全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...

Developments in Diagnosis and Antileishmanial Drugs

DOI: 10.1155/2012/626838

Full-Text   Cite this paper   Add to My Lib

Abstract:

Leishmaniasis ranks the third in disease burden in disability-adjusted life years caused by neglected tropical diseases and is the second cause of parasite-related deaths after malaria; but for a variety of reasons, it is not receiving the attention that would be justified seeing its importance. Leishmaniasis is a diverse group of clinical syndromes caused by protozoan parasites of the genus Leishmania. It is estimated that 350 million people are at risk in 88 countries, with a global incidence of 1–1.5 million cases of cutaneous and 500,000 cases of visceral leishmaniasis. Improvements in diagnostic methods for early case detection and latest combitorial chemotherapeutic methods have given a new hope for combating this deadly disease. The cell biology of Leishmania and mammalian cells differs considerably and this distinctness extends to the biochemical level. This provides the promise that many of the parasite’s proteins should be sufficiently different from hosts and can be successfully exploited as drug targets. This paper gives a brief overview of recent developments in the diagnosis and approaches in antileishmanial drug discovery and development. 1. Introduction Protozoan parasitic diseases remain a major concerned public health problem, especially in tropical regions. The major death toll is due to malaria, African and American trypanosomiasis, and leishmaniasis, whose high mortality rates in underdeveloped developing countries are associated to poor hygienic conditions and lack of efficient prophylactic measures [1]. For many years, the public health impacts of the parasitic diseases have been grossly underestimated, mainly due to lack of awareness of its serious impact on health. Protozoan parasites of the genus Leishmania cause severe diseases that threaten human beings, both for the high death rates involved and the economic loss resulting from morbidity, primarily in the tropical and subtropical areas [2]. It ranks the second only to malaria, and the control of leishmaniasis remains a serious problem with ever increasing cases worldwide. It has become a major focus of concern and a serious third world problem affecting the poorer sections of the society [3]. Leishmaniasis is included in the list of neglected tropical diseases (NTDs) [4] and has a strong link to poverty [5]. The disease has been reported in 88 countries in five continents—Africa, Asia, Europe, North America, and South America out of the seven continents (22 in the new world and 66 in the old world) [6], out of which 16 are developed countries, 72 are developing, and 13 of

References

[1]  R. Pink, A. Hudson, M. A. Mouriès, and M. Bendig, “Opportunities and challenges in antiparasitic drug discovery,” Nature Reviews Drug Discovery, vol. 4, no. 9, pp. 727–740, 2005.
[2]  H. K. Majumder, B. B. Das, and A. Ganguly, “DNA topoisomerases of Leishmania: the Potential targets for anti-leishmanial therapy,” Advances in Experimental Medicine and Biology, vol. 625, pp. 103–115, 2008.
[3]  WHO, The world health report—reducing risks, promoting healthy life, World Health Organization, Geneva, Switzerland, 2002.
[4]  J. Alvar, S. Yactayo, and C. Bern, “Leishmaniasis and poverty,” Trends in Parasitology, vol. 22, no. 12, pp. 552–557, 2006.
[5]  N. Feasey, M. Wansbrough-Jones, D. C. W. Mabey, and A. W. Solomon, “Neglected tropical diseases,” British Medical Bulletin, vol. 93, no. 1, pp. 179–200, 2010.
[6]  P. Desjeux, “Leishmaniasis: current situation and new perspectives,” Comparative Immunology, Microbiology and Infectious Diseases, vol. 27, no. 5, pp. 305–318, 2004.
[7]  TDR/WHO, “Leishmaniasis,” 2012, http://www.who.int/tdr/diseases/leish/info/en/index.html.
[8]  Leishmaniasis Control, 2007, http://www.who.int/topics/leishmaniasis/en/.
[9]  J. Alvar, I. D. Velez, C. Bern et al., “Leishmaniasis worldwide and global estimates of its incidence,” PloS One, vol. 7, no. 5, pp. e35671–e356712.
[10]  B. L. Herwaldt, “Leishmaniasis,” The Lancet, vol. 354, no. 9185, pp. 1191–1199, 1999.
[11]  H. W. Murray, J. Pépin, T. B. Nutman, S. L. Hoffman, and A. A. F. Mahmoud, “Recent advances: tropical medicine,” British Medical Journal, vol. 320, no. 7233, pp. 490–494, 2000.
[12]  P. J. Guerin, P. Olliaro, S. Sundar et al., “Visceral leishmaniasis: current status of control, diagnosis, and treatment, and a proposed research and development agenda,” Lancet Infectious Diseases, vol. 2, no. 8, pp. 494–501, 2002.
