Download UNIVERSITY OF DUBLIN TRINITY COLLEGE DUBLIN SCHOOL OF BIOCHEMISTRY &

Document related concepts

Drosophila melanogaster wikipedia , lookup

Lymphopoiesis wikipedia , lookup

T cell wikipedia , lookup

Phagocyte wikipedia , lookup

DNA vaccination wikipedia , lookup

Immune system wikipedia , lookup

Adaptive immune system wikipedia , lookup

Polyclonal B cell response wikipedia , lookup

Hygiene hypothesis wikipedia , lookup

Molecular mimicry wikipedia , lookup

Adoptive cell transfer wikipedia , lookup

Cancer immunotherapy wikipedia , lookup

Immunosuppressive drug wikipedia , lookup

Innate immune system wikipedia , lookup

Psychoneuroimmunology wikipedia , lookup

Immunomics wikipedia , lookup

Transcript
UNIVERSITY OF DUBLIN
TRINITY COLLEGE DUBLIN
SCHOOL OF BIOCHEMISTRY &
IMMUNOLOGY
SENIOR SOPHISTER
IMMUNOLOGY
COURSE DETAILS
2015-2016
1
This course is partially funded by the Irish government under the Human
Capital Investment Operational Programme 2007-2013 and aided by the
European Social Fund (ESF) under the 2007-2013 Community Support Framework
(CSF)
2
IMMUNOLOGY SENIOR SOPHISTER MODULES (60 ECTS)
BI4275 CELLS OF THE INNATE AND ADAPTIVE IMMUNE SYSTEM (5ECTS)
This module outlines the differentiation and roles of specific leukocyte populations in
mediating innate and adaptive immune responses
BI4055 MICROBIAL DISEASES’ (5 ECTS)
Protozoan, bacterial and viral diseases will be covered in detail in this module. BI4215 ORGAN-SPECIFIC IMMUNITY (5 ECTS)
This module describes characteristic and distinctive features of the innate and adaptive
immune responses at mucosal sites, the liver and the central nervous system.
BI4045 AUTOIMMUNE AND INFLAMMATORY CONDITIONS (5 ECTS)
This module covers basic and clinical aspects of autoinflammatory and autoimmune
conditions, including rheumatoid arthritis and multiple sclerosis and immunodeficiency
syndromes.
BI4235 IMMUNE SIGNALLING (5 ECTS).
This module covers immune-related signalling events including cell signalling in apoptotic cell
death, cytokine signalling, the molecular basis for immune signalling, PI3 kinase and MTOR
and T cell receptor signalling
BI4245 IMMUNITY TO PATHOGENS AND VACCINATION (5 ECTS)
This module covers immune responses to viruses and bacteria including tuberculosis and
Bordetella pertussis, vaccines and the danger theory
BI4255 IMMUNOTHERAPY, IMMUNOGENETICS AND IMMUNOMODULATION (5 ECTS)
This module covers key concepts and specific examples in immunogenetics, immunotherapy
and immunomodulation. It also covers transplantation, graft rejection and immunosuppressive
therapies.
BI4265 TUMOUR IMMUNOLOGY (5 ECTS)
This module addresses cancer, invasion and tumour immunity including stem cell therapy,
immunotherapy, vaccines and the immune response to tumours.
BI4295 RESEARCH PROJECT IN IMMUNOLOGY (15 ECTS)
The module comprises of an original research project in biochemistry and a research
thesis.
BI4015 DATA HANDLING (5 ECTS)
This module covers quantitative biochemical problems, bioinformatics and molecular
modelling.
NOTE: Learning outcomes for each of the modules can be found on the
School homepage: http://www.tcd.ie/Biochemistry/courses/senior_soph.php
3
Explanation of ECTS
The European Credit Transfer and Accumulation System (ECTS) is an academic
credit system based on the estimated student workload required to achieve the
objectives of a module or programme of study. It is designed to enable academic
recognition for periods of study, to facilitate student mobility and credit accumulation
and transfer. The ECTS is the recommended credit system for higher education in
Ireland and across the European Higher Education Area.
The ECTS weighting for a module is a measure of the student input or workload
required for that module, based on factors such as the number of contact hours, the
number and length of written or verbally presented assessment exercises, class
preparation and private study time, laboratory classes, examinations, clinical
attendance, professional training placements, and so on as appropriate. There is no
intrinsic relationship between the credit volume of a module and its level of difficulty.
The European norm for full-time study over one academic year is 60 credits.
The Trinity academic year is 40 weeks from the start of Michaelmas Term to the end
of the annual examination period. 1 ECTS credit represents 20-25 hours estimated
student input, so a 10-credit module will be designed to require 200-250 hours of
student input including class contact time and assessments.
ECTS credits are awarded to a student only upon successful completion of the
course year. Progression from one year to the next is determined by the course
regulations. Students who fail a year of their course will not obtain credit for that
year even if they have passed certain component courses. Exceptions to this rule are
one-year and part-year visiting students, who are awarded credit for individual
modules successfully completed.
For additional details see: http://www.tcd.ie/vp-cao/bd/vpbd3college_ects.php
Examinations and Breakdown of Marks:
Senior Sophister Module Name
ECTS Weighting
1) Cells of the innate and adaptive immune system
2) Microbial diseases
3) Organ-specific immunity
4) Autoimmune and inflammatory conditions
5) Immune Signalling
6) Immunity to pathogens and vaccination
7) Immunotherapy, immunogenetics, immunomodulation
8) Tumour immunology
9) Research Project in Immunology
10) Data handling
BI4275
BI4055
BI4215
BI4045
BI4235
BI4245
BI4255
BI4265
BI4295
BI4015
5 ECTS
5 ECTS
5 ECTS
5 ECTS
5 ECTS
5 ECTS
5 ECTS
5 ECTS
15 ECTS
5 ECTS
4
The Senior Sophister year is broken down into a total of 60 marks.
Annual Examination Papers. Value: 40 marks or 66.66% of SS year
There are four exam papers at the end of the SS year, each with equal weighting
(total 40 marks or 66.66% of SS year) as follows:
Paper 1
Value: 10 marks or 16.66% of SS year
Specific questions related to modules 1-4 above. Exam paper divided up into 4
sections (one module per section). Answer 4 questions, one from each section.
Paper 2
Value: 10 marks or 16.66% of SS year
Specific questions related to modules 5-8 above. Exam paper divided up into 4
sections (one module per section). Answer 4 questions, one from each section.
Papers 3 & 4 are indirectly linked to modules listed above, being general in nature.
Paper 3
Value: 10 marks or 16.66% of SS year
Exam paper divided up into two sections. Answer 4 questions at least one from each
section. The quantitative problems given in Section A will be related to those
practiced in the Junior Sophister AND Senior Sophister year.
Paper 4
Value: 10 marks or 16.66% of SS year
Exam paper divided up into two sections. Section A- question 1: short questions,
answer 10 out of 18 (compulsory question) -worth 50% of paper. Section B-answer 2
questions -worth 50% of paper
All answers from the above exam papers are double-marked.
Research Project in Immunology
11-week research project and thesis.
Value: 15 marks or 25% of SS year.
Data Handling Value: 5 marks or 8.34% of SS year.
This module covers quantitative biochemical problems, bioinformatics and molecular
modelling, animal handling and a presentation by the student on a biochemical
technique. Marks are awarded through continual assessment and in-course exams as
follows:
Quantitative biochemical problem topics:
3 sequence analysis exercises:
1 animal handling in-course exam:
1 biochemical techniques presentation:
2.5 marks
1.25 marks
0.625 mark
0.625 mark
__________
5 marks
The overall degree mark is comprised of 80% of SS year and 20% of JS year.
5
On completion of their annual examinations, selected students sit a viva voce
examination with the External Examiner. Students are considered
‘borderline’ if they are 2.5% or less off a grade and following the viva voce
examination, the External Examiner may recommend at the Examiners’
meeting that the students’ degree mark be brought up to the next grade.
Tutorials:
Tutors have been chosen randomly (see page 16). Please contact your tutor
during the first week of the first semester. You are expected to attend a
tutorial every fortnight. Times and dates of tutorials given on timetable are a rough
guide only. Your tutor will set various exercises and these should help you in your
final examinations.
Practise Vivas:
Vivas (oral exams) are held approximately two weeks following the completion of your
four exam papers. The maximum percentage marks that you can be brought up by is
2.5%. You cannot be marked down by a viva. You will not know your mark before
sitting the viva.
How can you prepare for the viva?
Practise vivas will be held once in the first semester and once in the second semester.
You will be assigned to a pair of academic staff members in the first semester and a
different pair of staff in the second semester. The vivas take approximately 20
minutes and you will be asked a variety of questions. There are no marks going for
this. Please regard these vivas not as a test of your knowledge but as useful practise.
They are also good interview experience!
When you are called for a viva in the summer, you should read over your project
thesis as the Extern often starts off by asking you about your project. They will want
to relax you and will generally start you off on a topic you know a lot about. The
Extern will probably cover about 4-6 topics during the viva and it is impossible to
second guess what they will ask. However, if you feel you did badly in one particular
exam question, it is a good idea to revise this topic. The Extern has access to all your
marks and if he/she sees a blip in an otherwise very consistent set of marks they may
wish to follow this up. The Extern may also ask you if there is a topic in Biochemistry
that you find particularly interesting and that you wish to talk about. It is therefore a
good idea to have something prepared but ensure that it is a specific topic. Do not be
too general and say that you’re interested in protein structure! The Extern may also
ask you on your views of the course; was there a part of the course you really
enjoyed or not as the case may be. The role of the Extern is not only to assess your
performance but also to assess our teaching capabilities and to identify
strengths/weaknesses and even omissions in the course so that they can make
recommendations for the following year.
Quantitative Problem Sessions:
All Quantitative Problems will be given out at introductory sessions by different staff
members (e.g. Prb 1 Intro on the timetable), at the times indicated. You will attempt
the problem in your own time and must bring the problem with you to a timetabled
6
tutorial session (e.g. Prb 1 Tutorial). You must be able to demonstrate that you have
attempted the problem to the staff member and failure to do so will result in being
returned as non-satisfactory. In the tutorial session, the staff member will go through
the solution with you.
Later on that same week you will sit an exam on this particular problem topic (e.g.
Prb 1 Exam). The mark for each problem topic will comprise of the in-course exam
mark.
Sequence Analysis Sessions:
There will be three three-hour Sequence Analysis Sessions (Dr Jerrard Hayes). Each
session will begin with a brief introduction in FRED (2pm). You will then move to the
East End Public Access Mac Room for the practical session. Submission dates for the
Sequence Analysis Exercises are indicated on the timetable. Dr Hayes will advise you
as to how and where you submit the exercises.
Course Feedback:
A Feedback Form for each course will be given out at the beginning of the term.
These (anonymous) forms are a mechanism whereby students can make comments
and suggestions that will help us to maintain and indeed improve the quality of the
teaching offered by the School of Biochemistry & Immunology. Please fill out the form
upon completing each course, do not wait until the end of term (you will forget!). Put
the forms into the box provided in the secretary’s office.
Addresses and Phone No's:
Please enter your College based address, e-mail address and telephone number (if
any) on the sheet provided at the Introductory Lecture. Please also include a home
(or other contact) address and telephone number. This will enable us to contact you
in an emergency or with important changes in such details as timetables, exam
venues, etc. If you do not enter these details you may not be informed of any
changes.
Careers Talk:
Sean Gannon will give a Careers Talk tailored for Immunology students, on Tuesday
6th October at 11 am in FRED.
Research Projects:
At the start of first semester you will be given Project Summaries of all available
projects. You then have two weeks to select your first choice for a project. Feel free to
discuss these projects with Staff. You may not select a project offered by a Staff
Member with whom you have done a Summer Project. Projects will then be
allocated by the following procedure: any project with only one taker will be allocated
to that student; any projects with more than one taker will be allocated by drawing
one name out of a hat; losers and unallocated projects will then be placed into a
second round; the allocation procedure will again take place, and will continue until all
students and projects are exhausted. In this manner it is hoped than an element of
7
choice may operate in selection of projects. You may then, if you wish, change
projects by mutual exchange between students. However, in the event you do not
gain your project of choice no additional projects will be set.
Once projects are selected you should then contact your project supervisor for
discussions.
Most importantly, you should check that essential chemicals and
equipment are, or will be, available when required.
Before projects begin, you will be expected to give a 15-20 min talk that explains your
project, its intended aims and your experimental approach (week beginning 2th
November in FRED). It is advisable to arrange at least one practise talk with your
project supervisor.
Project laboratory work will start on November 16th and terminate on the 26th
of February. After the end of laboratory work, you will be given approximately 4
weeks to present a thesis on your project. In this period you will also present a
Project Poster to the School at a poster session. There will be prizes for the best
posters. A deadline for handing in projects will operate. It is 5.00 pm on Thursday
the 24th of March. For every working day that your thesis is late, 2% of the mark
that you would have obtained will be subtracted from your mark.
Ms Roisin Cleere and Dr Audrey Carroll (Preparation Room) will advise you about the
presentation of your poster and print it for you. Further details on Project write-ups
and poster presentations will be given in the second semester.
The Margaret Ciotti Prize Merit Award is awarded to a Senior Sophister student for
excellence in undergraduate research. This award was initiated by Bruno Orsi to
honour his wife's achievements in biochemistry and will now be a memorial to her. It
is traditionally presented by Bruno on a date between the end of the exams and the
vivas. You will be advised of the date closer to the time. This award is independent of
the poster prizes.
Biochemistry Personnel and Contact Details:
The Senior Sophister Course Co-ordinator is Ed Lavelle (phone extension 2488,
email ([email protected]). The names of tutors are provided on page 17. A complete
list of staff in the school can be found at:
http://www.tcd.ie/Biochemistry/people/index.php
Health and Safety Management:
1) Registration with Safety Officer
Preliminary safety registration takes place during two mandatory health and safety
briefing sessions timetabled in Week 1 (see timetable). Later on you must register, in
person, with the Safety Officer during the week after you have been assigned your
project. This is necessary in order to record your next-of-kin details in the unlikely
8
event of an accident, to record where you will be working, to ascertain whether or not
you have to work with major hazards during your project work (carcinogens,
mutagens, cyto-toxics, biological agents, GMOs, radioactivity, etc), to ensure that you
and your supervisor understand that you have to conduct a HIRAC review (hazard
identification, risk assessment and risk control) of the proposed work and to discuss
your response to the Science Faculty’s Health Questionnaire.
2) Formal Health and Safety Briefings
Mr Liam McCarthy (Chief Technician) will describe the general management and
security features of the building on the first day of term. Dr Nóirín Nic a’ Bháird, the
School Safety Officer will give you a formal general Health and Safety briefing at 11
a.m. on Tuesday and Wednesday 29th & 30th of September. Dr Nic a’ Bháird will give
two more detailed Health and Safety workshops just before you start your project
work in the research laboratories. ATTENDANCE AT THESE BRIEFINGS AND ANY
ADDITIONAL TRAINING SESSIONS (e.g. Radiological Protection Workshop,
viewing safety videos, etc.) IS MANDATORY. Some of these actions are legal,
license or College's insurer's requirements that have to be complied with.
3) Safety Lab Coat & Spectacles
You must have at least one Howie-style laboratory safety coat, conforming to the
NISO 1993, or better, standard, along with a pair of safety spectacles with you at all
stages during active laboratory work.
4) Specific Aspects of Health and Safety Associated with Project Work
You are required to attend two scheduled and compulsory short briefings on aspects
of Health and Safety in laboratories given by the School Safety Officer, Dr. Audrey
Carroll, a few days before project work starts. Any hazardous materials, steps or
procedures (including off-site work connected with your research such as collecting
samples from other laboratories, etc.) involved in your project will have been
identified by, and discussed with you by your project supervisor. He/she is required,
by law, to perform this hazard identification, risk assessment and risk control (HIRAC)
on every experiment undertaken by you, but you have a role to play as well in making
sure that you record the conclusions of this procedure in you notebook. The control
measures necessary to reduce or eliminate risk must be written in your notebook for
each hazardous step or procedure. The law requires this to be done. You are still in
training so you cannot be classed as a competent biochemist and thus able to do this
yourself to ensure your safety. If in doubt about the proper procedures for any
experiment, do not perform that experiment.
Senior Sophisters must make themselves aware of the College's and School’s Safety
Statement which is displayed prominently in every laboratory in the School. [It can be
downloaded
from
the
School’s
Local
Home-Page
at
this
URL:
www.tcd.ie/biochemistry/]. You are still bound by the 'Science Faculty's Health and
Safety Guidance Manual' and the associated Health Questionnaire which you
completed at the start of JF year. In case your health status has changed since then
in terms of the categories listed (including pregnancy or lactation) you have to
complete a new Health Questionnaire. If your health status again changes during the
9
year you must consult, in confidence, with the Safety Officer. [This particularly applies
in the case of pregnancy.]
You are not permitted to work with unsealed radionuclide sources unless you have
attended and satisfactorily completed a Radiological Protection Workshop to be held
on Thursday 7th (9:30am-4:00pm) and Fri 8th (9:30am-1:00pm) of January 2014. Any
student working with human materials (blood, buffy coats, semen, CSF, dialysis fluid,
primary explants, etc.) must be vaccinated against Hepatitis B prior to commencing
your project. You are not permitted to work with any Category 3 biological agents
such as HIV, Hepatitis B and C, etc. or to culture Category 3 (or higher) pathogens.
