Memory T cells: promising biomarkers for evaluating protection and vaccine efficacy against leishmaniasis
Jazyk angličtina Země Švýcarsko Médium electronic-ecollection
Typ dokumentu časopisecké články, přehledy
PubMed
38469319
PubMed Central
PMC10925770
DOI
10.3389/fimmu.2024.1304696
Knihovny.cz E-zdroje
- Klíčová slova
- biomarkers, leishmaniasis, memory CD4+ T cells, tissue resident memory, vaccine,
- MeSH
- biologické markery MeSH
- CD4-pozitivní T-lymfocyty MeSH
- leishmanióza kožní * MeSH
- lidé MeSH
- paměťové T-buňky MeSH
- účinnost vakcíny MeSH
- vakcíny * MeSH
- Check Tag
- lidé MeSH
- Publikační typ
- časopisecké články MeSH
- přehledy MeSH
- Názvy látek
- biologické markery MeSH
- vakcíny * MeSH
Understanding the immune response to Leishmania infection and identifying biomarkers that correlate with protection are crucial for developing effective vaccines. One intriguing aspect of Leishmania infection is the persistence of parasites, even after apparent lesion healing. Various host cells, including dendritic cells, fibroblasts, and Langerhans cells, may serve as safe sites for latent infection. Memory T cells, especially tissue-resident memory T cells (TRM), play a crucial role in concomitant immunity against cutaneous Leishmania infections. These TRM cells are long-lasting and can protect against reinfection in the absence of persistent parasites. CD4+ TRM cells, in particular, have been implicated in protection against Leishmania infections. These cells are characterized by their ability to reside in the skin and rapidly respond to secondary infections by producing cytokines such as IFN-γ, which activates macrophages to kill parasites. The induction of CD4+ TRM cells has shown promise in experimental immunization, leading to protection against Leishmania challenge infections. Identifying biomarkers of protection is a critical step in vaccine development and CD4+ TRM cells hold potential as biomarkers, as their presence and functions may correlate with protection. While recent studies have shown that Leishmania-specific memory CD4+ T-cell subsets are present in individuals with a history of cutaneous leishmaniasis, further studies are needed to characterize CD4+ TRM cell populations. Overall, this review highlights the importance of memory T cells, particularly skin-resident CD4+ TRM cells, as promising targets for developing effective vaccines against leishmaniasis and as biomarkers of immune protection to assess the efficacy of candidate vaccines against human leishmaniasis.
Department of Medical Genetics 3rd Faculty of Medicine Charles University Prague Czechia
Department of Parasitology Pasteur Institute of Iran Tehran Iran
Zobrazit více v PubMed
Burza S, Croft SL, Boelaert M. Leishmaniasis. Lancet (London England) (2018) 392:951–70. doi: 10.1016/S0140-6736(18)31204-2 PubMed DOI
Ponte-Sucre A, Gamarro F, Dujardin JC, Barrett MP, López-Vélez R, García-Hernández R, et al. . Drug resistance and treatment failure in leishmaniasis: A 21st century challenge. PloS Negl Trop Dis (2017) 11:e0006052. doi: 10.1371/journal.pntd.0006052 PubMed DOI PMC
Kaye PM, Mohan S, Mantel C, Malhame M, Revill P, Le Rutte E, et al. . Overcoming roadblocks in the development of vaccines for leishmaniasis. Expert Rev Vaccines (2021) 20:1419–30. doi: 10.1080/14760584.2021.1990043 PubMed DOI PMC
Rostami MN, Khamesipour A. Potential biomarkers of immune protection in human leishmaniasis. Med Microbiol Immunol (2021) 210:81–100. doi: 10.1007/s00430-021-00703-8 PubMed DOI PMC
Sacks D, Noben-Trauth N. The immunology of susceptibility and resistance to Leishmania major in mice. Nat Rev Immunol (2002) 2:845–58. doi: 10.1038/nri933 PubMed DOI
Khamesipour A. Therapeutic vaccines for leishmaniasis. Expert Opin Biol Ther (2014) 14:1641–9. doi: 10.1517/14712598.2014.945415 PubMed DOI
Iborra S, Solana JC, Requena JM, Soto M. Vaccine candidates against leishmania under current research. Expert Rev Vaccines (2018) 17:323–34. doi: 10.1080/14760584.2018.1459191 PubMed DOI
Dinc R. Leishmania vaccines: the current situation with its promising aspect for the future. Korean J Parasitol (2022) 60:379–91. doi: 10.3347/kjp.2022.60.6.379 PubMed DOI PMC
Abdellahi L, Iraji F, Mahmoudabadi A, Hejazi SH. Vaccination in leishmaniasis: A review article. Iranian Biomed J (2022) 26:1–35. doi: 10.52547/ibj.26.1.35 PubMed DOI PMC
Kaye PM, Matlashewski G, Mohan S, Le Rutte E, Mondal D, Khamesipour A, et al. . Vaccine value profile for leishmaniasis. Vaccine (2023) 41 Suppl 2:S153–s75. doi: 10.1016/j.vaccine.2023.01.057 PubMed DOI
Younis BM, Osman M, Khalil EAG, Santoro F, Furini S, Wiggins R, et al. . Safety and immunogenicity of ChAd63-KH vaccine in post-kala-azar dermal leishmaniasis patients in Sudan. Mol therapy: J Am Soc Gene Ther (2021) 29:2366–77. doi: 10.1016/j.ymthe.2021.03.020 PubMed DOI PMC
Lacey C, Musa A, Khalil ET, Younis B, Osman M, Wiggins R, et al. . LEISH2b - A phase 2b study to assess the safety, efficacy, and immunogenicity of the Leishmania vaccine ChAd63-KH in post-kala azar dermal leishmaniasis. Wellcome Open Res (2022) 7:200. doi: 10.12688/wellcomeopenres PubMed DOI PMC
Duthie MS, MaChado BAS, Badaró R, Kaye PM, Reed SG. Leishmaniasis vaccines: applications of RNA technology and targeted clinical trial designs. Pathog (Basel Switzerland) (2022) 11(11):259. doi: 10.3390/pathogens11111259 PubMed DOI PMC
Coler RN, Duthie MS, Hofmeyer KA, Guderian J, Jayashankar L, Vergara J, et al. . From mouse to man: safety, immunogenicity and efficacy of a candidate leishmaniasis vaccine LEISH-F3+GLA-SE. Clin Trans Immunol (2015) 4:e35. doi: 10.1038/cti.2015.6 PubMed DOI PMC
Azami-Conesa I, Gómez-Muñoz MT, Martínez-díaz RA. A systematic review (1990-2021) of wild animals infected with zoonotic leishmania. Microorganisms (2021) 9(5):1101. doi: 10.3390/microorganisms9051101 PubMed DOI PMC
Shokri A, Fakhar M, Teshnizi SH. Canine visceral leishmaniasis in Iran: A systematic review and meta-analysis. Acta Tropica (2017) 165:76–89. doi: 10.1016/j.actatropica.2016.08.020 PubMed DOI
Marcolino Silva D, Passarella Teixeira AI, Sierra Romero GA. Socioeconomic status of guardians as a risk factor for canine visceral leishmaniasis: A cohort study in an endemic area of the federal district, Brazil. Am J Trop Med Hygiene (2023) 108:328–34. doi: 10.4269/ajtmh.21-1170 PubMed DOI PMC
Velez R, Gállego M. Commercially approved vaccines for canine leishmaniosis: a review of available data on their safety and efficacy. Trop Med Int Health: TM IH (2020) 25:540–57. doi: 10.1111/tmi.13382 PubMed DOI
Loría-Cervera EN, Andrade-Narváez FJ. Animal models for the study of leishmaniasis immunology. Rev do Instituto Medicina Trop Sao Paulo (2014) 56:1–11. doi: 10.1590/S0036-46652014000100001 PubMed DOI PMC
Janeway CA, Jr TP, Walport M, Shlomchik MJ. General properties of armed effector T cells. In: Immunobiology: The Immune System in Health and Disease. Garland Science, New York: (2001).