[13]  N. Berhe, D. Wolday, A. Hailu et al., “HIV viral load and response to antileishmanial chemotherapy in co-infected patients,” AIDS, vol. 13, no. 14, pp. 1921–1925, 1999.
[14]  K. Seifert, F. J. Pérez-Victoria, M. Stettler et al., “Inactivation of the miltefosine transporter, LdMT, causes miltefosine resistance that is conferred to the amastigote stage of Leishmania donovani and persists in vivo,” International Journal of Antimicrobial Agents, vol. 30, no. 3, pp. 229–235, 2007.
[15]  E. Cupolillo, L. R. Brahim, C. B. Toaldo et al., “Genetic polymorphism and molecular epidemiology of Leishmania (Viannia) braziliensis from different hosts and geographic areas in Brazil,” Journal of Clinical Microbiology, vol. 41, no. 7, pp. 3126–3132, 2003.
[16]  J. Dereure, J. A. Rioux, M. Gallego et al., “Leishmania tropica in Morocco: infection in dogs,” Transactions of the Royal Society of Tropical Medicine and Hygiene, vol. 85, no. 5, p. 595, 1991.
[17]  J. Dereure, J. A. Rioux, A. Khiami, F. Pratlong, J. Périères, and A. Martini, “Ecoepidemiology of leishmaniases in Syria. 2—Presence, in dogs, of Leishmania infantum Nicolle and Leishmania tropica (Wright) (Kinetoplastida-Trypanonomatidae),” Annales de Parasitologie Humaine et Comparee, vol. 66, no. 6, pp. 252–255, 1991.
[18]  H. Yahia, P. D. Ready, A. Hamdani, J. M. Testa, and N. Guessous-Idrissi, “Regional genetic differentiation of Phlebotomus sergenti in three Moroccan foci of cutaneous leishmaniasis caused by Leishmania tropica,” Parasite, vol. 11, no. 2, pp. 189–199, 2004.
[19]  R. Reithinger, J. C. Dujardin, H. Louzir, C. Pirmez, B. Alexander, and S. Brooker, “Cutaneous leishmaniasis,” Lancet Infectious Diseases, vol. 7, no. 9, pp. 581–596, 2007.
[20]  P. A. Bates, “Transmission of Leishmania metacyclic promastigotes by phlebotomine sand flies,” International Journal for Parasitology, vol. 37, no. 10, pp. 1097–1106, 2007.
[21]  M. E. Rogers, T. Ilg, A. V. Nikolaev, M. A. J. Ferguson, and P. A. Bates, “Transmission of cutaneous leishmaniasis by sand flies is enhanced by regurgitation of fPPG,” Nature, vol. 430, no. 6998, pp. 463–467, 2004.
[22]  E. Handman and D. V. R. Bullen, “Interaction of Leishmania with the host macrophage,” Trends in Parasitology, vol. 18, no. 8, pp. 332–334, 2002.
[23]  R. J. S. Burchmore and M. P. Barrett, “Life in vacuoles—nutrient acquisition by Leishmania amastigotes,” International Journal for Parasitology, vol. 31, no. 12, pp. 1311–1320, 2001.
[24]  P. Desjeux, “Leishmaniasis: current situation and new perspectives,” Comparative Immunology, Microbiology and Infectious Diseases, vol. 27, no. 5, pp. 305–318, 2004.
[25]  S. M. Collin, P. G. Coleman, K. Ritmeijer, and R. N. Davidson, “Unseen Kala-azar deaths in south Sudan (1999-2002),” Tropical Medicine and International Health, vol. 11, no. 4, pp. 509–512, 2006.
[26]  S. P. Singh, D. C. S. Reddy, M. Rai, and S. Sundar, “Serious underreporting of visceral leishmaniasis through passive case reporting in Bihar, India,” Tropical Medicine and International Health, vol. 11, no. 6, pp. 899–905, 2006.
[27]  D. Jacquet, M. Boelaert, J. Seaman et al., “Comparative evaluation of freeze-dried and liquid antigens in the direct agglutination test for serodiagnosis of visceral leishmaniasis (ITMA-DAT/VL),” Tropical Medicine and International Health, vol. 11, no. 12, pp. 1777–1784, 2006.
[28]  P. J. Hotez, J. H. F. Remme, P. Buss, G. Alleyne, C. Morel, and J. G. Breman, “Combating tropical infectious diseases: report of the Disease Control Priorities in Developing Countries Project,” Clinical Infectious Diseases, vol. 38, no. 6, pp. 871–878, 2004.
