).BU is Neglected Tropical Disease. The bacillus is the third most common mycobacteria disease-causing agent after Mycobacterium tuberculosis and Mycobacterium leprae. M. ulcerans produces the toxin-Mycolactone, which plays a key role in the pathophysiological features of the disease. Buruli ulcer has been reported in 34 countries, mainly in the tropics and subtropics. Tropical countries include Benin, Cameroon, Ghana, Democratic Republic of Congo and Nigeria. BU is also prevalent in Queensland, a subtropical region, and in Victoria, a temperate area, all within Australia. The exact mode of the transmission remains unclear. However, M. ulcerans is believed to have an aquatic niche. Initial diagnosis of BU is based on the experience of the clinician, but PCR targeting the M. ulcerans DNA, IS2404, isolation and culture of the bacillus and histopathology are used for confirmation. The current, commonly used methods for confirmatory diagnosis have logistic and resource challenges. Novel cell mediated immunity (CMI) and serology-based tests would be beneficial to provide a more accurate assessment of population exposure.

Keywords

  • Mycobacterium ulcerans
  • Buruli ulcer disease

1. Introduction

Mycobacterium ulcerans is the causative agent of the subcutaneous necrotic condition known as Buruli ulcer (Figure 1) [2, 3]. The bacillus is the third most common mycobacteria disease causing agent after Mycobacterium tuberculosis and Mycobacterium leprae [4].

Figure 1.

A severe Buruli ulcer on the upper limb. Image was taken from WHO website [1].

M. ulcerans produces the toxin-Mycolactone (Figure 2), which plays a key role in the pathophysiological features of the disease [6, 7, 8]. Buruli ulcer has been reported in 34 countries (Figure 3), mainly in the tropics and subtropics [10]. Tropical countries include Benin [11], Cameroon [12, 13, 14], Ghana [15], Democratic Republic of Congo [16] and Nigeria [17, 18]. BU is also prevalent in Queensland [19], a subtropical region, and in Victoria, a temperate area, all within Australia [20].

Figure 2.

Mycolactone structure [5].

Figure 3.

Distribution of Buruli ulcer, worldwide, 2020 [9].

The exact mode of the transmission remains unclear [10, 21]. However, M. ulcerans is believed to have an aquatic niche [22, 23, 24, 25, 26]. Aquatic insects, belonging to the Family: Belostomatidae and Naucoridae have been implicated as possible vectors in Africa [22, 27, 28]. In Australia, mosquitoes [20] and possum [29] have been suggested as vector and reservoir [20, 29] respectively. Initial diagnosis of BU is based on the experience of the clinician [30], but PCR targeting the M. ulcerans DNA, IS2404 [31], isolation and culture of the bacillus and histopathology are used for confirmation [32, 33, 34].

In Africa, the disease is common in the rural setting where access to medical facilities and other infrastructure is limited [35, 36]. The opposite is true in Australia [10]. More cases have been reported among people living near wetlands [21, 37]. Gender is not a risk factor [10, 38], however age has been identified as a non-environmental risk factor [10, 38]. BU is prevalence among children between 5 and 15 age group in Africa [38]. Whereas in Australia the >60 age group are most susceptible [39, 40, 41]. This has been attributed to the ageing population in Australia, as well as immunosenescence [41, 42, 43].

Advertisement

2. Mycobacterium ulcerans infection

The time of infection or exposure to M. ulcerans to showing clinical symptoms of BU ranges from 1 week to 9 months [44]. The median incubation period is 4.5 months [44]. Non-ulcerative and ulcerative are the two main forms of the disease. The non-ulcerative form manifests as either painless nodule, swelling, plaque or oedema [45].

Opened necrotic lesion on the skin describes the ulcerative form and is categorised into 3 forms. Category 1 ulcers are single lesion on the skin and 5 cm in diameter [46]. Ulcers with size 5 to 15 cm is characterised as category 2 [46]. Multiples ulcers on different part of the body and/or ulcers greater than 15 cm is clinically described as category 3 [46]. African cohort usually report category 3 and 2 ulcers whereas category 1 are common in Australia [46]. This has been attributed to differences in geographical strains and virulence factor of M. ulcerans [47]. Access to adequate medical services in rural endemic regions in Africa and late presentation of condition to medical centres is another reason [30, 48].

Rook et al., described 3 phases of immune response to M. ulcerans in murine model in 1975 [49]. The first phase involves leucocyte migration and delayed hypersensitivity response at the site of infection [49]. This response is usually cell-mediated and involves proliferation of T-cells, monocytes and macrophages. In the next phase, as M. ulcerans cells multiple at the site of infection, there is subsequent reduction in the migration of inflammatory biomarkers at this site [49]. Though, there appears to be reactive cells in the lymph nodes [49]. The last phase is marked by depletion of T-cells [49]. This is attributed to excessive exposure to the bacillus toxins (mycolactone) [49]. Studies in tuberculosis and BU patients also confirms that individuals could have a negative skin-test while their lymph tissues is filled with enormous responsive cells [50, 51]. These 3 phases describes the phenomena of “sensitization” and “desensitisation” in mycobacteria infection. The latter being the non-appearances of cell-mediated response to a previously encountered antigen [49, 52].