Scott P, Novais FO. Cutaneous leishmaniasis: immune responses in protection and pathogenesis. Nat Rev Immunol (2016) 16:581–92. doi: 10.1038/nri.2016.72 PubMed DOI
Nateghi Rostami M, Keshavarz Valian H, Eskandari SE, Miramin Mohammadi A, Shahrestani ST, Sarraf-Nejad A, et al. . Differential in vitro CD4+/CD8+ T-cell response to live vs. killed Leishmania major. Parasite Immunol (2010) 32:101–10. doi: 10.1111/pim.2010.32.issue-2 PubMed DOI
Sacks D, Anderson C. Re-examination of the immunosuppressive mechanisms mediating non-cure of Leishmania infection in mice. Immunol Rev (2004) 201:225–38. doi: 10.1111/j.0105-2896.2004.00185.x PubMed DOI
Alexander J, Bryson K. T helper (h)1/Th2 and Leishmania: paradox rather than paradigm. Immunol Lett (2005) 99:17–23. doi: 10.1016/j.imlet.2005.01.009 PubMed DOI
Gonçalves-de-Albuquerque SDC, Pessoa ESR, Trajano-Silva LAM, de Goes TC, de Morais RCS, da COCN, et al. . The equivocal role of th17 cells and neutrophils on immunopathogenesis of leishmaniasis. Front Immunol (2017) 8:1437. doi: 10.3389/fimmu.2017.01437 PubMed DOI PMC
Khatonier R, Ahmed G, Sarmah P, Narain K, Khan AM. Immunomodulatory role of Th17 pathway in experimental visceral leishmaniasis. Immunobiology (2021) 226:152148. doi: 10.1016/j.imbio.2021.152148 PubMed DOI
Lopez Kostka S, Dinges S, Griewank K, Iwakura Y, Udey MC, von Stebut E. IL-17 promotes progression of cutaneous leishmaniasis in susceptible mice. J Immunol (Baltimore Md: 1950) (2009) 182:3039–46. doi: 10.4049/jimmunol.0713598 PubMed DOI PMC
Gonzalez-Lombana C, Gimblet C, Bacellar O, Oliveira WW, Passos S, Carvalho LP, et al. . IL-17 mediates immunopathology in the absence of IL-10 following Leishmania major infection. PloS Pathog (2013) 9:e1003243. doi: 10.1371/journal.ppat.1003243 PubMed DOI PMC
Pitta MG, Romano A, Cabantous S, Henri S, Hammad A, Kouriba B, et al. . IL-17 and IL-22 are associated with protection against human kala azar caused by Leishmania donovani. J Clin Invest (2009) 119:2379–87. doi: 10.1172/JCI38813 PubMed DOI PMC
Nateghi Rostami M, Seyyedan Jasbi E, Khamesipour A, Mohammadi AM. Tumour Necrosis Factor-alpha (TNF-α) and its soluble receptor type 1 (sTNFR I) in human active and healed leishmaniases. Parasite Immunol (2016) 38:255–60. doi: 10.1111/pim.12305 PubMed DOI
Nateghi Rostami M, Seyyedan Jasbi E, Khamesipour A, Miramin Mohammadi A. Plasma levels of tumor necrosis factor-alpha (TNF-α), TNF-α soluble receptor type 1 (sTNFR I) and IL-22 in human leishmaniasis. Trop Biomed (2015) 32:478–84. PubMed
Asad M, Sabur A, Kamran M, Shadab M, Das S, Ali N. Effector functions of Th17 cells are regulated by IL-35 and TGF-β in visceral leishmaniasis. FASEB J (2021) 35:e21755. doi: 10.1096/fj.202002356RR PubMed DOI
Kemp M, Hey AS, Kurtzhals JA, Christensen CB, Gaafar A, Mustafa MD, et al. . Dichotomy of the human T cell response to Leishmania antigens. I. Th1-like response to Leishmania major promastigote antigens in individuals recovered from cutaneous leishmaniasis. Clin Exp Immunol (1994) 96:410–5. doi: 10.1111/j.1365-2249.1994.tb06043.x PubMed DOI PMC
Egui A, Ledesma D, Pérez-Antón E, Montoya A, Gómez I, Robledo SM, et al. . Phenotypic and functional profiles of antigen-specific CD4(+) and CD8(+) T cells associated with infection control in patients with cutaneous leishmaniasis. Front Cell Infect Microbiol (2018) 8:393. doi: 10.3389/fcimb.2018.00393 PubMed DOI PMC
Ajdary S, Riazi-Rad F, Alimohammadian MH, Pakzad SR. Immune response to Leishmania antigen in anthroponotic cutaneous leishmaniasis. J Infect (2009) 59:139–43. doi: 10.1016/j.jinf.2009.05.010 PubMed DOI
Ansari NA, Ramesh V, Salotra P. Interferon (IFN)-gamma, tumor necrosis factor-alpha, interleukin-6, and IFN-gamma receptor 1 are the major immunological determinants associated with post-kala azar dermal leishmaniasis. J Infect Dis (2006) 194:958–65. doi: 10.1086/506624 PubMed DOI
Bamorovat M, Sharifi I, Aflatoonian MR, Sadeghi B, Shafiian A, Oliaee RT, et al. . Host’s immune response in unresponsive and responsive patients with anthroponotic cutaneous leishmaniasis treated by meglumine antimoniate: A case-control study of Th1 and Th2 pathways. Int Immunopharmacol (2019) 69:321–7. doi: 10.1016/j.intimp.2019.02.008 PubMed DOI
Babaloo Z, Kaye PM, Eslami MB. Interleukin-13 in Iranian patients with visceral leishmaniasis: relationship to other Th2 and Th1 cytokines. Trans R Soc Trop Med Hygiene (2001) 95:85–8. doi: 10.1016/S0035-9203(01)90344-X PubMed DOI
Kemp M, Kurtzhals JA, Bendtzen K, Poulsen LK, Hansen MB, Koech DK, et al. . Leishmania donovani-reactive Th1- and Th2-like T-cell clones from individuals who have recovered from visceral leishmaniasis. Infect Immun (1993) 61:1069–73. doi: 10.1128/iai.61.3.1069-1073.1993 PubMed DOI PMC
Herath S, Kropf P, Müller I. Cross-talk between CD8(+) and CD4(+) T cells in experimental cutaneous leishmaniasis: CD8(+) T cells are required for optimal IFN-gamma production by CD4(+) T cells. Parasite Immunol (2003) 25:559–67. doi: 10.1111/j.0141-9838.2004.00668.x PubMed DOI
Müller I, Kropf P, Etges RJ, Louis JA. Gamma interferon response in secondary Leishmania major infection: role of CD8+ T cells. Infect Immun (1993) 61:3730–8. doi: 10.1128/iai.61.9.3730-3738.1993 PubMed DOI PMC
Uzonna JE, Joyce KL, Scott P. Low dose Leishmania major promotes a transient T helper cell type 2 response that is down-regulated by interferon gamma-producing CD8+ T cells. J Exp Med (2004) 199:1559–66. doi: 10.1084/jem.20040172 PubMed DOI PMC
Nateghi Rostami M, Keshavarz H, Edalat R, Sarrafnejad A, Shahrestani T, Mahboudi F, et al. . CD8+ T cells as a source of IFN-γ production in human cutaneous leishmaniasis. PloS Negl Trop Dis (2010) 4:e845. doi: 10.1371/journal.pntd.0000845 PubMed DOI PMC
Khamesipour A, Nateghi Rostami M, Tasbihi M, Miramin Mohammadi A, Shahrestani T, Sarrafnejad A, et al. . Phenotyping of circulating CD8+ T cell subsets in human cutaneous leishmaniasis. Microbes Infect (2012) 14:702–11. doi: 10.1016/j.micinf.2012.02.006 PubMed DOI
Faria DR, Souza PE, Durães FV, Carvalho EM, Gollob KJ, MaChado PR, et al. . Recruitment of CD8(+) T cells expressing granzyme A is associated with lesion progression in human cutaneous leishmaniasis. Parasite Immunol (2009) 31:432–9. doi: 10.1111/j.1365-3024.2009.01125.x PubMed DOI PMC
Belkaid Y. The role of CD4(+)CD25(+) regulatory T cells in Leishmania infection. Expert Opin Biol Ther (2003) 3:875–85. doi: 10.1517/eobt.3.6.875.21257 PubMed DOI
Nylén S, Sacks D. Interleukin-10 and the pathogenesis of human visceral leishmaniasis. Trends Immunol (2007) 28:378–84. doi: 10.1016/j.it.2007.07.004 PubMed DOI
Belkaid Y, Hoffmann KF, Mendez S, Kamhawi S, Udey MC, Wynn TA, et al. . The role of interleukin (IL)-10 in the persistence of Leishmania major in the skin after healing and the therapeutic potential of anti-IL-10 receptor antibody for sterile cure. J Exp Med (2001) 194:1497–506. doi: 10.1084/jem.194.10.1497 PubMed DOI PMC