[29]  C. Bern, A. W. Hightower, R. Chowdhury et al., “Risk factors for kala-azar in Bangladesh,” Emerging Infectious Diseases, vol. 11, no. 5, pp. 655–662, 2005.
[30]  S. Collin, R. Davidson, K. Ritmeijer et al., “Conflict and kala-azar: determinants of adverse outcomes of kala-azar among patients in southern Sudan,” Clinical Infectious Diseases, vol. 38, no. 5, pp. 612–619, 2004.
[31]  L. C. Rey, C. V. Martins, H. B. Ribeiro, and A. A. M. Lima, “American visceral leishmaniasis (kala-azar) in hospitalized children from an endemic area,” Jornal de Pediatria, vol. 81, no. 1, pp. 73–78, 2005.
[32]  E. E. Zijlstra, A. M. Musa, E. A. G. Khalil, I. M. El Hassan, and A. M. El-Hassan, “Post-kala-azar dermal leishmaniasis,” Lancet Infectious Diseases, vol. 3, no. 2, pp. 87–98, 2003.
[33]  P. R. V. Desjeux, “Post-kala-azar dermal leishmaniasis: facing the challenge of eliminating kala-azar from South Asia,” in Kala Azar in South Asia: Current Status and Challenges Ahead, pp. 111–124, Springer Science + Business Media B.V., 2011.
[34]  J. Alvar, P. Aparicio, A. Aseffa et al., “The relationship between leishmaniasis and AIDS: the second 10 years,” Clinical Microbiology Reviews, vol. 21, no. 2, pp. 334–359, 2008.
[35]  H. Reyburn, M. Rowland, M. Mohsen, B. Khan, and C. Davies, “The prolonged epidemic of anthroponotic cutaneous leishmaniasis in Kabul, Afghanistan: “Bringing down the neighbourgood”,” Transactions of the Royal Society of Tropical Medicine and Hygiene, vol. 97, no. 2, pp. 170–176, 2003.
[36]  H. Goto and J. A. L. Lindoso, “Current diagnosis and treatment of cutaneous and mucocutaneous leishmaniasis,” Expert Review of Anti-Infective Therapy, vol. 8, no. 4, pp. 419–433, 2010.
[37]  http://www.fp7-rapsodi.eu/150/.
[38]  P. D. Marsden and W. H. Lumsden, “Trypanosomiasis and leishmaniasis,” Practitioner, vol. 207, no. 238, pp. 181–185, 1971.
[39]  R. S. Bray, “Leishmania,” Annual Review of Microbiology, vol. 28, pp. 189–217, 1974.
[40]  D. M. Pratt and J. R. David, “Monoclonal antibodies that distinguish between New World species of Leishmania,” Nature, vol. 291, no. 5816, pp. 581–583, 1981.
[41]  L. Ryan, A. Vexenat, P. D. Marsden, R. Lainson, and J. J. Shaw, “The importance of rapid diagnosis of new cases of cutaneous leishmaniasis in pin-pointing the sandfly vector,” Transactions of the Royal Society of Tropical Medicine and Hygiene, vol. 84, no. 6, p. 786, 1990.
[42]  D. E. Arnot and D. C. Barker, “Biochemical identification of cutaneous leishmanias by analysis of kinetoplast DNA. II. Sequence homologies in Leishmania kDNA,” Molecular and Biochemical Parasitology, vol. 3, no. 1, pp. 47–56, 1981.
[43]  M. A. Miles, R. Lainson, J. J. Shaw, M. Povoa, and A. A. de Souza, “Leishmaniasis in Brazil: XV. Biochemical distinction of Leishmania mexicana amazonensis, L. braziliensis braziliensis and L. braziliensis guyanensis—aetiological agents of cutaneous leishmaniasis in the Amazon Basin of Brazil,” Transactions of the Royal Society of Tropical Medicine and Hygiene, vol. 75, no. 4, pp. 524–529, 1981.
[44]  A. A. L. De Ibarra, J. G. Howard, and D. Snary, “Monoclonal antibodies to Leishmania tropica major: specificities and antigen location,” Parasitology, vol. 85, no. 3, pp. 523–531, 1982.
[45]  E. Handman and J. M. Curtis, “Leishmania tropica: surface antigens of intracellular and flagellate forms,” Experimental Parasitology, vol. 54, no. 2, pp. 243–249, 1982.
