Team:Heidelberg/Project Measurement

From 2009.igem.org

(Difference between revisions)
(Real-time RT-PCR)
 
(357 intermediate revisions not shown)
Line 1: Line 1:
__NOTOC__
__NOTOC__
{{Template_HD_3}}
{{Template_HD_3}}
-
<html><body id="project_measure"></body></html>
+
<html><body id="project"></body></html>
{|
{|
Line 7: Line 7:
|width="650px" style="padding: 0 15px 15px 20px; background-color:#ede8e2"|
|width="650px" style="padding: 0 15px 15px 20px; background-color:#ede8e2"|
__NOTOC__
__NOTOC__
 +
= Measurement =
-
__NOTOC__
+
== Abstract ==
-
{{Template_HD}}
+
In order to achieve defined protein levels in a cell, promoters of defined strength are an obvious requirement.  [[Team:Heidelberg/Project_Synthetic_promoters|Such promoters]] can only be valuable to synthetic biology if they are well characterized. For future eukaryotic devices that require [http://www.partsregistry.org/PoPS PoPS (Polymerase per second)] as an input, our promoters will be very suitable since they deliver PoPS as an output. PoPS is ''the'' standard unit of synthetic biology, but it is very difficult to measure directly. For bacteria, relative measurements (relative promoter units, RPU) are most commonly used and it has been shown how to convert them to PoPS [[Team:Heidelberg/Project_Measurement#References|[1]]]. In this project, we identify and deal with several challenges to achieve the same in mammalian cells, suggest solutions and provide easy-to-use relative measures for application in mammalian cells - one based on RNA levels (Relative Mammalian Promoter Units, RMPU), the other based on folded protein levels (Relative Expression Units, REU). We apply those measurements on the characterization of CMV, an existing promoter from the registry. Finally, we discuss how to transform these units into PoPS.
-
== Synthetic Promoters ==
+
== Introduction ==
-
'''The central question of the synthetic promoter project is: Are we able to make specific promoters by predicting their sequence ''in silico''?'''
+
The need for standardized measurements of promoter activity ''in vivo'' has been widely accepted across the synthetic biology community [[Team:Heidelberg/Project_Measurement#References|[1]]]. Only if a part is well characterized initially, function of an engineered device or system can be predicted reliably. Most work of synthetic biology has focused on bacteria, especially ''Escherichia coli'', as a model system. Novel tasks in synthetic biology, especially for medical applications, will require synthetic biology of mammalian cells. Mammalian systems are the most complex biological systems, and therefore, little work has been done in the field of mammalian synthetic biology, leaving a huge potential for future research.<br>
-
Or, going even further: Are we able to develop a standard method for creating promoters of
+
[[Image:HD09_formula1.png|thumb|left|294px|<div style="text-align:justify;">'''Box 1: Calculation of PoPS''', where ''&gamma;M'' is the mRNA degradation rate, ''a'' is the GFP maturation rate, ''&gamma;I'' is the degradation rate of immature GFP, &rho; is the translation rate of immature GFP from mRNA and ''n'' is the number of copies of promoter per cell. [[Team:Heidelberg/Project_Measurement#References|[1]]]</div> ]] <div style="text-align:justify;">The holy grail of synthetic biology measurements is to express device input and output in "Polymerase per Second" (PoPS), an absolute unit which describes the rate of RNA polymerase molecules passing by the final base pair of the promoter [[Team:Heidelberg/Project_Measurement#References|[2]]]. If PoPS is to be measured from protein expression (e.g. GFP expression), in bacteria PoPS can be calculated from GFP synthesis rate, mRNA degradation, GFP maturation rate, GFP translation rate, plasmid copy number and degradation rate of immature GFP (see Box 1). Only GFP levels are easy to measure by fluorescence readouts, the other factors require complex experiments. Therefore, it has become a standard procedure to characterize promoters in RPUs. One RPU is the activity equivalent to BBa_J23101. [[Team:Heidelberg/Project_Measurement#References|[1]]] This  is possible because by cloning both the promoter and the reference separately but into the same plasmid backbone, promoters will create the same mRNA (as we always used GFP as a readout), thus having the same mRNA degradation and translation rate. In the same way, plasmid copy number, GFP maturation and degradation rates should be equal when using the one cell line at same conditions.</div><br>
-
* '''Defined strength'''
+
-
* '''Defined response'''
+
-
* '''Defined pathway integration'''
+
-
How this is supposed to work... Read on!
+
For mammalian cells, no such unit has been reported, nor is there a kit for measuring promoter activity. Mammalian cells have higher levels of complexity - the process from a gene to a functional protein involves transcription (regulated by the promoter itself and chromatin structure), RNA modification, RNA splicing, RNA export, RNA degradation, protein folding and protein degradation [[Team:Heidelberg/Project_Measurement#References|[3]]]. All of these processes are highly regulated, both generally and specifically for the individual gene. <br>
-
=== Abstract ===
+
A frequently used system is the dual luciferase assay. We dismissed the idea of using luciferase as it is an invasive technique - it requires killing the cells before measuring  [[Team:Heidelberg/Project_Measurement#References|[4]]]. This makes time-course experiments impossible. Moreover, GFP expression can be detected non-invasively with a wide variety of methods. As outlined [[Team:Heidelberg/Project_Measurement#Identifiaction_of_challenges_unique_to_higher_eukaryotes|below]], simple fluorescence measurements by fluorimeters cannot be the method of choice for measuring promoter activity in mammalian systems.<br>
-
