Tải bản đầy đủ (.pdf) (18 trang)

Đánh giá độc tính của các hợp chất phân tử nhỏ sử dụng hệ thống mô hình cá ngựa vằn

Bạn đang xem bản rút gọn của tài liệu. Xem và tải ngay bản đầy đủ của tài liệu tại đây (2.33 MB, 18 trang )

<span class='text_page_counter'>(1)</span><div class='page_container' data-page=1>

VIETNAM NATIONAL UNIVERSITY, HANOI



<b>INSTITUTE OF MICROBIOLOGY AND BIOTECHNOLOGY </b>


and



UNIVERSITY OF LIÈGE


<b>--- </b>



<b>Đinh Duy Thành </b>



<b>TOXICITY ASSESSMENT OF SMALL MOLECULES USING THE </b>


<b>ZEBRAFISH AS A MODEL SYSTEM </b>



Subject:

Biotechnology


Code:

60.42.02.01



MASTER’S THESIS


SUPERVISORS:


Prof. Marc Muller


Dr. Nguyễn Lai Thành



</div>
<span class='text_page_counter'>(2)</span><div class='page_container' data-page=2>

ACKNOWLEDGEMENT


<i><b>This thesis would not have been possible without all the support, </b></i>


<i><b>guidance, inspiration, and patience of the following people and </b></i>


<i><b>organisations during the course of my study. It is a privilege to </b></i>


<i><b>convey my gratefulness to them in my humble acknowledgements. </b></i>


<i><b>First and foremost, I own my deepest gratitude to Prof. Marc </b></i>


<i><b>Muller, who gave me the opportunity to pursue my own interests </b></i>


<i><b>as a trainee in the GIGA-Research. Your wisdom, guidance, </b></i>



<i><b>support, and endurance enable me to develop and improve my </b></i>


<i><b>expertise in both laboratory works and scientific writing. </b></i>


<i><b>Moreover, you did motivate me through my inner pressures as </b></i>


<i><b>well as outer obstacles. </b></i>



<i><b>I offer my thankfulness to my co-supervisor, Dr. Nguyễn Lai </b></i>


<i><b>Thành, for continuously encouraging me to explore my own ideas. </b></i>


<i><b>Your knowledge, gentleness, and trust have inspired me and other </b></i>


<i><b>students to keep following the scientific path. </b></i>



</div>
<span class='text_page_counter'>(3)</span><div class='page_container' data-page=3>

ii


<i><b>study with devoted professors and lectures within the course. </b></i>


<i><b>They not only gave me the knowledge but also a new vision to </b></i>


<i><b>perceive the Science of Life. </b></i>



<i><b>It is my great pleasure to thank Benoist, Yoann, and Audrey in </b></i>


<i><b>the Toxicology team as well as the Mullerians and members of </b></i>


<i><b>the BMGG: Thomas, Marie, David, and all others. Your supports </b></i>


<i><b>and helps during my stay in Liège crucially contributed to the </b></i>


<i><b>completion of my research. I would also like to express my </b></i>


<i><b>thanks to my friends and colleagues: Lung, Tuấn, An, Loan, and </b></i>


<i><b>others for their cares and encouragements in life and work. </b></i>



<i><b>My research trip was co-sponsored by the Wallonia-Brussels </b></i>


<i><b>International (WBI) and the Wallonia-Brussels delegation to </b></i>


<i><b>Vietnam. I would like to thank you for your commitment to </b></i>


<i><b>supporting scientific innovations as well as strengthening the </b></i>


<i><b>collaborations between the two laboratories and between our </b></i>


<i><b>countries. </b></i>




</div>
<span class='text_page_counter'>(4)</span><div class='page_container' data-page=4>