[46]  D. F. Wirth and D. M. Pratt, “Rapid identification of Leishmania species by specific hybridization of kinetoplast DNA in cutaneous lesions,” Proceedings of the National Academy of Sciences of the United States of America, vol. 79, no. 22, pp. 6999–7003, 1982.
[47]  G. C. Brainard and P. L. Podolin, “Near-ultraviolet radiation suppresses pineal melatonin content,” Endocrinology, vol. 119, no. 5, pp. 2201–2205, 1986.
[48]  S. M. Le Blancq and W. Peters, “Leishmania in the Old World: 2. Heterogeneity among L. tropica zymodemes,” Transactions of the Royal Society of Tropical Medicine and Hygiene, vol. 80, no. 1, pp. 113–119, 1986.
[49]  B. L. Travi, Y. Osorio, P. C. Melby, B. Chandrasekar, L. Arteaga, and N. G. Saravia, “Gender is a major determinant of the clinical evolution and immune response in hamsters infected with Leishmania spp,” Infection and Immunity, vol. 70, no. 5, pp. 2288–2296, 2002.
[50]  WHO, “Control of the Leishmaniasis,” Technical Report Series 793, 1990.
[51]  I. L. Mauricio, J. R. Stothard, and M. A. Miles, “The strange case of Leishmania chagasi,” Parasitology Today, vol. 16, no. 5, pp. 188–189, 2000.
[52]  N. L. Sharma, V. K. Mahajan, A. Kanga et al., “Localized cutaneous leishmaniasis due to Leishmania donovani and Leishmania tropica: preliminary findings of the study of 161 new cases from a new endemic focus in Himachal Pradesh, India,” American Journal of Tropical Medicine and Hygiene, vol. 72, no. 6, pp. 819–824, 2005.
[53]  A. L. Ba?uls, J. C. Dujardin, F. Guerrini et al., “Is Leishmania (Viannia) peruviana a distinct species? A MLEE/RAPD evolutionary genetics answer,” Journal of Eukaryotic Microbiology, vol. 47, no. 3, pp. 197–207, 2000.
[54]  J. Fraga, A. M. Montalvo, S. De Doncker, J. C. Dujardin, and G. Van der Auwera, “Phylogeny of Leishmania species based on the heat-shock protein 70 gene,” Infection, Genetics and Evolution, vol. 10, no. 2, pp. 238–245, 2010.
[55]  H. W. Murray, J. D. Berman, C. R. Davies, and N. G. Saravia, “Advances in leishmaniasis,” The Lancet, vol. 366, no. 9496, pp. 1561–1577, 2005.
[56]  S. Deborggraeve, T. Laurent, D. Espinosa et al., “A simplified and standardized polymerase chain reaction format for the diagnosis of leishmaniasis,” Journal of Infectious Diseases, vol. 198, no. 10, pp. 1565–1572, 2008.
[57]  F. Vega-Lopez, “Diagnosis of cutaneous leishmaniasis,” Current Opinion in Infectious Diseases, vol. 16, pp. 97–101, 2003.
[58]  S. N. Al-Hucheimi, B. A. Sultan, and M. A. Al-Dhalimi, “A comparative study of the diagnosis of Old World cutaneous leishmaniasis in Iraq by polymerase chain reaction and microbiologic and histopathologic methods,” International Journal of Dermatology, vol. 48, no. 4, pp. 404–408, 2009.
[59]  M. Siddig, H. Ghalib, D. C. Shillington, and E. A. Petersen, “Visceral leishmaniasis in the Sudan: comparative parasitological methods of diagnosis,” Transactions of the Royal Society of Tropical Medicine and Hygiene, vol. 82, no. 1, pp. 66–68, 1988.
[60]  G. Srividya, A. Kulshrestha, R. Singh, and P. Salotra, “Diagnosis of visceral leishmaniasis: developments over the last decade,” Parasitology Research, vol. 110, pp. 1065–1078, 2012.
[61]  A. M. Allahverdiyev, M. Bagirova, S. Uzun et al., “The value of a new microculture method for diagnosis of visceral leishmaniasis by using bone marrow and peripheral blood,” American Journal of Tropical Medicine and Hygiene, vol. 73, no. 2, pp. 276–280, 2005.
[62]  G. Sreenivas, N. A. Ansari, R. Singh et al., “Diagnosis of visceral leishmaniasis: comparative potential of amastigote antigen, recombinant antigen and PCR,” British Journal of Biomedical Science, vol. 59, no. 4, pp. 218–222, 2002.