Belkaid Y, Piccirillo CA, Mendez S, Shevach EM, Sacks DL. CD4+CD25+ regulatory T cells control Leishmania major persistence and immunity. Nature (2002) 420:502–7. doi: 10.1038/nature01152 PubMed DOI
Yurchenko E, Tritt M, Hay V, Shevach EM, Belkaid Y, Piccirillo CA. CCR5-dependent homing of naturally occurring CD4+ regulatory T cells to sites of Leishmania major infection favors pathogen persistence. J Exp Med (2006) 203:2451–60. doi: 10.1084/jem.20060956 PubMed DOI PMC
Rodrigues OR, Marques C, Soares-Clemente M, Ferronha MH, Santos-Gomes GM. Identification of regulatory T cells during experimental Leishmania infantum infection. Immunobiology (2009) 214:101–11. doi: 10.1016/j.imbio.2008.07.001 PubMed DOI
Eufrásio de Figueiredo WM, Heredia FF, Santos AS, da Rocha Braga R, Marciano Fonseca FR, Lúcia de Castro Rodrigues N, et al. . CXCL10 treatment promotes reduction of IL-10(+) regulatory T (Foxp3(+) and Tr1) cells in the spleen of BALB/c mice infected by Leishmania infantum. Exp Parasitol (2019) 207:107789. doi: 10.1016/j.exppara.2019.107789 PubMed DOI
Barros N, Vasquez N, Woll F, Sanchez C, Valencia B, Llanos-Cuentas A, et al. . Regulatory T-Cell Dynamics in Cutaneous and Mucocutaneous Leishmaniasis due to Leishmania Braziliensis. Am J Trop Med hygiene (2018) 98:753–8. doi: 10.4269/ajtmh.17-0624 PubMed DOI PMC
Oliveira WN, Ribeiro LE, Schrieffer A, MaChado P, Carvalho EM, Bacellar O. The role of inflammatory and anti-inflammatory cytokines in the pathogenesis of human tegumentary leishmaniasis. Cytokine (2014) 66:127–32. doi: 10.1016/j.cyto.2013.12.016 PubMed DOI PMC
Faria DR, Gollob KJ, Barbosa J, Jr., Schriefer A, MaChado PR, Lessa H, et al. . Decreased in situ expression of interleukin-10 receptor is correlated with the exacerbated inflammatory and cytotoxic responses observed in mucosal leishmaniasis. Infect Immun (2005) 73:7853–9. doi: 10.1128/IAI.73.12.7853-7859.2005 PubMed DOI PMC
Campanelli AP, Roselino AM, Cavassani KA, Pereira MS, Mortara RA, Brodskyn CI, et al. . CD4+CD25+ T cells in skin lesions of patients with cutaneous leishmaniasis exhibit phenotypic and functional characteristics of natural regulatory T cells. J Infect Dis (2006) 193:1313–22. doi: 10.1086/502980 PubMed DOI
Rodriguez-Pinto D, Navas A, Blanco VM, Ramírez L, Garcerant D, Cruz A, et al. . Regulatory T cells in the pathogenesis and healing of chronic human dermal leishmaniasis caused by Leishmania (Viannia) species. PloS Negl Trop Dis (2012) 6:e1627. doi: 10.1371/journal.pntd.0001627 PubMed DOI PMC
Bahrami F, Darabi H, Riazi-Rad F, Khaze V, Ajdary S, Alimohammadian MH. FOXP3 expression and frequency of regulatory T cells in healed individuals from Leishmania major infection and the asymptomatic cases. Hum Immunol (2014) 75:1026–33. doi: 10.1016/j.humimm.2014.08.204 PubMed DOI
Rai AK, Thakur CP, Singh A, Seth T, Srivastava SK, Singh P, et al. . Regulatory T cells suppress T cell activation at the pathologic site of human visceral leishmaniasis. PloS One (2012) 7:e31551. doi: 10.1371/journal.pone.0031551 PubMed DOI PMC
Nylén S, Maurya R, Eidsmo L, Manandhar KD, Sundar S, Sacks D. Splenic accumulation of IL-10 mRNA in T cells distinct from CD4+CD25+ (Foxp3) regulatory T cells in human visceral leishmaniasis. J Exp Med (2007) 204:805–17. doi: 10.1084/jem.20061141 PubMed DOI PMC
Bhattacharya P, Ghosh S, Ejazi SA, Rahaman M, Pandey K, Ravi Das VN, et al. . Induction of IL-10 and TGFβ from CD4+CD25+FoxP3+ T cells correlates with parasite load in Indian kala-azar patients infected with leishmania donovani. PloS Negl Trop Dis (2016) 10:e0004422. doi: 10.1371/journal.pntd.0004422 PubMed DOI PMC
Bacellar O, Faria D, Nascimento M, Cardoso TM, Gollob KJ, Dutra WO, et al. . Interleukin 17 production among patients with American cutaneous leishmaniasis. J Infect Dis (2009) 200:75–8. doi: 10.1086/599380 PubMed DOI PMC
Boaventura VS, Santos CS, Cardoso CR, de Andrade J, Dos Santos WL, Clarêncio J, et al. . Human mucosal leishmaniasis: neutrophils infiltrate areas of tissue damage that express high levels of Th17-related cytokines. Eur J Immunol (2010) 40:2830–6. doi: 10.1002/eji.200940115 PubMed DOI
Glennie ND, Yeramilli VA, Beiting DP, Volk SW, Weaver CT, Scott P. Skin-resident memory CD4+ T cells enhance protection against Leishmania major infection. J Exp Med (2015) 212:1405–14. doi: 10.1084/jem.20142101 PubMed DOI PMC
Glennie ND, Volk SW, Scott P. Skin-resident CD4+ T cells protect against Leishmania major by recruiting and activating inflammatory monocytes. PloS Pathog (2017) 13:e1006349. doi: 10.1371/journal.ppat.1006349 PubMed DOI PMC
Zhang WW, Karmakar S, Gannavaram S, Dey R, Lypaczewski P, Ismail N, et al. . A second generation leishmanization vaccine with a markerless attenuated Leishmania major strain using CRISPR gene editing. Nat Commun (2020) 11:3461. doi: 10.1038/s41467-020-17154-z PubMed DOI PMC
Ismail N, Karmakar S, Bhattacharya P, Sepahpour T, Takeda K, Hamano S, et al. . Leishmania major centrin gene-deleted parasites generate skin resident memory T-cell immune response analogous to leishmanization. Front Immunol (2022) 13:864031. doi: 10.3389/fimmu.2022.864031 PubMed DOI PMC
Rodrigues A, Claro M, Alexandre-Pires G, Santos-Mateus D, Martins C, Valério-Bolas A, et al. . Leishmania infantum antigens modulate memory cell subsets of liver resident T lymphocyte. Immunobiology (2017) 222:409–22. doi: 10.1016/j.imbio.2016.08.009 PubMed DOI
Colpitts SL, Scott P. The early generation of a heterogeneous CD4+ T cell response to Leishmania major. J Immunol (Baltimore Md: 1950) (2010) 185:2416–23. doi: 10.4049/jimmunol.1000483 PubMed DOI PMC
Zaph C, Uzonna J, Beverley SM, Scott P. Central memory T cells mediate long-term immunity to Leishmania major in the absence of persistent parasites. Nat Med (2004) 10:1104–10. doi: 10.1038/nm1108 PubMed DOI
Ismail N, Kaul A, Bhattacharya P, Gannavaram S, Nakhasi HL. Immunization with live attenuated leishmania donovani centrin(-/-) parasites is efficacious in asymptomatic infection. Front Immunol (2017) 8:1788. doi: 10.3389/fimmu.2017.01788 PubMed DOI PMC
Fiuza JA, Gannavaram S, Gaze ST, de Ornellas LG, Alves ÉA, Ismail N, et al. . Deletion of MIF gene from live attenuated LdCen(-/-) parasites enhances protective CD4(+) T cell immunity. Sci Rep (2023) 13:7362. doi: 10.1038/s41598-023-34333-2 PubMed DOI PMC
Sallusto F, Lenig D, Förster R, Lipp M, Lanzavecchia A. Two subsets of memory T lymphocytes with distinct homing potentials and effector functions. Nature (1999) 401:708–12. doi: 10.1038/44385 PubMed DOI