<b>TABLE OF CONTENTS</b>



<b>TABLE OF CONTENTS ... i</b>


<b>LIST OF TABLES AND FIGURES ... v</b>


<b>ABBREVIATIONS ... vii</b>


<b>PREFACE ... 1</b>


<b>Chapter 1: BACKGROUND INFORMATION ... 2</b>


<i><b>1.1. Small molecules: safety concerns ... 2 </b></i>


<i>1.1.1. Pharmaceuticals and personal care products (PPCPs) ... 3</i>


<i>1.1.2. Food additives ... 4</i>


<i>1.1.3. Household chemicals ... 5</i>


<i><b>1.2. The Zebrafish embryo toxicity test (ZET) ... 6 </b></i>


<b>Chapter 2: METHODS ... 11</b>


<i><b>2.1. Substances ... 11 </b></i>


<i><b>2.2. Zebrafish maintenance ... 12 </b></i>


<i><b>2.3. Chemical exposure and embryo observation ... 12 </b></i>



<i><b>2.4. Behavioural analysis ... 14 </b></i>


<i><b>2.5. Gene expression analysis ... 14 </b></i>


<i>2.5.1. Reverse transcription and quantitative polymerase chain reaction ... 14</i>


<i>2.5.2. Transgenic fluorescent lines ... 16</i>


<i><b>2.6. Statistical analysis ... 16 </b></i>


<i><b>2.7. Quality control ... 17 </b></i>


</div>
<span class='text_page_counter'>(5)</span><div class='page_container' data-page=5>

iv


<i><b>3.1. Morphological and lethal effects ... 18 </b></i>


<i><b>3.2. Locomotor defects ... 29 </b></i>


<i><b>3.3. Specific transgene expression in living embryos ... 33 </b></i>


<i><b>3.4. Reverse transcriptive – qPCR ... 38 </b></i>


<b>Chapter 4: CONCLUSIONS ... 41</b>


</div>
<span class='text_page_counter'>(6)</span><div class='page_container' data-page=6>

<b>LIST OF TABLES AND FIGURES </b>


<b>Tables</b>


<i><b>Table 2-1: List of studied chemicals ... 11</b></i>



<i><b>Table 2-2: Lethality endpoints ... 13</b></i>


<i><b>Table 2-3: Quantitative PCR primer set ... 15</b></i>


<i><b>Table 3-1: Concentration ranges selected for the main study... 18</b></i>


<i><b>Table 3-2: Lethal concentrations, effective concentrations, teratogenic indices, and </b></i>
<i><b>typical defects of studied substances ... 25</b></i>


<b>Figures</b>
<i><b>Figure 1.1: Orthologous genes shared among the zebrafish, human, mouse and chicken </b></i>
<i><b>genomes (reprinted from Howe et. al. [33]) ... 7</b></i>


<i><b>Figure 1.2: Literature analysis using the Scopus database in February 2014 ... 8</b></i>


<i><b>Figure 1.3: Comparisons between the ZET test and the classical acute fish toxicity test </b></i>
<i><b>(reprinted from Lammer et. al. [40]) ... 10</b></i>


<i><b>Figure 2.1: Normal morphological stages of zebrafish development at 28.5 </b></i><i><b>C (photos </b></i>
<i><b>excerpted from Kimmel et.al. [39]). Scale bars = 250 </b></i><i><b>M. ... 13</b></i>


<i><b>Figure 3.1: Morphological phenotypes in hatched zebrafish larvae ... 19</b></i>


<i><b>Figure 3.2: Concentration-response curves and frequency of typical phenotypes caused </b></i>
<i><b>by tested substances ... 22</b></i>


<i><b>Figure 3.3: LC50, EC50 Hill slope values of tested chemicals ... 27</b></i>


<i><b>Figure 3.4: Correlation between LC50s resulting from this study and those obtained </b></i>
<i><b>using the procedure described in the OECD 236 guideline [59] ... 28</b></i>



<i><b>Figure 3.5: Larval motion measurements during the dark/light cycles ... 30</b></i>


<i><b>Figure 3.6: Comparative analysis of larval activity ... 31</b></i>


</div>
<span class='text_page_counter'>(7)</span><div class='page_container' data-page=7>

vi


</div>
<span class='text_page_counter'>(8)</span><div class='page_container' data-page=8>

<b>ABBREVIATIONS </b>



DCA 3,4-Dichloroaniline


DMSO Dimethyl sulfoxide


dpf Day post fertilisation


EtOH Ethanol


hpf Hour post fertilisation


MSG Monosodium glutamate


OECD Organisation for Economic Co-operation and


Development


PPCPs Pharmaceuticals and Personal Care Products


qPCR Quantitative polymerase chain reaction


QY Quinoline yellow



SB Sodium Benzoate


TTZ Tartrazine


</div>
<span class='text_page_counter'>(9)</span><div class='page_container' data-page=9>