You must request or otherwise obtain Material Safety Data Sheets (MSDS) for any
toxic or dangerous chemicals or preparations that you are using in your project.
These MSDS's have to be requested at the point of ordering any material. The MSDS
must be stuck into your laboratory notebook. The guidance must be followed.
After 6:00 pm on working days, and at all times on weekends and public holidays, no
Senior Sophister may work in any laboratory without the close presence of a member
of the academic staff. It is the Senior Sophister's responsibility to ask that staff
member if he/she will consent to act in a supervisory capacity for the time the student
is working. During normal working hours no student may work alone in any
laboratory.
Failure to observe these rules/procedures will cause the offenders to be officially
warned, and be reported to the Head of School, school safety officer and project
supervisor. Normal College disciplinary procedures can be invoked (including fines
being levied as well as withdrawal of student i.d. card, etc.) Persistent failure to
observe these rules may result in that student being banned from laboratory work
with loss of those marks available for project work.
Students with Disabilities:
The University Policy Relating to students with disabilities is available at
www.tcd.ie/disability. The Student Disability Service is located in Room 2054 Arts
Building, phone = 8963111, email = [email protected]. The Student Disability Services
Committee provides the formal channel for raising issues affecting students with
disabilities.
Plagiarism:
The full statement of College policy on plagiarism can be found at: http://tcd-
ie.libguides.com/plagiarism
We recommend that you familiarise yourself with these new regulations. The College
Calendar defines plagiarism, gives examples of the kinds of actions that are deemed to constitute
plagiarism, and elaborates on the procedures for dealing with plagiarism cases. It is essential that
you read the Calendar entry that is relevant to you as an undergraduate or postgraduate student. You
should also look at the matrix that explains the different levels of plagiarism and how they are dealt
with. The calendar entry is presented below.
10
In addition members of staff of the School of Biochemistry & Immunology may
scan your written assignments using plagiarism-detecting software such as Turnitin
(additional information for which can be found at:
http://turnitin.com/static/index.html).
Calendar Statement on Plagiarism for Undergraduates - Part II, 82-91
Plagiarism
82 General
It is clearly understood that all members of the academic community use and build on the
work and ideas of others. It is commonly accepted also, however, that we build on the work
and ideas of others in an open and explicit manner, and with due acknowledgement.
Plagiarism is the act of presenting the work or ideas of others as one’s own, without due
acknowledgement.
Plagiarism can arise from deliberate actions and also through careless thinking and/or
methodology. The offence lies not in the attitude or intention of the perpetrator, but in the
action and in its consequences.
It is the responsibility of the author of any work to ensure that he/she does not commit
plagiarism.
Plagiarism is considered to be academically fraudulent, and an offence against academic
integrity that is subject to the disciplinary procedures of the University.
83 Examples of Plagiarism
Plagiarism can arise from actions such as:
(a)
copying another student’s work;
(b)
enlisting another person or persons to complete an assignment on the student’s behalf;
(c)
procuring, whether with payment or otherwise, the work or ideas of another;
(d) quoting directly, without acknowledgement, from books, articles or other sources, either
in printed, recorded or electronic format, including websites and social media;
(e)
paraphrasing, without acknowledgement, the writings of other authors.
Examples (d) and (e) in particular can arise through careless thinking and/or methodology
where students:
11
(i) fail to distinguish between their own ideas and those of others;
(ii) fail to take proper notes during preliminary research and therefore lose track of the
sources from which the notes were drawn;
(iii) fail to distinguish between information which needs no acknowledgement because it is
firmly in the public domain, and information which might be widely known, but which
nevertheless requires some sort of acknowledgement;
(iv) come across a distinctive methodology or idea and fail to record its source.
All the above serve only as examples and are not exhaustive.
84 Plagiarism in the context of group work
Students should normally submit work done in co-operation with other students only when
it is done with the full knowledge and permission of the lecturer concerned. Without this,
submitting work which is the product of collusion with other students may be considered to be
plagiarism.
When work is submitted as the result of a group project, it is the responsibility of all
students in the group to ensure, so far as is possible, that no work submitted by the group is
plagiarised.
85 Self plagiarism
No work can normally be submitted for more than one assessment for credit. Resubmitting
the same work for more than one assessment for credit is normally considered self-plagiarism.
86 Avoiding plagiarism
Students should ensure the integrity of their work by seeking advice from their lecturers,
tutor or supervisor on avoiding plagiarism. All schools and departments must include, in their
handbooks or other literature given to students, guidelines on the appropriate methodology for
the kind of work that students will be expected to undertake. In addition, a general set of
guidelines for students on avoiding plagiarism is available on http://tcdie.libguides.com/plagiarism.
87 If plagiarism as referred to in §82 above is suspected, in the first instance, the Director of
Teaching and Learning (Undergraduate), or their designate, will write to the student, and the
student’s tutor advising them of the concerns raised. The student and tutor (as an alternative to
the tutor, students may nominate a representative from the Students’ Union) will be invited to
attend an informal meeting with the Director of Teaching and Learning (Undergraduate), or
their designate, and the lecturer concerned, in order to put their suspicions to the student and
give the student the opportunity to respond. The student will be requested to respond in
writing stating his/her agreement to attend such a meeting and confirming on which of the
12
suggested dates and times it will be possible for them to attend. If the student does not in this
manner agree to attend such a meeting, the Director of Teaching and Learning
(Undergraduate), or designate, may refer the case directly to the Junior Dean, who will
interview the student and may implement the procedures as referred to under conduct and
college regulations §2.
88 If the Director of Teaching and Learning (Undergraduate), or designate, forms the view
that plagiarism has taken place, he/she must decide if the offence can be dealt with under the
summary procedure set out below. In order for this summary procedure to be followed, all
parties attending the informal meeting as noted in §87 above must state their agreement in
writing to the Director of Teaching and Learning (Undergraduate), or designate. If the facts of
the case are in dispute, or if the Director of Teaching and Learning (Undergraduate), or
designate, feels that the penalties provided for under the summary procedure below are
inappropriate given the circumstances of the case, he/she will refer the case directly to the
Junior Dean, who will interview the student and may implement the procedures as referred to
under conduct and college regulations §2.
89 If the offence can be dealt with under the summary procedure, the Director of Teaching
and Learning (Undergraduate), or designate, will recommend one of the following penalties:
(a) Level 1: Student receives an informal verbal warning. The piece of work in question is
inadmissible. The student is required to rephrase and correctly reference all plagiarised
elements. Other content should not be altered. The resubmitted work will be assessed and
marked without penalty;
(b) Level 2: Student receives a formal written warning. The piece of work in question is
inadmissable. The student is required to rephrase and correctly reference all plagiarised
elements. Other content should not be altered. The resubmitted work will receive a reduced or
capped mark depending on the seriousness/extent of plagiarism;
(c)
Level 3: Student receives a formal written warning. The piece of work in question is
inadmissible. There is no opportunity for resubmission.
90 Provided that the appropriate procedure has been followed and all parties in §87 above
are in agreement with the proposed penalty, the Director of Teaching and Learning
(Undergraduate) should in the case of a Level 1 offence, inform the course director and where
appropriate the course office. In the case of a Level 2 or Level 3 offence, the Senior Lecturer
must be notified and requested to approve the recommended penalty. The Senior Lecturer will
inform the Junior Dean accordingly. The Junior Dean may nevertheless implement the
procedures as referred to under conduct and college regulations §2.
91 If the case cannot normally be dealt with under the summary procedures, it is deemed to
be a Level 4 offence and will be referred directly to the Junior Dean. Nothing provided for
under the summary procedure diminishes or prejudices the disciplinary powers of the Junior
Dean under the 2010 Consolidated Statutes.
13
School of Biochemistry & Immunology Guidelines on Marking:
Scheme for marking of examination answers:
I
Excellent; full understanding of concepts with excellent knowledge of subject;
evidence of outside reading and thought beyond the content of specific
courses.
II-I
Very good answer demonstrating good understanding of concepts and broad
knowledge of the subject. Lapse of content tolerated at the lower end of the
scale.
II-II
Good answer that is generally sound but with limited scope. Lapses in detail.
III
Adequate but with significant shortcomings in content; containing errors in
detail and with poor structure.
F1
Weak answer containing some relevant information but lacking substance and
understanding.
F2
Poor answer; serious and absurd errors; contains few or no items relevant to
the question.
Scheme for marking of projects:
The project mark is comprised of the Supervisor’s mark and one other Examiner’s
marks for the project thesis. The Supervisor’s mark will be based on the student’s
performance within the laboratory (technical ability, understanding of the project and
literature pertaining to it, critical evaluation of results, demonstration of initiative and
independent thought) and on the content and presentation of the first draft of the
project thesis. The supervisor will also make the other Examiner of the project thesis
aware of any unforeseen difficulties that arose during the course of the project.
Class
Mark
Range
85100
I
70-84
Criteria
Exceptional project report showing broad understanding of the
project area and excellent knowledge of the relevant literature.
Exemplary presentation and analysis of results, logical
organisation and ability to critically evaluate and discuss results
coupled with insight and originality.
A very good project report showing evidence of wide reading,
with clear presentation and thorough analysis of results and an
ability to critically evaluate and discuss research findings. Clear
indication of some insight and originality. A very competent and
well presented report overall but falling short of excellence in
each and every aspect.
14
II-1
60-69
II-2
50-59
III
40-49
20-39
Fail
0-19
A good project report, which shows a reasonably good
understanding of the problem and some knowledge of the
relevant literature. Mostly sound presentation and analysis of
results but with occasional lapses. Some relevant interpretation
and critical evaluation of results, though somewhat limited in
scope. General standard of presentation and organisation
adequate to good.
A moderately good project report which shows some
understanding of the
problem but limited knowledge and appreciation of the relevant
literature. Presentation, analysis and interpretation of the results
at a basic level and showing little or no originality or critical
evaluation.
Insufficient
attention
to
organization
and
presentation of the report.
A weak project report showing only limited understanding of the
problem and superficial knowledge of the relevant literature.
Results presented in a confused or inappropriate manner and
incomplete or erroneous analysis. Discussion and interpretation
of
result
severely
limited,
including
some
basic
misapprehensions, and lacking any originality or critical
evaluation. General standard of presentation poor.
An unsatisfactory project containing substantial errors and
omissions. Very
limited understanding, or in some cases misunderstanding of the
problem and very restricted and superficial appreciation of the
relevant literature. Very poor, confused and, in some cases,
incomplete presentation of the results and limited analysis of the
results including some serious errors. Severely limited discussion
and interpretation of the results revealing little or no ability to
relate experimental results to the existing literature. Very poor
overall standard of presentation.
A very poor project report containing every conceivable error
and fault. Showing virtually no real understanding or
appreciation of the problem and of the literature pertaining to it.
Chaotic presentation of results, and in some cases incompletely
presented and virtually non-existent or inappropriate or plainly
wrong analysis. Discussion and interpretation seriously confused
or wholly erroneous revealing basic misapprehensions.
_________________
Dr Ed Lavelle
September 2015
15
SS IMMUNOLOGY TUTORIAL GROUPS 2015-2016
STUDENT
NUMBER
NAME
12302743
Amet, Rebecca
12304512
Coleman, Amy
12303530
Diskin, Ciana
12302499
Delaney, Grainne Mary
12302659
Gorby, Claire
11500287
12305741
Case, Sarah
Mahon, Fearghus
McAlpine David
12300689
Kelly, Alanna
12300915
12306480
White, Graeme
O Leary, Fionn Timothy
Noel
12304832
Grennan, Olivia
12302931
12308123
Feehan, Karen
MacDonald, James
Fraser
12303030
O Hare, John
12307742
Barber, Gillian
12310699
Reid, Caoimhe
12308668
Shanahan, Katharine
Ann
12327336
Fleming, Aaron
12310109
Hennessy, Naoise
12311515
Doran, Ciara
EMAIL
TUTOR
LUKE O’NEILL
CLAIR GARDINER
KINGSTON MILLLS
ANDREW BOWIE
RACHEL MCLOUGHLIN
ED LAVELLE
CLIONA O’FARRELLY
16
SS IMMUNOLOGY PRACTICE VIVA VOCE GROUPS 2014-2015
First Semester
Second Semester
A. Bowie & C. Gardiner
R McLoughlin & E. Lavelle
Amet, Rebecca
Coleman, Amy
Diskin, Ciana
Grainne Delaney
Fionn O’Leary
Sarah Case
Fearghus Mahon
Alanna Kelly
Ciara Doran
Graeme White
Claire Gorby
Karen Feehan
James MacDonald
R McLoughlin & E. Lavelle
C. O’Farrelly & K Mills
Fearghus Mahon
Alanna Kelly
Ciara Doran
Graeme White
Claire Gorby
Karen Feehan
James MacDonald
John O Hare
Gillian Barber
Naoise Hennessy
Katharine Shanahan
Aaron Fleming
Caoimhe Reid
Olivia Grennan
C. O’Farrelly & K Mills
A. Bowie & C.Gardiner
John O Hare
Gillian Barber
Naoise Hennessy
Katharine Shanahan
Aaron Fleming
Caoimhe Reid
Olivia Grennan
Amet, Rebecca
Coleman, Amy
Diskin, Ciana
Grainne Delaney
Fionn O’Leary
Sarah Case
17
Immunology: Breakdown of SS Papers I and II
2015-2016
Paper I
Section 1: BI4275 ‘Cells of the innate and adaptive immune system’
Myeloid cells (EC)
Natural killer cells (CG)
T cells (KM)
B cells and humoral immunity (MC)
2 questions
Section 2: BI4055 ‘Microbial Diseases’
Trypanosomiases (DN)
Prokaryotic pathogens (HW)
Helminths (PF)
2 questions
Section 3: BI4215 ‘Organ specific immunity’
Reproductive and liver immunology (COF)
Gastrointestinal tract (EL)
Respiratory tract (RMcL)
Neuroimmunology (CC, AD)
2 questions
Section 4: BI4225 ‘Autoimmune and inflammatory conditions’
Biochemistry of the inflammatory process (JB)
Rheumatoid arthritis (LON)
Autoinflammatory diseases (EC)
Immunodeficiency/clinical immunology (DD)
2 questions
Answer one question from each section (4 OUT OF 8)
18
Paper II
Section 1: BI4235 ‘Immune signalling’
Apoptosis (DZ)
Cytokine Signalling (LON)
Molecular basis of immune signalling (AK)
T cell receptor signalling (AB)
PI3K and mTOR signalling (DF)
2 questions
Section 2: BI4245 ‘Immunity to pathogens and vaccination’
Immunity to viruses (NS)
Viral evasion of innate and adaptive immunity (AB)
Antimicrobial resistance and host response to infection (RMcL)
Immune response to tuberculosis (JK)
Vaccines, adjuvants and danger hypothesis (EL)
2 questions
Section 3: BI4225 ‘Immunogenetics, immunomodulation and immunotherapy
Transgenics and animal models of disease (VK)
Immunogenetics (RMcM)
Immunotherapy and transplantation (COF)
2 questions
Section 4: BI4265 ‘Tumour Immunology’
Initiation & Progression (VK)
Metastasis & Treatment (VK/KM)
Cellular imaging (DN)
2 questions
Answer one question from each section (4 OUT OF 8)
19
SS Course Summaries: 2015-2016
BI4275
CELLS OF THE INNATE AND ADAPTIVE IMMUNE SYSTEM (5ECTS)
Myeloid cells (3 lectures) Emma Creagh)
Lecture 1: Summary of haematopoiesis from Myeloid progenitor cell; overview of
Granulocyte, Macrophage and DC cell types, maturation & cell specific functions in
innate immunity. Myeloid derived suppressor cells (MDSCs).
Lecture 2: Pattern recognition receptors (PRRs), Complement Rs - recognition of
infection & injury. Phagocytosis by cellular subtypes, vesicular trafficking, lysosome
formation & degradation mechanisms.
Lecture 3: Granulocyte migration, activation, degranulation. Exocytosis & Secretory
pathways - release of preformed molecules from granules (eg. Histamine) and newly
synthesised molecules (eg. TNFalpha).
Natural Killer Cells
(5 lectures) Clair Gardiner
Lecture 1: General features of NK cells including cell surface phenotypes, cytokine
production and main functions.
Lecture 2: Cytotoxicity mechanisms used by NK cells: necrosis and apoptosis (fas-L,
granzymes, TNF-α and LT). Benefits of these; NK cells as effectors of graft-rejection,
GVHD and GVL. Recognition of target cells; missing self hypothesis
Lecture 3: Receptors for MHC class I; human vs. mouse; lectin like receptors and Ig
superfamily receptors. Inhibitory vs. stimulatory receptors. Diversity inherent within
these receptors.
Lecture 4: Ligand specificity of human NK cell receptors – KIR and CD94/NKG2. Role of
peptide in ligand recognition; general mechanisms of NK cell recognition of virally inected
cells; viral escape strategies.
20
Lecture 5: Role of NK cell and receptors in particular diseases - a research perspective.
T cell differentiation and regulation (2 lectures) Kingston Mills
Lecture 1. T cell subtypes, antigen presentation and T cell differentiation’.
Lecture 2. Natural and induced regulatory T cells. Regulatory T cells in infectious
diseases. Role of anti-inflammatory cytokines produced by innate cells and T cells in
subversion of immunity to infection.
T cell immunity to bacterial and viral infection (2 lectures) Kingston Mills
Lecture 1: The bridge between innate and adaptive immunity. Pathogen activation of
macrophages and dendritic cells through pattern recognition receptors. Role of dendritic
cells in directing T cell subtypes.
Lecture 2: Role of Th1/Th2 cells in immunity to infection, including HIV, hepatitis C virus
and Bordetella pertussis.
B cells and humoral immunity (4 lectures) Michael Carty
Lecture 1: Discovery and history of B cells will be given as will a detailed description
on the activation of B cells. B cell subtypes and regulation will also be described in
detail.
Lecture 2: A detailed description of antibody production will be given. Dysregulation
of this system will be described in disease processes. Therapeutic manipulation of B