Keshavarz Valian H, Nateghi Rostami M, Tasbihi M, Miramin Mohammadi A, Eskandari SE, Sarrafnejad A, et al. . CCR7+ central and CCR7- effector memory CD4+ T cells in human cutaneous leishmaniasis. J Clin Immunol (2013) 33:220–34. doi: 10.1007/s10875-012-9788-7 PubMed DOI
de Oliveira BC, da Silva AA, de Andrade Cavalcante MK, de Brito MEF, de Castro M, de Medeiros VLS, et al. . Central and Effector Memory Human CD4+ and CD8+ T Cells during Cutaneous Leishmaniasis and after In Vitro Stimulation with Leishmania (Viannia) Braziliensis Epitopes. Vaccines (2023) 11(1):158. doi: 10.3390/vaccines11010158 PubMed DOI PMC
de Oliveira Mendes-Aguiar C, Vieira-Gonçalves R, Guimarães LH, de Oliveira-Neto MP, Carvalho EM, Da-Cruz AM. Effector memory CD4(+) T cells differentially express activation associated molecules depending on the duration of American cutaneous leishmaniasis lesions. Clin Exp Immunol (2016) 185:202–9. doi: 10.1111/cei.12798 PubMed DOI PMC
Rodrigues-Neto JF, Monteiro GR, Keesen TSL, Lacerda HG, Carvalho EM, Jeronimo SMB. CD45RO+ T Cells and T Cell Activation in the Long-Lasting Immunity after Leishmania infantum Infection. Am J Trop Med hygiene (2018) 98:875–82. doi: 10.4269/ajtmh.16-0747 PubMed DOI PMC
Rodrigues LS, Barreto AS, Bomfim LGS, Gomes MC, Ferreira NLC, da Cruz GS, et al. . Multifunctional, TNF-α and IFN-γ-secreting CD4 and CD8 T cells and CD8(High) T cells are associated with the cure of human visceral leishmaniasis. Front Immunol (2021) 12:773983. doi: 10.3389/fimmu.2021.773983 PubMed DOI PMC
Olivier M, Gregory DJ, Forget G. Subversion mechanisms by which Leishmania parasites can escape the host immune response: a signaling point of view. Clin Microbiol Rev (2005) 18:293–305. doi: 10.1128/CMR.18.2.293-305.2005 PubMed DOI PMC
Mukbel RM, Patten C, Jr., Gibson K, Ghosh M, Petersen C, Jones DE. Macrophage killing of Leishmania amazonensis amastigotes requires both nitric oxide and superoxide. Am J Trop Med hygiene (2007) 76:669–75. doi: 10.4269/ajtmh.2007.76.669 PubMed DOI
Sadeghi S, Seyed N, Rafati S, Taheri T. Optimization of the timing of induction for the assessment of nitric oxide production in leishmania major infected macrophage cells. Iranian J Parasitol (2016) 11:325–31. PubMed PMC
Van Assche T, Deschacht M, da Luz RA, Maes L, Cos P. Leishmania-macrophage interactions: insights into the redox biology. Free Radical Biol Med (2011) 51:337–51. doi: 10.1016/j.freeradbiomed.2011.05.011 PubMed DOI
Brandonisio O, Panaro MA, Fumarola I, Sisto M, Leogrande D, Acquafredda A, et al. . Macrophage chemotactic protein-1 and macrophage inflammatory protein-1 alpha induce nitric oxide release and enhance parasite killing in Leishmania infantum-infected human macrophages. Clin Exp Med (2002) 2:125–9. doi: 10.1007/s102380200017 PubMed DOI
Gomez MA, Contreras I, Hallé M, Tremblay ML, McMaster RW, Olivier M. Leishmania GP63 alters host signaling through cleavage-activated protein tyrosine phosphatases. Sci Signal (2009) 2:ra58. doi: 10.1126/scisignal.2000213 PubMed DOI
Isnard A, Shio MT, Olivier M. Impact of Leishmania metalloprotease GP63 on macrophage signaling. Front Cell Infect Microbiol (2012) 2:72. doi: 10.3389/fcimb.2012.00072 PubMed DOI PMC
Hermoso T, Fishelson Z, Becker SI, Hirschberg K, Jaffe CL. Leishmanial protein kinases phosphorylate components of the complement system. EMBO J (1991) 10:4061–7. doi: 10.1002/embj.1991.10.issue-13 PubMed DOI PMC
Sen S, Roy K, Mukherjee S, Mukhopadhyay R, Roy S. Restoration of IFNγR subunit assembly, IFNγ signaling and parasite clearance in Leishmania donovani infected macrophages: role of membrane cholesterol. PloS Pathog (2011) 7:e1002229. doi: 10.1371/journal.ppat.1002229 PubMed DOI PMC
Bhattacharyya S, Ghosh S, Jhonson PL, Bhattacharya SK, Majumdar S. Immunomodulatory role of interleukin-10 in visceral leishmaniasis: defective activation of protein kinase C-mediated signal transduction events. Infect Immun (2001) 69:1499–507. doi: 10.1128/IAI.69.3.1499-1507.2001 PubMed DOI PMC
Lodge R, Descoteaux A. Phagocytosis of Leishmania donovani amastigotes is Rac1 dependent and occurs in the absence of NADPH oxidase activation. Eur J Immunol (2006) 36:2735–44. doi: 10.1002/eji.200636089 PubMed DOI
Blanchette J, Racette N, Faure R, Siminovitch KA, Olivier M. Leishmania-induced increases in activation of macrophage SHP-1 tyrosine phosphatase are associated with impaired IFN-gamma-triggered JAK2 activation. Eur J Immunol (1999) 29:3737–44. doi: 10.1002/(ISSN)1521-4141 PubMed DOI
Fernández-Figueroa EA, Imaz-Rosshandler I, Castillo-Fernández JE, Miranda-Ortíz H, Fernández-López JC, Becker I, et al. . Down-regulation of TLR and JAK/STAT pathway genes is associated with diffuse cutaneous leishmaniasis: A gene expression analysis in NK cells from patients infected with leishmania mexicana. PloS Negl Trop Dis (2016) 10:e0004570. doi: 10.1371/journal.pntd.0004570 PubMed DOI PMC
Yilmaz IC, Dunuroglu E, Ayanoglu IC, Ipekoglu EM, Yildirim M, Girginkardesler N, et al. . Leishmania kinetoplast DNA contributes to parasite burden in infected macrophages: Critical role of the cGAS-STING-TBK1 signaling pathway in macrophage parasitemia. Front Immunol (2022) 13:1007070. doi: 10.3389/fimmu.2022.1007070 PubMed DOI PMC
Martínez-Valencia AJ, Daza-Rivera CF, Rosales-Chilama M, Cossio A, Casadiego Rincón EJ, Desai MM, et al. . Clinical and parasitological factors in parasite persistence after treatment and clinical cure of cutaneous leishmaniasis. PloS Negl Trop Dis (2017) 11:e0005713. doi: 10.1371/journal.pntd.0005713 PubMed DOI PMC
Romero I, Téllez J, Suárez Y, Cardona M, Figueroa R, Zelazny A, et al. . Viability and burden of Leishmania in extralesional sites during human dermal leishmaniasis. PloS Negl Trop Dis (2010) 4(9):e819. doi: 10.1371/journal.pntd.0000819 PubMed DOI PMC
Moll H, Flohé S, Röllinghoff M. Dendritic cells in Leishmania major-immune mice harbor persistent parasites and mediate an antigen-specific T cell immune response. Eur J Immunol (1995) 25:693–9. doi: 10.1002/eji.1830250310 PubMed DOI
Blank C, Fuchs H, Rappersberger K, Röllinghoff M, Moll H. Parasitism of epidermal Langerhans cells in experimental cutaneous leishmaniasis with Leishmania major. J Infect Dis (1993) 167:418–25. doi: 10.1093/infdis/167.2.418 PubMed DOI
Bogdan C, Donhauser N, Döring R, Röllinghoff M, Diefenbach A, Rittig MG. Fibroblasts as host cells in latent leishmaniosis. J Exp Med (2000) 191:2121–30. doi: 10.1084/jem.191.12.2121 PubMed DOI PMC
McConville MJ, Naderer T. Metabolic pathways required for the intracellular survival of Leishmania. Annu Rev Microbiol (2011) 65:543–61. doi: 10.1146/annurev-micro-090110-102913 PubMed DOI
Mendez S, Reckling SK, Piccirillo CA, Sacks D, Belkaid Y. Role for CD4(+) CD25(+) regulatory T cells in reactivation of persistent leishmaniasis and control of concomitant immunity. J Exp Med (2004) 200:201–10. doi: 10.1084/jem.20040298 PubMed DOI PMC