[63]  D. Jacquet, M. Boelaert, J. Seaman et al., “Comparative evaluation of freeze-dried and liquid antigens in the direct agglutination test for serodiagnosis of visceral leishmaniasis (ITMA-DAT/VL),” Tropical Medicine and International Health, vol. 11, no. 12, pp. 1777–1784, 2006.
[64]  S. Sundar, R. K. Singh, R. Maurya et al., “Serological diagnosis of Indian visceral leishmaniasis: direct agglutination test versus rK39 strip test,” Transactions of the Royal Society of Tropical Medicine and Hygiene, vol. 100, no. 6, pp. 533–537, 2006.
[65]  M. Boelaert, S. El-Safi, A. Hailu et al., “Diagnostic tests for kala-azar: a multi-centre study of the freeze-dried DAT, rK39 strip test and KAtex in East Africa and the Indian subcontinent,” Transactions of the Royal Society of Tropical Medicine and Hygiene, vol. 102, no. 1, pp. 32–40, 2008.
[66]  R. Kumar, K. Pai, K. Pathak, and S. Sundar, “Enzyme-linked immunosorbent assay for recombinant K39 antigen in diagnosis and prognosis of Indian visceral leishmaniasis,” Clinical and Diagnostic Laboratory Immunology, vol. 8, no. 6, pp. 1220–1224, 2001.
[67]  P. Srivastava, A. Dayama, S. Mehrotra, and S. Sundar, “Diagnosis of visceral leishmaniasis,” Transactions of the Royal Society of Tropical Medicine and Hygiene, vol. 105, no. 1, pp. 1–6, 2011.
[68]  P. Srivastava, S. Mehrotra, P. Tiwary, J. Chakravarty, and S. Sundar, “Diagnosis of indian visceral leishmaniasis by nucleic acid detection using PCR,” PLoS ONE, vol. 6, no. 4, article e19304, 2011.
[69]  R. Maurya, R. K. Singh, B. Kumar, P. Salotra, M. Rai, and S. Sundar, “Evaluation of PCR for diagnosis of Indian kala-azar and assessment of cure,” Journal of Clinical Microbiology, vol. 43, no. 7, pp. 3038–3041, 2005.
[70]  K. Kuhls, L. Keilonat, S. Ochsenreither et al., “Multilocus microsatellite typing (MLMT) reveals genetically isolated populations between and within the main endemic regions of visceral leishmaniasis,” Microbes and Infection, vol. 9, no. 3, pp. 334–343, 2007.
[71]  A. Dey and S. Singh, “Genetic heterogeneity among visceral and post-Kala-Azar dermal leishmaniasis strains from eastern India,” Infection, Genetics and Evolution, vol. 7, no. 2, pp. 219–222, 2007.
[72]  K. W. Q. Tintaya, X. Ying, J. P. Dedet, S. Rijal, X. De Bolle, and J. C. Dujardin, “Antigen genes for molecular epidemiology of leishmaniasis: polymorphism of cysteine proteinase B and surface metalloprotease glycoprotein 63 in the Leishmania donovani complex,” Journal of Infectious Diseases, vol. 189, no. 6, pp. 1035–1043, 2004.
[73]  I. L. Mauricio, J. R. Stothard, and M. A. Miles, “Leishmania donovani complex: genotyping with the ribosomal internal transcribed spacer and the mini-exon,” Parasitology, vol. 128, no. 3, pp. 263–267, 2004.
[74]  S. De Doncker, V. Hutse, S. Abdellati et al., “A new PCR-ELISA for diagnosis of visceral leishmaniasis in blood of HIV-negative subjects,” Transactions of the Royal Society of Tropical Medicine and Hygiene, vol. 99, no. 1, pp. 25–31, 2005.
[75]  S. Khosravi, S. H. Hejazi, M. Hashemzadeh, G. Eslami, and H. Y. Darani, “Molecular diagnosis of old world leishmaniasis: real-time PCR based on tryparedoxin peroxidase gene for the detection and identification of Leishmania spp,” Journal of Vector Borne Diseases, vol. 49, no. 1, pp. 15–18, 2012.
[76]  E. E. Zijlstra, M. S. Ali, A. M. El-Hassan et al., “Kala-azar: a comparative study of parasitological methods and the direct agglutination test in diagnosis,” Transactions of the Royal Society of Tropical Medicine and Hygiene, vol. 86, no. 5, pp. 505–507, 1992.
[77]  A. M. Allahverdiyev, M. Bagirova, S. Uzun et al., “The value of a new microculture method for diagnosis of visceral leishmaniasis by using bone marrow and peripheral blood,” American Journal of Tropical Medicine and Hygiene, vol. 73, no. 2, pp. 276–280, 2005.