1


<b>PREFACE </b>



The human population are increasingly exposed to various chemicals whose
beneficial or deleterious properties often remain unexplored. The rising public
concern about hazardous substances existing in foods and consumer products has
forced legislators to tighten chemical management policy that requires extensive
toxicity testing. However, assessment of chemical toxicity is a challenging task,
especially in terms of reliability and efficiency. Ethical issues over the use of animal
testing also add further complication to the task.


The zebrafish (<i>Danio rerio</i>) embryo is an emerging model system for
chemical testing that is attracting scientific and legal attention. Its advantages
including rapid development, high availability, and easy observation have made the
model amenable to high-throughput assays. Moreover, as a complex and
independent organism retaining the “non-animal” status, the zebrafish embryo is the
ideal vertebrate testing model.


</div>
<span class='text_page_counter'>(10)</span><div class='page_container' data-page=10>

<b>Chapter 1: BACKGROUND INFORMATION </b>


<b>1.1. Small molecules: safety concerns </b>


Chemicals have become an integral part of modern daily life. They play an
important role in almost all industries and economic sectors. Consumer goods of our
everyday-use are either containing chemicals, or involving them during production.


Global chemical production has increased from 1 million tonnes in 1930 to 400
million tonnes in 2001 [25], with more than 143,000 substances in the European
market*. It is undeniable that these chemicals are progressively benefiting people’s
life and economy.


However, many chemicals are also posing potential deleterious effects on
human and environment health, especially those with small molecular size (<900
Daltons). Amongst the most well-known examples is the <i>thalidomide </i>scandal which
involved thousands of cases of stillborn and extreme congenital deformity [38], or
the carcinogenic <i>benzene</i> [73] which may have claimed thousands of deaths around
the world. Another case is <i>DDT</i>, the insecticide whose extensive use and high
accumulation have greatly threatened both wildlife species and human health [83].
A common theme in these three instances is that large-scale application of these
chemicals was conducted without having sufficient knowledge on their adverse
impacts, and measures to restrict the uses were taken too late to prevent irreversible
damages.


Ironically, despite efforts to achieve the world governments’ agreement to
use and produce chemicals “…in ways that do not lead to significant adverse effects
on human health and the environment…” by 2020 using scientific assessment
procedures [85], the number of compounds and the complexity of the issue lead to
the situation that unrecognised or unacknowledged toxic compounds in domestic




*


</div>
<span class='text_page_counter'>(11)</span><div class='page_container' data-page=11>

43


<b>REFERENCES </b>




1. Aboel-Zahab H., el-Khyat Z., Sidhom G., Awadallah R., Abdel-al W., and
Mahdy K. (1997), “Physiological effects of some synthetic food colouring
additives on rats”, <i>Bollettino chimico farmaceutico</i>, 136 (10), pp. 615-627.
2. Ali M.M., Bawari M., Misra U.K., and Babu G.N. (2000), “Locomotor and


learning deficits in adult rats exposed to monosodium-l-glutamate during
early life”, <i>Neuroscience Letters</i>, 284 (1–2), pp. 57-60.


3. Ali S., Champagne D.L., Alia A., and Richardson M.K. (2011), “Large-scale
analysis of acute ethanol exposure in zebrafish development: a critical time
window and resilience”, <i>PLoS One</i>, 6 (5), pp. e20037.


4. Ali S., Champagne D.L., Spaink H.P., and Richardson M.K. (2011),
“Zebrafish embryos and larvae: a new generation of disease models and drug
screens”, <i>Birth defects research. Part C, Embryo today: Reviews</i>, 93 (2), pp.
115-133.


5. Anastasaki C., Rauen K.A., and Patton E.E. (2012), “Continual low-level
MEK inhibition ameliorates cardio-facio-cutaneous phenotypes in
zebrafish”, <i>Disease models & mechanisms</i>, 5 (4), pp. 546-552.


