Open Access


Read more
image01

Online Manuscript Submission


Read more
image01

Submitted Manuscript Trail


Read more
image01

Online Payment


Read more
image01

Online Subscription


Read more
image01

Email Alert



Read more
image01

Original Research Article | OPEN ACCESS

Luteolin as a potent anti-leishmanial agent against intracellular Leishmania tropica parasite

Kashif Igbal1,3 , Qaiser Jamal4, Javeid Igbal1, Maria Sadaf Afreen5, Muhammad Zeeshan Ahmed Sandhu2, Eshwa Dar2, Umar Farooq2, Mohammad Fahd Mushtaq2, Numera Arshad2, Muhammad Mohsin Iqbal2

1Department of Pharmacology, Faculty of Pharmacy and Health Sciences, University of Balochistan, Quetta; 2Islam College of Pharmacy, Sialkot, Punjab; 3School of Pharmacy, University of Lahore-Islamabad Campus, Islamabad, Pakistan; 4Department of Zoology, University of Peshawar, Peshawar; 5Gomal University, Dera Ismail Khan, KP, Pakistan.

For correspondence:-  Kashif Igbal   Email: kashifiqbal321@gmail.com   Tel:+923356951284

Received: 1 November 2016        Accepted: 16 January 2017        Published: 26 February 2017

Citation: Igbal K, Jamal Q, Igbal J, Afreen MS, Sandhu MZ, Dar E, et al. Luteolin as a potent anti-leishmanial agent against intracellular Leishmania tropica parasite. Trop J Pharm Res 2017; 16(2):337-342 doi: 10.4314/tjpr.v16i2.11

© 2017 The authors.
This is an Open Access article that uses a funding model which does not charge readers or their institutions for access and distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0) and the Budapest Open Access Initiative (http://www.budapestopenaccessinitiative.org/read), which permit unrestricted use, distribution, and reproduction in any medium, provided the original work is properly credited..

Abstract

Purpose: To examine the anti-leishmanial and cytotoxic effects of five naturally occurring phenolic compounds: luteolin (1), lalioside (2), luteolin-4’-O-β-D-glucopyranoside (3), apigenin 4-O-β-D-glucopyranoside (4) and apigenin (5) on Leishmania tropica KWH23 amastigotes .
Methods: The compounds were isolated from the leaves of Lawsonia Inermis via hyphenated high performance liquid chromatography-high resolution mass spectrometry coupled with solid phase extraction-tube transfer nuclear magnetic resonance technique. The isolated compounds were given intraperitoneally to L. tropica KWH23 amastigotes-infected albino mice at a dose of ≥ 3 mg/kg for 5 days. Amphotericin-B was used as standard (reference) drug. Lymphocytes were used to analyze their cytotoxicity. 
Results: For compound 1, mean lesion size decreased from 0.82 ± 0.12 to 0.10 ± 0.01 after 120 days, with 97 % cure of intracellular L. tropica amastigotes at a dose of 15 mg/kg, compared to amphotericin B which produced 95 % cure at a dose of 30 mg/kg. Half-maximal concentration (IC50) for compound 1 was 4.15 µg/ml against lymphocytes.
Conclusion: The results indicate that luteolin is a potent inhibitor of L. tropica amastigotes, with a higher cytotoxic activity against lymphocytes, compared with luteolin-4’-O-β-D-glucopyranoside.

Keywords: Leishmania tropica, Luteolin, Lalioside, Luteolin-4’-O-^6;-D-glucopyranoside, Apigenin 4-O- ^6;-D-glucopyranoside, Apigenin

Introduction

Leishmaniasis, caused by parasites belonging to the genus Leishmania (Family Trypanoso-matidae), is a major public health problem in tropical and sub-tropical regions. The parasite is transmitted by the sand fly (vector), with dogs, sheep, rats, horses, and cats being common animal hosts. The World Health Organization, WHO has reported that people from 98 countries in 5 continents, are at high risk of leishmaniasis, and it is estimated that approximately 12 million people are currently infected [1-3]. Cutaneous leishmaniasis is caused by different Leishmania species, e.g. Leishmania tropica, Leishmania major, Leishmania amazonensis, and Leishmania brazillensis. In Pakistan, L. tropica and L. major are the main causes of cutaneous leishmaniasis [4,5]. First line therapy for cutaneous leishmaniasis in Europe, Asia and Africa is pentavalent antimonials, i.e., sodium stibogluconate and meglumine antimoniate [2]. However, antimonials have severe side effects like myalgia, pancreatitis, cardiac arrhythmia, hepatitis, and drug accumulation in liver and spleen. Thus, there is an urgent need for new chemical entities for non-toxic and effective treatment of leishmaniasis [2,4].