[78]  F. Chappuis, S. Rijal, A. Soto, J. Menten, and M. Boelaert, “A meta-analysis of the diagnostic performance of the direct agglutination test and rK39 dipstick for visceral leishmaniasis,” British Medical Journal, vol. 333, no. 7571, pp. 723–726, 2006.
[79]  M. Gari-Toussaint, A. Leliévre, P. Marty, and Y. Le Fichoux, “Contribution of serological tests to the diagnosis of visceral leishmaniasis in patients infected with the human immunodeficiency virus,” Transactions of the Royal Society of Tropical Medicine and Hygiene, vol. 88, no. 3, pp. 301–302, 1994.
[80]  H. D. F. H. Schallig, G. J. Schoone, C. C. M. Kroon, A. Hailu, F. Chappuis, and H. Veeken, “Development and application of 'simple' diagnostic tools for visceral leishmaniasis,” Medical Microbiology and Immunology, vol. 190, no. 1-2, pp. 69–71, 2001.
[81]  S. Sundar, R. K. Singh, R. Maurya et al., “Serological diagnosis of Indian visceral leishmaniasis: direct agglutination test versus rK39 strip test,” Transactions of the Royal Society of Tropical Medicine and Hygiene, vol. 100, no. 6, pp. 533–537, 2006.
[82]  C. B. Palatnik-de-Sousa, E. M. Gomes, E. Paraguai-de-Souza, M. Palatnik, K. Luz, and R. Borojevic, “Leishmania donovani: titration of antibodies to the fucose-mannose ligand as an aid in diagnosis and prognosis of visceral leishmaniasis,” Transactions of the Royal Society of Tropical Medicine and Hygiene, vol. 89, no. 4, pp. 390–393, 1995.
[83]  R. Kumar, K. Pai, K. Pathak, and S. Sundar, “Enzyme-linked immunosorbent assay for recombinant K39 antigen in diagnosis and prognosis of Indian visceral leishmaniasis,” Clinical and Diagnostic Laboratory Immunology, vol. 8, no. 6, pp. 1220–1224, 2001.
[84]  R. L. Houghton, M. Petrescu, D. R. Benson et al., “A cloned antigen (recombinant K39) of Leishmania chagasi diagnostic for visceral leishmaniasis in human immunodeficiency virus type 1 patients and a prognostic indicator for monitoring patients undergoing drug therapy,” Journal of Infectious Diseases, vol. 177, no. 5, pp. 1339–1344, 1998.
[85]  T. M. Mohapatra, D. P. Singh, M. R. Sen, K. Bharti, and S. Sundar, “Comparative evaluation of rK9, rK26 and rK39 antigens in the serodiagnosis of Indian visceral leishmaniasis,” Journal of Infection in Developing Countries, vol. 4, no. 2, pp. 114–117, 2010.
[86]  F. Chappuis, Y. Mueller, A. Nguimfack et al., “Diagnostic accuracy of two rK39 antigen-based dipsticks and the formol gel test for rapid diagnosis of visceral leishmaniasis in northeastern Uganda,” Journal of Clinical Microbiology, vol. 43, no. 12, pp. 5973–5977, 2005.
[87]  F. Chappuis, S. Sundar, A. Hailu et al., “Visceral leishmaniasis: what are the needs for diagnosis, treatment and control?” Nature Reviews Microbiology, vol. 5, no. 11, pp. 873–882, 2007.
[88]  M. J. Pérez-Alvarez, R. Larreta, C. Alonso, and J. M. Requena, “Characterisation of a monoclonal antibody recognising specifically the HSP70 from Leishmania,” Parasitology Research, vol. 87, no. 11, pp. 907–910, 2001.
[89]  R. Piarroux, F. Gambarelli, H. Dumon et al., “Comparison of PCR with direct examination of bone marrow aspiration, myeloculture, and serology for diagnosis of visceral leishmaniasis in immunocompromised patients,” Journal of Clinical Microbiology, vol. 32, no. 3, pp. 746–749, 1994.
[90]  S. De Doncker, V. Hutse, S. Abdellati et al., “A new PCR-ELISA for diagnosis of visceral leishmaniasis in blood of HIV-negative subjects,” Transactions of the Royal Society of Tropical Medicine and Hygiene, vol. 99, no. 1, pp. 25–31, 2005.
[91]  R. K. Singh, H. P. Pandey, and S. Sundar, “Visceral leishmaniasis (kala-azar): challenges ahead,” Indian Journal of Medical Research, vol. 123, no. 3, pp. 331–344, 2006.