cells and humoral immunity will also be provided in inflammatory diseases and other
conditions.
Lecture 3 Discovery of complement and the proteolytic cleavage pathways leading to the
activation of the terminal effector proteins of the complement system. Activation of this
system results in pathogen opsonisation, phagocytosis, recruitment of inflammatory cells
21
and pathogen killing. In addition mechanisms by which these pathways are regulated will
also be described.
Lecture 4: Diseases in which complement components are thought to be involved will
be described. These diseases include those in which components of the complement
system are altered by mutation such as Systemic lupus erythematosus and conditions
where there is inappropriate and excessive activation of complement such as
glomerulonephritis. Finally therapeutic opportunities which target proteins of the
complement system will also be explored.
22
BI4055
MICROBIAL DISEASES (5ECTS)
African trypansomes
(8 lectures) Derek Nolan
The aim of these lectures is to provide an introduction to African trypanosomes, parasitic
protozoans that cause sleeping sickness in humans and a related disease, Nagana, in cattle.
These parasites are a major problem for human and veterinary health throughout sub Saharan
Africa and serious barrier to economic development of the region. Perhaps the most striking
feature of these parasites is that that they are exclusively extracellular. They grow and divide
in the mammalian vasculature and consequently exposed the adaptive and innate defence
responses of their mammalian hosts. In addition, for a variety of reasons, African
trypanosomes have been come a favourite model organism for molecular and cell biologists
and many discoveries of broad significance have emerged from studies on these model
unicellular eukaryotes. Areas where such discoveries have been reported will be illustrated in
the lectures where appropriate. The course is organized into two parts.
Trypanosomes Part 1: Stealth strategies of an elusive parasite
1.
How are trypanosomes, such as Trypanosoma brucei, able to evade the host humoral
immune response given that they are constantly exposed to this arm of the immune response?
2.
What other strategies do trypanosomes employ to circumvent the innate immune
responses?
3.
How are these parasites able to acquire essential macromolecular growth factors from
their hosts without attracting a response?
Trypanosomes Part 2: What is the molecular basis of human sleeping sickness?
The focus in part II is on the innate immunity that humans and other primates have to infection
by all but a few trypanosomes. In effect in this part we will consider the molecular basis of
African human sleeping sickness. We will consider the nature of the trypanolytic toxin present
in human serum and how this toxin kills these parasites. We will see an amazing link between
the toxin and an unsuspected programmed cell death pathway. Finally, we will see how two
strains of trypanosomes have responded by developing independent mechanisms to resist this
toxin and how in turn certain human populations are able to overcome this resistance and the
price they pay for this capacity.
Reading List:
Additional specific references for key experiments will be provided within the lectures which
are available on the school website.
Trypanosomes Part I
(1) Cross, G.A.M. (2001) African trypanosomes in the 21st century: what is their future in
science and health? Int. J. Parasitol. 31: 427-433
(2) Borst, P. (2002) Antigenic variation and alleleic exclusion Cell 109: -8.
(3) Pays, E. (2005) Regulation of antigen gene expression in Trypanosoma brucei Trends
Parasitol. 21: 517-520.
23
(4) Pays E. (2006) The variant surface glycoprotein as a tool for adaptation in African
trypanosomes. Microbes and infection 8: 30-937.
(5) Field, MC & Carrington, M. (2004) Intracellular membrane transport systems in
Trypanosoma brucei. Traffic 5:1-9
(6) Nolan, DP, Garcia-Salcedo, J.A., Geuskens, M., Salmon, D., Paturiaux-Hanocq, F.,
Pays,A., Tebabi, P. and Pays, E. (2001)
Endocytosis of macromolecules by African trypanosomes. pp127-141 In “World Class
Parasites Volume 1: The African Trypanosomes” Eds. Seed, R. & Black, S.J. (Kluwer
Academic Publishers)
(7) Stockdale C. et al (2008) PLoS biology Vol6 issue 7 e185 “Antigenic Variation in
Trypanosoma brucei: Joining the DOTs”
(8) Navarro M. et al (2007)TRENDS in Microbiology Vol.15 No.6
doi:10.1016/j.tim.2007.04.004
Nuclear architecture underlying gene expression in Trypanosoma brucei
Trypanosomes Part II
(1) Pays, E. Vanhamme, L., Vanhollebeke, B., Nolan, D. P.. and Perez-Morga, D. (2006)
The trypanolytic factor of human serum. Nat Rev Microbiol. 6: 477-86.
(2) Vanhollebeke B & Pays E (2010) Mol. Microbiol. 76: 806-814
The trypanolytic factor of human serum, many ways to enter the parasites, a single way to kill
it.
(3) Pays E & Vanhollebeke B (2008) Microbes Infect 10: 985-989
Mutual self-defence: the trypanolytic story
(4) Genovese et al. (2010) Science 329: 841-845.
Association of trypanolytic ApoL1 variants with kidney disease in African Americans
(5) Pays E. et al. (2014) The molecular arms race between African trypanosomes and humans
Nature Reviews Microbiology VOLUME 12 575-584.
(6) Vanwalleghem G. et al. (2015) NATURE COMMUNICATIONS | 6:8078 | DOI:
10.1038/ncomms9078 Coupling of lysosomal and mitochondrial membrane permeabilization in
trypanolysis by APOL1
Helminths of Human Importance (4 lectures)
Padraic Fallon
A third of the world’s population is infected with parasitic worms. These lectures will address
the major parasitic worms that are of medical importance.
Lecture 1-2:
Introduction to the major helminth parasites that infect man. Medical and economic impact of
helminth parasites on society.
Lecture 3-5:
Wormy people: genetic predisposition to helminth infection. Co-evolution of man and parasitic
worms: molecular and biochemical adaptation. The schistosome tegument or nematode
24
cuticle as model membranes. Helminth proteome and genome projects. Gastro-intestinal
versus systemic (tissue or blood dwelling) worm infections. Endosymbiotic infections.
Modulation of immunity by helminth parasites: implications for designing vaccines. Molecular
and biochemical targets for current and future drugs to treat helminth infections.
A reading list will be given out during the course
Prokaryotic pathogens (4 lectures)
Henry Windle
Lecture 1:. Introduction to prokaryotic pathogens of medical importance. Emerging and reemerging diseases.
Lecture 2: Overview of molecular mechanisms of bacterial induced disease - modulation of
host cell signalling responses and pathogenesis. Strategies to identify vaccine
candidates/therapeutic targets.
Lecture 3: Bacterial pathogens as a paradigm for chronic infection I. Infection and cancer –
the Helicobacter pylori connection: molecular basis of pathogenesis.
Lecture 4: Bacterial pathogens as a paradigm for chronic infection II. Animal models of
disease. Microbiomes, metagenomics and engineering microbes for our benefit. Mixed
microbial populations and disease.
General Reading:
Insights into host responses against pathogens from transcriptional profiling. Jenner et al. Nat
Rev Microbiol (2005)3 (4), 281-94
The impact of the microbiota on the pathogenesis of IBD: lessons from mouse infection
models S Nell, S Suerbaum & C Josenhans NAT REV MICROBIOL (2010) 8 (8) 564-77.
What are the consequences of the disappearing human microbiota? MJ. Blaser & S
Falkow NAT REV MICROBIOL (2009) 7 (12) 887-94.
Helicobacter pylori: gastric cancer and beyond. Polk DB, Peek RM
(2010)10(6):403-14.
Nat Rev Cancer.
25
BI4245
IMMUNITY TO PATHOGENS AND VACCINATION (5 ECTS)
Immunity to viruses (3 lectures) Nigel Stephenson
Lecture 1: Viruses and intracellular immunity
This lecture will provide an introduction to viruses and the key intracellular innate
immune events involved in viral detection and subsequent anti-viral Interferon
induction.
Lecture 2: Anti-viral Interferons
This lecture will describe Interferons, their effects and signalling pathways required for
anti-viral activity. The function of key Interferon Stimulated Genes will be outlined.
The role of Interferons as therapeutics will be discussed with specific focus on
Hepatitis C.
Viral Evasion of innate and adaptive immunity (6 lectures) Dr Andrew Bowie)
Lecture 1:
Introduction to viruses and immune evasion. Virus classification and life cycle. Six key
concepts in viral evasion: (1) Molecular recognition, (2) Nature of the viral-host
interaction, (3) Viral evasion and pathogenicity – what makes a good virus? (4)
Learning from viruses about the immune response, (5) Different types of viral proteins
involved, and apparent contradictions, (6) Targetting innate vs adaptive immunity.
Lecture 2:
Theanti-viral immune response I: Viral PAMPs and Pattern Recognition
Receptors(PRRs) – PKR, TLRs, RLRs, ALRs such as IFI16 and other DNA sensors
(cGAS). How PRRs signal.
Lecture 3:
Theanti-viral immune response II: Anti-viral PRRs trigger type I IFN induction. The
type I IFN system. IFN-stimulated genes (ISGs). Natural Killer Cells, Cytokines,
Chemokines & Inflammation, Compliment, Antibody, T Cells.
Lecture 4:
Overview of evasion of innate and adaptive defense mechanisms. Avoiding and
blocking recognition by PRRs. NFkB interactions – subversion or detection?
Lecture 5:
Interfering with Interferon. Inhibition of antigen presentation. Disrupting NK cell
function
26
Lecture 6:
Modulating chemokines and cytokines. Viral proteins and peptides as biotherapeutics.
Example of vaccinia virus protein A46 and VIPER, a peptide derived from A46.
Antimicrobial resistance and the host response to bacterial infection
(2 lectures) Rachel McLoughlin
Lecture 1: Mechanisms of antimicrobial resistance in bacteria. Epidemology of
antimicrobial resistant bacterial infections. The limitations of antibiotics as therapeutic
strategies.
Lecture 2: Targeting the host immune response for the development of vaccines and
immunomodulatory therapies to treat bacterial infection. Case study Staphylococcus
aureus (MRSA) infections.
The immune response to tuberculosis (3 lectures) Joe Keane)
Lecture 1: The immune response to tuberculosis; a model for pathogen evasion of
the human host response.
The innate immune response to tuberculosis;
macrophages and dendritic cells
a. The first contact; Inhaled M. tuberculosis bacillus is phagocytosed by a
macrophage; receptors for macrophage uptake; the signals of invasions; the phagolysosomal maturation arrest; TACO
b. The host fights back; The ROI and RNI reactions to invasion; Cytokine and
chemokine release; macrophage apoptosis and bug killing
c. Time to go clonal; Antigen presentation; proliferation and recruitment of T and B
cells; CMI; Delayed type hypersensitivity
Lecture 2: The granuloma; prison for the live bug
a. The granulomatous response; TB , sarcoid, leprosy, Wegeners and
Schistosomiasis
b. What holds it together? TNF, IFN gamma, somatostatin
c. What breaks it apart?; Immunosupression, HIV, steroids, TNF blockers, antiinterferon Ab or interferon receptor defects
Lecture 3: TNF blockers and reactivation of tuberculosis; more questions and clinical
proof of scientific principle
27
The role of TNF in tuberculosis disease. The role of TNF in preventing reactivation of
latent infection. Mouse evidence translated into human tuberculosis observations.
The consequence of TNF blocking for the disease and the host. The differences in
TNF blockers; sTNFR v. monoclonals. Other effects of monoclonals. Preventing
reactivation with TNF blockade.
Vaccines, adjuvants and the danger hypothesis (5 lectures) Dr Ed Lavelle)
Lecture 1: Basic concepts in vaccine development. Traditional approaches to
vaccination. Nature and mode of action of vaccines in current use.
Lecture 2: Vaccine adjuvants. Inert particulate and live bacterial and viral
delivery systems. Toxin/lectin and toll like receptor-based adjuvants.
Lecture 3: Mucosal vaccines. Distinctive features of the mucosal immune system and
implications for vaccination. Mucosal vaccine adjuvants and delivery systems.
Lecture 4: Vaccines for neonatal immunisation. Therapeutic vaccines.
Lecture 5: Danger theory. Endogenous danger signals in innate immune activation,
role of danger signals in efficacy of vaccine adjuvants.
BI4045
AUTOIMMUNE AND INFLAMMATORY CONDITIONS (5 ECTS)
Biochemistry of the Inflammatory Process (4 lectures) Dr Jack Bloomfield)
Lecture 1:
Definition of inflammation and inflammatory diseases. Difference between
inflammation in infection and inflammatory diseases.Mechanism of the inflammatory
process: respiratory burst, prostaglandin and leukotriene production. Degranulation
28
causes release of inflammatory mediators. Treatment of inflammatory diseases.
Anti-inflammatory drugs.
Lecture 2:
Psoriasis as a model of inflammatory skin disease. Role of neutrophil in the
perpetuation of chronic inflammation. Role of leukotrienes in pathogenesis of
psoriasis. Neutral proteinases in psoriasis.
Lecture 3:
Helicobacter pylori in peptic ulcer disease. Effect of H2-receptor antagonists on peptic
ulcers. Study of prostaglandin and leukotriene production in peptic ulcer disease.
Antiinflammatory effects of H2-receptor antagonists.
Lecture 4:
Role of prostaglandins and leukotrienes in inflammation. Effect of non-steroidal antiinflammatory drugs on prostaglandin and leukotriene production. Pathways of
prostaglandin and leukotriene production.
Rheumatoid Arthritis (2 lectures) Luke O’Neill
Lecture 1: What is rheumatoid arthritis? Clinical, molecular and cellular definitions.
Early concepts: connective tissue structure and degradation. Rheumatoid Factor.
And B cells. HLA associations and the genetic component. Autoantigens. Role of
inflammation – prostaglandins and tissue degrading enzymes.
Lecture 2: Key role of cytokines – IL-1, TNF, IL6. Current therapies – NSAIDs,
steroids, biologic therapies (anti-TNF, anti-IL-1, anti-IL-6, anti-CD20 and CTLA-4 Ig).
Prospect for future therapies.
Multiple Sclerosis and EAE (3 lectures) Jean Fletcher, Kingston Mills
Lecture 1: Breakdown of tolerance in autoimmunity. Risk factors, pathogenesis,
diagnosis and monitoring of MS
Lecture 2. MS therapies: Mechanisms of action, efficacy, side effects.
Lecture 3: EAE. Role of innate and adaptive immunity in pathogenesis of autoimmune
diseases. Role of regulatory T cells in preventing autoimmune diseases.
Autoinflammmatory diseases (2 lectures) Emma Creagh
29
Lecture 1: Key features of systemic autoinflammatory disorders. Classic hereditary
'Periodic Fever Syndromes' - FMF (Familial Mediterranean Fever), TRAPS (TNF
Receptor Associated Periodic Syndrome) and HIDS (Hyperimmunoglobulinemia-D
with periodic fever syndrome).
Lecture 2: NLRP3/Cryopyrin-associated periodic syndromes (CAPS): Familial Cold
Inflammatory Syndrome (FCAS); Muckle-Wells Syndrome (MWS) and Neonatal onset
multisystem inflammatory disease (NOMID). Autoinflammatory disorders associated
with skin pustules, such as DIRA (deficiency of IL-1R antagonist), CARD14 mediated
psoriasis (CAMPS) and early onset inflammatory bowel diseases (EO-IBD).
Immunodeficiency/ Clinical immunology (4 lectures) Derek Doherty
Lecture 1: Primary immunodeficiencies
This lecture will cover the genetic bases, clinical presentations, diagnoses and
treatments of primary immunodeficiencies, including antibody, complement, MHC and
lymphocyte deficiencies.
Lecture 2: Acquired immunodeficiencies
This lecture will cover the different causes of acquired immunodeficiencies but will
focus mainly on HIV-associated disease, including the virology, immunology, clinical
features and recent progress in vaccine development. The significance of HIV in the
developing world, where many other infectious disease are also endemic, will be
emphasized.
Lecture 3: Autoimmune disease
The lecture will start with an overview of autoimmune diseases, emphasizing the
heterogeneity of such diseases and the roles of T cells, B cells and other cells of the
immune system in the pathogenesis. We will then focus on the example of coeliac
disease as an illustration of how multiple pathogenic hits can result in disease and
how the disease can be diagnosed and treated.
Lecture 4: Antibody-mediated autoimmune diseases
This lecture will focus on autoimmune diseases whose pathologies are mainly
mediated by antibodies, such as systemic lupus erythematosus, myasthenia gravis
and autoimmune vasculitis.
30
BI4235
IMMUNE SIGNALLING (5 ECTS)
Apoptosis (5 lectures) Daniela Zisterer
Lecture 1: Introduction to apoptosis. Role in development, maturation of the immune
system and in cell turnover. Morphological features of apoptosis. Comparison with
necrosis. Biochemical methods used for examination of apoptosis e.g. Annexin V
staining. Aberrations in apoptosis: implicated in cancer and neurodegenerative disesases
e.g. Alzheimer's. Genetic studies into nematode C. elegans provides key insights into
molecular mechanisms regulating apoptosis.
Lecture 2: Caspases: family of cysteine proteases: 'death executioners' in apoptosis. 14
caspases identified to date. Caspases subdivided into 3 categories: substrate specificity,
prodomain length and prodomain sequence. How are caspases activated? By
autoactivation, transactivation or proteolysis by other proteases. Experimental evidence
that caspases are important in apoptosis. Biochemical measurement of caspases:
fluorigenic assays and Western Blotting assays.
Lecture 3: Apoptotic signal linked to caspases through 'sensor' and 'adapter'. Model for
regulation of apoptosis by APAF1: cytochrome c released from mitochondria as 'sensor'
and APAF1 (apoptosis activating factor 1) as 'adapter'. Formation of apoptotsome. IAPs
(inhibitor of apoptosis proteins) which bind to and inhibit caspase activity. Smac/DIABLO
which binds to and neutralises IAPs inhibitory activity. Subcellular localisation of
caspases: cytosol, nuclei, mitochondria and ER. Caspase substrates e.g. PARP (poly
ADP ribose polymerase), Lamins and CAD (caspase activated DNAase). Caspase 12:
link with Alzheimer's disease? Caspase-independent cell death. AIF, Endo G, Omi/HtrA2.
Lecture 4: The Bcl-2 protein family. Primary structure. Subdivided into 'pro-survival' and
'pro-apoptotic' proteins. 3-D structure. Channel forming activity. Subcellular localisation.
Model for regulation of apoptosis by Bcl-2. How is cytochrome c released from
mitochondria during apoptosis? Two competing models: PTP (permeability transition
pore) opening and movement through VDAC (voltage-dependent anion channel). Posttranslational modification of Bcl-2 family: 2 types- proteolysis and phophorylation.
Example of each type. Regulation of cell survival by Akt pathway.
Lecture 5: Death Receptors: signalling and modulation. Examples of death receptors
and signalling mechanisms involved: Fas, TNFR1 (tumour necrosis factor receptor1),
DR3 (death receptor3), DR4 and DR5. Death domains (DDs), Death effector domains
(DEDs), caspase-recruitment domains (CARDs); DISC (death inducing signalling
complex). Experimental evidence for TNFR1 signalling pathway. Modulation of apoptosis
by decoy receptors e.g. DcR1 and DcR2 in TRAIL signalling. Apoptosis induction by
Granzyme B. Mitogen-activated protein kinases and apoptosis. Induction of apoptosis by
cancer chemotherapy. Mechanisms of evasion of apoptosis by tumour cells.
31
Cytokine Signalling (5 lectures) Luke O'Neill
Lecture 1: Cytokine families:
interleukins, interferons, tumour necrosis factors,
chemokines, colony stimulating factors. Properties and functions: inflammation,
hemopoeisis, immune cell activation, anti-inflammatory cytokines. Class I cytokine
receptors: JAKs and STATs. Specificity in signalling. WSWS motif. gp130 as second
chain. Common and unique receptor chains. Complexity of IL2 signalling: PI3 kinase,
IRS-1.
Lecture 2: Type II cytokine receptors: Interferon receptor signalling: discovery of ISGFs
and Tyk. Use of JAK and STAT nomenclature. JAK and STAT knock-out mice: key