Anderson CF, Oukka M, Kuchroo VJ, Sacks D. CD4(+)CD25(-)Foxp3(-) Th1 cells are the source of IL-10-mediated immune suppression in chronic cutaneous leishmaniasis. J Exp Med (2007) 204:285–97. doi: 10.1084/jem.20061886 PubMed DOI PMC
Gonzalez K, Calzada JE, Tomokane TY, Pacheco CMS, Flores GVA, Castro Gomes CM, et al. . In situ study of cellular immune response in human cutaneous lesions caused by Leishmania (Viannia) panamensis in Panama. Parasite Immunol (2021) 43:e12801. doi: 10.1111/pim.12801 PubMed DOI
Jafarzadeh A, Jafarzadeh S, Sharifi I, Aminizadeh N, Nozari P, Nemati M. The importance of T cell-derived cytokines in post-kala-azar dermal leishmaniasis. Cytokine (2021) 147:155321. doi: 10.1016/j.cyto.2020.155321 PubMed DOI
de Freitas EO, Leoratti FM, Freire-de-Lima CG, Morrot A, Feijó DF. The contribution of immune evasive mechanisms to parasite persistence in visceral leishmaniasis. Front Immunol (2016) 7:153. doi: 10.3389/fimmu.2016.00153 PubMed DOI PMC
Arumugam S, Scorza BM, Petersen C. Visceral leishmaniasis and the skin: dermal parasite transmission to sand flies. Pathog (Basel Switzerland) (2022) 11(6):610. doi: 10.3390/pathogens11060610 PubMed DOI PMC
Aslan H, Oliveira F, Meneses C, Castrovinci P, Gomes R, Teixeira C, et al. . New Insights Into the Transmissibility of Leishmania infantum From Dogs to Sand Flies: Experimental Vector-Transmission Reveals Persistent Parasite Depots at Bite Sites. J Infect Dis (2016) 213:1752–61. doi: 10.1093/infdis/jiw022 PubMed DOI PMC
Peixoto RF, Gois BM, Martins M, Palmeira PHS, Rocha JC, Gomes JAS, et al. . Characterization of regulatory T cells in patients infected by leishmania infantum. Trop Med Infect Dis (2022) 8(1):18. doi: 10.3390/tropicalmed8010018 PubMed DOI PMC
Kumar P, Misra P, Thakur CP, Saurabh A, Rishi N, Mitra DK. T cell suppression in the bone marrow of visceral leishmaniasis patients: impact of parasite load. Clin Exp Immunol (2018) 191:318–27. doi: 10.1111/cei.13074 PubMed DOI PMC
Gasim S, Elhassan AM, Khalil EA, Ismail A, Kadaru AM, Kharazmi A, et al. . High levels of plasma IL-10 and expression of IL-10 by keratinocytes during visceral leishmaniasis predict subsequent development of post-kala-azar dermal leishmaniasis. Clin Exp Immunol (1998) 111:64–9. doi: 10.1046/j.1365-2249.1998.00468.x PubMed DOI PMC
Santos MF, Alexandre-Pires G, Pereira MA, Gomes L, Rodrigues AV, Basso A, et al. . Immunophenotyping of peripheral blood, lymph node, and bone marrow T lymphocytes during canine leishmaniosis and the impact of antileishmanial chemotherapy. Front Vet Sci (2020) 7:375. doi: 10.3389/fvets.2020.00375 PubMed DOI PMC
Mandell MA, Beverley SM. Continual renewal and replication of persistent Leishmania major parasites in concomitantly immune hosts. Proc Natl Acad Sci USA (2017) 114:E801–e10. doi: 10.1073/pnas.1619265114 PubMed DOI PMC
Peters NC, Pagán AJ, Lawyer PG, Hand TW, Henrique Roma E, Stamper LW, et al. . Chronic parasitic infection maintains high frequencies of short-lived Ly6C+CD4+ effector T cells that are required for protection against re-infection. PloS Pathog (2014) 10:e1004538. doi: 10.1371/journal.ppat.1004538 PubMed DOI PMC
Uzonna JE, Wei G, Yurkowski D, Bretscher P. Immune elimination of Leishmania major in mice: implications for immune memory, vaccination, and reactivation disease. J Immunol (Baltimore Md: 1950) (2001) 167:6967–74. doi: 10.4049/jimmunol.167.12.6967 PubMed DOI
Scott P. Long-lived skin-resident memory T cells contribute to concomitant immunity in cutaneous leishmaniasis. Cold Spring Harbor Perspect Biol (2020) 12(10):a038059. doi: 10.1101/cshperspect.a038059 PubMed DOI PMC
Viana da Costa A, Huerre M, Delacre M, Auriault C, Correia Costa JM, Verwaerde C. IL-10 leads to a higher parasite persistence in a resistant mouse model of Leishmania major infection. Parasitol Int (2002) 51:367–79. doi: 10.1016/S1383-5769(02)00039-9 PubMed DOI
Okwor I, Uzonna J. Persistent parasites and immunologic memory in cutaneous leishmaniasis: implications for vaccine designs and vaccination strategies. Immunologic Res (2008) 41:123–36. doi: 10.1007/s12026-008-8016-2 PubMed DOI
Hohman LS, Mou Z, Carneiro MB, Ferland G, Kratofil RM, Kubes P, et al. . Protective CD4+ Th1 cell-mediated immunity is reliant upon execution of effector function prior to the establishment of the pathogen niche. PloS Pathog (2021) 17:e1009944. doi: 10.1371/journal.ppat.1009944 PubMed DOI PMC
Costa-Madeira JC, Trindade GB, Almeida PHP, Silva JS, Carregaro VT. Lymphocyte exhaustion during human and experimental visceral leishmaniasis. Front Immunol (2022) 13:835711. doi: 10.3389/fimmu.2022.835711 PubMed DOI PMC
Diupotex M, Zamora-Chimal J, Gajón JA, Bonifaz LC, Becker I. CXCR5 and TIM-3 expressions define distinct exhausted T cell subsets in experimental cutaneous infection with Leishmania mexicana. Front Immunol (2023) 14:1231836. doi: 10.3389/fimmu.2023.1231836 PubMed DOI PMC
Berard M, Tough DF. Qualitative differences between naïve and memory T cells. Immunology (2002) 106:127–38. doi: 10.1046/j.1365-2567.2002.01447.x PubMed DOI PMC
Stockinger B, Kassiotis G, Bourgeois C. CD4 T-cell memory. Semin Immunol (2004) 16:295–303. doi: 10.1016/j.smim.2004.08.010 PubMed DOI
Sallusto F, Geginat J, Lanzavecchia A. Central memory and effector memory T cell subsets: function, generation, and maintenance. Annu Rev Immunol (2004) 22:745–63. doi: 10.1146/annurev.immunol.22.012703.104702 PubMed DOI
Wu CY, Kirman JR, Rotte MJ, Davey DF, Perfetto SP, Rhee EG, et al. . Distinct lineages of T(H)1 cells have differential capacities for memory cell generation in vivo. Nat Immunol (2002) 3:852–8. doi: 10.1038/ni832 PubMed DOI
Kaech SM, Ahmed R. Memory CD8+ T cell differentiation: initial antigen encounter triggers a developmental program in naïve cells. Nat Immunol (2001) 2:415–22. doi: 10.1038/87720 PubMed DOI PMC
Reinhardt RL, Khoruts A, Merica R, Zell T, Jenkins MK. Visualizing the generation of memory CD4 T cells in the whole body. Nature (2001) 410:101–5. doi: 10.1038/35065111 PubMed DOI
Wherry EJ, Teichgräber V, Becker TC, Masopust D, Kaech SM, Antia R, et al. . Lineage relationship and protective immunity of memory CD8 T cell subsets. Nat Immunol (2003) 4:225–34. doi: 10.1038/ni889 PubMed DOI
Okada R, Kondo T, Matsuki F, Takata H, Takiguchi M. Phenotypic classification of human CD4+ T cell subsets and their differentiation. Int Immunol (2008) 20:1189–99. doi: 10.1093/intimm/dxn075 PubMed DOI
Wherry EJ, Barber DL, Kaech SM, Blattman JN, Ahmed R. Antigen-independent memory CD8 T cells do not develop during chronic viral infection. Proc Natl Acad Sci USA (2004) 101:16004–9. doi: 10.1073/pnas.0407192101 PubMed DOI PMC
Seder RA, Ahmed R. Similarities and differences in CD4+ and CD8+ effector and memory T cell generation. Nat Immunol (2003) 4:835–42. doi: 10.1038/ni969 PubMed DOI