[92]  T. K. Jha, “Evaluation of diamidine compound (pentamidine isethionate) in the treatment of resistant cases of kala-azar occurring in North Bihar, India,” Transactions of the Royal Society of Tropical Medicine and Hygiene, vol. 77, no. 2, pp. 167–170, 1983.
[93]  S. N. Jha, N. K. Singh, and T. K. Jha, “Changing response to diamidine compounds in cases of kala-azar unresponsive to antimonial,” The Journal of the Association of Physicians of India, vol. 39, no. 4, pp. 314–316, 1991.
[94]  S. Sundar, “Treatment of visceral leishmaniasis,” Medical Microbiology and Immunology, vol. 190, no. 1-2, pp. 89–92, 2001.
[95]  S. L. Croft and V. Yardley, “Chemotherapy of leishmaniasis,” Current Pharmaceutical Design, vol. 8, no. 4, pp. 319–342, 2002.
[96]  A. Jhingran, B. Chawla, S. Saxena, M. P. Barrett, and R. Madhubala, “Paromomycin: uptake and resistance in Leishmania donovani,” Molecular and Biochemical Parasitology, vol. 164, no. 2, pp. 111–117, 2009.
[97]  N. Singh, M. Kumar, and R. K. Singh, “Leishmaniasis: current status of available drugs and new potential drug targets,” Asian Pacific Journal of Tropical Medicine, vol. 5, no. 6, pp. 485–497, 2012.
[98]  A. Rochette, F. Raymond, J. M. Ubeda et al., “Genome-wide gene expression profiling analysis of Leishmania major and Leishmania infantum developmental stages reveals substantial differences between the two species,” BMC Genomics, vol. 9, pp. 255–281, 2008.
[99]  S. Krieger, W. Schwarz, M. R. Arlyanayagam, A. H. Fairlamb, R. L. Krauth-Siegel, and C. Clayton, “Trypanosomes lacking trypanothione reductase are avirulent and show increased sensitivity to oxidative stress,” Molecular Microbiology, vol. 35, no. 3, pp. 542–552, 2000.
[100]  D. B. Castro-Pinto, M. Genestra, G. B. Menezes et al., “Cloning and expression of trypanothione reductase from a New World Leishmania species,” Archives of Microbiology, vol. 189, no. 4, pp. 375–384, 2008.
[101]  R. F. Rodrigues, D. Castro-Pinto, A. Echevarria et al., “Investigation of trypanothione reductase inhibitory activity by 1, 3, 4- thiadiazolium-2-aminide derivatives and molecular docking studies,” Bioorganic & Medicinal Chemistry, vol. 20, no. 5, pp. 1760–1766, 2012.
[102]  P. Ascenzi, A. Bocedi, P. Visca, G. Antonini, and L. Gradoni, “Catalytic properties of cysteine proteinases from Trypanosoma cruzi and Leishmania infantum: a pre-steady-state and steady-state study,” Biochemical and Biophysical Research Communications, vol. 309, no. 3, pp. 659–665, 2003.
[103]  S. O. Lorente, R. Gómez, C. Jiménez et al., “Biphenylquinuclidines as inhibitors of squalene synthase and growth of parasitic protozoa,” Bioorganic and Medicinal Chemistry, vol. 13, no. 10, pp. 3519–3529, 2005.
[104]  P. Bhargava, K. Kumar, S. S. Chaudhaery, A. K. Saxena, and U. Roy, “Cloning, overexpression and characterization of Leishmania donovani squalene synthase,” FEMS Microbiology Letters, vol. 311, no. 1, pp. 82–92, 2010.
[105]  J. A. Urbina, J. L. Concepcion, S. Rangel, G. Visbal, and R. Lira, “Squalene synthase as a chemotherapeutic target in Trypanosoma cruzi and Leishmania mexicana,” Molecular and Biochemical Parasitology, vol. 125, no. 1-2, pp. 35–45, 2002.
[106]  C. Jiménez-Jiménez, J. Carrero-Lérida, M. Sealey-Cardona, L. M. Ruiz-Pérez, J. A. Urbina, and D. González-Pacanowska, “Δ24 (25)-sterol methenyltransferase: intracellular localization and azasterol sensitivity in Leishmania major promastigotes overexpressing the enzyme,” Molecular and Biochemical Parasitology, vol. 160, no. 1, pp. 52–59, 2008.