Advertisement

3. Role of mycolactone in disease progression

Mycolactone is responsible for the necrotic nature of the disease [6, 7]. It is essentially a lipid-like toxin [6]. The exact role in the bacillus is unknown [6]. However, it is speculated to offer predatory protection from eukaryotic microorganisms [6]. Currently, 5 variants of mycolactone has been described and is grouped into A/B, C, D, E and F [53, 54]. Mycolactone A/B which is the most virulent variant is produced by M. ulcerans strains of African, Japan and Malaysia origin [55]. Ulcers are therefore, the most severe and difficult to treat [55]. Mycolactone C and D are localised to strains in the pacific regions, specifically Australia and China, respectively [55]. Mycobacterium liflandii, a West African frog pathogen produces the “E” variants [56, 57]. Mycolactone F is synthesised by Mycobacterium pseudoshottsii and Mycobacterium marinum [55].

Mycolactone has been observed to have cytopathic effect on host cells both in vitro and in vivo [6, 7, 58]. M. ulcerans upon entry into the body establishes a localised niche and multiplies along with mycolactone release [49]. The host mounts an initial cell-mediated response to clear the infection [49]. However, mycolactone functions as immune modulation and immunosuppressive agent [49, 59]. Thus preventing communication and recruitment of other important immune cells to the site of trauma [49, 60, 61]. Bacillus overload and dead inflammatory cells is usually seen as closed nodule-like or painless swellings (non-ulcerative). At this early stage, infection may clear or resolve naturally but persistent production of mycolactone eventually leads to necrotic and open ulcers [46, 49, 51].

Advertisement

4. Mycobacteria genus

Mycobacteria species are among well-studied genus of microorganism and M. tuberculosis has been an exemplar [62, 63]. The ubiquitous nature of organisms in this genus has huge medical implication [64]. Over 100 species have been identified and characterised from soil and water [64]. Mycobacteria could be divided into two main categories namely, tuberculous and non-tuberculous mycobacteria (NTM) or atypical [64]. Tuberculous mycobacteria causes diseases such as pulmonary tuberculous disease and leprosy [63]. Non-tuberculous mycobacterial species include M. ulcerans, M. avium, M. intracellularae, M. kansasii, M. xenopi, M. chelonae, M. marinum and M. fortuitum [63].

Advertisement

5. General overview of immune response in mammals

The natural and initial response of mammals to invading pathogens, including mycobacteria is the innate immune response [65, 66]. This involves recruitment of inflammatory cells (granulocytes) to the site of infection and subsequent ingestion and disruption of the harmful agent, which then is engulfed by macrophages [67, 68]. These are necessitated by innate immune cells, hence the name cell-mediated immunity [66]. Cell-mediate response is transient (acute) and last 1 day to 2 weeks [66]. However, humoral (antibodies) response or immunity which forms after initial expose to a pathogen is long lasting [66]. In adaptive immunity, two cells play key role [66]. These are the T (T-cell) and B (B-cell) lymphocytes [66]. T-cells develop in the bone marrow and matures in the thymus, they recognises and fights viral particulates. B-cells on the other hand matures in the bone marrow and work to eliminate bacteria and viruses from the body [66].

5.1 T-cells

T-cell is divided into “helper T cell (Th)”, “cytotoxic T cell (Tc)”, “natural killer T cell (NKT)” and “memory T cell (Tm)” [66]. Th cells are responsible for activation and proliferation of cytokines. They also assist in the maturation of B cells and activation of Tc and macrophages [66]. Helper T cell is divided into Type 1 (Th1) and Type 2 (Th2) T-helper cells [66]. Cell-mediated response is usually aided by Th1. Resulting in the recruitment of macrophages to an infected site for action [66]. Interferon gamma (IFNγ) and macrophages are the main effector biomarkers of Th1 immune response [66]. Th1 response is usually mounted against intracellular bacteria and protozoa [66]. Th2 on the other hand aids in the activation and proliferation of B cells, and is a component of humoral immunity [66]. Th2 cells functions to clear extracellular bacteria from the body [66]. Eosinophils basophils, and mast cells are important effector agents of Th2 immune response [66]. Cytotoxic T cell (Tc) is responsible for the disruption and cytolysis of host cells infected by virus [66]. They also play key role in tumour and cancer immunity [66]. Natural killer T cell (NKT) also assist in B cell activation and microbial defence [66].

5.2 B-cells

B cells is a component of the white blood cells and humoral immunity [66]. B lymphocytes secrete antibodies which are critical in pathogen specific recognition and elimination [66]. These cells are essential in the production of antigen-specific immunoglobulin (Ig) which are the agents that binds to pathogens [66]. Major sites of B cells in human include the blood and the lymph nodes [66]. Spleen and tonsil are other areas B cell are be found [66]. B cells and T cells work in concert to eliminate pathogens from the body [66]. After production and maturation in the bone marrow, B cells circulate through the blood and lymphoid system. Upon encounter with a specific pathogen, binding occurs. Subsequent assistance from the T cells lead to the elimination of the invading cell from the host [66].