features. Interferon responsive genes and anti-viral effects. IL10 signalling. Suppresors
of Cytokine signalling.
Lecture 3: Type III cytokine receptor family: TNF receptors. Homology between TNFR,
NGFR, Fas and CD40. TNF signalling: TRADD, RIP, FADD and caspases. TRAFs.
Pathways to NFkB and apoptosis. Mechanism of activation of NFkB. IKK complex.
CARD-containing proteins.
Lecture 4: Type IV cytokine receptors: IL1 family. IL1 receptor signalling: IL1 pathway
as prototypical 'stress' response in plants and animals. The TIR domain: structure and
function. Toll-like receptors in mammals and innate immunity. LPS and IL18 receptors/
MyD88 as key adaptor. Roles of TLR-1 to TLR-10: recognition of PAMPs by PRRs.
Primacy of TLRs in innate immunity.
Lecture 5: Signal transduction pathways activated by the TIR domain. MyD88, IRAK1 –
IRAK-4. TAB1/TAK-1. Traf-6 and ubiquitination. Regulation Stress activated protein
kinases: p38 MAP kinase and JNK. Comparison to classical MAP kinases. IKK
activation by TAK-1. Lessons from knock-out mice: Specific adapters for different TLRs?
The role of Mal in LPS signalling. NALPs and NODs. Regulation of caspase-1
Molecular basis for immune signalling (2 lectures) Amir Khan
Lecture 1: Structural properties of TCR/MHC complexes and the immunological synapse
Lecture 2: Thermodynamics and kinetics of TcR/MHC complexes: models for
immune signaling
32
T cell receptor signalling (2 lectures) Andrew Bowie
Lecture 1 TCR signalling I: Signal transduction in T cells, overview of signalling
molecules involved, T cell activation via the T cellreceptor: Src family kinases,
adaptor proteins, PLCgamma, G proteins and GEFs, MAP kinases, transcription
factors, feedback signals.
Lecture 2. TCR signalling II: The contribution of signal 2 via co-stimulatory
molecules to T cell activation, roles of CD28 co-stimulation, motifs on CD28 and key
signallingmolecules involved in CD28 signalling, role of CTLA4 and ICOS, signalling
basis of Th1 vs Th2 polarisation, role of lipid rafts in TCR signalling,
NFkappaBactivation in T cells.
PI3K, mTOR and immune cell metabolism (5 lectures) Dr David Finlay (DKF)
Lecture 1: Phosphoinositide 3-kinase (PI3K) signalling (DKF)
This lecture will introduce PI3K as a key signalling molecule in the immune system,
describe PI3K’s enzymatic activity and discuss the various PI3K isoforms and their
relative expression patterns. How the different PI3K isoforms are activated will also
be discussed.
Lecture 2: mammalian Target of Rapamycin (mTOR) signalling (DKF)
The composition of the two distinct mTOR complexes, mTORC1 and mTORC2 will be
described. Mechanisms that regulate mTORC1 activity will be explained. The
relationship between PI3K and mTOR will be addressed using some experimental
data.
Lecture 3: PI3K and mTOR: immune cell function and metabolism (DKF)
This lecture will discuss the role for PI3K and mTOR signalling in controlling the
differentiation and function of immune cells. PI3K and mTORC1 control of immune
cell metabolism will be introduced.
Lecture 4: Cellular metabolism in immune cells (DKF)
Recap on cellular metabolism, in particular glycolysis and oxidative phosphorylation.
How cellular metabolism can be configured to match the demands of immune cells for
energy, biosynthesis and longevity will be discussed.
Lecture 5: Metabolic regulation of immune cell function (DKF)
33
This lecture will address the ways that cellular metabolism can directly impact upon
immune cell function. Metabolic regulators and enzymes with important
immunoregulatory functions will be described. Cellular metabolites that impact upon
cellular signalling and epigenetic programming will be considered.
34
BI4255
IMMUNOGENETICS, IMMUNOMODULATION AND IMMUNOTHERAPY
(5 ECTS)
Transgenics and animal models of immune mediated disease (5 lectures)
Vincent Kelly and Derek Nolan
Lecture 1. Mutagenic, transgenic & cloning technology (VK): The concept of
forward and reverse genetics in understanding gene function will be considered and
how these mutations are physically introduced into the genome through random
mutagenesis, viral mutagenesis, gene replacement and gene-targeting strategies.
The process of microinjection to create transgenic animals, gene knockouts and
cloned animal will be covered and the generation and use of induced pluripotant stem
cells (iPS) in biomedical research applications.
Lecture 2. Design and development of transgenic constructs (VK): The design of
targeting vectors relies on a detailed structural/functional understanding of the gene
under study. Various strategies for controlling the activity of the gene are available
including the creation of knock-outs, knock-ins, conditional knockout and reporter
systems. Gene-trap technology has, in recent times, gained significantly in popularity
and the methodology will be examined in some detail.
Lecture 3. Zinc Finger Nucleases and Talen Nucleases(VK): These state-of-theart technologies have the potential to revolutionise the manipulation of the eukaryotic
genome, from cells in culture to mice, rats, rabbits, pigs etc. This lecture will cover the
principles of this technology and how it is being currently exploited in research.
Lectures 4 & 5. RNA interference (DN): The discovery of the classical RNA
interference pathway involving siRNA will be described. The lectures will consider the
concept of regulation of expression through siRNA and microRNAs along with the use
and design of RNAi based approaches in functional genomics. The advantages and
limitation of such approaches will investigated through the use of specific examples.
The potential use of RNAi in therapeutic approaches will be outlined.
Immunogenetics (5 lectures) Ross McManus
Lecture 1. Identifying Disease Susceptibility Genes
Lecture 2) Introduction to Genetic Diseases
Inherited diseases cover a spectrum of disorders and can result from mutations in
35
single genes (Mendelian inheritance) or from the additive effects of common
polymorphisms in complex networks of genes giving rise to complex genetic
disorders. These lectures discuss the modes of inheritance of genetically determined
diseases and the mechanisms used to identify chromosomal regions associated with
disease susceptibility.
Lecture 3. Genetics of Inflammatory Bowel Disease
Inflammatory bowel disease afflicts almost 0.5% of western populations and is
characterised by a chronic, relapsing, intestinal inflammation. It is subdivided primarily
into two broad phenotypes, ulcerative colitis and Crohn’s disease. There is a clear
tendency for these conditions to be inherited, although the pattern of inheritance is
complex, indicating that both are multifactorial diseases resulting from the interplay of
genetic and environmental factors. This lecture will review recent finding regarding the
genetics of IBD.
Lecture 4. Genetics of Coeliac Disease
Coeliac Disease is a prevalent disease in Caucasian populations with a prevalence of
susceptible individuals of approximately 1%, although the actual frequency of individuals
displaying clinical symptoms is lower at about 1:300. Coeliac disease affects the small
intestine where dietary proteins derived from cereals provoke an immunological reaction
which leads to extensive damage to the gut wall and atrophy of the small intestinal villi.
This lecture willcover what has been learned in the recent past regarding the genetics of
this disease and what this tells us about the cell biological processes at play.
Lecture 5. MHC & Inherited Diseases
The Major Histocompatibility Complex is situated on the short arm of chromosome 6 and
is one of the most gene- dense regions of the human genome. Many of these genes
code for proteins which play a role in the function of the immune system with genes in
both the HLA Class I and II regions involved in antigen presentation and genes in the
Class III/IV encoding many other proteins with diverse functions in immunity and
inflammation. Furthermore, many autoimmune or inflammatory diseases have been
genetically linked to the MHC and it is clear the inherited variation in the MHC is a major
determinant of susceptibility to these diseases. This lecture will describe the structure
and content of the MHC and evidence of a causal association with inherited disease.
Immunotherapy and transplantation (4 lectures) Cliona O’Farrelly
Growth factors, interferons, monoclonal antibodies, steroids, personalised therapies)
immunosuppressive agents (FK506, cyclosporine A).
Allo recognition; the major histocompatibility complex; NK cells; T cells; bone marrow,
liver, kidney transplantation
36
Immunotherapy and transplantation (4 lectures) Cliona O’Farrelly
Growth factors, interferons, monoclonal antibodies, steroids, personalised therapies)
immunosuppressive agents (FK506, cyclosporine A).
Allo recognition; the major histocompatibility complex; NK cells; T cells; bone marrow,
liver, kidney transplantation
37
BI4215 ORGAN-SPECIFIC IMMUNITY (5 ECTS)
Introduction to regional immunity, reproductive and liver immunology (6
lectures) Cliona O’Farrelly)
Reproductive Immunology
This will introduce students to the basics of reproductive immunology. Against a
background of some basic anatomy, physiology and endocrinology of the human
male and female reproductive tracts, current understanding of local immune
mechanisms and their regulation will be presented. The effects of immunoregulatory
abnormalities on related pathologies will be introduced, in particular endometriosis,
infertility, sexually transmitted infection and cervical cancer. In this context, the
potential for immunotherapeutic interventions will be explored. Students will have the
opportunity to visit the National Maternity Hospital at Holles St where the Director of
the Merrion Fertility Clinic will give some insight into current major clinical challenges.
Liver Immunology Cliona O’Farrelly
This will introduce students to the fundamentals of liver immunology. Against a
background of some basic anatomy and physiology of human liver, current
understanding of local immune mechanisms and their regulation will be presented.
The effects of immunoregulatory abnormalities on related pathologies will be
introduced, in particular liver metastasis, transplant rejection and HCV infection. In
this context, current immunotherapeutic interventions and the potential for new
developments will be explored. Students will have the opportunity to visit the
National Liver Transplant Centre at St.Vincent's University Hospital, where one of
the hepatobiliary surgeons or pathologists will give some insight into current major
clinical challenges facing hepatology.
Gastrointestinal tract (3 lectures) Ed Lavelle
Lecture 1: Overview of gut associated lymphoid tissue, Peyer’s patches, inductive
and effector sites. Uptake of antigens across epithelial surfaces.
Lecture 2: Dendritic cells and T cells in the gastrointestinal tract. Homing of gut T
cells
Lecture 3: Mucosal humoral immunity. IgA responses and their regulation.
38
Respiratory tract (3 lectures) Rachel McLoughlin
Lecture 1: Introduction to the basic biology of the respiratory tract: conducting
airways, mucosal surface, lung parenchyma and organization of the lung immune
system. Understanding the concept that the lung is continually exposed to foreign
antigens and must discriminate between recognition of innocuous environmental
antigens and pathogenic antigens.
Lecture 2: Roles played by individual cells in regulating immune response in the
lung: airway epithelial cells, alveolar macrophages, regulatory T-cells, T-cell homing
to lung, innate lymphoid cells
Lecture 3: Immunological challenges faced by the lungs: Infection (???), Allergic
disease (Asthma), inflammatory disease (COPD), toxin exposure (Cigarette smoke)
Neuroimmunology (8 lectures) Aisling Dunne, Colm Cunningham
I Colm Cunningham (4 lectures)
Brain as an immune privileged organ, multiple sclerosis
Multiple sclerosis and treatment
Acute neuroinflammation in infection/stroke/TBI.
CNS inflammation induced by systemic inflammation (Sickness Behaviour)
II. Aisling Dunne (3 lectures)
Aspects of microglial activation:
downregulators
microglial
phenotypes,
PAMPs,
Microglial
Sterile inflammation, DAMPs in the context of neurodegeneration (including
ischaemic preconditioning).
Discussion session
39
BI4265
TUMOUR IMMUNOLOGY (5 ECTS)
Initiation & Progression (4 Lectures) Vincent Kelly
Lecture 1. Underlying causes of cancer (VK): The characteristics that are used to
classify cancers and their stage of development will be described. A number of
examples will be given of how environmental factors, i.e. xenobiotics, radiation and
oxidative damage contribute to multistep carcinogenesis. The means by which cancer
is limited by DNA damage sensing, DNA repair and cellular adaptation to
oxygen/radical damage will be covered.
Lecture 2. Oncogenes and tumour suppressor genes (VK): Many of the original
discoveries on oncogenes were derived from work on viruses. The concepts of
onocgenes and proto-ocogenes will discussed such as src and the Rous sarcoma
virus and there will be an in dept examination of the ras oncoprotein pathway an the
function of other oncogenes including abl, sis, c-myc and how they influence cellular
proliferation. Suppressor genes play an important role in limiting cancer formation and
a number of models were put forward from original studies including Knodson’s twohit model and haploinsufficiency. The mode of action of tumour suppressors such as
APC, MSH2, MLH1, BRCA1, p53 will be examined with particular focus on p53, Rb
and APC.
Lecture 3. Cancer epigenetics (VK): Changes in the genetic code is but one means
to arrive at a pre-malignant crossroads. Epigenetics changes in gene expression
have been found to alter tumor suppessor gene activity through. These epigenetic
changes may occur as a consequence of altered DNA methylation status at CpG
promoter regions of aberrant histone modification. In fact, cooperative suppression by
both mechanisms has recently become the focus of new anti-cancer therapies
through the development of DNMT and histone deacetylase inhibitors.
Lecture 4. Cancer metabolism & the tumor microenvironment (VK): Many of the
control points of cancer, oncogenes, tumor suppressor genes (including mTOR, PI3K,
Akt, p53, AMPK) are intimately linked to metabolism, especially glycolysis, which
provides the cancer with the building blocks for growth. The tumor cell
microenvironment is invariably acidic and hypoxic causing the transcription factor
HIF1a to set in place protective responses including unregulating the production of
monocarboxylate transporters, VEGF, matrix metalloproteinases and angiogenic
factors.
Metastasis and Cancer Treatments (6 Lectures) Vincent Kelly & Kingston Mills
Lecture 1. Angiogenesis and metastasis (VK): The process by which cancer cells
develop new blood supplies (angiogensis) is reliant on being able to remodel the
tumor environment and the extracellular matrix. A discussion of how this remodelling
occurs through matrix metalloproteinases and plasminogen will be given along with
40
the cause and consequences of breaking cell-cell interactions. The means used by
cancer cells to physically move from the primary tumor (e.g. epithelial-mesenchymal
transition) and how the immune system promotes this process will be described.
Breast cancer will be used as a model of how cancer cells choose secondary sites for
proliferation, especially the bone marrow; ‘the vicious cycle’.
Lecture 2. Colon cancer, genetics and epigenetics (VK): Arguably, colon cancer is
one of the best studied cancers in terms of its formation and progression. This lecture
will discuss the contribution of chromosomal instability in terms of changes to APC,
COX2 and Smad4 and microsatellite instability caused by epigenetic suppression of
mis-match repair enzymes including MSH2 & MLH1. The contribution of inflammation
to colon caner will be considered and how NSAIDS and IL-10 mediate polyp
formation.
Lecture 3. Stem cell theory of cancer, focusing on colon cancer (VK): The
intestinal crypt stem cells are maintained in a specialized compartment of the
intestinal crypt through the Ephrin receptors. The maintenance and proliferation of
these stems cells will be covered including the various signals used to control their
proliferation, such as hedgehog, WNT, PDGF, Eph, NOTCH and BMP. The
importance of the intestinal stems cells to cancer development and treatment will be
considered.
Lecture 4. Cancer treatment (VK): Classical anti-cancer drugs such as
antimetabolites, alkylating agents and antimytotic agents are still widely used in
therapy today despite severe side-effects. Newer ‘magic bullets, hold promise of more
specific cancer treatment strategies such as Imatinab in the treatment of CML.
However, drug resistance is a problem and has revealed the phenomenon of
oncogene addition. Recent drug strategies have begun to focus on targeting tumor
cell metabolism, its environment and the cancer initiating cells (cancer stem cells) that
perpetuate proliferation even after treatment.
Lecture 5. Cellular and humoral Immune responses to tumors (KM): These
lectures include the role of antibody, cytotoxic T lymphocytes, macrophages, NK cells
and Th1 cells; Evasion and subversion of immune responses by tumors - antiinflammatory cytokine production and regulatory T cell induction; Tumor-specific
antigens and breaking tolerance to self antigens
Lecture 6. Tumor immunotherapy (KM): Antibodies, Toll-like receptor agonists and
cell-based therapies; Tumor vaccines - killed tumor cells, tumor specific peptides and
antigens, heat shock proteins and dendritic cell vaccines
Cellular Imaging (4 lectures) Derek Nolan
41
Lecture 1: Introduction to imaging and the concept of resolution. Application of electron
microscopy in cell imaging.
Lecture 2: EM tomography and specialized techniques. Introduction to light microscopy.
Lecture 3: Advanced light microscopy: wide field and confocal microscopy.
Lecture 4: Application of fluorescent proteins and probes in multidimensional imaging in
fixed and live cells.
42
BI4015 DATA HANDLING (5 ECTS)
SEQUENCE ANALYSIS & MOLECULAR MODELLING
Sequence Analysis Jerrard Hayes
The course will provide an introduction into Bioinformatics. Part I of the course consists of
three lectures and three exercise sessions. Topics covered include:
-
DNA (including genomic) and protein databases
Accessing sequence information from databases using the Internet
Sequence similarity searches (i.e. BLAST, FASTA)
Identification of homologous proteins
Multiple sequence alignments (i.e. Clustal W)
Searches for protein motifs, domain, patterns
Students will carry out three exercises (marked as problems):
Exercise 1: Accessing databases from the Internet, retrieval of sequences (DNA and
protein), extracting relevant sequence information, presentation and annotation of a
chosen sequence
Exercise 2: Sequence similarity search (BLAST), identification of homologous proteins,
multiple sequence alignment (Clustal W)
Exercise 3: Sequence analysis of membrane proteins,
identification of transmembrane helices and signal peptides
hydrophobicity
plots,
Reading list:
*essential reading
# recommended
*Bioinformatics: Sequence, structure, and databanks. A practical approach. D. Higgins and
W. Taylor (eds.) Oxford University Press, 2000.
*Trends guide to bioinformatics. Elsevier Science, 1998
#Benson, D. A. et al. 1999. GenBank. Nucleic Acid Research, 27: 12-17
#Bairoch, A. and R. Apweiler. 2000. The SWISS-PROT protein sequence database and its
supplement TrEMBL in 2000. Nucleic Acid Research, 28: 45-48.
43
#Altschul et al. 1990. Basic local alignment search tool. J. Mol. Biol. 215:403-410.
#Needleman, S. B. and Wunsch, C. D. 1970. A general method applicable to the search
for similarities in the amino acid sequence of two proteins. J. Mol. Biol., 48: 443-453.
#Smith, T. F. and Waterman, M. S. 1981. Identification of common molecular
subsequences. J. Mol. Biol., 147: 195-197.