[107]  C. W. Roberts, R. McLeod, D. W. Rice, M. Ginger, M. L. Chance, and L. J. Goad, “Fatty acid and sterol metabolism: potential antimicrobial targets in apicomplexan and trypanosomatid parasitic protozoa,” Molecular and Biochemical Parasitology, vol. 126, no. 2, pp. 129–142, 2003.
[108]  O. Heby, L. Persson, and R. Madhubala, “Targeting the polyamine biosynthetic enzymes: a promising approach to therapy of African sleeping sickness, Chagas' disease, and leishmaniasis,” Amino Acids, vol. 33, no. 2, pp. 359–366, 2007.
[109]  V. Iniesta, L. C. Gómez-Nieto, and I. Corraliza, “The inhibition of arginase by Nω-hydroxy-L-arginine controls the growth of Leishmania inside macrophages,” Journal of Experimental Medicine, vol. 193, no. 6, pp. 777–783, 2001.
[110]  R. Balana-Fouce, E. Calvo-álvarez, R. álvarez-Velilla, C. F. Prada, Y. Pérez-Pertejo, and R. M. Reguera, “Role of trypanosomatid's arginase in polyamine biosynthesis and pathogenesis,” Molecular and Biochemical Parasitology, vol. 181, no. 2, pp. 85–93, 2012.
[111]  B. M. Bakker, F. I. C. Mensonides, B. Teusink, P. Van Hoek, P. A. M. Michels, and H. V. Westerhoff, “Compartmentation protects trypanosomes from the dangerous design of glycolysis,” Proceedings of the National Academy of Sciences of the United States of America, vol. 97, no. 5, pp. 2087–2092, 2000.
[112]  P. A. M. Michels, V. Hannaert, and F. Bringaud, “Metabolic aspects of glycosomes in Trypanosomatidae—new data and views,” Parasitology Today, vol. 16, no. 11, pp. 482–489, 2000.
[113]  P. Hassan, D. Fergusson, K. M. Grant, and J. C. Mottram, “The CRK3 protein kinase is essential for cell cycle progression of Leishmania mexicana,” Molecular and Biochemical Parasitology, vol. 113, no. 2, pp. 189–198, 2001.
[114]  K. M. Grant, M. H. Dunion, V. Yardley et al., “Inhibitors of Leishmania mexicana CRK3 cyclin-dependent kinase: chemical library screen and antileishmanial activity,” Antimicrobial Agents and Chemotherapy, vol. 48, no. 8, pp. 3033–3042, 2004.
[115]  E. Xingi, D. Smirlis, V. Myrianthopoulos et al., “6-Br-5methylindirubin-3′oxime (5-Me-6-BIO) targeting the leishmanial glycogen synthase kinase-3 (GSK-3) short form affects cell-cycle progression and induces apoptosis-like death: exploitation of GSK-3 for treating leishmaniasis,” International Journal for Parasitology, vol. 39, no. 12, pp. 1289–1303, 2009.
[116]  P. H. Liang and K. S. Anderson, “Substrate channeling and domain-domain interactions in bifunctional thymidylate synthase-dihydrofolate reductase,” Biochemistry, vol. 37, no. 35, pp. 12195–12205, 1998.
[117]  O. Senkovich, N. Schormann, and D. Chattopadhyay, “Structures of dihydrofolate reductase-thymidylate synthase of trypanosoma cruzi in the folate-free state and in complex with two antifolate drugs, trimetrexate and methotrexate,” Acta Crystallographica Section D, vol. 65, no. 7, pp. 704–716, 2009.
[118]  F. Zuccotto, A. C. R. Martin, R. A. Laskowski, J. M. Thornton, and I. H. Gilbert, “Dihydrofolate reductase: a potential drug target in trypanosomes and leishmania,” Journal of Computer-Aided Molecular Design, vol. 12, no. 3, pp. 241–257, 1998.
[119]  I. D. Kuntz, “Structure-based strategies for drug design and discovery,” Science, vol. 257, no. 5073, pp. 1078–1082, 1992.
[120]  P. S. T. Veras, C. I. Brodskyn, F. M. P. Balestieri et al., “A dhfr-ts- Leishmania major Knockout Mutant Cross-protects against Leishmania amazonensis,” Memorias do Instituto Oswaldo Cruz, vol. 94, no. 4, pp. 491–496, 1999.
[121]  F. Fonseca-Silva, J. D. F. Inacio, M. M. Canto-Cavalheiro, and E. E. Almeida-Amaral, “Reactive oxygen species production and mitochondrial dysfunction contribute to quercetin induced death in Leishmania amazonensis,” PLoS ONE, vol. 6, no. 2, article e14666, 2011.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133

WeChat 1538708413