6. Arkhipova V., Wendik B., Devos N., Ek O., Peers B., and Meyer D. (2012),
“Characterization and regulation of the hb9/mnx1 beta-cell progenitor
specific enhancer in zebrafish”, <i>Developmental biology</i>, 365 (1), pp.
290-302.


7. Balbi H.J. (2004), “Chloramphenicol: A review”, <i>Pediatrics in Review</i>, 25
(8), pp. 284-288.



8. Ballentine C. (1981), “Taste of raspberries, taste of death: The 1937 elixir
sulfanilamide incident”, <i>FDA Consumer magazine</i>, 15 (5).


9. Bilotta J., Barnett J.A., Hancock L., and Saszik S. (2004), “Ethanol exposure
alters zebrafish development: A novel model of fetal alcohol syndrome”,


<i>Neurotoxicology and teratology</i>, 26 (6), pp. 737-743.


10. Boris M. and Mandel F.S. (1994), “Foods and additives are common causes
of the attention deficit hyperactive disorder in children”, <i>Annals of allergy</i>,
72 (5), pp. 462-468.


11. Boxall A.B., Rudd M.A., et al. (2012), “Pharmaceuticals and personal care
products in the environment: what are the big questions?”, <i>Environmental </i>
<i>health perspectives</i>, 120 (9), pp. 1221-1229.


12. Brown W., Buist N.M., Cory Gipson H., Huston R., and Kennaway N.
(1982), “Fatal benzyl alcohol poisoning in a neonatal intensive care unit”,


</div>
<span class='text_page_counter'>(12)</span><div class='page_container' data-page=12>

13. Cavaletti G., Oggioni N., et al. (2000), “Effect on the peripheral nervous
system of systemically administered dimethylsulfoxide in the rat: a
neurophysiological and pathological study”, <i>Toxicology Letters</i>, 118 (1–2),
pp. 103-107.


14. Chen Q., Huang N.N., et al. (2009), “Sodium benzoate exposure
downregulates the expression of tyrosine hydroxylase and dopamine
transporter in dopaminergic neurons in developing zebrafish”, <i>Birth defects </i>
<i>research. Part B, Developmental and reproductive toxicology</i>, 86 (2), pp.
85-91.



15. Chen T.H., Wang Y.H., and Wu Y.H. (2011), “Developmental exposures to
ethanol or dimethylsulfoxide at low concentrations alter locomotor activity in
larval zebrafish: implications for behavioral toxicity bioassays”, <i>Aquatic </i>
<i>toxicology</i>, 102 (3-4), pp. 162-166.


16. Committee on Toxicity Testing and Assessment of Environmental Agents
(2007), <i>Toxicity testing in the 21st Century: A vision and a strategy</i>, The
National Academies Press.


17. Criep L.H. (1971), “Allergic vascular purpura”, <i>Journal of Allergy and </i>
<i>Clinical Immunology</i>, 48 (1), pp. 7-12.


18. Dann A.B. and Hontela A. (2011), “Triclosan: environmental exposure,
toxicity and mechanisms of action”, <i>Journal of applied toxicology : JAT</i>, 31
(4), pp. 285-311.


19. Daughton C.G. and Ternes T.A. (1999), “Pharmaceuticals and personal care
products in the environment: Agents of subtle change?”, <i>Environmental </i>
<i>health perspectives</i>, 107 Suppl 6, pp. 907-938.


20. de Jong E., Barenys M., et al. (2011), “Comparison of the mouse Embryonic
Stem cell Test, the rat Whole Embryo Culture and the Zebrafish
Embryotoxicity Test as alternative methods for developmental toxicity
testing of six 1,2,4-triazoles”, <i>Toxicology and applied pharmacology</i>, 253
(2), pp. 103-111.


21. Dipalma J.R. (1990), “Tartrazine sensitivity”, <i>American family physician</i>, 42
(5), pp. 1347-1350.


22. Dodson R.E., Nishioka M., Standley L.J., Perovich L.J., Brody J.G., and


Rudel R.A. (2012), “Endocrine disruptors and asthma-associated chemicals
in consumer products”, <i>Environmental health perspectives</i>, 120 (7), pp.
935-943.