Harari A, Vallelian F, Meylan PR, Pantaleo G. Functional heterogeneity of memory CD4 T cell responses in different conditions of antigen exposure and persistence. J Immunol (Baltimore Md: 1950) (2005) 174:1037–45. doi: 10.4049/jimmunol.174.2.1037 PubMed DOI
Imhof BA, Dunon D. Leukocyte migration and adhesion. Adv Immunol (1995) 58:345–416. doi: 10.1016/S0065-2776(08)60623-9 PubMed DOI
Künzli M, Masopust D. CD4(+) T cell memory. Nat Immunol (2023) 24:903–14. doi: 10.1038/s41590-023-01510-4 PubMed DOI PMC
Todryk SM. T cell memory to vaccination. Vaccines (2018) 6(4):184. doi: 10.3390/vaccines6040084 PubMed DOI PMC
Boesteanu AC, Katsikis PD. Memory T cells need CD28 costimulation to remember. Semin Immunol (2009) 21:69–77. doi: 10.1016/j.smim.2009.02.005 PubMed DOI PMC
Schiött A, Lindstedt M, Johansson-Lindbom B, Roggen E, Borrebaeck CA. CD27- CD4+ memory T cells define a differentiated memory population at both the functional and transcriptional levels. Immunology (2004) 113:363–70. doi: 10.1111/j.1365-2567.2004.01974.x PubMed DOI PMC
Scott P, Artis D, Uzonna J, Zaph C. The development of effector and memory T cells in cutaneous leishmaniasis: the implications for vaccine development. Immunol Rev (2004) 201:318–38. doi: 10.1111/j.0105-2896.2004.00198.x PubMed DOI
Titus RG, Gueiros-Filho FJ, de Freitas LA, Beverley SM. Development of a safe live Leishmania vaccine line by gene replacement. Proc Natl Acad Sci USA (1995) 92:10267–71. doi: 10.1073/pnas.92.22.10267 PubMed DOI PMC
Scott P. Immunologic memory in cutaneous leishmaniasis. Cell Microbiol (2005) 7:1707–13. doi: 10.1111/j.1462-5822.2005.00626.x PubMed DOI
Rivino L, Messi M, Jarrossay D, Lanzavecchia A, Sallusto F, Geginat J. Chemokine receptor expression identifies Pre-T helper (Th)1, Pre-Th2, and nonpolarized cells among human CD4+ central memory T cells. J Exp Med (2004) 200:725–35. doi: 10.1084/jem.20040774 PubMed DOI PMC
Uzonna JE, Späth GF, Beverley SM, Scott P. Vaccination with phosphoglycan-deficient Leishmania major protects highly susceptible mice from virulent challenge without inducing a strong Th1 response. J Immunol (Baltimore Md: 1950) (2004) 172:3793–7. doi: 10.4049/jimmunol.172.6.3793 PubMed DOI
Kaech SM, Wherry EJ, Ahmed R. Effector and memory T-cell differentiation: implications for vaccine development. Nat Rev Immunol (2002) 2:251–62. doi: 10.1038/nri778 PubMed DOI
Pereira-Carvalho R, Mendes-Aguiar CO, Oliveira-Neto MP, Covas CJ, Bertho AL, Da-Cruz AM, et al. . Leishmania Braziliensis-reactive T cells are down-regulated in long-term cured cutaneous Leishmaniasis, but the renewal capacity of T effector memory compartments is preserved. PloS One (2013) 8:e81529. doi: 10.1371/journal.pone.0081529 PubMed DOI PMC
Willemsen M, Linkutė R, Luiten RM, Matos TR. Skin-resident memory T cells as a potential new therapeutic target in vitiligo and melanoma. Pigment Cell melanoma Res (2019) 32:612–22. doi: 10.1111/pcmr.12803 PubMed DOI PMC
Wu H, Liao W, Li Q, Long H, Yin H, Zhao M, et al. . Pathogenic role of tissue-resident memory T cells in autoimmune diseases. Autoimmun Rev (2018) 17:906–11. doi: 10.1016/j.autrev.2018.03.014 PubMed DOI
Chen L, Shen Z. Tissue-resident memory T cells and their biological characteristics in the recurrence of inflammatory skin disorders. Cell Mol Immunol (2020) 17:64–75. doi: 10.1038/s41423-019-0291-4 PubMed DOI PMC
Clark RA. Resident memory T cells in human health and disease. Sci Trans Med (2015) 7:269rv1. doi: 10.1126/scitranslmed.3010641 PubMed DOI PMC
Jameson SC, Masopust D. Understanding subset diversity in T cell memory. Immunity (2018) 48:214–26. doi: 10.1016/j.immuni.2018.02.010 PubMed DOI PMC
Szabo PA, Miron M, Farber DL. Location, location, location: Tissue resident memory T cells in mice and humans. Sci Immunol (2019) 4(34):eaas9673. doi: 10.1126/sciimmunol.aas9673 PubMed DOI PMC
Mackay LK, Minnich M, Kragten NA, Liao Y, Nota B, Seillet C, et al. . Hobit and Blimp1 instruct a universal transcriptional program of tissue residency in lymphocytes. Sci (New York NY) (2016) 352:459–63. doi: 10.1126/science.aad2035 PubMed DOI
Kumar BV, Ma W, Miron M, Granot T, Guyer RS, Carpenter DJ, et al. . Human tissue-resident memory T cells are defined by core transcriptional and functional signatures in lymphoid and mucosal sites. Cell Rep (2017) 20:2921–34. doi: 10.1016/j.celrep.2017.08.078 PubMed DOI PMC
Milner JJ, Toma C, Yu B, Zhang K, Omilusik K, Phan AT, et al. . Runx3 programs CD8(+) T cell residency in non-lymphoid tissues and tumours. Nature (2017) 552:253–7. doi: 10.1038/nature24993 PubMed DOI PMC
Jiang X, Clark RA, Liu L, Wagers AJ, Fuhlbrigge RC, Kupper TS. Skin infection generates non-migratory memory CD8+ T(RM) cells providing global skin immunity. Nature (2012) 483:227–31. doi: 10.1038/nature10851 PubMed DOI PMC
Carlson CM, Endrizzi BT, Wu J, Ding X, Weinreich MA, Walsh ER, et al. . Kruppel-like factor 2 regulates thymocyte and T-cell migration. Nature (2006) 442:299–302. doi: 10.1038/nature04882 PubMed DOI
Shiow LR, Rosen DB, Brdicková N, Xu Y, An J, Lanier LL, et al. . CD69 acts downstream of interferon-alpha/beta to inhibit S1P1 and lymphocyte egress from lymphoid organs. Nature (2006) 440:540–4. doi: 10.1038/nature04606 PubMed DOI
Mackay LK, Braun A, Macleod BL, Collins N, Tebartz C, Bedoui S, et al. . Cutting edge: CD69 interference with sphingosine-1-phosphate receptor function regulates peripheral T cell retention. J Immunol (Baltimore Md: 1950) (2015) 194:2059–63. doi: 10.4049/jimmunol.1402256 PubMed DOI
Sathaliyawala T, Kubota M, Yudanin N, Turner D, Camp P, Thome JJ, et al. . Distribution and compartmentalization of human circulating and tissue-resident memory T cell subsets. Immunity (2013) 38:187–97. doi: 10.1016/j.immuni.2012.09.020 PubMed DOI PMC
Gebhardt T, Whitney PG, Zaid A, Mackay LK, Brooks AG, Heath WR, et al. . Different patterns of peripheral migration by memory CD4+ and CD8+ T cells. Nature (2011) 477:216–9. doi: 10.1038/nature10339 PubMed DOI
Mackay LK, Rahimpour A, Ma JZ, Collins N, Stock AT, Hafon ML, et al. . The developmental pathway for CD103(+)CD8+ tissue-resident memory T cells of skin. Nat Immunol (2013) 14:1294–301. doi: 10.1038/ni.2744 PubMed DOI
Thome JJ, Yudanin N, Ohmura Y, Kubota M, Grinshpun B, Sathaliyawala T, et al. . Spatial map of human T cell compartmentalization and maintenance over decades of life. Cell (2014) 159:814–28. doi: 10.1016/j.cell.2014.10.026 PubMed DOI PMC
Watanabe R, Gehad A, Yang C, Scott LL, Teague JE, Schlapbach C, et al. . Human skin is protected by four functionally and phenotypically discrete populations of resident and recirculating memory T cells. Sci Trans Med (2015) 7:279ra39. doi: 10.1126/scitranslmed.3010302 PubMed DOI PMC