Advertisement

6. Immune profile of active BU patients, exposed and unexposed controls

Screening for cytokines and antibodies specific responses in BU patients is still a matter of intense research [59, 60, 69, 70, 71, 72, 73, 74, 75]. This is because individuals with active disease are incapable of mounting an effective specific immune response to M. ulcerans [63]. It is known that patients seem to have reduced expression of Th1 cytokines [49, 69, 70]. In comparison, Th2 response is common among active ulcerative patients and those with healed ulcers [69, 70]. The Th2 cytokine cells include interleukin (IL) 4, IL-5, IL-6, and IL-10. Exposed household contact with no clinical symptoms also have Th2 response [69, 70]. However, BU patients are more likely to express IL-4 and IL-10 [69, 70, 76]. Th1 immune response is characterised by expression of IFNγ and IL-12, critical mediators for macrophages recruitments [66, 69, 70, 76]. Interestingly, previous studies have shown that histopathology of ulcerative tissue biopsy shows macrophages with acid fast bacteria overload (likely to be M. ulcerans) [73, 77]. Mycolactone functions to either supress or deviate immune cells that could lead to the effective clearance of M. ulcerans from the host [61, 78]. Therefore, first responder cytokines such as tumour necrotic factors (TNF) is downregulated. TNF is responsible for septic shock and it works in concert with IL-17 in the release of inflammation infiltrates to infected areas and induces fever [60, 79].

Advertisement

7. Immune profile of BU patients undergoing antibiotic treatment

BU is curable [80], but treatment could take months if not diagnosed early [80]. Like many bacterial infection, appropriate antimicrobial medications is required in killing M. ulcerans to wane off mycolactone production [81, 82]. Currently, 8 weeks of oral rifampicin, is the preferred antibiotic regimen in many medical centres [46, 83]. Like mycolactone, rifampicin is a polyketide, and functions to inhibit the ribonucleic acid (RNA) polymerase activity of bacteria [84]. Thus leading to the shutdown of important amino acids, protein synthesis and inhibiting the growth of the bacillus [84]. Other medications/treatment tested or used in combination with rifampicin include Streptomycin, Oral Fluoroquinolones [85], Amikacin [86], Clofazimine [87], Co-trimoxazole [87], Ofloxacin [87], Cephalosporin [87], Penicillin [87], Metronidazol [87], Minocyclin [87], dapsone [87], Phenytoin powder [88], Topical nitrogen oxides, mycobacteriophage D29 [89], clay and local heat [87]. Cytokine profile of patients undergoing antibiotic treatment have shown that there is gradual re-establishment of Th1 immune response during the healing process [90]. These inflammatory biomarkers include IL-4, IL-7 and IL-8 [90].

Advertisement

8. Paradoxical reactions in BU treatment

Paradoxical reaction in the healing process has also been reported [91]. This describes a stage where instead of the ulcers healing during antibiotic treatment, it rather becomes worst [91]. The exact mechanism underlining paradoxical reaction in BU healing is poorly understood. It has been speculated that immediate restoration of cytokine response to M. ulcerans antigens after mycolactone wash out could be the plausible explanation.

Advertisement

9. BU vaccines

There is currently no specific vaccine for BU [92]. But Bacillus Calmette–Guérin (BCG) vaccine has been used to check for cross-protection in M. ulcerans infection in Uganda [50]. BCG is a purified protein derivative (PPD) from Mycobacterium bovis [63]. It is the main vaccination regimen for TB and some non-tuberculous mycobacteria (NTM) [63, 64]. It stimulates a strong Th1 immune response. This include IFNγ-secreting CD4+ T cell response in recipient [64]. It was observed that BCG offered a protective effect between 47 to 50% among those tested in Uganda [50, 93]. However, this protection was short-lived, the reason is unclear [50, 94]. The main protective agent in BCG vaccine is the peptide Ag85A [92]. It is a highly conserved motif and plays a critical role in cell wall synthesis [95, 96]. Ag85A homologue from M. ulcerans have also been screened for cross-protection in BU disease and have shown prospect [96]. M. ulcerans surface protein −18 kDa small heat shock protein (MUL_2232) [97] and MUL_3720 [98] have also been used in vaccine formulation against M. ulcerans [99]. In a murine experiment, a recombinant (r) vaccine formulation (rMUL2232 and rMUL3720) failed to protect against BU even though antibodies were present [99]. It appears that M. ulcerans specific antibodies fail to protect against developing overt disease [100]. Although mycolactone is the culprit in this debilitating disease [6], vaccine based on it has been thought off as a “goose chase”. The reason being, mycolatone is a polyketide, a lipid-like derivative and not necessary immunogenic [92]. Animal experiment by Robert et al 1997 did not find mycolactone an ideal candidate for diagnosis or vaccine [58].