</div>
<span class='text_page_counter'>(13)</span><div class='page_container' data-page=13>

45


24. Emran F., Rihel J., and Dowling J.E. (2008), “A behavioral assay to measure
responsiveness of zebrafish to changes in light intensities”, <i>Journal of </i>
<i>visualized experiments : JoVE</i>, (20), pp. e923.


25. European Commission (2001), <i>White Paper, Strategy for a Future Chemicals </i>
<i>Policy</i>, Office for Official Publications of the European Communities.


26. European Council (1989), <i>Directive 89/107/EEC of 21 December 1988 on </i>
<i>the approximation of the laws of the Member States concerning food </i>
<i>additives authorized for use in foodstuffs intended for human consumption</i>.
27. European Council (1992), <i>Directive 92/32/EEC of 30 April 1992 amending </i>


<i>for the seventh time Directive 67/548/EEC on the approximation of the laws, </i>
<i>regulations and administrative provisions relating to the classification, </i>
<i>packaging and labelling of dangerous substances</i>.


28. Gaj T., Gersbach C.A., and Barbas C.F., 3rd (2013), “ZFN, TALEN, and
CRISPR/Cas-based methods for genome engineering”, <i>Trends in </i>
<i>biotechnology</i>, 31 (7), pp. 397-405.


29. Gultekin F. and Doguc D.K. (2013), “Allergic and immunologic reactions to
food additives”, <i>Clinical reviews in allergy & immunology</i>, 45 (1), pp. 6-29.
30. Gỹngửrmỹ C. and Klỗ A. (2012), “The safety assessment of food additives



by reproductive and developmental toxicity studies”, in <i>Food Additive</i>,
InTech, pp. 31-48.


31. Hill A., Mesens N., Steemans M., Xu J.J., and Aleo M.D. (2012),
“Comparisons between in vitro whole cell imaging and in vivo
zebrafish-based approaches for identifying potential human hepatotoxicants earlier in
pharmaceutical development”, <i>Drug metabolism reviews</i>, 44 (1), pp.
127-140.


32. Hill A.J., Teraoka H., Heideman W., and Peterson R.E. (2005), “Zebrafish as
a model vertebrate for investigating chemical toxicity”, <i>Toxicological </i>
<i>sciences</i>, 86 (1), pp. 6-19.


33. Howe K., Clark M.D., et al. (2013), “The zebrafish reference genome
sequence and its relationship to the human genome”, <i>Nature</i>, 496 (7446), pp.
498-503.


34. International Organization for Standardization, "ISO 15088:2007 Water
quality-Determination of the Acute toxicity of Waste water to Zebrafish eggs
(Danio rerio)", in <i>ISO/TC 147 - Water quality</i>.


35. Irons T.D., MacPhail R.C., Hunter D.L., and Padilla S. (2010), “Acute
neuroactive drug exposures alter locomotor activity in larval zebrafish”,


</div>
<span class='text_page_counter'>(14)</span><div class='page_container' data-page=14>

36. Isogai S., Horiguchi M., and Weinstein B.M. (2001), “The vascular anatomy
of the developing zebrafish: an atlas of embryonic and early larval
development”, <i>Developmental biology</i>, 230 (2), pp. 278-301.


37. Kanungo J., Lantz S., and Paule M.G. (2011), “In vivo imaging and
quantitative analysis of changes in axon length using transgenic zebrafish


embryos”, <i>Neurotoxicology and teratology</i>, 33 (6), pp. 618-623.


38. Kim J.H. and Scialli A.R. (2011), “Thalidomide: the tragedy of birth defects
and the effective treatment of disease”, <i>Toxicological sciences</i>, 122 (1), pp.
1-6.


39. Kimmel C.B., Ballard W.W., Kimmel S.R., Ullmann B., and Schilling T.F.
(1995), “Stages of embryonic development of the zebrafish”, <i>Developmental </i>
<i>Dynamics</i>, 203 (3), pp. 253-310.


40. Lammer E., Carr G.J., Wendler K., Rawlings J.M., Belanger S.E., and
Braunbeck T. (2009), “Is the fish embryo toxicity test (FET) with the
zebrafish (Danio rerio) a potential alternative for the fish acute toxicity
test?”, <i>Comparative biochemistry and physiology. Toxicology </i> <i>& </i>
<i>pharmacology : CBP</i>, 149 (2), pp. 196-209.