Phenolic compounds comprise different groups including coumarins, flavonoids, tannins, naphthalenes, naphthoquinones, xanthones, lignans and alkylphenones [6]. Flavonoids and alkylphenones possess a variety of biological and pharmacological activities, including antioxidant [7], antibacterial, antiulcer [8], antifungal [9], antiviral [10], HIV - inhibiting [11], and anti-herpes simplex type 1 (HSV-1) [12]. They also inhibit electron transfer in mitochondrial inner membrane [13]. Flavonoids and alkylphenones are chemically well investigated, and a large number of member compounds have been reported. These include flavonoids and luteolin [14]; luteolin-4’-O-β-D-glucopyranoside [15]; apigenin 4-O-β-D-glucopyranoside [16]; apigenin [15]; xanhoangelol [8]; macaranggin [7]; artelastin [10]; 5,7-Dihydroxy-6,8-diphenyl flavonoids [11]; leachianone G [12] and alkylphenones such as Lalioside [17], 2,4,6-trihydroxyacetophenone-2-O-β-D-glucopyranoside [18] and lawsoniaside [19]. 

However, the current work deals with anti-leishmanial and cytotoxic effects of naturally occurring plant phenolic compounds having four flavonoids and one alkylphenone.

Methods

Chemicals

Fetal bovine serum, dimethylsulphoxide, Luteolin, Lalioside, luteolin-4’-O-β-D-glucopyra-noside, apigenin 4-O-β-D-glucopyranoside, apigenin and RPMI-1640 medium were purchased from Sigma-Aldrich (St. Louis, MO), whereas penicillin and streptomycin were purchased from MERCK, U.S.A. Amphotericin B was supplied by AmB-Fungizone, Bristol-Myers Squibb, UK. Water used for in vivo test solutions was purified by deionization and 0.22 µm membrane filtration (Millipore, Billerica, MA).

In vivo test

To study the pathogenesis of leishmania strain, 9 groups of male BALB/c mice (aged 6 - 8 weeks, and weighing 20 - 32 g) were used. Drug administration was through cardiac route. Promastigotes of L. Tropica KWH23 [21] were cultured in RPMI-1640 medium along with 10 % fetal bovine serum, penicillin (200 U/ml) and streptomycin (0.2 mg/ml). The parasite was cultured at 26 °C for 4 days in BOD incubator (Gallenkamp, Size 1, UK), and then harvested parasite. The harvested promastigotes were taken in a sterile tube and counted in a haemocytometer (REICHERT, N.Y, USA) under upright microscope (CX31, OLYMPUS, Tokyo, Japan). The promastigotes were centrifuged at 4 °C for 10 min at 2000 rpm; the supernatant liquid was discarded while pellet was left in tube. Fresh RPMI-1640 medium with 10 % FBS was added to get the required volume (10ml). The required volume (10 µl) of promastigotes (containing 1.4 x 106 promastigotes/ml) was injected into the cardiac cavity (intraperitoneally) of the BALB/c mice. Developed lesions were measured weekly with dial micrometer (Mitutoyo, Japan) during the infection period. Infection was well established and clearly visible lesions were evident to the naked eye after 36 days. Then treatment process was started. Dose of test compounds given to Groups I, II, III, IV and V were 3.0 mg/kg for 5 days (total dose = 15 mg/kg) in DMSO up to final volume of 3 ml. Amphotericin-B was used as standard drug (positive control) at a dose of 15 mg/kg. No drug agent was used in VII group (negative control). The injection dose of 10 µl was given five times with 3-day intervals, and lesion measurements were recorded regularly. Dial micrometer was used to note the difference between size of the lesion in infected and uninfected mice weekly. Before and after treatment, needle aspirations were taken from the lesions [22]. To detect amastigotes under upright microscope, Giemsa stain was used, and the samples were examined under oil immersions. On the 30th, 60th, 90th and 120th day of infection, 60 mg of tissue sample was taken from the lesion for biopsy. In the identification of amastigotes, sample was smeared on the slides stained with Giemsa and upright microscope was used for examination.