Advertisement

10. BU and host susceptibility

M. ulcerans is an environmental pathogen [64]. Several studies have reported M. ulcerans DNA target both in BU endemic and non-endemic regions [21]. In addition, Sero-epidemiological studies have also shown that antibodies for the immuno-dominant protein, 18 kDa shsp is present in healthy controls [100]. This has raised numerous questions and hypotheses on host genetics and susceptibility [36]. From previous reports, it appears not all those exposed to M. ulcerans develop overt disease [36]. Some genetic marks such as solute carrier family 11, member 1 (SLC11A1) [36], autophagy-related genes E3 ubiquitin-protein ligase (PARK2) and autophagy-related protein 16–1 (ATG16L1) [36] nucleotide-binding oligomerization domain-containing 2 (NOD2) have been suggested as key players in BU susceptibility [36]. If host susceptibility is at play, it could change our perception on the mode of transmission and treatment of BU.

References

  1. 1. World Health Organization (Buruli Ulcer) [Internet]. 2018. Available from: https://www.who.int/news-room/facts-in-pictures/detail/buruli-ulcer#:~:text=Buruli%20ulcer%20is%20caused%20by,develop%20severe%20scars%20and%20deformities [Accessed: 2022-04-07]
  2. 2. Clancey JK. Mycobacterial skin ulcers in Uganda: Description of a new Mycobacterium (Mycobacterium Buruli). The Journal of Pathology and Bacteriology. 1964;88:175-187
  3. 3. Hayman J, McQueen A. The pathology of Mycobacterium ulcerans infection. Pathology. 1985;17(4):594-600
  4. 4. Weir E. Buruli ulcer: The third most common mycobacterial infection. CMAJ. 2002;166(13):1691
  5. 5. Matthias G, Karl-Heinz A. The chemistry and biology of mycolactones. Beilstein Journal of Organic Chemistry. 2017;13:1596-1660
  6. 6. George KM, Chatterjee D, Gunawardana G, Welty D, Hayman J, Lee R, et al. Mycolactone: A polyketide toxin from Mycobacterium ulcerans required for virulence. Science. 1999;283(5403):854-857
  7. 7. George KM, Pascopella L, Welty DM, Small PL. A Mycobacterium ulcerans toxin, mycolactone, causes apoptosis in guinea pig ulcers and tissue culture cells. Infection and Immunity. 2000;68(2):877-883
  8. 8. van der Werf TS, Stinear T, Stienstra Y, van der Graaf WT, Small PL. Mycolactones and Mycobacterium ulcerans disease. Lancet. 2003;362(9389):1062-1064
  9. 9. WHO. Distribution of Buruli Ulcer, Worldwide, 2020. 2020
  10. 10. O'Brien DP, Jeanne I, Blasdell K, Avumegah M, Athan E. The changing epidemiology worldwide of Mycobacterium ulcerans. Epidemiology and Infection. 2018;147:e19. DOI: 10.1017/S0950268818002662
  11. 11. Muelder K, Nourou A. Buruli ulcer in Benin. Lancet. 1990;336(8723):1109-1111
  12. 12. Boisvert H. Skin ulcer caused by Mycobacterium ulcerans in Cameroon. II. Bacteriological study. Bulletin de la Société de Pathologie Exotique et de ses Filiales. 1977;70(2):125-131
  13. 13. Ravisse P. Skin ulcer caused by Mycobacterium ulcerans in Cameroon. I. Clinical, epidemiological and histological study. Bulletin de la Societe de pathologie Exotique. 1977;70(2):109-124
  14. 14. Noeske J, Kuaban C, Rondini S, Sorlin P, Ciaffi L, Mbuagbaw J, et al. Buruli ulcer disease in Cameroon rediscovered. The American Journal of Tropical Medicine and Hygiene. 2004;70(5):520-526
  15. 15. van der Werf TS, van der Graaf WT, Groothuis DG, Knell AJ. Mycobacterium ulcerans infection in Ashanti region, Ghana. Transactions of the Royal Society of Tropical Medicine and Hygiene. 1989;83(3):410-413
  16. 16. Bafende AE, Phanzu MD, Imposo BB. Buruli ulcer in the Democratic Republic of Congo: Epidemiology, presentation and outcome. Tropical Doctor. 2004;34(2):82-84
  17. 17. Oluwasanmi JO, Solankee TF, Olurin EO, Itayemi SO, Alabi GO, Lucas AO. Mycobacterium ulcerans (Buruli) skin ulceration in Nigeria. The American Journal of Tropical Medicine and Hygiene. 1976;25(1):122-128
  18. 18. Chukwuekezie O, Ampadu E, Sopoh G, Dossou A, Tiendrebeogo A, Sadiq L, et al. Buruli ulcer, Nigeria. Emerging Infectious Diseases. 2007;13(5):782-783
  19. 19. Abrahams EW. Mycobacterium Ulcerans infection in Queensland. The Medical Journal of Australia. 1964;1:334-335
  20. 20. Johnson PD, Azuolas J, Lavender CJ, Wishart E, Stinear TP, Hayman JA, et al. Mycobacterium ulcerans in mosquitoes captured during outbreak of Buruli ulcer, southeastern Australia. Emerging Infectious Diseases. 2007;13(11):1653-1660