41. Lammer E., Kamp H.G., et al. (2009), “Development of a flow-through
system for the fish embryo toxicity test (FET) with the zebrafish (Danio
rerio)”, <i>Toxicology in vitro</i>, 23 (7), pp. 1436-1442.


42. Lange F.T., Scheurer M., and Brauch H.J. (2012), “Artificial sweeteners--a
recently recognized class of emerging environmental contaminants: a
review”, <i>Analytical and bioanalytical chemistry</i>, 403 (9), pp. 2503-2518.
43. Lau K., McLean W.G., Williams D.P., and Howard C.V. (2006),


“Synergistic interactions between commonly used food additives in a
developmental neurotoxicity test”, <i>Toxicological sciences</i>, 90 (1), pp.
178-187.


44. Lawson N.D. and Weinstein B.M. (2002), “In vivo imaging of embryonic


vascular development using transgenic zebrafish”, <i>Developmental Biology</i>,
248 (2), pp. 307-318.


45. Lessman C.A. (2011), “The developing zebrafish (Danio rerio): a vertebrate
model for high-throughput screening of chemical libraries”, <i>Birth defects </i>
<i>research. Part C, Embryo today: Reviews</i>, 93 (3), pp. 268-280.


46. Lieschke G.J. and Currie P.D. (2007), “Animal models of human disease:
Zebrafish swim into view”, <i>Nature reviews Genetics</i>, 8 (5), pp. 353-367.
47. Lu X., Le Noble F., et al. (2004), “The netrin receptor UNC5B mediates


</div>
<span class='text_page_counter'>(15)</span><div class='page_container' data-page=15>

47


48. MacPhail R.C., Brooks J., Hunter D.L., Padnos B., Irons T.D., and Padilla S.
(2009), “Locomotion in larval zebrafish: Influence of time of day, lighting
and ethanol”, <i>Neurotoxicology</i>, 30 (1), pp. 52-58.


49. Maiolini E., Ferri E., et al. (2014), “Bisphenol A determination in baby
bottles by chemiluminescence enzyme-linked immunosorbent assay, lateral
flow immunoassay and liquid chromatography tandem mass spectrometry”,


<i>The Analyst</i>, 139 (1), pp. 318-324.


50. Marza E., Barthe C., Andre M., Villeneuve L., Helou C., and Babin P.J.
(2005), “Developmental expression and nutritional regulation of a zebrafish
gene homologous to mammalian microsomal triglyceride transfer protein
large subunit”, <i>Developmental dynamics</i>, 232 (2), pp. 506-518.


51. McCann D., Barrett A., et al. (2007), “Food additives and hyperactive
behaviour in 3-year-old and 8/9-year-old children in the community: A


randomised, double-blinded, placebo-controlled trial”, <i>The Lancet</i>, 370
(9598), pp. 1560-1567.


52. McCurley A.T. and Callard G.V. (2008), “Characterization of housekeeping
genes in zebrafish: male-female differences and effects of tissue type,
developmental stage and chemical treatment”, <i>BMC Molecular Biology</i>, 9,
pp. 102.


53. Miller K. (1982), “Sensitivity to tartrazine”, <i>British medical journal (Clinical </i>
<i>research ed.)</i>, 285 (6355), pp. 1597-1598.


54. Murray K.E., Thomas S.M., and Bodour A.A. (2010), “Prioritizing research
for trace pollutants and emerging contaminants in the freshwater
environment”, <i>Environmental pollution</i>, 158 (12), pp. 3462-3471.


55. Muth-Kohne E., Wichmann A., Delov V., and Fenske M. (2012), “The
classification of motor neuron defects in the zebrafish embryo toxicity test
(ZFET) as an animal alternative approach to assess developmental
neurotoxicity”, <i>Neurotoxicology and teratology</i>, 34 (4), pp. 413-424.


56. OECD (2002), <i>Harmonised Integrated Classification System for Human </i>
<i>Health and Environmental Hazards of Chemical Substances and Mixtures</i>,
OECD Series on Testing and Assessment No. 33, OECD Publishing.