Takamura S, Yagi H, Hakata Y, Motozono C, McMaster SR, Masumoto T, et al. . Specific niches for lung-resident memory CD8+ T cells at the site of tissue regeneration enable CD69-independent maintenance. J Exp Med (2016) 213:3057–73. doi: 10.1084/jem.20160938 PubMed DOI PMC
Iijima N, Iwasaki A. T cell memory. A local macrophage chemokine network sustains protective tissue-resident memory CD4 T cells. Sci (New York NY) (2014) 346:93–8. doi: 10.1126/science.1257530 PubMed DOI PMC
Strutt TM, Dhume K, Finn CM, Hwang JH, Castonguay C, Swain SL, et al. . IL-15 supports the generation of protective lung-resident memory CD4 T cells. Mucosal Immunol (2018) 11:668–80. doi: 10.1038/mi.2017.101 PubMed DOI PMC
Collins N, Jiang X, Zaid A, Macleod BL, Li J, Park CO, et al. . Skin CD4(+) memory T cells exhibit combined cluster-mediated retention and equilibration with the circulation. Nat Commun (2016) 7:11514. doi: 10.1038/ncomms11514 PubMed DOI PMC
McCully ML, Ladell K, Andrews R, Jones RE, Miners KL, Roger L, et al. . CCR8 expression defines tissue-resident memory T cells in human skin. J Immunol (Baltimore Md: 1950) (2018) 200:1639–50. doi: 10.4049/jimmunol.1701377 PubMed DOI PMC
Clark RA, Chong B, Mirchandani N, Brinster NK, Yamanaka K, Dowgiert RK, et al. . The vast majority of CLA+ T cells are resident in normal skin. J Immunol (Baltimore Md: 1950) (2006) 176:4431–9. doi: 10.4049/jimmunol.176.7.4431 PubMed DOI
Nolz JC, Richer MJ. Control of memory CD8(+) T cell longevity and effector functions by IL-15. Mol Immunol (2020) 117:180–8. doi: 10.1016/j.molimm.2019.11.011 PubMed DOI PMC
Shin MS, Kim D, Yim K, Park HJ, You S, Dong X, et al. . IL-7 receptor alpha defines heterogeneity and signature of human effector memory CD8(+) T cells in high dimensional analysis. Cell Immunol (2020) 355:104155. doi: 10.1016/j.cellimm.2020.104155 PubMed DOI PMC
Mackay LK, Wynne-Jones E, Freestone D, Pellicci DG, Mielke LA, Newman DM, et al. . T-box transcription factors combine with the cytokines TGF-β and IL-15 to control tissue-resident memory T cell fate. Immunity (2015) 43:1101–11. doi: 10.1016/j.immuni.2015.11.008 PubMed DOI
Adachi T, Kobayashi T, Sugihara E, Yamada T, Ikuta K, Pittaluga S, et al. . Hair follicle-derived IL-7 and IL-15 mediate skin-resident memory T cell homeostasis and lymphoma. Nat Med (2015) 21:1272–9. doi: 10.1038/nm.3962 PubMed DOI PMC
Goncalves R, Zhang X, Cohen H, Debrabant A, Mosser DM. Platelet activation attracts a subpopulation of effector monocytes to sites of Leishmania major infection. J Exp Med (2011) 208:1253–65. doi: 10.1084/jem.20101751 PubMed DOI PMC
Romano A, Carneiro MBH, Doria NA, Roma EH, Ribeiro-Gomes FL, Inbar E, et al. . Divergent roles for Ly6C+CCR2+CX3CR1+ inflammatory monocytes during primary or secondary infection of the skin with the intra-phagosomal pathogen Leishmania major. PloS Pathog (2017) 13:e1006479. doi: 10.1371/journal.ppat.1006479 PubMed DOI PMC
Miramin-Mohammadi A, Javadi A, Eskandari SE, Nateghi-Rostami M, Khamesipour A. Immune Responses in Cutaneous Leishmaniasis: In vitro Thelper1/Thelper2 Cytokine Profiles Using Live Versus Killed Leishmania major. J Arthropod-borne Dis (2021) 15:126–35. doi: 10.18502/jad.v15i1.6491 PubMed DOI PMC
Bahrami F, Harandi AM, Rafati S. Biomarkers of cutaneous leishmaniasis. Front Cell Infect Microbiol (2018) 8:222. doi: 10.3389/fcimb.2018.00222 PubMed DOI PMC
Reiner SL, Locksley RM. Cytokines in the differentiation of Th1/Th2 CD4+ subsets in leishmaniasis. J Cell Biochem (1993) 53:323–8. doi: 10.1002/jcb.240530409 PubMed DOI
Kellina OI. [Differences in the sensitivity of inbred mice of different lines to Leishmania tropica major]. Meditsinskaia parazitologiia i parazitarnye bolezni (1973) 42:279–85. PubMed
Mitchell GF, Curtis JM, Scollay RG, Handman E. Resistance and abrogation of resistance to cutaneous leishmaniasis in reconstituted BALB/c nude mice. Aust J Exp Biol Med Sci (1981) 59:539–54. doi: 10.1038/icb.1981.47 PubMed DOI
Heinzel FP, Sadick MD, Holaday BJ, Coffman RL, Locksley RM. Reciprocal expression of interferon gamma or interleukin 4 during the resolution or progression of murine leishmaniasis. Evidence for expansion of distinct helper T cell subsets. J Exp Med (1989) 169:59–72. doi: 10.1084/jem.169.1.59 PubMed DOI PMC
Mosmann TR, Cherwinski H, Bond MW, Giedlin MA, Coffman RL. Two types of murine helper T cell clone. I. Definition according to profiles of lymphokine activities and secreted proteins. J Immunol (Baltimore Md: 1950) (1986) 136:2348–57. doi: 10.4049/jimmunol.136.7.2348 PubMed DOI
Cher DJ, Mosmann TR. Two types of murine helper T cell clone. II. Delayed-type hypersensitivity is mediated by TH1 clones. J Immunol (Baltimore Md: 1950) (1987) 138:3688–94. doi: 10.4049/jimmunol.138.11.3688 PubMed DOI
Locksley RM, Heinzel FP, Holaday BJ, Mutha SS, Reiner SL, Sadick MD. Induction of Th1 and Th2 CD4+ subsets during murine Leishmania major infection. Res Immunol (1991) 142:28–32. doi: 10.1016/0923-2494(91)90007-6 PubMed DOI
Coffman RL, Chatelain R, Leal LM, Varkila K. Leishmania major infection in mice: a model system for the study of CD4+ T-cell subset differentiation. Res Immunol (1991) 142:36–40. doi: 10.1016/0923-2494(91)90009-8 PubMed DOI
Sadick MD, Locksley RM, Tubbs C, Raff HV. Murine cutaneous leishmaniasis: resistance correlates with the capacity to generate interferon-gamma in response to Leishmania antigens in vitro. J Immunol (Baltimore Md: 1950) (1986) 136:655–61. doi: 10.4049/jimmunol.136.2.655 PubMed DOI
Darzi F, Davoudian R, Nateghi Rostami M. Differential inflammatory responses associated with Leishmania major and L tropica in culture. Parasite Immunol (2021) 43:e12841. doi: 10.1111/pim.12841 PubMed DOI
Bodhale NP, Pal S, Kumar S, Chattopadhyay D, Saha B, Chattopadhyay N, et al. . Inbred mouse strains differentially susceptible to Leishmania donovani infection differ in their immune cell metabolism. Cytokine (2018) 112:12–5. doi: 10.1016/j.cyto.2018.06.003 PubMed DOI
Childs GE, Lightner LK, McKinney L, Groves MG, Price EE, Hendricks LD. Inbred mice as model hosts for cutaneous leishmaniasis. I. Resistance and susceptibility to infection with Leishmania Braziliensis, L. mexicana and L. aethiopica. Ann Trop Med Parasitol (1984) 78:25–34. doi: 10.1080/00034983.1984.11811769 PubMed DOI
Nabors GS, Farrell JP. Site-specific immunity to Leishmania major in SWR mice: the site of infection influences susceptibility and expression of the antileishmanial immune response. Infect Immun (1994) 62:3655–62. doi: 10.1128/iai.62.9.3655-3662.1994 PubMed DOI PMC
Baldwin TM, Elso C, Curtis J, Buckingham L, Handman E. The site of Leishmania major infection determines disease severity and immune responses. Infect Immun (2003) 71:6830–4. doi: 10.1128/IAI.71.12.6830-6834.2003 PubMed DOI PMC
Fromm PD, Kling JC, Remke A, Bogdan C, Körner H. Fatal leishmaniasis in the absence of TNF despite a strong th1 response. Front Microbiol (2015) 6:1520. doi: 10.3389/fmicb.2015.01520 PubMed DOI PMC