  21. 21. Merritt RW, Walker ED, Small PL, Wallace JR, Johnson PD, Benbow ME, et al. Ecology and transmission of Buruli ulcer disease: A systematic review. PLoS Neglected Tropical Diseases. 2010;4(12):e911
  22. 22. Marsollier L, Robert R, Aubry J, Saint Andre JP, Kouakou H, Legras P, et al. Aquatic insects as a vector for Mycobacterium ulcerans. Applied and Environmental Microbiology. 2002;68(9):4623-4628
  23. 23. Marsollier L, Stinear T, Aubry J, Saint Andre JP, Robert R, Legras P, et al. Aquatic plants stimulate the growth of and biofilm formation by Mycobacterium ulcerans in axenic culture and harbor these bacteria in the environment. Applied and Environmental Microbiology. 2004;70(2):1097-1103
  24. 24. Marsollier L, Severin T, Aubry J, Merritt RW, Saint Andre JP, Legras P, et al. Aquatic snails, passive hosts of Mycobacterium ulcerans. Applied and Environmental Microbiology. 2004;70(10):6296-6298
  25. 25. Silva MT, Portaels F, Pedrosa J. Aquatic insects and Mycobacterium ulcerans: An association relevant to Buruli ulcer control? PLoS Medicine. 2007;4(2):e63
  26. 26. Benbow ME, Williamson H, Kimbirauskas R, McIntosh MD, Kolar R, Quaye C, et al. Aquatic invertebrates as unlikely vectors of Buruli ulcer disease. Emerging Infectious Diseases. 2008;14(8):1247-1254
  27. 27. Marsollier L, Aubry J, Coutanceau E, Andre JP, Small PL, Milon G, et al. Colonization of the salivary glands of Naucoris cimicoides by Mycobacterium ulcerans requires host plasmatocytes and a macrolide toxin, mycolactone. Cellular Microbiology. 2005;7(7):935-943
  28. 28. Mosi L, Williamson H, Wallace JR, Merritt RW, Small PL. Persistent association of Mycobacterium ulcerans with west African predaceous insects of the family belostomatidae. Applied and Environmental Microbiology. 2008;74(22):7036-7042
  29. 29. Fyfe JA, Lavender CJ, Handasyde KA, Legione AR, O'Brien CR, Stinear TP, et al. A major role for mammals in the ecology of Mycobacterium ulcerans. PLoS Neglected Tropical Diseases. 2010;4(8):e791
  30. 30. Buntine J, Crofts K, World Health Organization, & Global Buruli Ulcer Initiative. Buruli ulcer: Management of Mycobacterium ulcerans disease: A manual for health care providers (No. WHO/CDS/CPE/GBUI/2001.3). World Health Organization; 2001
  31. 31. Ablordey A, Kotlowski R, Swings J, Portaels F. PCR amplification with primers based on IS2404 and GC-rich repeated sequence reveals polymorphism in Mycobacterium ulcerans. Journal of Clinical Microbiology. 2005;43(1):448-451
  32. 32. Fukunishi Y. Histopathological findings of Buruli ulcer: Cases from Benin in West Africa. Nihon Hansenbyō Gakkai Zasshi. 2002;71(3):179-186
  33. 33. Sakyi SA, Aboagye SY, Darko Otchere I, Yeboah-Manu D. Clinical and laboratory diagnosis of Buruli ulcer disease: A systematic review. Canadian Journal of Infectious Diseases and Medical Microbiology. 2016
  34. 34. Beissner M, Herbinger KH, Bretzel G. Laboratory diagnosis of Buruli ulcer disease. Future Microbiology. 2010;5(3):363-370
  35. 35. Giles-Vernick T, Owona-Ntsama J, Landier J, Eyangoh S. The puzzle of Buruli ulcer transmission, ethno-ecological history and the end of "love" in the Akonolinga district, Cameroon. Social Science & Medicine. 2015;129:20-27
  36. 36. Röltgen K, Pluschke G. Buruli ulcer: History and disease burden. Buruli Ulcer. 2019:1-41
  37. 37. Johnson PDR, Stinear T, Small PLC, Plushke G, Merritt RW, Portaels F, et al. Buruli ulcer (M-ulcerans infection): New insights, new hope for disease control. PLoS Medicine. 2005;2(4):282-286
  38. 38. Debacker M, Aguiar J, Steunou C, Zinsou C, Meyers WM, Scott JT, et al. Mycobacterium ulcerans disease: Role of age and gender in incidence and morbidity. Tropical Medicine & International Health. 2004;9(12):1297-1304
  39. 39. Quek TY, Henry MJ, Pasco JA, O'Brien DP, Johnson PD, Hughes A, et al. Mycobacterium ulcerans infection: Factors influencing diagnostic delay. The Medical Journal of Australia. 2007;187(10):561-563
  40. 40. O’Brien DP, Walton A, Hughes AJ, Friedman ND, McDonald A, Callan P, et al. Risk factors for recurrent Mycobacterium ulcerans disease after exclusive surgical treatment in an Australian cohort. The Medical Journal of Australia. 2013;198(8):436-439