57. OECD (2005), <i>Guidance Document on the Validation and International </i>
<i>Acceptance of New or Updated Test Methods for Hazard Assessment</i>, OECD
Series on Testing and Assessment No. 34, OECD Publishing.


58. OECD (2012), <i>Validation Report (Phase 2) for the Zebrafish Embryo </i>
<i>Toxicity Test: Part I and Part II (Annexes).</i> OECD Series on Testing and


Assessment No. 179, OECD Publishing.


</div>
<span class='text_page_counter'>(16)</span><div class='page_container' data-page=16>

60. Onesios K.M., Yu J.T., and Bouwer E.J. (2009), “Biodegradation and
removal of pharmaceuticals and personal care products in treatment systems:
a review”, <i>Biodegradation</i>, 20 (4), pp. 441-466.


61. Parodi G., Parodi A., and Rebora A. (1985), “Purpuric Vasculitis due to
Tartrazine”, <i>Dermatology</i>, 171 (1), pp. 62-63.


62. Peterson R.T. and Fishman M.C. (2011), “Designing zebrafish chemical
screens”, <i>Methods in cell biology</i>, 105, pp. 525-541.


63. Peterson R.T. and Macrae C.A. (2012), “Systematic approaches to
toxicology in the zebrafish”, <i>Annual review of pharmacology and toxicology</i>,
52, pp. 433-453.


64. Pfaffl M.W. (2001), “A new mathematical model for relative quantification
in real-time RT–PCR”, <i>Nucleic Acids Research</i>, 29 (9), pp. e45.


65. Pichler F.B., Laurenson S., Williams L.C., Dodd A., Copp B.R., and Love
D.R. (2003), “Chemical discovery and global gene expression analysis in
zebrafish”, <i>Nature biotechnology</i>, 21 (8), pp. 879-883.


66. Pruvot B., Jacquel A., et al. (2011), “Leukemic cell xenograft in zebrafish
embryo for investigating drug efficacy”, <i>Haematologica</i>, 96 (4), pp.
612-616.


67. Pruvot B., Quiroz Y., et al. (2012), “Zebrafish (Danio rerio) behavioral
analysis: A new tool in toxicological assays”, <i>Toxicology Letters</i>, 211,
Supplement (0), pp. S153.



68. Pruvot B., Quiroz Y., et al. (2012), “A panel of biological tests reveals
developmental effects of pharmaceutical pollutants on late stage zebrafish
embryos”, <i>Reproductive toxicology</i>, 34 (4), pp. 568-583.


69. Quirce S. and Barranco P. (2010), “Cleaning agents and asthma”, <i>Journal of </i>
<i>investigational allergology & clinical immunology</i>, 20 (7), pp. 542-550.
70. Quiroz Y., Lopez M., et al. (2012), “The HMG-box transcription factor


Sox4b is required for pituitary expression of gata2a and specification of
thyrotrope and gonadotrope cells in zebrafish”, <i>Molecular endocrinology</i>, 26
(6), pp. 1014-1027.


71. Raldúa D., Barata C., et al. (2012), “Zebrafish as a vertebrate model to assess
sublethal effects and health risks of emerging pollutants”, in <i>Emerging </i>
<i>Organic Contaminants and Human Health</i>, Springer Berlin Heidelberg, pp.
395-414.


</div>
<span class='text_page_counter'>(17)</span><div class='page_container' data-page=17>

49


73. Rinsky R.A., Smith A.B., et al. (1989), “Benzene and leukemia: An
epidemiologic risk assessment”, <i>New England Journal of Medicine</i>, 316
(17), pp. 1044-1050.


74. Rubinstein A.L. (2006), “Zebrafish assays for drug toxicity screening”,


<i>Expert Opinion on Drug Metabolism & Toxicology</i>, 2 (2), pp. 231-240.
75. Scheil V., Kienle C., Osterauer R., Gerhardt A., and Kohler H.R. (2009),


“Effects of 3,4-dichloroaniline and diazinon on different biological


organisation levels of zebrafish (Danio rerio) embryos and larvae”,


<i>Ecotoxicology</i>, 18 (3), pp. 355-363.


76. Schena M., Shalon D., Davis R.W., and Brown P.O. (1995), “Quantitative
monitoring of gene expression patterns with a complementary DNA
microarray”, <i>Science</i>, 270 (5235), pp. 467-470.