Ethics statement

BALB/c mice were supplied by Department of Pharmacology (Animal center), University of Peshawar, KPK, Pakistan and this study was approved by Animal and Ethics Committee, Faculty of Pharmacy and Health Sciences, University of Balochistan (UOB), Quetta (approval ref. no. 093/FOPHS/UOB). The animals were maintained in accordance with UOB, Quetta Policy and international guidelines on the care and use of laboratory animals [20]. Standard diet along with water were given ad libitum to the BALB/c mice during experiments.

Cytotoxicity test

Fresh blood (10 ml) from a healthy volunteer was taken in BD vacutainer K2E (EDTA) to get mammalian cells (lymphocytes). Cytotoxic assay of test compounds was carried out by an adoption of the method described by Iqbal et al [22]. PBS was passed through 0.2 µm filter under laminar flow hood (kept sterile conditions) and then equal volumes of PBS and blood were taken in a sterile tube. Ficol solution (volume ratio 1:2) was carefully added at 165° angle to the mixture of PBS and blood. The tube was centrifuged at 2000 rpm at 4 °C for 30 minutes. The lower transparent portion was punctured with a syringe, and the liquid carefully removed and added to 5 ml of RPMI-1640 medium. The number of lymphocytes was counted in a haemocytometer under upright microscope. In the next step, 100 µl of the lymphocyte media was put into each well of a 96-well culture plate. The test compounds were added at doses of 100, 50, 25 and 10 µg/ml in DMSO, each in a final volume of 3 ml. Amphotericin-B (25 µg/ml, positive control) was used as reference drug, while negative control was L. tropica KWH23 promastigotes. Using a multipipette, 10 µl of promastigotes (containing 1.4 x 106 promastigotes/ml) was added to 12 wells of the culture plate and placed in an incubator at 26 °C for at least 48 h. Haemocytometer was used to count viable lymphocytes and promastigotes under light microscope (lens 40x) at 24 and 48 hours. The cytotoxic tests were done in triplicate and the IC50 for each compound was calculated [23].

Statistical analysis

Results of in vivo anti-leishmanial assay of plant extract were expressed as mean % inhibition of parasite growth ± SD (n = 3). Cytotoxicity values were expressed as 50 % Inhibitory concentration (IC50) and analysed by non-linear regression analysis. For in vivo assays, mean lesion size (mm) and percentage cure were analysed by GraphPad Prism 5 software (GraphPad software, San Diego, CA) at 95 % confidence limit.

Results

Results of in vivo anti-leishmanial activities of compounds 1 - 5 analyzed in albino mice infected with 0.02 ml of clinically isolated L. tropica KWH23 having 1.4 x 106 promastigotes, via intraperitoneal route, are shown . Mean lesion size decreased significantly from 0.29 ±  0.21 mm to 0.10 ± 0.01 mm after treatment with test compounds but that of the negative group reached 1.5 ± 0.02 mm, whereas it decreased in the Amphotericin (standard drug) group from 0.85 ± 0.05 mm to 0.17 ± 0.01mm after 120 days. Luteolin showed the strongest anti-leishmanial activity. The mice groups that received apigenin, luteolin-4’-O-β-D-glucopyranoside, and apigenin 4-O-β-D-glucopyranoside had mean lesion sizes of 0.29 ±  0.21 mm, 0.25 ±  0.09 mm and 0.21 ±  0.03 mm respectively, while corresponding percentage cure were 75.02, 78.51 and 80.19 % respectively. With Luteolin and lalioside mean lesion sizes were decreased to 0.10 ± 0.01 and 0.19 ± 0.03 mm, corresponding to 97.02 and 85.27 % cure, respectively.

Cytotoxic effect

Results of cytotoxic activities of compounds 1-5 are summarized in . Compounds 3, 4 and 5 showed highest cytotoxic activities 10.27, 9.50 and 8.30 µg/ml respectively. Compound 3 exhibited the greatest cytotoxicity against lymphocytes, with IC50 value of 10.27 µg/ml (95 % C.I = 6.215-15.38). Compound 1 gave the lowest cytotoxic activity (IC50 = 4.15 µg/ml; 95 % C.I = 2.287-5.941), whereas compound 2 showed IC50 value of 5.02 µg/ml (95 % C.I = 2.136-7.087). In comparison, amphotericin-B (standard drug) displayed IC50 of 1.072 µg/ml (95 % C.I = 0.823-2.358).