  41. 41. Loftus MJ, Tay EL, Globan M, Lavender CJ, Crouch SR, Johnson PDR, et al. Epidemiology of Buruli ulcer infections, Victoria, Australia, 2011-2016. Emerging Infectious Diseases. 2018;24(11):1988-1997
  42. 42. O'Brien DP, Friedman ND, Cowan R, Pollard J, McDonald A, Callan P, et al. Mycobacterium ulcerans in the elderly: More severe disease and suboptimal outcomes. PLoS Neglected Tropical Diseases. 2015;9(12):e0004253
  43. 43. O'Brien DP, Wynne JW, Buultjens AH, Michalski WP, Stinear TP, Friedman ND, et al. Exposure risk for infection and lack of human-to-human transmission of Mycobacterium ulcerans disease, Australia. Emerging Infectious Diseases. 2017;23(5):837-840
  44. 44. Trubiano JA, Lavender CJ, Fyfe JA, Bittmann S, Johnson PD. The incubation period of Buruli ulcer (Mycobacterium ulcerans infection). PLoS Neglected Tropical Diseases. 2013;7(10):e2463
  45. 45. Hayman J. Clinical features of Mycobacterium ulcerans infection. The Australasian Journal of Dermatology. 1985;26(2):67-73
  46. 46. WHO. Laboratory Diagnosis of Buruli Ulcer Disease. 2014
  47. 47. Tai AYC, Athan E, Friedman ND, Hughes A, Walton A, O’Brien DP. Increased severity and spread of Mycobacterium ulcerans, Southeastern Australia. Emerging Infectious Diseases. 2018;24(1):58-64
  48. 48. Roltgen K, Pluschke G. Epidemiology and disease burden of Buruli ulcer: A review. Research and Reports in Tropical Medicine. 2015;6:59-73
  49. 49. Rook GA. The immunological consequences of antigen overload in experimental mycobacterial infections of mice. Clinical and Experimental Immunology. 1975;19(1):167-177
  50. 50. Group UB. BCG vaccination against mycobacterium ulcerans infection (Buruli ulcer). First results of a trial in Uganda. Lancet. 1969;1(7586):111-115
  51. 51. Rook GA, Carswell JW, Stanford JL. Preliminary evidence for the trapping of antigen-specific lymphocytes in the lymphoid tissue of ‘anergic’ tuberculosis patients. Clinical and Experimental Immunology. 1976;26(1):129-132
  52. 52. Shield MJ, Stanford JL, Paul RC, Carswell JW. Multiple skin testing of tuberculosis patients with a range of new tuberculins, and a comparison with leprosy and Mycobacterium ulcerans infection. Epidemiology & Infection. 1977;78(3):331-348
  53. 53. Ranger BS, Mahrous EA, Mosi L, Adusumilli S, Lee RE, Colorni A, et al. Globally distributed mycobacterial fish pathogens produce a novel plasmid-encoded toxic macrolide, mycolactone F. Infection and Immunity. 2006;74(11):6037-6045
  54. 54. Kishi Y. Chemistry of mycolactones, the causative toxins of Buruli ulcer. Proceedings of the National Academy of Sciences of the United States of America. 2011;108(17):6703-6708
  55. 55. Pidot SJ, Hong H, Seemann T, Porter JL, Yip MJ, Men A, et al. Deciphering the genetic basis for polyketide variation among mycobacteria producing mycolactones. BMC Genomics. 2008;9(1):1-15
  56. 56. Trott KA, Stacy BA, Lifland BD, Diggs HE, Harland RM, Khokha MK, et al. Characterization of a Mycobacterium ulcerans-like infection in a colony of African tropical clawed frogs (Xenopus tropicalis). Comparative Medicine. 2004;54(3):309-317
  57. 57. Tobias NJ, Doig KD, Medema MH, Chen H, Haring V, Moore R, et al. Complete genome sequence of the frog pathogen Mycobacterium ulcerans ecovar Liflandii. Journal of Bacteriology. 2013;195(3):556-564
  58. 58. Roberts B, Hirst RG. Diagnostic potential of a serological assay for the diagnosis of ulcerans disease based on the putative Mycobacterium ulcerans toxin. Journal of Medical Microbiology. 1997;46(4):333-339
  59. 59. Coutanceau E, Decalf J, Martino A, Babon A, Winter N, Cole ST, et al. Selective suppression of dendritic cell functions by Mycobacterium ulcerans toxin mycolactone. The Journal of Experimental Medicine. 2007;204(6):1395-1403
  60. 60. Pahlevan AA, Wright DJ, Andrews C, George KM, Small PL, Foxwell BM. The inhibitory action of Mycobacterium ulcerans soluble factor on monocyte/T cell cytokine production and NF-kappa B function. Journal of Immunology. 1999;163(7):3928-3935
  61. 61. Coutanceau E, Marsollier L, Brosch R, Perret E, Goossens P, Tanguy M, et al. Modulation of the host immune response by a transient intracellular stage of Mycobacterium ulcerans: The contribution of endogenous mycolactone toxin. Cellular Microbiology. 2005;7(8):1187-1196