#Kyte, J. and Doolttle, R. F. 1982. A simple method for displaying the hydropathic
character of a protein. J. Mol. Biol., 157: 105-132.
#Persson, B. and Argos, P. 1994. Prediction of transmembrane segments in proteins
utilising multiple sequence alignments. J. Mol. Biol., 237: 182-192.
#Rost, B. et al. 1995. Transmembrane helices predicted at 95% accuracy. Protein
Science, 4: 521-533.
#Von Heijne, G. 1992. Membrane protein structure prediction. Hydrophobicity analysis and
the positive-inside rule. J. Mol. Biol., 225:487-494.
#Sonnhammer, E. L. L. et al. 1998. A hidden Markov model for predicting transmembrane
helices in protein sequences. In J. Glasgow et al. (eds.) Proc. Sixth Int. Conf. On Intelligent
Systems for Molecular Biology, 175-182. AAAI Press.
#Von Heijne, G. 1986. A new method for predicting signal sequence cleavage sites.
Nucleic Acid Research, 14: 4683-90.
#Nielsen, H. et al. 1997. Identification of prokaryotic and eukaryotic signal peptides and
prediction of their cleavage sites. Protein Engineering, 10:1-6.
Molecular Modelling
Dr Darren Fayne
The course consists of one main exercise and multiple mini-problems during the workshop
(Marked as one problem). Topics covered include:
1 - Introduction to the PDB database / information contained in crystal structure files.
2 - Use of the MOE software package for macromolecular visualisation of a protein/
inhibitor complex - here students will familiarise themselves with molecular displays, and
will get used to 'thinking in 3D' there will be an associated (in session) task to do with
displaying protein-ligand distances on screen.
3 - Use of the MOE software package – binding site analysis.
- Students will use MOE to display the tertiary structure of the chosen protein complex in
the popular `cartoon' rendering style. Here the students will see MOE in operation and will
be walked through protein binding site analysis tools.
4 – Introduction to homology modelling in MOE
44
Exercise:
a) Become familiarised with usage of the MOE software package.
b) Use of MOE to create small drug-like molecules.
c) Examination of protein-molecule binding complex
d) Identifying potential drug binding sites on a novel protein
e) Visualisation of the electrostatics of the protein binding site
f) Compare the complementarity between the protein binding site and a potential new drug
g) Homology modelling – generate the 3D structure of a new protein based on the known
X-ray structure of a related protein
h) A 30 minute exam based on the techniques covered during the workshop
All the tasks are electronic in nature and will not require paper submissions.
Background reading: http://www.chemcomp.com/software.htm
BioResources Unit Introductory Animal Course Peter Nowlan
The Purpose of this lecture course is to introduce Students to the basic requirements for
working with animals. This is necessary if a full appreciation of animal related work is to
be got from the projects. It is also a legal requirement that anybody involved in the use of
animals for scientific purposes has appropriate training (EC directive 86/609)
This module is not intended to be a comprehensive training course. To do this would
require a much more detailed and extensive series of talks. Most of the training which will
be required by students will be obtained by working in close contact with a technician and
with experienced supervisors.
The golden rule should be always 'if you don't know ask somebody'.
The welfare of the animal and often the success of your Project will depend on
using a correct approach to animals involved in your project.
Even if you do not intend choosing a project, which involves live animals, you may do so
in your future career. It will not be possible for anybody who does not pass the
assessment to choose a project involving animals.
Introduction to Laboratory Animal Science
The Law and Application for a licence
Animal House Design; Its effect on Research
Characteristics of Individual species
Experimental design Choice of species
45
Injections and tissue sampling
Health Considerations
Alternatives to live animal experimentation
Handling Video, Safety, Local arrangements
Video and discussion 'Ethics of Animal research'
The Scientists Viewpoint
Assessment
Reading List:
Laboratory animals an introduction for new experimenters
Handbook of laboratory animal management and care
M. Lloyd
Introduction to laboratory animal science and technology
Humane experimental technique
Experimental and surgical technique in the rat
P. Flecknell
Animals and alternatives in toxicology
; present and future prospects
In vitro toxicology
UFAW handbook on the care
& management of laboratory animals
Laboratory animals anaesthesia
Handbook of rodent and rabbit medicine
M. Swindle, P. Flecknell
The biology and medicine of rabbits and rodents
Wagner
The laboratory animals, principles and practice
A. Pearson
Man and mouse, animals in medical research
Lives in the balance;
the ethics of using animals in biomedical research
Vivisection in historical prospective
A. A. Tuffery
S. Wolefensohn,
J. Inglis
W. Russell, R. Burch
H. Wayneforth,
M. Balls, J. Bridges,
J. Southee
S. Cox Gad
T. Poole
P. Flecknell
K Laber-Laird,
J. Harkness J.
W. Lane-Petter,
W. Paton
J. Smith,K. Boyd
R. Rupke
46
SENIOR SOPHISTER PROJECTS
2015-2016
47
SS PROJECT NO.1
Supervisor: Mark Hughes and Prof Luke O’Neill
Title: EXPLORING THE ROLE OF GSTO1 AS A CRITICAL MEDIATOR OF TLR4
SIGNAL TRANSDUCTION
Background:
Antioxidants protect the cell from oxidative stress thereby maintaining the redox balance,
utilizing glutathione as the main antioxidant in the cell. Glutathione maintains cytoplasmic
redox potential and aids protein integrity via the glutathionylation of redox-sensitive amino
acids1. Toll-like receptors (TLR) are critical pattern recognition receptors in innate immunity,
playing pivotal roles in immune and infectious disease2. We have found evidence of
glutathionylation as a regulator of TLR signaling via an enzyme termed glutathione
transferase omega 1 (GSTO1). GSTO1 is able to both glutathionylate and deglutathionylate
key substrates. We have found that GSTO1 is critical for signaling by the TLR4 agonist LPS,
which is a critical mediator of septic shock3. Preliminary experiments on GSTO1 knockout
bone marrow derived macrophages (BMDMs) have identified a strong phenotype in TLR4
signaling. Our evidence suggests GSTO1 may act upstream of NF-kB signaling, and we are
currently assessing potential targets of GSTO1.
Mal is the main adaptor protein utilized by TLR1/2, TLR2/6 and TLR4 along with MyD88 to
elicit TLR signal transduction and induction of pro-inflammatory cytokines, including IL6,
IL12p40 and TNFα4. Mal contains unusual free cysteines that are not in disulfide bonds and
have the potential to be redox regulated. It is possible that GSTO1 is deglutathionylating these
cysteines in order for Mal to signal. This project will examine this possibility, in order to
determine the role of GSTO1 in signaling by TLR4.
Project Aims:
The aim of this project is to characterize the targeting of Mal by GSTO1 in order to further
implicate GSTO1 as a critical regulator of TLR4 signaling. There will be 3 main aims:
1.
Mutant forms of Mal in which critical amino acids that flank the key glutathionylated
cysteines will be tested in various functional assays, notably the ability to bind another key
adapter in TLR4 signaling MyD88, and also for NF-kB activation. In other proteins, flanking
amino acids have been shown to be required for cysteine glutathionylation.
2.
Whether the mutants are unable to undergo glutathionylation will be tested.
3.
A GSTO1 inhibitor (ML175) will be tested in TLR4 signaling, particularly on the
induction of cytokines by LPS.
Methods:
48
Several techniques will be used, including cell culture with immortalized and primary cell
lines, co-immunoprecipitations, NF-kB luciferase assays, bacterial cell culture, SDS-PAGE,
western blotting, plasmid generation, cytotoxicity assays and qPCR.
Predicted Outcome:
This project could strongly support the critical role of GSTO1 in signaling by TLR4. Since
TLR4 is a key therapeutic target in diseases such as sepsis and also inflammatory diseases
such as rheumatoid arthritis, this project might therefore provide new possibilities for
therapeutic intervention in these diseases where unmet medical need is great.
References:
1.
2.
3.
4.
Menon, D. et al. (2014). Free Radicals Biology and Medicine. 73, 318-327.
O’Neill LA. et al. (2013). Nature Reviews Immunology. 13, 453-460.
Menon, D. et al. (2015). Journal of Cell Science. 128, 1982-1990.
Bernard, J and O’Neill LA. (2013). IUBMB Life. 65, 777-786
49
SS PROJECT NO.2
Supervisor: Dr. Zbigniew Zaslona and Prof. Luke O’Neill
Title: REGULATION OF M2 MACROPHAGES BY
BMAL1 – RELEVANCE TO ASTHMA/ALLERGY
THE CIRCADIAN PROTEIN
Background
Our body clock drives rhythms in the expression of genes that have a 24 hour periodicity. A
number of physiological processes including metabolism, cardiovascular and most importantly
for this project immune function [1] have a time dependancy that is modulated by the
circadian clock. The in-vivo response to an immune challenge, such as LPS (the gram
negative bacterial product that acts via TLR4), varies greatly with time [2] and we recently
published a paper demonstrating a reduction in macrophages in clock gene expression,
specifically in the gene encoding Bmal1, with LPS challenge [2]. LPS stimulated
macrophages are also called M1 macrophages, while those stimulated with the cytokine IL-4
are termed M2 macrophages. The molecular pathway responsible for M2 polarization is not
fully understood, and the role of the body clock in the process of M2 regulation has not been
explored. M2 macrophages are known to be critical for anti-parasitic responses and play roles
in allergy and asthma [3]. Asthma is of particular relevance to the area of circadian control of
immunity, since it is a disease with very strong clinical evidence suggesting regulation by
ciracadian variation [4]. This project will explore the role of the clock protein Bmal1 in the
regulation of M2 macrophages in order to provide added molecular insights into the control of
macrophage function by the clock. We have preliminary evidence indicating that a model of
allergic asthma in mice is more severe in Bmal1-deficient mice. This suggests that Bmal1
might limit M2 macrophage polarisation, since M2 macrophages are thought to play a role in
the disease process in this model. We will therefore explore the role of Bmal1 in M2
macrophage polarisation to provide further information on the role of the circadian clock in
asthma.
Project Aims
1. Initially, we will use bone marrow derived macrophages from wildtype and Bmal1-deficient
mice (BMAL1) and stimulate them with IL-4 for 24 h. Using qPCR we will estimate the
expression of M2 markers such as ym-1 or arginase to determine the role of Bmal1 in the M2
phenotype.
2. We will also test for IL-4 signalling in the macrophages, measuring STAT6
phosphorylation as a key process regulated by IL-4.
3. M2 macrophages are also capable of phagocytosis and this will be also be measured in wild
type and Bmal1-deficient cells.
4. If the availability of Bmal1-deficient mice is limiting, pharmacological approaches can be
taken, in particular the use of a Rev-Erb-alpha agonist [5], since Rev-Erb-alpha is a possible
downstream target for Bmal1 that could be responsible for the effects of Bmal1 here.
50
Methods: Key techniques that will be deployed will include cell culture, SDS-PAGE and
Western blotting, RNA extraction and gene expression analysis by SybrGreen. Also flow
cytometry analysis will be used to detect phagocytosis.
Possible outcome: This project will aim to reveal an importance of Bmal1 in the activation of
M2 macropages and their phagocytic capacity. This will offer new insights into the cross talk
between inflammatory and circadian systems in diseases such as asthma/allergy.
References
1. Curtis AM, Bellet MM, Sassone-Corsi P, O'Neill LA, Circadian clock proteins and
immunity. Immunity. 2014 Feb 20;40(2):178-86
2. Curtis, A.M., et al., Circadian control of innate immunity in macrophages by miR-155
targeting Bmal1. Proc Natl Acad Sci U S A, 2015. 112(23): p. 7231-6.
3.Byers DE, Holtzman MJ. Alternatively activated macrophages and airway disease. Chest.
2011 Sep;140(3):768-74
4. Durrington, H.J., et al., The circadian clock and asthma. Thorax, 2014. 69(1): p. 90-2.
5. Gibbs JE, Blaikley J, Beesley S, Matthews L, Simpson KD, Boyce SH, Farrow SN, Else KJ,
Singh D, Ray DW, Loudon AS. The nuclear receptor REV-ERBα mediates circadian
regulation of innate immunity through selective regulation of inflammatory cytokines. Proc
Natl Acad Sci U S A. 2012 Jan 10;109(2):582-7
51
SS PROJECT NO.3
Supervisor: Dr Marcin Baran and Prof. Andrew Bowie.
Title: DETERMINING THE ROLE OF THE ELONGATOR COMPLEX IN CYTOKINE
INDUCTION BY PATTERN RECOGNITION RECEPTORS.
Background
The Pyrin and HIN200 domain-containing (PYHIN) protein family consists of five proteins in
human and 13 proteins in mouse. Mouse AIM2 and IFI204 have been previously described to
be intracellular pattern recognition receptors (PRRs) for dsDNA, leading to inflammasome
activation and IFNβ induction respectively1, 2. However the function of most mouse PYHIN
proteins remains unknown. We have shown that the mouse PYHIN protein IFI207 is highly
expressed in myeloid cells, and in contrast to IFI204, has an intrinsic role in regulation of gene
induction of specific cytokines, independent of DNA sensing. Interestingly, IFI207
interactome analysis by mass spectrometry revealed that it interacted with the Elongator
complex, a protein complex conserved from yeast to human which has been implicated in
transcriptional regulation, but has not previously been shown to regualate inducible
cytokines3. Using RNA interference to knock down the expression of Elongator subunits such
as Elp2 in mouse cells we showed for the first time a role for Elongator in TNF induction.
Preliminary results also showed a role for Elongator in LPS-stimulated TNF induction in the
human monocytic cell line THP-1, even though there is no human ortholog of mouse IFI207.
Thus the Elongator complex may have a general, but previously unappreciated, role in
cytokine gene induction. Therefore the aim of this project is to charaterise the role of the
Elongator complex in TNF gene induction in human monocytic cells.
Specific Aims:
1.
Use RNA interference (shRNA) to determine the role of Elongator subunits in PRRstimulated TNF induction in THP-1 cells.
2.
Examine whether human PYHIN proteins cooperate with Elongator to induce the TNF
gene.
3.
Determine which human PYHIN proteins, if any, interact with the Elongator complex.
Methods:
THP-1 cells stably expressing control shRNA, or shRNA targeting Elp2 and Elp3 will be
stimulated with TLR ligands, or transfected with RNA or DNA, and TNF mRNA induction
and protein production measured by qRT-PCR and ELISA respectively. Cells will also be
immunoblotted for Elp expression in order to demonstrate knockdown of shRNA targets. The
effect of Elp knock-down on induction of other PRR-stimulated genes will also be determined.
In a reporter gene assay which directly measures the induction of the TNF promoter, the
ability of human PYHIN proteins, when overexpressed, to cooperate with Elps in inducing
TNF will be determined. RNA interference can also be used to determine whether any of the
four known human PYHIN proteins (AIM2, IFI16, MNDA and PYHIN1) are involved in
PRR-stimulated TNF induction. Finally, co-immunoprecipitation assays will be used in order
to determine whether, as in the case of mouse IFI207, human PYHIN proteins can interact
with the Elongator complex.
52
Overall this project will reveal new mechanistic insights into how the pro-inflammatory
cytokine TNF is regulated by PRR pathways in human cells.
References:
1. Unterholzner, L., Keating, S.E., Baran, M., Horan, K.A., Jensen, S.B., Sharma, S., Sirois,
C.M., Jin, T., Latz, E., Xiao, T.S., Fitzgerald, K.A., Paludan, S.R. & Bowie, A.G. 2010. Nat.
Immunol. 11, 997-1005.
2. Connolly, D & Bowie A.G. 2014. The emerging role of human PYHIN proteins in innate
immunity: implications for health and disease. Biochem Pharmacol. 92, 405-414.
3. Creppe, C & Buschbeck, M. 2011. Elongator: an ancestral complex driving transcription
and migration through protein acetylation. J Biomed Biotechnol. doi: 10.1155/2011/924898.
53
SS PROJECT NO.4
Supervisor: Dr Dympna Connolly and Prof. Andrew Bowie
Title: EXPLORING THE ROLE OF PYHIN-INTERACTING PROTEINS IN PYHIN
FUNCTION AND INNATE IMMUNITY
Background
The innate immune response depends on the ability of immune cells to detect pathogens
through germline-encoded pattern recognition receptors (PRRs). Recently discovered PRRs
include some members of the Pyrin and HIN domain (PYHIN) family, which are encoded on
an interferon-inducible gene cluster located on chromosome 1q231, 2. There are five human
PYHIN proteins; Absent in melanoma 2 (AIM2), IFN-g inducible protein 16 (IFI16), Myeloid
cell nuclear differentiation antigen (MNDA), Pyrin and HIN domain family member 1
(PYHIN1) and the recently identified Pyrin domain only protein 3 (POP3). Early studies
reported roles for these proteins in cell cycle control, tumour suppression and transcriptional
regulation. AIM2 and IFI16 have now been shown to be immune sensors of non-self DNA,
such as that produced by viruses in infected cells2, 3. AIM2 binds DNA to activate the
inflammasome, while IFI16 detection of DNA can lead to the up-regulation of type I
interferons (IFNs). Furthermore, we have recently shown that MNDA is required for the type I
IFN response to both pathogens, RNA and DNA in monocytes, since it transcriptionally
regulates IRF7 expression. These diverse functions of PYHIN proteins are likely mediated by
specific PYHIN interactions with distinct proteins, to regulate protein signalling and effector
networks within cells. In order to begin to elucidate mechanisms whereby PYHIN proteins
signal to altered gene expression, we performed unbiased affinity purification and mass
spectroscopy to identify PYHIN-interacting proteins. Many of the proteins discovered in these
PYHIN interactomes are predicted to have roles in gene regulation, and some such interactors
have also been discovered in a recent paper using a similar approach4. Thus the aim of the
project is to test the potential role of specific PYHIN interacting proteins in mediating
PYHIN function.
Specific Aims:
1.
Select shortlist of PYHIN interactors to examine.
2.
Determine which interactors are expressed in THP-1 and HT1080 cells (cells where
PYHIN function can be examined).
3.
Use loss-of-function approaches (siRNA) to examine the role of selected interactors in
AIM2, IFI16 and MNDA functions.
Methods:
Up to five PYHIN interactors which have been found to interact with specific subsets of
PYHIN proteins will be selected. For example SKIV2L2 interacts with MNDA and IFI16 but
not AIM2, while LAMTOR1 interacts with MNDA and AIM2 but not IFI16, and VPRBP
interacts with MNDA but not IFI16 nor AIM2. The expression of selected interactors in THP1 and HT1080 cells will then be examined, with and without cell stimulation, by qRT-PCR. In
cells where these interactors are shown to be expressed, siRNA oligonucleotides targeting
them will be designed and tested for efficacy. These siRNAs will then be used in loss-of-
54
function experiments to knock down specific interactors in order to assay their potential role
in PYHIN functions. To assay AIM2 function, DNA-stimulated IL-1b release will be
measured in THP-1 cells. To assay IFI16 function, DNA-stimulated type I IFN production will
be measured in THP-1 and/or HT1080 cells by bioassay. To assay MNDA function, IRF7
protein expression and IFNa induction after cell stimulation of THP-1 cells will be measured
by immunoblot and qRT-PCR respectively.
Overall this project will provide novel mechanistic insights into how PYHIN proteins