Discussion

Compounds 1-5 were selected due to their promising in vitro anti-leishmanial effects [22] against L. tropica. All tested compounds showed a cure rate between 97.02 and 75.02 % at 3 mg/kg concentration (for 5 days) whereas cytotoxic effects against lymphocytes in terms of IC50 values ranging between 4.15 and 10.27 µg/ml. All the mice were cured at or after 120th day of infection, when compounds 1 - 5 were given; whereas after 60th day, mean lesion size was decreased up to 0.4 ± 0.07 mm in mice. Luteolin achieved a higher cure rate and decrease in lesion size than amphotericin B. Interspecies variation in sensitivity against luteolin have been observed, e.g., it showed in vitro IC50 of 12.5 µM against L. donovani [27,28] and reduced splenic parasitic load up to 80 % when 3.5 mg/kg (twice a week for 1 month) was given to L. donovani infected hamsters [29].

Luteolin decreased cell viability of human hepatoma HepG2 cells, result in decrease in cytotoxic activity up to 41 % [24]. In this study, lalioside, apigenin, luteolin-4’-O-β-D-glucopyranoside, and apigenin 4-O-β-D-glucopyranoside showed significant anti-leishmanial properties. Similar results have been previously reported. Tasdemir reported that apigenin showed in vitro anti-leishmanial activity against L. donovani with IC50 values 1.9 µg/ml [28]. Apigenin showed cytotoxic activity at 8.30 µg/ml and induced hepatotoxicity at 100 and 200 mg/kg whereas it was non-toxic in 25 and 50 mg/kg [25]. In another study, apigenin decreased viability of human hepatoma HepG2 cells up to 42 % [24]. Apigenin 4-O-β-D-glucopyranoside showed cytotoxic activity at a concentration of 9.50 µg/ml and exhibited antitumor activity towards Hep G2, Hep 3B, MCF-7, A549 and MDA-MB-231 [26]. In the present study, lalioside and luteolin-4’-O-β-D-glucopyranoside showed greater cytotoxicity against lymphocytes, with IC50 values of 5.02 µg/ml (95 % CL = 2.136-7.087) and 10.27 µg/ml (95 % CI = 6.215-15.38) respectively, compared with amphotericin B (IC50 values = 1.072 µg/ml ;95 % CL = 0.823-2.358). The cytotoxic effects of these compounds may be due to the presence of phenolic hydroxyl groups [28]. Hydroxyl groups have affinity for proteins and may result in inhibition of microbial enzymes and plant NADH dehydrogenase [30,31]. It is evident that luteolin and luteolin-4’-O-β-D-glucopyranoside merit further investigations as suitable drug candidates against L. tropica.

Although different activities pertaining to the compounds studied are available in literature, the current study is the first report on the anti-leishmanial activities of compounds 2 – 4 and the first comprehensive in vivo study of cytotoxic activities of compounds 1-5 against L. tropica and mammalian cells.

Conclusion

Luteolin is a potent anti-leishmanial agent, but is cytotoxic against lymphocytes. Luteolin-4’-O-β-D-glucopyranoside possesses significant anti-leishmanial activity, and is least toxic against lymphocytes. Lalioside exert anti-leishmanial activity while apigenin 4-O-β-D-glucopyranoside and apigenin have a moderate inhibitory effect on intracellular amastigotes of L. tropica strain. Further studies on luteolin, luteolin-4’-O-β-D-glucopyranoside and other isolated compounds are necessary to investigate their mechanisms of action, specificity and structure-activity relationships.

Declarations

Acknowledgement

The authors would like to thank Prof Dr Fazal Subhan of Department of Pharmacy, University of Peshawar, Peshawar, Pakistan for providing the albino mice and other technical support.