  62. 62. Beer V, Bonifas V. Mycobacteria. Schweizerische Medizinische Wochenschrift. 1975;105(31):984-987
  63. 63. Barksdale L, Kim SK. Mycobaterium. Bacteriological Reviews. 1977;41(1):217-372
  64. 64. Kazda J, Pavlik I, Falkinham III JO, Hruska K. The Ecology of Mycobacteria: Impact on Animal’s and Human’s Health. Springer Science+ Business Media BV; 2009
  65. 65. Rastogi N, Legrand E, Sola C. The mycobacteria: An introduction to nomenclature and pathogenesis. Revue Scientifique et Technique. 2001;20(1):21-54
  66. 66. Riley E, Franks SA. Immunology and Evolution of Infectious Disease. Princeton University Press; 2002, 2006. p. 360. Hardback US 85.00,paperbackUS 37.95. ISBN 0 691 09595 7
  67. 67. Connor DH, Lunn HF. Mycobacterium ulcerans infection (with comments on pathogenesis). International Journal of Leprosy. 1965;33(Suppl. 3):698-709
  68. 68. Arango Duque G, Descoteaux A. Macrophage cytokines: Involvement in immunity and infectious diseases. Frontiers in Immunology. 2014;5:491
  69. 69. Gooding TM, Johnson PD, Smith M, Kemp AS, Robins-Browne RM. Cytokine profiles of patients infected with Mycobacterium ulcerans and unaffected household contacts. Infection and Immunity. 2002;70(10):5562-5567
  70. 70. Gooding TM, Kemp AS, Robins-Browne RM, Smith M, Johnson PD. Acquired T-helper 1 lymphocyte anergy following infection with Mycobacterium ulcerans. Clinical Infectious Diseases. 2003;36(8):1076-1077
  71. 71. Prevot G, Bourreau E, Pascalis H, Pradinaud R, Tanghe A, Huygen K, et al. Differential production of systemic and intralesional gamma interferon and interleukin-10 in nodular and ulcerative forms of Buruli disease. Infection and Immunity. 2004;72(2):958-965
  72. 72. Westenbrink BD, Stienstra Y, Huitema MG, Thompson WA, Klutse EO, Ampadu EO, et al. Cytokine responses to stimulation of whole blood from patients with Buruli ulcer disease in Ghana. Clinical and Diagnostic Laboratory Immunology. 2005;12(1):125-129
  73. 73. Kiszewski AE, Becerril E, Aguilar LD, Kader IT, Myers W, Portaels F, et al. The local immune response in ulcerative lesions of Buruli disease. Clinical and Experimental Immunology. 2006;143(3):445-451
  74. 74. Phillips R, Horsfield C, Kuijper S, Sarfo SF, Obeng-Baah J, Etuaful S, et al. Cytokine response to antigen stimulation of whole blood from patients with Mycobacterium ulcerans disease compared to that from patients with tuberculosis. Clinical and Vaccine Immunology. 2006;13(2):253-257
  75. 75. Peduzzi E, Groeper C, Schutte D, Zajac P, Rondini S, Mensah-Quainoo E, et al. Local activation of the innate immune system in Buruli ulcer lesions. The Journal of Investigative Dermatology. 2007;127(3):638-645
  76. 76. Zavattaro E, Mesturini R, Dossou A, Melensi M, Johnson RC, Sopoh G, et al. Serum cytokine profile during Mycobacterium ulcerans infection (Buruli ulcer). International Journal of Dermatology. 2010;49(11):1297-1302
  77. 77. Torrado E, Fraga AG, Logarinho E, Martins TG, Carmona JA, Gama JB, et al. IFN-gamma-dependent activation of macrophages during experimental infections by Mycobacterium ulcerans is impaired by the toxin mycolactone. Journal of Immunology. 2010;184(2):947-955
  78. 78. Phillips R, Sarfo FS, Guenin-Mace L, Decalf J, Wansbrough-Jones M, Albert ML, et al. Immunosuppressive signature of cutaneous Mycobacterium ulcerans infection in the peripheral blood of patients with Buruli ulcer disease. The Journal of Infectious Diseases. 2009;200(11):1675-1684
  79. 79. Yeboah-Manu D, Peduzzi E, Mensah-Quainoo E, Asante-Poku A, Ofori-Adjei D, Pluschke G, et al. Systemic suppression of interferon-gamma responses in Buruli ulcer patients resolves after surgical excision of the lesions caused by the extracellular pathogen Mycobacterium ulcerans. Journal of Leukocyte Biology. 2006;79(6):1150-1156
  80. 80. Yotsu RR, Murase C, Sugawara M, Suzuki K, Nakanaga K, Ishii N, et al. Revisiting Buruli ulcer. The Journal of Dermatology. 2015;42(11):1033-1041
  81. 81. Dega H, Robert J, Bonnafous P, Jarlier V, Grosset J. Activities of several antimicrobials against Mycobacterium ulcerans infection in mice. Antimicrobial Agents and Chemotherapy. 2000;44(9):2367-2372
  82. 82. Owusu E, Newman MJ, Addo KK, Addo P. In vitro susceptibility of Mycobacterium ulcerans isolates to selected antimicrobials. Canadian Journal of Infectious Diseases and Medical Microbiology. 2017;2017:5180984