function in innate immunity.
References:
1. Dempsey, A. and Bowie, A.G. (2015) Virology 479-480: 146-152
2. Connolly, D & Bowie A.G. (2014) Biochem Pharmacol. 92, 405-414.
3. Unterholzner, L. et al (2010) Nat. Immunol. 11, 997-1005.
4. Diner et al (2015) Molecular Systems Biology 11, 787.
55
SS PROJECT NO.5
Supervisor: Dr Michael Carty & Prof Andrew Bowie
Title: DOES SARM OLIGOMERISATION HAVE A ROLE IN INHIBITION OF HIV-1
TRANSCRIPTION?
Background
Toll-like receptors (TLRs) are pattern recognition receptors that recognise the presence of
pathogen associated molecular patterns such as viral RNA and DNA within host cells1. TLRs
signal to altered gene expression via Toll-IL-1R (TIR) domain containing adaptor proteins
such as MyD88 and TRIF. Like MyD88 and TRIF, SARM is a cytosolic protein with a TIR
domain2. SARM is the most evolutionarily conserved TIR domain protein, and has been
shown to have roles both in TLR signaling but also more broadly in TLR-indpendent
functions such as gene regulation and cell death2. For example, we showed a role for human
SARM in the regulation of TRIF-dependent signalling3, while other roles for mammalian
SARM include promotion of neuronal and axonal death, restriction of West Nile virus
pathogenesis in the brain4, and in the induction of the chemokine CCL55.
Other anti-viral roles for SARM are likely to emerge and very recently we showed that SARM
also inhibits transcription of the HIV long terminal repeat (LTR). One mechanism whereby
the HIV protein TAT fulfils an essential role in the HIV life cycle, is by transcriptionally
regulating the HIV LTR, and we showed that overexpression of SARM inhibited TATdependent HIV LTR transcription, while knockdown of endogenous SARM by siRNA
enhanced TAT-mediated LTR activity in HEK293 cells. Thus SARM may be a novel inhibitor
of HIV infection, which may have relevance to virus-host interactions in HIV-1-infected cells.
Work from other groups has suggested that SARM may oligomerise after cell activation in
order to cause cell death2, however the mechanism whereby SARM inhibits TAT is unclear.
Therefore the aim of this project is to explore the mechanism whereby SARM inhibits
HIV-1 transcription and to test whether SARM oligomerisation is involved.
Specific Aims:
1.
Confirm the role of SARM in TAT inhibition using gain-of-function and loss-offunction approaches.
2.
Determine the protein domains in SARM required to inhibit TAT.
3.
Test whether the SARM domains required to inhibit TAT are required for SARM
oligomerisation.
Methods:
The effect of SARM overexpression (gain of function) and RNA interference targeting
endogenous SARM (loss of function) on the ability of TAT to enhance HIV transcription will
be assessed in a reporter gene assay which examines TAT-stimulated activation of a HIV-1LTR-Luc reporter. This will be confirmed in HEK293 cells and tested in an independent cell
type, namely HeLa. SARM has three types of defined protein motifs, namely two SAM
motifs, ARM repeats and a TIR domain2. In order to map the domains of SARM required to
inhibit TAT, different truncated forms of SARM will be tested for expression, and assessed
56
for the ability to inhibit TAT in the HIV LTR reporter gene assay. Thereafter, both full length
and truncated forms of SARM will be tested for oligomerisation using a cross-linking assay,
whereby protein crosslinking captures oligomeric protein complexes which can then be
visualised on SDS-PAGE by immunoblotting.
Overall this project will determine for the first time how SARM inhibits TAT-dependent
HIV LTR transcription.
References:
1.
Gürtler, C. & Bowie, A.G. (2013) Trends Microbiol. 21: 413-20.
2.
Panneerselvam and Ding (2015) International Reviews of Immunology, Early Online 113.
3.
Carty, M. et al (2006) Nat Immunol 7: 1074-81.
4.
Carty, M. et al (2014) Trends Immunol. 35: 79-87.
5.
Gürtler, C. et al (2014) J Immunol. 192: 4821-32.
57
SS PROJECT NO.6
Supervisor: Mag Needham, Rachel McLoughlin and Cliona O’ Farrelly
Title: IL8 AND HCV INFECTION
Project Outline
Background
Irish women who have been exposed to hepatitis C virus through contaminated anti-D
immunoglobulin products and who resisted infection provide a unique opportunity to study
possible innate immune pathways involved in resistance to HCV infection. Previous work
within the lab has discovered elevated levels of the IL8 cytokine in plasma samples from a
small cohort of these uninfected anti-D women. We believe these women to be innately
resistant to HCV infection and hypothesise that the increased expression of IL8 (a proinflammatory cytokine involved in neutrophil recruitment and activation) plays an integral
role in this innate immune resistance.
The human IL8 gene is well characterised and SNPs within its promoter region have
previously been shown to directly influence gene expression and have been linked with the
progression and outcome in different diseases. An IL8 gene promoter polymorphism (-251A/T
) has been associated with an increased risk of developing liver cirrhosis in people with
hepatitis B (Qin et al, 2012) and with susceptibility to bacterial infection (Garey et al. 2010).
Previously we have reported that genetic variation within the bovine IL8 promotor
differentially regulates IL8 expression and may have an impact on bovine susceptibility to
infectious disease and inflammation (Meade et al. 2012).
Hypothesis
We propose that IL8 has a role in HCV infection and that SNPs within the human IL8
promoter region are associated with HCV resistance possibly by influencing transcription
factor binding sites
Technologies
We will use sequence analysis to identify any IL8 promoter SNPs in our sample cohort and to
characterise the influence of any promoter haplotypes on IL8 expression levels using TLR
agonist and interferon stimulation of PBMCs. This project will also use bioinformatics based
studies to predict the possible impact of promoter region SNPs on transcriptional factor
binding in the human IL8 gene.
Finally, to explore the functional implications of elevated IL8 expression this project will
investigate the impact of IL8 on neutrophil function, specifically the role IL8 plays in inducing
neutrophil extracellular traps (NETs), which have recently been shown to have important antiviral roles (Jenne et al. 2015).
Implications
The findings from this project will help identify novel innate immune mechanisms responsible
for resistance to HCV infection which could influence development of new vaccination and
therapeutic strategies.
58
References:
Qin X. et al. The IL-8 gene polymorphisms and the risk of the hepatitis B virus/infected
patients. (2012) DNA Cell Biology 31, 1125-1130.
Garey K. W. et al. A common polymorphism in the interleukin-8 gene promoter is associated
with an increased risk for recurrent Clostridium difficile infection. (2010) Clinical Infectious
Diseases (51)1406-1410.
Meade K. G. et al. Functional characterisation of bovine interleukin 8 promoter haplotypes in
vitro (2012) Molecular Immunology 50, 108-116.
Jenne C. N. et al. Virus-Induced NETs – Critical Component of Host Defense or Pathogenic
Mediator? (2015) PLoS Pathogens 11, e1004546.
59
SS PROJECT NO.7
Supervisor: Prof Cliona O’ Farrelly
Title: THE EFFECT OF THE IMMUNOSUPPRESSIVE DRUG TACROLIMUS ON
HEPATIC NATURAL KILLER CELL PHENOTYPE AND FUNCTION: RELEVANCE
TO GRAFT TOLERANCE.
Introduction:
Liver transplantation is a life saving operation for patients with end stage liver disease.
Recipients must take powerful immunosuppressive medication in order to prevent graft
rejection, side effects of which include malignancy, cardiovascular disease and renal failure.
Tacrolimus is the most commonly used of these drugs. Belonging to the calcineurin inhibitor
family, it prevents T cell activation by blocking the phosphorylation of NFAT. However it is
unclear how this drug affects innate lymphoid cells which play important roles in transplant
rejection and tolerance. The adult human liver contains significant populations of innate
immune cells, especially NK cells. The liver is a naturally tolerogenic organ and can be
transplanted without HLA matching. It is hypothesised that immune cells in the donor organ
mediate this tolerance. We believe hepatic NK cells from the donor organ are key. However, it
is unclear how immunosuppression might interfere.
Objectives:
To identify the effects of tacrolimus on hepatic NK cell phenotype and function.
Methodology:
Molecular and cellular techniques will be used in the project, including cell culture, in vitro
cytotoxicity assays and flow cytometry. These techniques will be optimised using NKL cells,
a cell line derived from leukemic NK cells and then used to test stored hepatic NK cell
populations from liver transplants. The effect of tacrolimus on hepatic NK cell cytotoxicity
and IFN-g production will be assessed by a CD107a assay. The expression of natural
cytotoxicity receptors (NCRs) will also be assessed to determine if immunosuppression has an
effect on hepatic NK cell phenotype.
Likely outcome:
This is the first time that the effect of immunosuppression on liver resident NK cells has been
assessed. The data will be an important component of an ongoing research project into the role
of hepatic NK cells in graft tolerance post transplantation.
References:
1.
Harmon C et al. Am J Transplantation 2015 in press
2.
Lysakova-Devine T, O’Farrelly C. J Leukoc Biol 2014 Dec 96(6):981–90.
3.
Nemeth, E., Baird, A. W., & O’Farrelly, C. Seminars in Immunopathology, 2009
31(3), 333–43.
4.
Moroso V et al. Liver Transplant 2010;16:895–908.
5.
Benitez C et al. Hepatology. 2013;58(5):1824–35.
60
6.
7.
Meehan AC et al.. PLoS One. 2013;8(3):e60144.
Mukherjee & Mukherjee. Journal of Transplantation 2009 1-20
61
SS PROJECT NO.8
Supervisor: Ms. Anne Barry-Reidy and Prof. Cliona O’Farrelly
Title: HOST DEFENCE PROTEINS IN THE FEMALE REPRODUCTIVE TRACT
Introduction:
Viral, bacterial, yeast and parasitic infection of the female reproductive tract is the most
common cause of pathology in young females. The reproductive tract is equipped with a
complex and diverse immune repertoire to provide protection against these challenges. Our
group is particularly interested in the role of host defence peptides, important innate immune
molecules, and have focused in the past on defensins
The multifaceted nature of these peptides means their activity in the female reproductive tract
(FRT) has received attention in recent years (Narciandi, Lloyd, Meade, & O’Farrelly, 2013;
Tribe, 2015). The female tract is immunologically complex- it must tolerate the influx of
foreign sperm and the implantation of an embryo, yet respond to pathogens which enter as a
result of intercourse and labour. In addition, labour represents a significant level of tissue
injury which must be resolved efficiently. Elafin, secretory leukocyte protease inhibitor (SLPI)
and human epididymal protein 4 (HE4) members of the WFDC family are expressed
throughout the human female reproductive tract (FRT) (Frew & Stock, 2011; Yarbrough,
Winkle, & Herbst-Kralovetz, 2015). Elafin mRNA is increased in human endometrial
epithelial cells treated with IL-1β and TNFα (King, Critchley, Sallenave, & Kelly, 2003). We
aim to examine expression on these genes in endometrium from women with endometriosis.
In the first instance, technologies will be optimised using bovine tissues. Our group has
demonstrated that elafin and WFDC18 are upregulated in uterine biopsies taken from cows 7
days post-partum (dpp) relative to 21 dpp. Elafin is also upregulated in animals with a uterine
infection relative to healthy animals 21 dpp. This suggests these proteins may be involved in
pathogen clearance, wound repair and the re-establishment of homeostasis after labour. As
prolonged post-partum inflammation is particularly common in the bovine and delays the
return to a fertile state, we are interested in the role of WFDC proteins in the bovine FRT.
Quantitative PCR will be used to detect expression of selected WFDC proteins in a biobank of
bovine FRT tissues including vagina, cervix, uterus, fallopian tube and ovary. In addition,
material will be made available from primary bovine endometrial stromal and epithelial
culture experiments and from women with endometriosis. We also propose to use publiclyavailable genome databases to characterise the WFDC locus in the human and bovine
genomes.
Objectives:
To determine the presence of WFDC proteins within a range of female reproductive tract
tissues and within endometrial stromal and epithelial cells.
Methodology:
RNA extraction and cDNA synthesis from bovine reproductive tract tissues.
62
Primer design and qPCR for whey acidic protein genes.
Likely outcome:
Whey acid proteins are expressed in several tissues from the bovine female reproductive tract
References:
Frew, L., & Stock, S. J. (2011). Antimicrobial peptides and pregnancy. Reproduction, 141(6),
725–735. http://doi.org/10.1530/REP-10-0537
King, A. E., Critchley, H. O. D., Sallenave, J.-M., & Kelly, R. W. (2003). Elafin in human
endometrium: an antiprotease and antimicrobial molecule expressed during menstruation. The
Journal
of
Clinical
Endocrinology
and
Metabolism,
88(9),
4426–4431.
http://doi.org/10.1210/jc.2003-030239
Narciandi, F., Lloyd, A., Meade, K. G., & O’Farrelly, C. (2013). A novel subclass of bovine
β-defensins links reproduction and immunology. Reproduction, Fertility and Development.
Retrieved from http://dx.doi.org/10.1071/RD13153
Tribe, R. M. (2015). Small Peptides with a Big Role: Antimicrobial Peptides in the Pregnant
Female Reproductive Tract. American Journal of Reproductive Immunology, 74(2), 123–125.
http://doi.org/10.1111/aji.12379
Yarbrough, V. L., Winkle, S., & Herbst-Kralovetz, M. M. (2015). Antimicrobial peptides in
the female reproductive tract: a critical component of the mucosal immune barrier with
physiological and clinical implications. Human Reproduction Update, 21(3), 353–377.
http://doi.org/10.1093/humupd/dmu065
63
SS PROJECT NO.9
Supervisor: Dr. Stephanie Longet and Dr. Ed Lavelle
Title: ANALYSIS OF INTESTINAL IMMUNE RESPONSES FOLLOWING ORAL
ADMINISTRATION OF THE INTK CELL ACTIVATOR Α-GALACTOSYLCERAMIDE
Introduction:
Enteric infections are a major worldwide health problem causing high levels of mortality and
morbidity especially in developing countries (1). Oral vaccination is regarded as the optimal
means to fight intestinal infections as it elicits local immune responses including the secretion
of IgA, shown to be particularly important in protection from enteric infections. One of the
most important challenges in oral vaccination is the generally poor immunogenicity of orally
delivered antigens (2, 3). In order to increase immunogenicity, the presence of an adjuvant is
required. Even though many oral adjuvants have been evaluated, as yet none has been licenced
for human use (4). Consequently, it remains crucial to find suitable oral adjuvants capable of
enhancing intestinal antigen-specific IgA responses that could be potentially used in humans.
The glycolipid α-GalactosylCeramide (α-GalCer), an invariant natural killer cell activator, was
shown to be a powerful mucosal adjuvant in animal models (5). In the lab, we have
demonstrated the ability of α-GalCer to increase intestinal antigen-specific IgA responses in
mice in the context of an oral vaccine against enterotoxigenic Escherichia Coli (ETEC) that
we are developing (6-9). However, we lack a comprehensive understanding of how orally
administered α-GalCer increases antibody responses and this project aims at addressing this
deficiency. The effects of α-GalCer on B cells in the principal intestinal lymphoid tissues,
Peyer’s patches and mesenteric lymph nodes will be determined in vivo and in vitro. In
addition, the release of key cytokines involved in IgA class switching and secretion including
IL-17 and TGF-β will be measured post-administration of oral α-GalCer in intestinal tissues
and washes. Finally, the impact of oral administered α-GalCer on the expression and
localisation of polymeric Ig receptor (pIgR) on intestinal epithelial cells will be assessed. This
receptor is directly involved in IgA transcytosis into the intestinal lumen.
Objectives:
•
To assess modifications in B cell activation in gut associated lymphoid tissues
following oral administration of α-GalCer or in the presence of α-GalCer in vitro.
•
To examine changes in secretion of specific IgA promoting cytokines in intestinal
tissues and washes post-administration of oral α-GalCer.
•
To evaluate the expression and localisation of pIgR on intestinal epithelial cells
following oral administration of α-GalCer.
Methodology:
A range of state-of-the art immunological techniques will be used in the project. At several
time-points after oral administration of α-GalCer in mice, cells will be isolated from Peyer’s
patches and mesenteric lymph nodes. For comparison and to address systemic immune
responses, the B cell analysis will also be performed with splenocytes. Modifications in
64
expression of B cell activation markers will be evaluated by FACS analysis. The same type of
experiments will be performed with cells isolated from Peyer’s patches, mesenteric lymph
nodes and spleen of naïve mice that will be stimulated with α-GalCer in vitro. IL-17 and
TGF-β released in intestinal tissues and washes will be measured after oral administration of
α-GalCer by ELISA. For comparison, the release of these cytokines will also be measured in
serum. The expression of pIgR will be evaluated on intestinal tissues collected after oral
administration of α-GalCer by immunohistochemistry.
Likely outcome:
The project will give us a better understanding of the effects of α-GalCer on intestinal
responses related to IgA production/secretion after oral administration. It will be the first study
towards a detailed understanding of how orally administered α-GalCer increases intestinal IgA
responses.
References:
1) Cheng, A.C. et al.Infectious diarrhea in developed and developing countries. J
ClinGastroenterol, 2005. 39(9): p. 757-73.
2) Czerkinsky, C. and Holmgren, J. Enteric vaccines for the developing world: a challenge for
mucosal immunology. Mucosal Immunol, 2009.2(4): p. 284-7.
3) Pasetti, M.F. et al. Immunology of gut mucosal vaccines. Immunol Rev, 2011.239(1): p.
125-48.
4) Rhee, J.H. et al.Mucosalvaccineadjuvantsupdate. ClinExpVaccine Res, 2012 Jul;1(1):5063.
5) Courtney, A.N. Characterization of alpha-galactosylceramide as a mucosal adjuvant. UT
GSBS Dissertations and Theses (Open Access), Texas Medical Center Library, Digital
Commons@The
Texas
Medical
Center,
2010.
Available
online
at:http://digitalcommons.library.tmc.edu/cgi/viewcontent.cgi?article=1085&context=utgsbs_di
ssertations
6) Quadri, F. et al. EnterotoxigenicEscherichia coli in Developing Countries: Epidemiology,
Microbiology, Clinical Features, Treatment, and Prevention, ClinMicrobiol Rev, 2005
Jul;18(3):465-83.
7) Croxen, M.A. andFinlay B.B.Molecularmechanisms of Escherichiacolipathogenicity.Nat
Rev Microbiol, 2010 Jan;8(1):26-38.
8) Tobias, J., et al., Construction of non-toxic Escherichia coli and Vibrio cholerae strains
expressing high and immunogenic levels of enterotoxigenic E. coli colonization factor I
fimbriae. Vaccine, 2008.26(6): p. 743-52.
9) Lundgren, A., et al., Clinical trial to evaluate safety and immunogenicity of an oral
inactivated enterotoxigenic Escherichia coli prototype vaccine containing CFA/I
overexpressing bacteria and recombinantly produced LTB/CTB hybrid protein. Vaccine,
2013.31(8): p. 1163-70.
65
SS PROJECT NO. 10
Supervisor: Dulce Bento, Filipa Lebre and Dr. Ed Lavelle
Title: ACTIVATION OF DENDRITIC CELLS USING POROUS NANOPARTICLES TO
TARGET IMMUNOMODULATORS
Introduction
The use of particles as vaccine adjuvants offers several advantages including enhanced uptake
by antigen presenting cells, protection of antigen from degradation, promotion of a depot
effect with gradual release of the antigen and the ability to facilitate antigen cross-presentation