References

  1. Alvar J, Ve´lez ID, Bern C, Herrero M, Desjeux P, Cano J, Jannin J, Boer MD. Leishmaniasis Worldwide and Global Estimates of Its Incidence. PLoS ONE 2012; 7(5): e35671. [PubMed]
  2. Markle WH, Makboul K. Cutaneous leishmaniasis Recognition and Treatment. Am Fam Physician 2004; 69: 455-460
  3. Pearson RD, Sousa ADQ. Clinical spectrum of leishmaniasis. Clin Infect Dis 1996; 22 (1): 1-13.
  4. Brooker S, Mohammed N, Adil K, Agha S, Reithinger R, Rowland M, Ali I, Kolaczinski J. Leishmaniasis in refugee and local Pakistani populations. Emerg Infect Dis 2004; 10(9): 1681-1684.
  5. Rowland M, Munir A, Durrani N, Noyes H, Reyburn H. An outbreak of cutaneous leishmaniasis in an Afghan refugee settlement in north-west Pakistan. Trans Roy Soc Trop Med Hyg 1999; 93(2):133-136.
  6. Semwal RB, Semwal DK, Combrinck S, Cartwright-Jones C, Viljoen, A. Lawsonia Inermis L. (henna): Ethnobotanical, Phytochemical and pharmacological aspects. J Ethnopharmacol 2014; 155: 80-103. DOI:10.1016/j.jep.2014.05.042
  7. Sutthivaiyakit S, Unganont S, Sutthivaiyakit P, Suksamrarn A. Diterpenylated and prenylated flavonoids from Macaranga denticulata. Tetrahedron 2002; 58; 3619-3622. DOI:10.1016/S0040-4020(02)00296-X
  8. Baba K, Taniguchi M, Nakata K. Studies on Angelica keiskei "Ashitaba". Foods Food Ingred J Japan 1998; 178: 52-60.
  9. Fontenelle RO, Marais SM, Brito EH, Brilhante RS, Cordeiro RA, Lima YC, Brasil NV, Monteiro AJ, Sidrim JJ, Rocha MF. Alkylphenol activity against candida spp. Microsporum canis: A focus on the antifungal activity of thymol, eugenol and O-methyl derivatives. Molecules 2011; 16: 6422-6431.
  10. Cidade HM, Nacimento MS, Pinto MM, Kijjoa A, Silva AM, Herz W. Artelastocarpin and carpelastofuran, two new flavones and cytotoxicities of prenyl flavonoids from artocarpus elasticus against three cancer cell lines. Planta med 2001; 67 (9): 867-870.
  11. Meragelman KM, McKee TC, Boyd MR. Anti-HIV prenylated flavonoids from Monotes africanus. J Nat Prod 2001; 64(4): 546-548.
  12. Dufall KG, Ngadjui BT, Simeon KF, Abegaz BM, Croft KD. Antioxidant activity of prenylated flavonoids from the West African medicinal plant Dorstenia mannii. J Ehtnopharmacol 2003; 87(1): 67-72.
  13. Barron D, Balland C, Possety F, Ravanel P, Desfougeres A. Prenyl flavonoids and renyl flavonoids and memebrane-permeability, Acta botanica gallica 1996; 143(6): 509-520.
  14. Mahmoud ZF, Abdel Salam NA, Khafagy SM. Constituents of henna leaves (Lawsonia inermis L.) growing in Egypt. Fitoterapia 1980; 51: 153–155.
  15. Liou JR, ElShazly M, Du YC, Tseng CN, Hwang TL, Chuang YL, Hsu YM, Hsieh PW, Wu CC, Chen SL, et al., 1,5-Diphenylpent-3-en-1-ynes and methyl naphthalene carboxylates from Lawsonia inermis and their anti-inflammatory activity. Phytochem 2013; 88: 67–73. DOI:10.1016/j.phytochem.2012.11.010
  16. Afzal M, Al-Oriquat G, Al-Hassan JM. Flavone glycosides from Lawsonia inermis. Heterocycles 1980; 14: 1973–1976.
  17. Takeda Y, Fatope MO. New phenolic glucosides from Lawsonia inermis. J Nat Prod 1988; 51: 725–729.
  18. Hsouna AB, Trigui M, Culioli G, Blache Y, Jaoua S. Antioxidant’s constituents from Lawsonia Inermis leaves: Isolation, structure elucidation and antioxidative capacity. Food Chem 2011; 125: 193-200. DOI: 10.1016/j.foodchem.2010.08.060
  19. Cuong NX, Nhiem NX, Thao NP, Nam NH, Dat NT, Anh HLT, Huong LM, Kiem PV, Minh CV, Won J-H, et al, Inhibitors of osteoclastogenesis from Lawsonia inermis leaves. Bioorg Med Chem Lett 2010; 20: 4782–4784.
  20. National Research Council of The National Academy of Sciences. Guide for the Care and Use of Laboratory Animals: 8th edn. Washington, D.C.: The National Academies Press; 2010.
  21. Jamal Q, Khan NH, Wahid S, Awan MM, Sutherland C, Shah A. In-vitro sensitivity of Pakistani Leishmania tropica field isolate against buparvaquone in comparison to standard anti-leishmanial drugs. Exp Parasitol 2015; 154: 93-97.
  22. Iqbal K, Iqbal J, Umair M, Farooq U, Iqbal MM, Qamar S, Bashir M. Anti-leishmanial and cytotoxic activities of extracts from three Pakistani Plants. Trop J Pharm Res 2016; 15(10): 2113-2119.
  23. Iqbal K, Iqbal J, Afreen MS. Comparative Study on Antileishmanial and Cytotoxic activity    of Lawsonia Inermis bark and Aloe Vera leaves. Int J Biol Pharm Allied Sci 2016, 5(6), 1490-150.
  24. Lin JH, Lin YT, Huang YJ, Wen KC, Chen RM, Ueng TH, Liao CH. Isolation and cytotoxicity of flavonoids from Daphnis Genkwae Flos. J Food Drug Anal 2001; 9 (1): 6-11.
  25. Singh P, Mishra SK, Noel S, Sharma S, Rath SK. Acute Exposure of Apigenin Induces Hepatotoxicity in Swiss Mice. PLoS ONE 2012; 7(2): e31964. DOI:10.1371/journal.pone.0031964
  26. Lin AS, Chang FR, Wu CC, Liaw CC, Wu YC. New cytotoxic flavonoids from Thelypteris torresiana. Planta Med 2005; 71 (9): 867-870. DOI: 10.1055/s-2005-871292
  27. Mittra B, Saha A, Chowdhury AR, Pal C, Mandal S, Mukhopadhyay S, Bandyopadhyay S, Majumder HK. Luteolin, an abundant dietary component is a potent anti-leishmanial agent that acts by inducing topoisomerase II-mediated kinetoplast DNA cleavage leading to apoptosis. Mol Med 2000; 6: 527–541.
  28. Tasdemir D, Kaiser M, Brun R, Yardley V, Schmidt TJ, Tosun F, Rüedi P. Antitrypanosomal and Antileishmanial Activities of Flavonoids and Their Analogues: In Vitro, In Vivo, Structure-Activity Relationship, and Quantitative Structure-Activity Relationship Studies. Antimicrob Agents Chemother 2006; 50(4): 1352-1364.
  29. Salem MM, Werbovetz KA. Natural Products from Plants as Drug Candidates and Lead Compounds against Leishmaniasis and Trypanosomiasis. Curr Med Chem 2006; 13: 2571-2598.
  30. Prusky D. Keen NT. Involvement of preformed antifungal compounds in the resistance of subtropical fruits to fungal decay. Plant Dis 1993; 77: 114-119.
  31. Ravanel P, Creuzet S, Tissut M. Inhibitory effect of hydroxyflavones on the exogenous NADH dehydrogenase of plant mitochondrial inner membranes. Phytochem 1990; 29 (2): 441-445. DOI:10.1016/0031-9422(90)85093-U
Impact Factor
Thompson Reuters (ISI): 0.523 (2021)
H-5 index (Google Scholar): 39 (2021)