  83. 83. Phillips RO, Sarfo FS, Landier J, Oldenburg R, Frimpong M, Wansbrough-Jones M, et al. Combined inflammatory and metabolic defects reflected by reduced serum protein levels in patients with Buruli ulcer disease. PLoS Neglected Tropical Diseases. 2014;8(4):e2786
  84. 84. Campbell EA, Korzheva N, Mustaev A, Murakami K, Nair S, Goldfarb A, et al. Structural mechanism for rifampicin inhibition of bacterial RNA polymerase. Cell. 2001;104(6):901-912
  85. 85. O’Brien DP, McDonald A, Callan P, Robson M, Friedman ND, Hughes A, et al. Successful outcomes with oral fluoroquinolones combined with rifampicin in the treatment of Mycobacterium ulcerans: An observational cohort study. PLoS Neglected Tropical Diseases. 2012;6(1):e1473
  86. 86. Marsollier L, Prevot G, Honore N, Legras P, Manceau AL, Payan C, et al. Susceptibility of Mycobacterium ulcerans to a combination of amikacin/rifampicin. International Journal of Antimicrobial Agents. 2003;22(6):562-566
  87. 87. Sizaire V, Nackers F, Comte E, Portaels F. Mycobacterium ulcerans infection: Control, diagnosis, and treatment. The Lancet Infectious Diseases. 2006;6(5):288-296
  88. 88. Adjei O, Evans MR, Asiedu A. Phenytoin in the treatment of Buruli ulcer. Transactions of the Royal Society of Tropical Medicine and Hygiene. 1998;92(1):108-109
  89. 89. Trigo G, Martins TG, Fraga AG, Longatto-Filho A, Castro AG, Azeredo J, et al. Phage therapy is effective against infection by Mycobacterium ulcerans in a murine footpad model. PLoS Neglected Tropical Diseases. 2013;7(4):e2183
  90. 90. Martins TG, Gama JB, Fraga AG, Saraiva M, Silva MT, Castro AG, et al. Local and regional Re-establishment of cellular immunity during curative Antibiotherapy of murine Mycobacterium ulcerans infection. PLoS One. 2012;7(2)
  91. 91. Barogui YT, Klis SA, Johnson RC, Phillips RO, van der Veer E, van Diemen C, et al. Genetic susceptibility and predictors of paradoxical reactions in Buruli ulcer. PLoS Neglected Tropical Diseases. 2016;10(4):e0004594
  92. 92. Huygen K. Prospects for vaccine development against Buruli disease. Expert Review of Vaccines. 2003;2(4):561-569
  93. 93. Smith PG, Revill WD, Lukwago E, Rykushin YP. The protective effect of BCG against Mycobacterium ulcerans disease: A controlled trial in an endemic area of Uganda. Transactions of the Royal Society of Tropical Medicine and Hygiene. 1976;70(5-6):449-457
  94. 94. Stanford JL, Paul RC. A preliminary study of the effect of contact with environmental mycobacteria on the pattern of sensitivity to a range of new tuberculins amongst Ugandan adults. J Hyg (Lond). 1976;76(2):205-214
  95. 95. Tanghe A, Content J, Van Vooren JP, Portaels F, Huygen K. Protective efficacy of a DNA vaccine encoding antigen 85A from Mycobacterium bovis BCG against Buruli ulcer. Infection and Immunity. 2001;69(9):5403-5411
  96. 96. Hart BE, Hale LP, Lee S. Recombinant BCG expressing Mycobacterium ulcerans Ag85A imparts enhanced protection against experimental Buruli ulcer. PLoS Neglected Tropical Diseases. 2015;9(9):e0004046
  97. 97. Diaz D, Dobeli H, Yeboah-Manu D, Mensah-Quainoo E, Friedlein A, Soder N, et al. Use of the immunodominant 18-kiloDalton small heat shock protein as a serological marker for exposure to Mycobacterium ulcerans. Clinical and Vaccine Immunology. 2006;13(12):1314-1321
  98. 98. Dreyer A, Roltgen K, Dangy JP, Ruf MT, Scherr N, Bolz M, et al. Identification of the Mycobacterium ulcerans protein MUL_3720 as a promising target for the development of a diagnostic test for Buruli ulcer. PLoS Neglected Tropical Diseases. 2015;9(2)
  99. 99. Bolz M, Benard A, Dreyer AM, Kerber S, Vettiger A, Oehlmann W, et al. Vaccination with the surface proteins MUL_2232 and MUL_3720 of Mycobacterium ulcerans induces antibodies but fails to provide protection against Buruli ulcer. PLoS Neglected Tropical Diseases. 2016;10(2):e0004431
  100. 100. Yeboah-Manu D, Roltgen K, Opare W, Asan-Ampah K, Quenin-Fosu K, Asante-Poku A, et al. Sero-epidemiology as a tool to screen populations for exposure to Mycobacterium ulcerans. PLoS Neglected Tropical Diseases. 2012;6(1)

Written By

Michael S. Avumegah

Submitted: 14 January 2022 Reviewed: 21 February 2022 Published: 14 May 2022