and co-deliver of antigens and immunomodulators to the same cell population [1]. Properties
including particle size, surface chemistry, charge and shape are important for the
immunomodulatory effects of particulates [2].
Recently, mesoporous silica nanoparticles (MSNs) have received significant attention for drug
and vaccine delivery [3]. Their advantages include biocompatibility, surface functionalization,
the ability to fine-tune particle and pore size and geometry, a high surface area that allows the
efficient loading of drugs and antigens molecules and increased stability when compared with
other polymer-based carriers. However, the effect of particle pore size in the induction of
innate and adaptive immune responses has not been addressed.
In this project, the influence of pore size of silica nanoparticles on the innate immune response
will be assessed using 200 nm porous and non-porous particles. Additionally, the ability of
these nanoparticles to improve the immunomodulatory properties of TLR agonists will be
tested. Nanoparticle size was selected based on data from our lab showing that showed that of
a range of particle sizes (50 nm to 100 µm), the smaller particles were optimal for induction of
both CD4+ T and CD8+ T cell response.
Objectives:
•
To assess whether MSN can activate dendritic cells and macrophages and determine
the impact of pore size of MSN on cytokine production and maturation
•
To evaluate the capacity of MSN to efficiently delivery TLR ligands to dendritic cells
and to compare ligands for plasma membrane and endosomal TLRs
•
To assess the innate immune response induced by the injection of unmodified or TLR
ligand-loaded MSN
Methodology: Cell culture techniques will be employed to stimulate murine bone-marrow
derived dendritic cells (BMDCs) and bone-marrow derived macrophages (BMDMs) with
particles. The cell supernatants will be collected and cytokines measured by ELISA. Cell
maturation and cell death will be analysed by flow cytometry. The innate immunomodulatory
effects of the most promising strategies will be evaluated 24 h after intraperitoneal injection in
vivo. Peritoneal exudate and cells, draining lymph nodes and spleens will be collected and
innate immune response will be assessed by flow cytometry analysis assessed by FACs,
cytokine production after restimulation with various stimuli (e.g. TLR agonists) by ELISA.
66
Likely Outcome: This study will provide valuable new information on how the pore size of
porous particulate adjuvants impacts on the induction of innate immune responses and of how
mesoporous particles synergize with immunomodulators. This has relevance for the rational
design of novel particle based adjuvants.
References
[1] V.E. Schijns, E.C. Lavelle, Trends in vaccine adjuvants, Expert review of vaccines, 10
(2011) 539-550.
[2] D.M. Smith, J.K. Simon, J.R. Baker, Jr., Applications of nanotechnology for immunology,
Nature reviews. Immunology, 13 (2013) 592-605.
[3] H. Vallhov, N. Kupferschmidt, S. Gabrielsson, S. Paulie, M. Stromme, A.E. GarciaBennett, A. Scheynius, Adjuvant properties of mesoporous silica particles tune the
development of effector T cells, Small, 8 (2012) 2116-2124.
67
SS PROJECT NO. 11
Supervisor: Dr Natalia Muñoz-Wolf and Dr Ed Lavelle
Title: INFLUENCE OF CHARGE IN THE ADJUVANTICITY OF NANOCONJUGATES
BASED ON TLR LIGANDS.
Introduction:
Vaccine industry has shifted from attenuated or inactivated vaccines towards safer subunit
formulations based on purified antigens. Although safer, subunit vaccines are less
immunogenic and require adjuvants to boost its efficacy. Particularly, there is a pressing need
for generation of adjuvants able to trigger robust Th1 and Th17 cell-mediated immunity
necessary to protect against pathogens like TB, HIV or pneumococcus [1]. Nanotechnology is
a valuable tool for adjuvant development since nanoparticles (NPs) can display intrinsic
adjuvanticity, act as antigen delivery systems and be tailored for specific cell-targeting. These
systems are attractive because they allow the delivery of antigens to specific immune cells
enhancing cellular responses [2]. Particulates can activate the NLRP3 inflammasome inducing
secretion of the Th17-polarising cytokine IL-1 [3]. Pathogen-associated molecular patterns
(PAMPS) such as nucleic acid sensor, Toll-like or NOD-like receptor agonists can also act as
immunopotentiators [4]. Therefore, generation of tailored NPs which can simultaneously
target and activate immune cells is of great interest for adjuvant development.
We have generated a new candidate adjuvant based on conjugation of the TLR5/NLRC4
agonist Flagellin (FliC) to negatively charged nanoparticles. The nanoconjugate NP-FliC
showed enhanced ability to activate antigen-presenting cells by enhancing secretion of
proinflammatory cytokines, expression of costimulatory molecules and inflammasome
activation. NP-FliC also activated human mononuclear cells, pointing to the potential of this
novel nanoparticulate adjuvant for injectable and mucosal vaccines.
Physicochemical characteristics of particles such as size, charge, shape, polymer composition,
hydrophobicity, define hoe the particles interact with the immune system [5] and
understanding this is crucial for rational design of adjuvants and vaccines. Our current
understanding is that cationic NPs are more immunogenic than negative particles. On the other
hand, positively charged NPs are generally more toxic. Hence we aim to explore how NP
charge can affect adjuvanticity and immunostimulatory properties of our novel adjuvant NPFliC.
Objectives:
1.
To study the effect of surface charge on adjuvant potential of flagellin-coated
nanoparticles by assessing their capacity to induce maturation and activation of antigen
presenting cells (APCs).
2.
To study the importance of conjugation vs adsorption of flagellin in the
immunostimulatory effect of the candidate adjuvant.
3.
To evaluate functional capacity of APCs stimulated with particles of different charges
to drive differentiation of Th1 and Th17 cells in vitro.
68
Methodology:
This project will involve a number of state-of-the-art techniques including cell culture of
primary cells, evaluation of cytokine gene expression by retrotranscription/real-time PCR and
flow cytometry for evaluation of surface marker expression and cell death. Secretion of
cytokines will be also measured by ELISA in culture supernatants.
Likely outcome:
The results obtained from this project will contribute to our understanding in the molecular
mechanisms involved in adjuvanticity of particulate adjuvants. Also they will complement the
in vivo studies that will be carried out in parallel to address the adjuvanticity of FliC-NPs
when administered in combination with relevant antigens.
References:
1.
Rappuoli, R. and A. Aderem, A 2020 vision for vaccines against HIV, tuberculosis and
malaria. Nature, 2011. 473(7348): p. 463-9.
2.
Zhao, L., et al., Nanoparticle vaccines. Vaccine, 2014. 32(3): p. 327-37.
3.
McNeela, E.A. and E.C. Lavelle, Recent advances in microparticle and nanoparticle
delivery vehicles for mucosal vaccination. Curr Top Microbiol Immunol, 2012. 354: p. 75-99.
4.
Fujita, Y. and H. Taguchi, Overview and outlook of Toll-like receptor ligand-antigen
conjugate vaccines. Ther Deliv, 2012. 3(6): p. 749-60.
5.
Yue, H. and G. Ma, Polymeric micro/nanoparticles: Particle design and potential
vaccine delivery applications. Vaccine, 2015.
69
SS PROJECT NO. 12
Supervisor: Vanessa Ziaitz Bittencourt, Dr David Finlay and Dr Clair Gardiner
Title: TO DEFINE THE IMPACT OF INHIBITOR RECEPTOR SIGNALLING ON
CYTOKINE INDUCED GLYCOLYSIS IN NK CELLS
Background: It is becoming clear that immune cells undergo dynamic and dramatic
metabolic changes in response to stimulation. Furthermore, metabolism can impact on the
immune response of cells and regulate their effector functions(1).
Natural Killer (NK) cells are lymphocytes that kill virally infected and cancer cells. Their
functions are regulated by cytokines, and by a range of cell surface receptors that serve to
either activate or inhibit NK cells. Our recent work (and that of others) on NK cells has
shown that they can upregulate both glycolysis and oxidative phosphorylation (oxphos) in
response to cytokine stimulation and that mTORC1 is important in the glycolytic response (2,
3). We have also recently found that in the mouse, co-stimulation of activating receptors
alongside cytokine primes NK cells for IL2 activation of metabolism (unpublished). In this
project, we now to wish to investigate the impact of engaging activating receptors on cytokine
induced metabolic changes in human NK cells.
Objectives:
To investigate the hypothesis that engagement of activating receptors on NK cells attenuates
cytokine induced changes in glycolysis.
Methods:
Peripheral blood mononuclear cells (PBMC) containing NK cells will be isolated from buffy
coat preparation. Cells will be stimulated with different cytokine combinations in the presence
or absence of antibody to engage activating receptors. The effect on phenotype and
metabolism will be investigated. Outputs measured will include CD71 and CD98 expression,
glucose uptake, glucose receptor expression, IL2R expression and potentially some detailed
metabolic analysis using the Seahorse analyser.
Outputs:
These experiments will define if specific activating signals to NK cells modulate metabolic
changes induced in response to cytokine.
References:
1.
Pearce, E. L., and E. J. Pearce. 2013. Metabolic pathways in immune cell activation
and quiescence. Immunity 38: 633-643.
2.
Donnelly, R. P., R. M. Loftus, S. E. Keating, K. T. Liou, C. A. Biron, C. M. Gardiner,
and D. K. Finlay. 2014. mTORC1-dependent metabolic reprogramming is a prerequisite for
NK cell effector function. Journal of immunology 193: 4477-4484.
3.
Marcais, A., J. Cherfils-Vicini, C. Viant, S. Degouve, S. Viel, A. Fenis, J. Rabilloud,
K. Mayol, A. Tavares, J. Bienvenu, Y. G. Gangloff, E. Gilson, E. Vivier, and T. Walzer. 2014.
70
The metabolic checkpoint kinase mTOR is essential for IL-15 signaling during the
development and activation of NK cells. Nature immunology 15: 749-757.
71
SS PROJECT NO. 13
Supervisor: Clair Gardiner, Orla Sheils and John O’Leary
Title: ELUCIDATING THE ROLE OF CD96 IN HUMAN NK CELLS
Natural Killer (NK) cells are lymphocytes that kill cancer cells and virally infected cells.
Their functions are regulated by activating and inhibiting receptors that they express. Among
the recently described signalling pathways described is the CD155/CD112CD226/CD96/TIGIT axis. CD155 and CD112 are expressed on target cells while CD226,
CD96 and TIGIT are expressed on NK cells (1). These molecules provide ligands for CD226,
CD96 and TIGIT. CD226 is known to function as an activating receptor after ligation with
CD155 while evidence supports that TIGIT inhibits NK cell functions. However, it is not yet
clear the role that CD96 plays in human NK cells. It has been described as a co-stimulatory
receptor in human NK cells and as an inhibitory receptor in murine NK cells (1). In this series
of experiments, we aim to define the function of CD96 in human NK cells and investigate if it
can be regulated by the pleiotropic cytokine, TGFβ.
Objectives:
o
Using PBMC from buffy coats, we will cross-link the CD96 receptor, CD226 or TIGIT
on human NK cells and measure the impact on direct activation using CD69 and functional
read outs.
o
We will titre cytokine to determine suboptimal IL2 or IL12 cytokine stimulation of
IFNγ production by NK cells and then look for the ability of CD96 to co-activate or indeed,
inhibit NK cells. CD226 and TIGIT will function as control molecules.
o
Use of rCD155 will be used to confirm results obtained with cross-linking antibodies.
o
A role of TGFβ in the system will be investigated by testing if rTGFβ modulates CD96
(or other important receptors) expression on human NK cells.
Likely Outcome:
These data will identify a function for CD96 as either activating or inhibitory in human
NK cells.
References
1.
Chan, C. J., L. Martinet, S. Gilfillan, F. Souza-Fonseca-Guimaraes, M. T. Chow, L.
Town, D. S. Ritchie, M. Colonna, D. M. Andrews, and M. J. Smyth. 2014. The receptors
CD96 and CD226 oppose each other in the regulation of natural killer cell functions. Nat
Immunol 15: 431-438.
72
SS PROJECT NO. 14
Supervisor: Dr Caroline Sutton, Dr Annie Curtis and Prof Kingston Mills
Title: THE ROLE OF THE CLOCK GENE BMAL1 IN DENDRITIC CELL ACTIVATION
Introduction: Our internal body clock drives expression of cassettes of mRNA and protein
that have a 24 h periodicity, and this is termed circadian expression. The central clock resides
in the suprachiasmatic nucleus but functioning clocks have been identified in the heart, liver,
vasculature and more recently in immune compartments such as the spleen. It has recently
been demonstrated that macrophages (Mθ) have circadian gene expression controlled by clock
genes and that these 24 h cycles within the Mθ regulate the inflammatory nature of these cells
(1), inflammatory cytokines and the pro-inflammatory microRNA MiR-155 (2). These
oscillations in innate immune cell activation coincide with fluctuations in pro-inflammatory
markers such as IL-1 and IL-6. One such clock gene, Bmal1 appears to limit pro-inflammatory
signalling in Mθ. Bmal1Lox/Lox Lyz2Cre mice (Bmal1-/-), in which the clock gene Bmal1 has
been knocked out of innate immune cells, show a hyper inflammatory environment associated
with accumulation of inflammatory monocytes (1). Furthermore, another clock gene has been
shown to be important for the development of inflammatory adaptive Th17 responses (3). We
have shown that under inflammatory conditions, such as stimulation with Mycobacterium
tuberculosis (Mtb), that the absence of Bmal1 in innate cells leads to hyper activation of both
Mθ and dendritic cells (DC) and in the case of DC that this is associated with a significant
increase in the Th1 and Th17 polarising cytokines IL-12p70 and IL-23p19 (Sutton, Mills and
Curtis unpublished). Furthermore, this increase promotes IL-17 and IFN-γ production from
CD4 T cells. We have also found that the loss of Bmal1 from innate immune cells can lead to
the exacerbation of experimental autoimmune encephalomyelitis (EAE), a mouse model for
multiple sclerosis.
Objectives:
•
To examine whether bone marrow derived dendritic cells (BMDC) from Bmal1-/- have
an enhanced pro-inflammatory phenotype, such as upregulated co-stimulatory molecule
expression and chemokine production, compared with WT BMDC at both a basal level and
under inflammatory conditions.
•
To evaluate whether anti-inflammatory pathways, such as PD-L1 and IL-10
expression, is down regulated in Bmal1-/- BMDC.
Methodology:
FACS analysis, ELISA and real time PCR will be used to examine CD11c+ BMDC for their
expression of both pro and anti-inflammatory associated markers under inflammatory and
non-inflammatory conditions. The project with require cell culture, RNA isolation, FACS
staining as well as FACS sorting of CD11c+ cells.
Likely outcome;
The results of this study should provide new information on how clock genes may influence
the immune response to infection and will have implications on best timing of treatment with
anti-inflammatory therapeutics.
73
References:
1)
Nguyen, K. D. et al, Circadian gene Bmal1 regulates diurnal oscillations of Ly6Chi
inflammatory monocytes. Science, Vol 341; 1483-1488. 2013.
2)
Curtis, A.M et al, Circadian control of innate immunity in macrophages by miR-155
targeting Bmal1. PNAS, Vol 112; 7231-7236. 2015
3)
Yu, X et al, TH17 cell differentiation is regulated by the circadian clock. Science. Vol
342; 727-730. 2013.
74
SS PROJECT NO. 15
Supervisor: Dr Alicja Misiak, Ms Aideen Allen and Professor Kingston Mills
Title: SYNERGY BETWEEN TLR AND STING AGONISTS IN PROMOTING INNATE
RESPONSES, WHICH DRIVE TH1 AND TH17 CELLS THAT PROTECT AGAINST
BACTERIAL INFECTION
Introduction:
Innate immune responses that help to control infection are activated through pattern
recognition receptors (PRR) that recognize a range of pathogen-associated molecular patterns
(PAMPs) expressed by microbial pathogens (1). These PPRs include Toll-like receptors
(TLR), such as TLR4 that recognizes lipopolysaccharides (LPS) from Gram-negative bacteria,
TLR9 that binds bacterial CpG, TLR2 that bind bacterial lipoproteins, and intracellular nucleic
acid receptors, including stimulator of IFN genes protein (STING), that bind nucleic acids and
their derivatives. STING plays a critical role in response to cytosolic DNA and unique
bacterial nucleic acids called cyclic dinucleotides and is involved in promoting type I IFN
production during infection (2, 3).
The bacterial pathogen Bordetella pertussis causes whooping cough (pertussis), a severe
respiratory infection in infants and young children. The incidence of infection has been
increasing in recent years with epidemics in many countries including Ireland (4). This is
thought to reflect a failure of the current acellular pertussis (aP) vaccine to induce optimum
immunity, especially Th1 and Th17 cells that are crucial for clearing bacteria from the
respiratory tract (5). The current aP vaccine is composed of 3 pertussis antigens formulated
with alum as the adjuvant. Studies in the Mills lab have shown that formulating the antigens
with TLR9 or TLR2 agonists enhances efficacy of the vaccine by promoting Th1 responses (5,
6). Preliminary data has suggested that it may be possible to enhance this further by
combining TLR and STING agonists as adjuvants (Allen et al unpublished). Although
activation of nucleic acid sensing pathway has traditionally been associated with type I IFN
production, we have evidence that STING, in combination with TLR agonists, can induce
potent IL-1, IL-23 and IL-12 production and co-stimulatory molecule expression on dendritic
cells (DCs) and thereby promote Th1 and Th17 responses.
Objectives:
•
To examine the synergy between STING and TLR agonists in activating DCs and
macrophages to produce IL-1, IL-12 and IL-23 production and CD80 and MHC class II
expression.
•
To examine the signalling pathways in DCs and macrophages activated by TLR and
STING agonists.
•
To examine the capacity of TLR and STING co-activated DCs to promote Th1 and
Th17 responses in vitro
Methodology:
A range of immunology techniques will be used in the project, including culture of dendritic
cells and macrophages, ELISA, real time PCR, Western blotting and intracellular staining and
75
FACS analysis. Small molecule inhibitors or siRNA specific for signalling
molecules/pathways (NFκB, ERK, MAP kinase, IRF1, IRF8, NLRP3) will be utilized to
assess the possible pathway involved in the synergy between TLR and STING agonism. T
cells from T cell receptor transgenic mice (cells prepared by licensed Postdoc in the lab) will
be used to assess the capacity of TLR and STING activated DCs to promote antigen-specific
Th1 and Th17 responses.
Likely outcome:
The long term objective is to design a more effective vaccine against whopping cough. This
project will assist in this goal by providing important information on the mechanism of
activity of two PAMPs that work in synergy as adjuvants to promote protective Th1 and Th17
responses against B. pertussiss.
References
1.
Kawai, T., and S. Akira. 2009. The roles of TLRs, RLRs and NLRs in pathogen
recognition. Int Immunol.
2.
Burdette, D. L., K. M. Monroe, K. Sotelo-Troha, J. S. Iwig, B. Eckert, M. Hyodo, Y.
Hayakawa, and R. E. Vance. 2011. STING is a direct innate immune sensor of cyclic di-GMP.
Nature 478: 515-518.
3.
Barker, J. R., B. J. Koestler, V. K. Carpenter, D. L. Burdette, C. M. Waters, R. E.
Vance, and R. H. Valdivia. 2013. STING-dependent recognition of cyclic di-AMP mediates
type I interferon responses during Chlamydia trachomatis infection. MBio 4: e00018-00013.
4.
Mills, K. H., P. J. Ross, A. C. Allen, and M. M. Wilk. 2014. Do we need a new vaccine