77. Simon R.A. (2003), “Adverse reactions to food additives”, <i>Current Allergy </i>
<i>and Asthma Reports</i>, 3 (1), pp. 62-66.


78. Sipes N.S., Padilla S., and Knudsen T.B. (2011), “Zebrafish-As an
integrative model for twenty-first century toxicity testing”, <i>Birth defects </i>
<i>research. Part C, Embryo today: Reviews</i>, 93 (3), pp. 256-267.


79. Sylvain N.J., Brewster D.L., and Ali D.W. (2010), “Zebrafish embryos
exposed to alcohol undergo abnormal development of motor neurons and
muscle fibers”, <i>Neurotoxicology and Teratology</i>, 32 (4), pp. 472-480.


80. Tan J.L. and Zon L.I. (2011), “Chemical screening in zebrafish for novel
biological and therapeutic discovery”, <i>Methods in cell biology</i>, 105, pp.
493-516.


81. Tang R., Dodd A., Lai D., McNabb W.C., and Love D.R. (2007),
“Validation of zebrafish (Danio rerio) reference genes for quantitative
real-time RT-PCR normalization”, <i>Acta Biochimica et Biophysica Sinica</i>, 39 (5),
pp. 384-390.


82. Tsay H.J., Wang Y.H., Chen W.L., Huang M.Y., and Chen Y.H. (2007),
“Treatment with sodium benzoate leads to malformation of zebrafish larvae”,



<i>Neurotoxicology and teratology</i>, 29 (5), pp. 562-569.


83. Turusov V., Rakitsky V., and Tomatis L. (2002),


“Dichlorodiphenyltrichloroethane (DDT): Ubiquity, Persistence, and Risks”,


<i>Environmental Health Perspectives</i>, 110 (2), pp. 125-128.


84. Voelker D., Vess C., et al. (2007), “Differential gene expression as a
toxicant-sensitive endpoint in zebrafish embryos and larvae”, <i>Aquatic </i>
<i>toxicology</i>, 81 (4), pp. 355-364.


</div>
<span class='text_page_counter'>(18)</span><div class='page_container' data-page=18>

86. Voncken A., Piot A., et al. (2010), "Zebrafish as model in
toxicology/pharmacology", in <i>Biomedica 2010</i>, Aachen, Germany.


87. Wallace K.N. and Pack M. (2003), “Unique and conserved aspects of gut
development in zebrafish”, <i>Developmental Biology</i>, 255 (1), pp. 12-29.
88. Westerhoff P., Yoon Y., Snyder S., and Wert E. (2005), “Fate of


endocrine-disruptor, pharmaceutical, and personal care product chemicals during
simulated drinking water treatment processes”, <i>Environmental Science & </i>
<i>Technology</i>, 39 (17), pp. 6649-6663.


89. Wittassek M., Koch H.M., Angerer J., and Bruning T. (2011), “Assessing
exposure to phthalates - the human biomonitoring approach”, <i>Molecular </i>
<i>nutrition & food research</i>, 55 (1), pp. 7-31.


90. Yu L., Zhang Y., Ma R., Bao L., Fang J., and Yu T. (2006), “Potent
protection of ferulic acid against excitotoxic effects of maternal intragastric


administration of monosodium glutamate at a late stage of pregnancy on
developing mouse fetal brain”, <i>European Neuropsychopharmacology</i>, 16 (3),
pp. 170-177.


91. Zhang X. and Gong Z. (2013), “Fluorescent transgenic zebrafish
Tg(nkx2.2a:mEGFP) provides a highly sensitive monitoring tool for
neurotoxins”, <i>PLoS One</i>, 8 (2), pp. e55474.


</div>

<!--links-->
NGHIÊN CỨU PHÂN LẬP, XÁC ĐỊNH CẤU TRÚC VÀ KHẢO SÁT HOẠT TÍNH SINH HỌC CỦA CÁC HỢP CHẤT PHENOLIC TỪ CÂY ALPINIA PINNANENSIS T. L. WU ET SENJEN (ZINGIBERACEAE)
  • 80
  • 891
  • 4
  • ×