Sharma U, Singh S. Immunobiology of leishmaniasis. Indian J Exp Biol (2009) 47:412–23. PubMed
Bomfim G, Nascimento C, Costa J, Carvalho EM, Barral-Netto M, Barral A. Variation of cytokine patterns related to therapeutic response in diffuse cutaneous leishmaniasis. Exp Parasitol (1996) 84:188–94. doi: 10.1006/expr.1996.0104 PubMed DOI
Sundar S, Reed SG, Sharma S, Mehrotra A, Murray HW. Circulating T helper 1 (Th1) cell- and Th2 cell-associated cytokines in Indian patients with visceral leishmaniasis. Am J Trop Med hygiene (1997) 56:522–5. doi: 10.4269/ajtmh.1997.56.522 PubMed DOI
Castellano LR, Filho DC, Argiro L, Dessein H, Prata A, Dessein A, et al. . Th1/Th2 immune responses are associated with active cutaneous leishmaniasis and clinical cure is associated with strong interferon-gamma production. Hum Immunol (2009) 70:383–90. doi: 10.1016/j.humimm.2009.01.007 PubMed DOI
da Silva Santos C, Brodskyn CI. The role of CD4 and CD8 T cells in human cutaneous leishmaniasis. Front Public Health (2014) 2:165. doi: 10.3389/fpubh.2014.00165 PubMed DOI PMC
Darrah PA, Hegde ST, Patel DT, Lindsay RW, Chen L, Roederer M, et al. . IL-10 production differentially influences the magnitude, quality, and protective capacity of Th1 responses depending on the vaccine platform. J Exp Med (2010) 207:1421–33. doi: 10.1084/jem.20092532 PubMed DOI PMC
Gomes-Silva A, de Cássia Bittar R, Dos Santos Nogueira R, Amato VS, da Silva Mattos M, Oliveira-Neto MP, et al. . Can interferon-gamma and interleukin-10 balance be associated with severity of human Leishmania (Viannia) Braziliensis infection? Clin Exp Immunol (2007) 149:440–4. doi: 10.1111/j.1365-2249.2007.03436.x PubMed DOI PMC
Khalil EA, Ayed NB, Musa AM, Ibrahim ME, Mukhtar MM, Zijlstra EE, et al. . Dichotomy of protective cellular immune responses to human visceral leishmaniasis. Clin Exp Immunol (2005) 140:349–53. doi: 10.1111/j.1365-2249.2005.02768.x PubMed DOI PMC
Botana L, Matía B, San Martin JV, Romero-Maté A, Castro A, Molina L, et al. . Cellular markers of active disease and cure in different forms of leishmania infantum-induced disease. Front Cell Infect Microbiol (2018) 8:381. doi: 10.3389/fcimb.2018.00381 PubMed DOI PMC
Verma S, Kumar R, Katara GK, Singh LC, Negi NS, Ramesh V, et al. . Quantification of parasite load in clinical samples of leishmaniasis patients: IL-10 level correlates with parasite load in visceral leishmaniasis. PloS One (2010) 5:e10107. doi: 10.1371/journal.pone.0010107 PubMed DOI PMC
Mesquita I, Ferreira C, Barbosa AM, Ferreira CM, Moreira D, Carvalho A, et al. . The impact of IL-10 dynamic modulation on host immune response against visceral leishmaniasis. Cytokine (2018) 112:16–20. doi: 10.1016/j.cyto.2018.07.001 PubMed DOI
Alimohammadian MH, Jones SL, Darabi H, Riazirad F, Ajdary S, Shabani A, et al. . Assessment of interferon-γ levels and leishmanin skin test results in persons recovered for leishmaniasis. Am J Trop Med hygiene (2012) 87:70–5. doi: 10.4269/ajtmh.2012.11-0479 PubMed DOI PMC
Volpedo G, Bhattacharya P, Gannavaram S, Pacheco-Fernandez T, Oljuskin T, Dey R, et al. . The history of live attenuated centrin gene-deleted leishmania vaccine candidates. Pathog (Basel Switzerland) (2022) 11(4):431. doi: 10.3390/pathogens11040431 PubMed DOI PMC
Volpedo G, Pacheco-Fernandez T, Holcomb EA, Zhang WW, Lypaczewski P, Cox B, et al. . Centrin-deficient Leishmania mexicana confers protection against New World cutaneous leishmaniasis. NPJ Vaccines (2022) 7:32. doi: 10.1038/s41541-022-00449-1 PubMed DOI PMC
Karmakar S, Volpedo G, Zhang WW, Lypaczewski P, Ismail N, Oliveira F, et al. . Centrin-deficient Leishmania mexicana confers protection against Old World visceral leishmaniasis. NPJ Vaccines (2022) 7:157. doi: 10.1038/s41541-022-00574-x PubMed DOI PMC
Oja AE, van Lier RAW, Hombrink P. Two sides of the same coin: Protective versus pathogenic CD4(+) resident memory T cells. Sci Immunol (2022) 7:eabf9393. doi: 10.1126/sciimmunol.abf9393 PubMed DOI
Muruganandah V, Sathkumara HD, Navarro S, Kupz A. A systematic review: the role of resident memory T cells in infectious diseases and their relevance for vaccine development. Front Immunol (2018) 9:1574. doi: 10.3389/fimmu.2018.01574 PubMed DOI PMC
Lefebvre MN, Harty JT. You shall not pass: memory CD8 T cells in liver-stage malaria. Trends Parasitol (2020) 36:147–57. doi: 10.1016/j.pt.2019.11.004 PubMed DOI PMC
Nakamae S, Miyagawa S, Ogawa K, Kamiya M, Taniguchi M, Ono A, et al. . Induction of liver-resident memory T cells and protection at liver-stage malaria by mRNA-containing lipid nanoparticles. Front Immunol (2023) 14:1116299. doi: 10.3389/fimmu.2023.1116299 PubMed DOI PMC
Fernandez-Ruiz D, Ng WY, Holz LE, Ma JZ, Zaid A, Wong YC, et al. . Liver-resident memory CD8(+) T cells form a front-line defense against malaria liver-stage infection. Immunity (2016) 45:889–902. doi: 10.1016/j.immuni.2016.08.011 PubMed DOI
Hoffman SL, Goh LM, Luke TC, Schneider I, Le TP, Doolan DL, et al. . Protection of humans against malaria by immunization with radiation-attenuated Plasmodium falciparum sporozoites. J Infect Dis (2002) 185:1155–64. doi: 10.1086/339409 PubMed DOI
Landrith TA, Sureshchandra S, Rivera A, Jang JC, Rais M, Nair MG, et al. . CD103(+) CD8 T cells in the toxoplasma-infected brain exhibit a tissue-resident memory transcriptional profile. Front Immunol (2017) 8:335. doi: 10.3389/fimmu.2017.00335 PubMed DOI PMC
Olivera GC, Vetter L, Tesoriero C, Del Gallo F, Hedberg G, Basile J, et al. . Role of T cells during the cerebral infection with Trypanosoma brucei. PloS Negl Trop Dis (2021) 15:e0009764. doi: 10.1371/journal.pntd.0009764 PubMed DOI PMC
Louis L, Clark M, Wise MC, Glennie N, Wong A, Broderick K, et al. . Intradermal Synthetic DNA Vaccination Generates Leishmania-Specific T Cells in the Skin and Protection against Leishmania major. Infect Immun (2019) 87(8):e00227–19. doi: 10.1128/IAI.00227-19 PubMed DOI PMC
Gurunathan S, Prussin C, Sacks DL, Seder RA. Vaccine requirements for sustained cellular immunity to an intracellular parasitic infection. Nat Med (1998) 4:1409–15. doi: 10.1038/4000 PubMed DOI
Volpedo G, Huston RH, Holcomb EA, Pacheco-Fernandez T, Gannavaram S, Bhattacharya P, et al. . From infection to vaccination: reviewing the global burden, history of vaccine development, and recurring challenges in global leishmaniasis protection. Expert Rev Vaccines (2021) 20:1431–46. doi: 10.1080/14760584.2021.1969231 PubMed DOI
Longet S, Paul S. Pivotal role of tissue-resident memory lymphocytes in the control of mucosal infections: can mucosal vaccination induce protective tissue-resident memory T and B cells? Front Immunol (2023) 14:1216402. doi: 10.3389/fimmu.2023.1216402 PubMed DOI PMC