to control the re-emergence of pertussis? Trends Microbiol 22: 49-52.
5.
Ross, P. J., C. E. Sutton, S. Higgins, A. C. Allen, K. Walsh, A. Misiak, E. C. Lavelle,
R. M. McLoughlin, and K. H. Mills. 2013. Relative contribution of Th1 and Th17 cells in
adaptive immunity to Bordetella pertussis: towards the rational design of an improved
acellular pertussis vaccine. PLoS Pathog 9: e1003264.
6.
Dunne, A., L. A. Mielke, A. C. Allen, C. E. Sutton, R. Higgs, C. C. Cunningham, S. C.
Higgins, and K. H. Mills. 2015. A novel TLR2 agonist from Bordetella pertussis is a potent
adjuvant that promotes protective immunity with an acellular pertussis vaccine. Mucosal
Immunol 8: 607-617.
76
SS PROJECT NO. 16
Supervisor: Dr Joseph DeCourcey, Dr Mieszko Wilk and Prof Kingston Mills
Title: THE ROLE OF EPITHELIAL CELLS IN THE IMMUNE RESPONSE TO
BACTERIA AND THEIR PRODUCTS.
Introduction:
The mucosal surfaces in the body are lined with epithelial cells that are often the targets for
binding of invading pathogens but also play a role in protective immunity to infection. They
recognise bacteria or their product via pattern recognition receptors (PRRs) located on, or
within the cell. PRRs recognise pathogen associated molecular patterns (PAMPs) which are
highly conserved structures from the cell wall or nucleic acids of invading pathogens1, 2. Upon
activation of PRRs, a number of signalling pathways are stimulated leading to activation of
innate immune cells and secretion of cytokines and chemokines. These innate immune
responses also direct the induction of the adaptive immune system which mediates clearance
of the invading pathogen. During inflammation these pathways can become dysregulated
leading to prolonged inflammation and inflammatory disease3.
Bordetella pertussis is a Gram negative bacterium that infects the respiratory system. It is the
cause of pertussis (whooping cough), which mainly affects young children. WHO estimated
that in 2008, about 16 million cases of pertussis occurred worldwide, 95% of which were in
developing countries, and that some 195 000 patients died from this disease4. B. pertussis
binds to epithelial cells, infects the the cell and may also modulate the immune response in
order to hinder its clearance. Our lab has shown that B. pertussis subvert immune responses by
modulating activation of dendritic cells and macrophages and thereby subvert protective Th1
and Th17 responses that are required to eliminate the bacteria4. It has also been reported that
tracheal cytotoxin (a product of bacterial peptidoglycan) is an agonist for the NOD-like
receptor (NLR) NOD-1 and can bind to epithelial cells and enhance IL-1 and nitric oxide
(NO) production, thereby causing damage to the epithelial lining of the respiratory tract5. We
have also shown that synthetic NOD-1 agonists promote IL-10 by macrophages thereby
exerting anti-inflammatory function (Unpublished). However, the effect of B. pertussis and
the immunomodulatory effects of NOD-1 agonists on epithelial cells has received little
attention. This project will investigate the effect of live versus heat killed B. pertussis on
epithelial cells in order to investigate the possible role of these cells in the immune response
and their modulation by B. pertussis. It will also assess the effects of synthetic PAMPs, such
as the cell wall component lipopolysaccharide (LPS), CpG, lipoproteins or peptidoglycans and
their derivatives, NOD-1 and NOD-2 agonists, on epithelial cell activation.
Objectives:
•
Investigate the effect of live versus heat killed B. pertussis on cytokine and chemokine
production by epithelial cells.
•
To examine the effect of TLR and NOD agonists on epithelial cell activation
•
To identify the PRR pathways in epithelial cells activated, or modulated by exposure
to bacteria or their products
77
Methodology:
The student will receive training in a range of immunology techniques, including culture of
cell line and primary cells, ELISA, intracellular cytokine staining and flow cytometry,
quantitative real time PCR and Western blotting.
Likely outcome:
This work will lead to a better understanding of the mechanisms by which B. pertussis is able
to infect the host, modulate the immune response and persist in the respiratory tract. It could
also generate vital new information on how NOD-like receptors may be involved in antinflammatory responses and can be exploited as therapeutics for inflammatory disorders.
References:
1.
De Nardo D. Toll-like receptors: Activation, signalling and transcriptional modulation.
Cytokine. 2015 Aug;74(2):181-9.
2.
Caruso R, Warner N, Inohara N, Núñez G. NOD1 and NOD2: signaling, host defense,
and inflammatory disease. Immunity. 2014 Dec 18;41(6):898-908.
3.
Mills KH. TLR-dependent T cell activation in autoimmunity. Nat Rev Immunol. 2011
Nov 18;11(12):807-22.
4.
Higgs R, Higgins SC, Ross PJ, Mills KH. Immunity to the respiratory pathogen
Bordetella pertussis. Mucosal Immunol. 2012 Sep;5(5):485-500.
5.
Flak TA, Heiss LN, Engle JT, Goldman WE. Synergistic epithelial responses to
endotoxin and a naturally occurring muramyl peptide. Infect Immun. 2000 Mar;68(3):123542.
78
SS PROJECT NO. 17
Supervisor: Dr Rachel McLoughlin
Title: THE ROLE OF IL-22 IN REGULATING STAPHYLOCOCCUS AUREUS
ADHERENCE TO HUMAN KERATINOCYTES.
This project will investigate the regulatory role of the proinflammatory cytokine IL-22 during
Staphylococcus aureus colonisation of the epithelium. S. aureus is an opportunistic pathogen
that is responsible for a wide variety of diseases, ranging from superficial skin and soft tissue
infections to invasive diseases such as endocarditis, osteomyelitis and sepsis. In addition to its
invasive infective potential, S. aureus is commonly found as part of the normal human
microflora and is carried persistently and asymptomatically by 20% of the population with the
remainder colonised intermittently1. Colonisation is an important risk factor for subsequent
infection particularly in a nosocomial setting and persistent carriers are most likely to be
infected by their resident strain2. Due to the increasing prevalence of meticilin-resistant S.
aureus (MRSA) carriage by at-risk individuals, novel therapies that activate or enhance the
local host immune response to S. aureus in the nose are needed and may provide new
strategies for patient decolonisation.
S. aureus nasal colonisation is a multifactorial process with many host and bacterial factors
implicated. Bacterial attachment to keratinocytes in the nasal epithelium is facilitated by a
specific interaction between S. aureus and structural keratinocyte cell envelope proteins, most
notably loricrin and cytokeratin 103, 4. As part of the host tissue healing response, the
proinflammatory cytokine IL-22 can downregulate the expression of loricrin and cytokeratin
10 at the epithelium5, 6. Unpublished data from our lab indicates that IL-22 is expressed at the
nasal epithelium during S. aureus nasal colonisation in mice and may mediate the
downregulation of murine loricrin and cytokeratin 10 protein expression in vivo, facilitating a
host defence mechanism whereby staphylococcal attachment to the nasal epithelium is limited.
Based on these results, this project aims to investigate the direct ability of IL-22 to influence
attachment of S. aureus to human keratinocytes in vitro.
Specifically, this research project will:
1.
Investigate the ability of S. aureus to adhere to human keratinocytes.
In vitro cultures of the HaCaT human keratinocyte cell line will be established and co-cultured
with fluorescently-labelled S. aureus. Bacterial adherence will be visualised using fluorescent
microscopy and will quantified by enumerating colony forming units (CFU) per field of view
and by measuring mean fluorescence.
2.
Examine the effect of IL-22 on bacterial adherence.
Bacterial adherence to HaCaT cells (as outlined in 1) will be assessed following incubation of
HaCaT cells with increasing concentrations of recombinant IL-22. Changes in bacterial
adherence will be determined visually using fluorescent microscopy and by measuring mean
fluorescence. Cell culture supernatants will be collected and pro-inflammatory cytokine (IL6/IL-8) production quantified by ELISA.
79
3.
Determine the availability of staphylococcal ligands in the presence of IL-22.
HaCaT cells will be co-cultured with S. aureus with and without the presence of IL-22 (as
outlined in 2). HaCaT cells will be collected and proteins extracted. The presence of loricrin
and cytokeratin 10 proteins will be determined using Western immunoblotting and will be
quantified using densitometry.
References:
1.
van Belkum A, Verkaik NJ, de Vogel CP, Boelens HA, Verveer J, Nouwen JL et al.
Reclassification of Staphylococcus aureus nasal carriage types. The Journal of infectious
diseases 2009; 199(12): 1820-1826.
80
SS PROJECT NO. 18
Supervisor: Dr Rachel McLoughlin
Title: THE ROLE OF STAPHYLOCOCCUS AUREUS SURFACE PROTEINS IN
ATTACHMENT AND IMMUNE ACTIVATION DURING SKIN INFECTION.
This project will investigate the ability of Staphyloccocus aureus to activate the host innate
immune system at the initial stages of skin infection. S. aureus is one of the leading causes of
skin and soft tissue infections (SSTIs) globally. Meticilin-resistant S. aureus (MRSA)
accounted for 59% of purulent SSTIs presented in emergency departments across North
America in 20081. The host innate response is the first line of defence against staphylococcal
infection in the skin. Antimicrobial peptides are expressed upon bacterial insult and promote
bacterial elimination as well as amplifying the immune response by mediating
proinflammatory cytokine production2.
The incidence of skin infection relies on the ability of S. aureus to attach successfully to the
epithelium and evade host innate immune defences. S. aureus expresses several
multifunctional and functionally redundant surface proteins that act as virulence factors to
mediate these processes at the onset of infection. These include clumping factor (Clf)A and
ClfB which may facilitate adherence to matrix molecules in the epithelium such as fibrinogen,
loricrin and cytokeratin3, 4, 5 and staphylococcal protein A (Spa) which plays an important role
in staphylococcal immune evasion6. ClfA, ClfB and Spa are covalently linked to the bacterial
cell wall in a process facilitated by the enzyme sortase A9. Recent evidence suggests that Spa
can bind to tumor necrosis factor receptor-1 (TNFR1) on airway epithelial cells and inhibit the
activity of TNF-α7. TNF-α has been shown to mediate the production of antimicrobial
peptides and inflammatory factors in keratinocyte cultures in vitro8. This study aims to
examine the role of staphylococcal surface proteins in attachment to the epithelial surface and
activation of the innate immune response (specifically the production of antimicrobial
peptides) at the onset of skin infection.
Specifically, this project will:
1.
Assess the adherence and virulence potential of mutant strains of MRSA to
human keratinocytes.
In vitro cultures of the HaCaT human keratinocyte cell line will be established and infected
with bioluminescent MRSA USA300 strain LAC:lux and compared to mutants generated in
this strain that are deficient in ClfA, ClfB, Spa and sortase A. Bacterial attachment will be
compared and quantified visually using fluorescent microscopy and by measuring mean
fluorescence. Keratinocyte cell survival will be determined by collecting cell culture
supernatants and measuring LDH release.
2.
Compare the ability of each strain to activate the kerainocyte innate immune
response.
Following infection with mutant MRSA strains (outlined in 1), infected HaCaT cells will be
isolated and RNA extracted. RNA expression of antimicrobial peptides will be determined
using quantitative real-time PCR. Cell culture supernatants will be collected and proinflammatory cytokine producion (IL-8, IL-6, TNFa) quantified by ELISA.
81
3.
Investigate the virulence of mutant strains in an in vivo model of staphylococcal
subcutaneous infection.
Based on results obtained from 1 and 2, mutant MRSA strains that are defective in epithelial
adherence and/or innate immune activation will be assessed in vivo in a murine model of
subcutaneous skin infection and virulence will be compared to a WT strain. Bacterial burden
will be measured by quantifying bacterial bioluminescence using a photon imager over the
course of infection.
References:
1.
Edelsberg, J., C. Taneja, M. Zervos, N. Haque, C. Moore, K. Reyes, J. Spalding, J.
Jiang, and G. Oster. 2009. Trends in US hospital admissions for skin and soft tissue infections.
Emerging infectious diseases 15: 1516-1518.
2.
Schauber J, Gallo RL. Antimicrobial peptides and the skin immune defense system. J
Allergy Clin Immunol. 2008;122:261–266.
3.
Mcdevitt, D., Nanavaty, T., House-Pompeo, K., Bell, E., Turner, N., Mcintire, L.,
Foster, T. and HööK, M. (1997), Characterization of the Interaction Between
the Staphylococcus Aureus Clumping Factor (ClfA) and Fibrinogen. European Journal of
Biochemistry, 247: 416–424. doi: 10.1111/j.1432-1033.1997.00416.x
4.
Mulcahy ME, Geoghegan JA, Monk IR, O'Keeffe KM, Walsh EJ, Foster TJ et al.
Nasal colonisation by Staphylococcus aureus depends upon clumping factor B binding to the
squamous epithelial cell envelope protein loricrin. PLoS pathogens 2012; 8(12): e1003092.
5.
O'Brien LM, Walsh EJ, Massey RC, Peacock SJ, Foster TJ. Staphylococcus aureus
clumping factor B (ClfB) promotes adherence to human type I cytokeratin 10: implications for
nasal colonization. Cellular microbiology 2002; 4(11): 759-770
6.
Gemmell, C., Tree, R., Patel, A., O’Reilly, M., Foster, T. J. Susceptibility to
opsonophagocytosis of protein A, α-haemolysin and β-toxin deficient mutants of
Staphylococcus aureus isolated by allele-replacement. Zentralbl. Bakteriol. 21 (Suppl.), 273–
277 (1991).
7.
Gomez, M. I., A. Lee, B. Reddy, A. Muir, G. Soong, A. Pitt, A. Cheung, and A.
Prince. 2004. Staphylococcus aureus protein A induces airway epithelial inflammatory
responses by activating TNFR1. Nat. Med. 10:842-848.
8.
Guilloteau, K., Paris, I., Pedretti, N., Boniface, K., Juchaux, F., Huguier, V., Guillet,
G., Bernard, F. X., Lecron, J. C., and Morel, F. (2010) Skin inflammation induced by the
synergistic action of IL-17A, IL-22, oncostatin M, IL-1, and TNF-a recapitulates some
features of psoriasis. J. Immunol. 184, 5263–5270
9.
Mazmanian S. K., Liu G., Jensen E. R., Lenoy E. & Schneewind O. Staphylococcus
aureus sortase mutants defective in the display of surface proteins and in the pathogenesis of
animal infections. Proc. Natl. Acad. Sci. U. S. A. 97, 5510–5515 (2000).
82
SS PROJECT NO. 19
Supervisor: Dr. Nigel Stevenson
Title: DOES HEPATITIS B VIRUS (HBV) ENHANCE ANTI-VIRAL RESPONSES VIA
THE IFN-Α JAK/STAT PATHWAY?
Summary:
Hepatitis B Virus (HBV) infects the liver and is a major global health problem, with ~360
million people chronically infected worldwide and >1 million dying from associated liver
disease per year [1]. The cytokine interferon-alpha (IFN-α) signals via the Janus kinase/Signal
transducer and activator of transcription (JAK/STAT) pathway, leading to the upregulation of
numerous anti-viral mediators [2]. Interestingly, it is estimated that up to 95% of patients clear
HBV naturally, for reasons that remain undetermined. We have found that several viruses,
including Respiratory syncytial virus (RSV) [3] and HCV [4] block the IFN pathway by
targeting STAT proteins for degradation via the proteasome, but our preliminary discoveries
indicate that HBV actually enhances STAT signalling, which may explain why so many
patients spontaneously clear infection. Therefore, to further investigate the effect of HBV
upon anti-viral responses this project will analyse the effect of specific HBV proteins upon
IFN-α-mediated anti-viral gene induction and the activation of the IFN-stimulated response
element (ISRE).
Techniques: 1) Tissue culture, 2) Transfection 3) Real-Time-PCR, 4) Reporter gene assays.
References:
[1] Thimme R, Dandri M. Dissecting the divergent effects of interferon-alpha on immune
cells: time to rethink combination therapy in chronic hepatitis B? J Hepatol. 2013
Feb;58(2):205-9. doi: 10.1016/j.jhep.2012.11.007. Epub 2012 Nov 14.
[2] Horner S, Gale MJ (2009) Intracellular innate immune cascades and interferon defenses
that control hepatitis C virus. J Interferon CytokineRes29:489-498.
[3] Elliott J, Lynch OT, Suessmuth Y, Qian P, Boyd CR ,Burrows JF, Buick R, Stevenson NJ,
Touzelet O, Gadina M, Power UF, and Johnston JA. Respiratory Syncytial Virus NS1 Protein
Degrades STAT2 by Using the Elongin-Cullin E3 Ligase. J Virol, 2007; 81 (7): 3428–3436
[4] Stevenson NJ, Bourke NM, Ryan EJ, Binder M, Fanning L, Johnston JA, Hegarty JE, Long
A, and O’Farrelly C. Hepatitis C Virus targets the Interferon-α JAK/STAT pathway by
promoting proteosomal degradation in immune cells and hepatocytes. FEBS Lett. 2013;
587(10):1571-8.
83
SS PROJECT NO. 20
Supervisor: Dr. Nigel Stevenson
Title:
DOES TRIAD3A PLAY A ROLE IN HIV-VIF-MEDIATED STAT
DEGRADATION?
Summary:
HIV is a major world health problem, affecting more than 40 million people worldwide.
Although anti-retroviral drugs are used to control the spread of the virus there is currently no
cure for the disease. The anti-viral cytokine, Interferon (IFN)-α signals via the Janus
kinase/Signal transducer and activator of transcription (JAK/STAT) pathway, leading to the
production of anti-viral mediators, such as protein kinase R (PKR), OAS, MxA and MxB,
which disrupt viral replication [1]. Even though IFN-α is induced upon HIV infection it is not
understood why this natural anti-viral drug fails to clear the virus. We have found that several
viruses, including Respiratory syncytial virus (RSV) [2] and Hepatitis C Virus (HCV) [3]
block the IFN pathway by targeting STAT proteins for ubiquitination and subsequent
degradation by the proteasome. Our preliminary investigations indicate that HIV protein Vif is
also targeting STAT proteins for degradation. TRIAD3A is a ubiquitin ligase which has a role
in innate immune signaling pathways and has been shown to interact with HIV Vif [4].
TRIAD3A has been shown to ubiquitinate TRAF3, TLR4 and TLR9 and promote their
proteasomal degradation [5]. To determine the mechanism by which Vif degrades STAT
proteins we will investigate if TRIAD3A plays a role. This will be achieved by investigating
if over-expression of TRIAD3A can affect Vif-mediated STAT degradation. In parallel to the
over-expression studies, STAT degradation by Vif will be investigated in cells where
endogenous TRIAD3A expression is suppressed by shRNA. Finally, the effect of Vif and
TRIAD3A on downstream IFN stimulated genes (ISGs) will be assessed by quantitative real
time PCR.
Techniques: 1) Tissue culture, 2) Transfection, 3) Western blotting and 4) Real-Time-PCR.
References:
[1] Horner S, Gale MJ (2009) Intracellular innate immune cascades and interferon defenses
that control hepatitis C virus. J Interferon CytokineRes29:489-498.
[2] Elliott J, Lynch OT, Suessmuth Y, Qian P, Boyd CR ,Burrows JF, Buick R, Stevenson NJ,
Touzelet O, Gadina M, Power UF, and Johnston JA. Respiratory Syncytial Virus NS1 Protein
Degrades STAT2 by Using the Elongin-Cullin E3 Ligase. J Virol, 2007; 81 (7): 3428–3436
[3] Stevenson NJ, Bourke NM, Ryan EJ, Binder M, Fanning L, Johnston JA, Hegarty JE, Long
A, and O’Farrelly C. Hepatitis C Virus targets the Interferon-α JAK/STAT pathway by
promoting proteosomal degradation in immune cells and hepatocytes. FEBS Lett. 2013;
587(10):1571-8.
[4] Feng F., Davis A., Lake JA., Carr J., Xia W., Burrell C., Li P. Ring finger protein ZIN
interacts with human immunodeficiency virus type 1 Vif. J. Virol. 2004. 78(19):10574-81
84
[5] Nakhaei P1, Mesplede T, Solis M, Sun Q, Zhao T, Yang L, Chuang TH, Ware CF, Lin
R, Hiscott J. The E3 ubiquitin ligase Triad3A negatively regulates the RIG-I/MAVS signaling
pathway by targeting TRAF3 for degradation. 2009. PLoS. Pathog.
85