Article Tools

Share this article with



Article status: Free
Fulltext in PDF
Similar articles in Google
Similar article in this Journal:

Archives

2024; 23: 
1,   2,   3,   4
2023; 22: 
1,   2,   3,   4,   5,   6,   7,   8,   9,   10,   11,   12
2022; 21: 
1,   2,   3,   4,   5,   6,   7,   8,   9,   10,   11,   12
2021; 20: 
1,   2,   3,   4,   5,   6,   7,   8,   9,   10,   11,   12
2020; 19: 
1,   2,   3,   4,   5,   6,   7,   8,   9,   10,   11,   12
2019; 18: 
1,   2,   3,   4,   5,   6,   7,   8,   9,   10,   11,   12
2018; 17: 
1,   2,   3,   4,   5,   6,   7,   8,   9,   10,   11,   12
2017; 16: 
1,   2,   3,   4,   5,   6,   7,   8,   9,   10,   11,   12
2016; 15: 
1,   2,   3,   4,   5,   6,   7,   8,   9,   10,   11,   12
2015; 14: 
1,   2,   3,   4,   5,   6,   7,   8,   9,   10,   11,   12
2014; 13: 
1,   2,   3,   4,   5,   6,   7,   8,   9,   10,   11,   12
2013; 12: 
1,   2,   3,   4,   5,   6
2012; 11: 
1,   2,   3,   4,   5,   6
2011; 10: 
1,   2,   3,   4,   5,   6
2010; 9: 
1,   2,   3,   4,   5,   6
2009; 8: 
1,   2,   3,   4,   5,   6
2008; 7: 
1,   2,   3,   4
2007; 6: 
1,   2,   3,   4
2006; 5: 
1,   2
2005; 4: 
1,   2
2004; 3: 
1
2003; 2: 
1,   2
2002; 1: 
1,   2

News Updates