Promoters are the key regulators of gene expression. Possessing promoters which are active under a desired condition, at a desired strength and in a specified tissue is of great value for Plant Biotechnology, Gene Therapy and fundamental research in Bioscience. Therefore, it has become a desire to synthetically construct promoters responsive to a variety of pathways. We explore two ways to the synthesis of promoters: On one hand, we have developed a bioinformatical model and database (HEARTBEAT) describing the structure of promoters responsive to user-defined inputs. On the other hand, we have developed a biochemical method for the synthesis of randomized promoter libraries. Using this method, we have created a library of constitutive promoters of varying strength. Also, we have created libraries of promoters putatively responsive to a variety of pathways. We have screened these libraries for functional, pathway responsive promoters and present a detailed characterization of a NF-&kappa;B responsive promoter of our making. We finally discuss ways to combine randomized biochemical synthesis and bioinformatical modeling to propose a method towards the generation of promoters of complex regulation (i.e. by multiple pathways).
+
For the quantification of reporter gene expression in each transfected cell, we used a dual assay with GFP and mCherry. Our cells are co-transfected with a reference plasmid containing our reference promoter in front of mCherry. The fluorescence intensities were measured by flow cytometry and fluorescence microscopy followed by ImageJ processing.  
-
=== Introduction ===
+
On the other hand, we attempted to measure the promoter strength through quantification of gene expression on mRNA level. Hereby, we applied the novel technique real-time RT-PCR.  The method consists of 2 steps, a reverse transcription, which synthesizes cDNA from extracted mRNA, and a real-time PCR, which is based on normal PCR, but collects the data throughout the PCR process, thus in real-time. Signal detection is achieved using fluorescent dye, which can <!--either be added to mastermix (SYBR Green approach), or be--> be attached to a probe  . The accumulation of PCR products over cycles results in the increase of fluorescence. <!--The cycle number where the fluorescence passes a certain threshold is known as Ct value. This values correlates directly to initial amount of mRNA present after RNA extraction.--> By comparing of cycles required to achieve a certain amount of fluorescence <!--values detected by different samples,--> the difference in mRNA expression profile can be illustrated. <!--In our approach to measure promoter strength using real-time RT-PCR, promoters of interest were cloned into the same backbone which contains eGFP as reporter gene. --> Through quantification of GFP mRNA expression driven by the test promoters, the difference in promoter strength can be easily illustrated. This approach measures directly on mRNA level, which means, translation and maturation of the GFP molecule do not have to be taken into account as in the flow cytometry approach.
-
Promoters are the key regulators of gene expression. Possessing promoters which are active under a desired condition, at a desired strength and in a specified tissue is of great value for Plant Biotechnology, Gene Therapy and fundamental research in Bioscience. Most efforts of obtaining such promoters focus on cloning them from Nature. This approach is, in eukaryotes, flawed for three reasons: First, promoters in eukaryotic cells are very complexly regulated by a wide variety of transcription factors, and thus, pathways [[Team:Heidelberg/Project_Synthetic_promoters#References|[1]]]. Therefore, natural promoters cannot be used reliably as transcriptional assays. Second, a promoter might be required to be active under a set of conditions for which no natural promoter exists. Third, for precise control of gene expression levels, promoters of human-defined transfer functions and expression strengths are required.<br>
+
However, a common problem when measuring any construct or device in mammalian cells is the fact that the transfection rate is not always constant. Thus, one never knows how many copies of a construct are actually in the cell. This is why we attempted to create a system that allows a controlled integration of our constructs into the genome. To accomplish this we used the FRT/Flp system which is based on homologous recombination by the enzyme flippase at a specific sequence – the FRT site [[Team:Heidelberg/Project_Measurement#References|[20]]]. [[Team:Heidelberg/stables|Read more about our stable cell lines]]
-
Therefore, efforts have emerged to synthetically construct promoters. Two concepts of synthetic promoters in mammalian cells co-exist independently from each other. One is the concept of "genetic switches" (see [[Team:Heidelberg/Project_Synthetic_promoters#References|[10]]] for a recent review) - promoters which can specifically be induced by a stimulus mammalian cells are usually insensitive to, e.g. tetracycline [[Team:Heidelberg/Project_Synthetic_promoters#References|[11]]]). Much fewer efforts have been put into developing promoters sensitive to ''endogenous'' signals (referred to as "synthetic promoters" in the rest of this article). Such promoters are of very high value for a broad variety of applications. Three examples should demonstrate this. First, in virotherapy for cancer and other diseases, it has become a desire to express toxic genes only in affected cells (reviewd in [[Team:Heidelberg/Project_Synthetic_promoters#References|[12]]]). For example, breast cancer cells are characterized by high levels of estrogen receptor. Constructing a promoter which is active only at high estrogen receptor levels (plus, maybe, only in cells which are irradiated, as ER can be very active in other tissues of the female reproductive tract also) might therefore help developing novel breast cancer therapies. Second, biologists studying pathway interactions are in need for transcriptional assays, that is promoters which are specifically activated by a single transcription factor. Third, the concept can be transferred to plants, where synthetic promoters can be very valuable, as plant biotechnology is always in need for novel tissue- or development-specific promoters.<br>
+
[[Team:Heidelberg/Project_Measurement#Measurement|[TOP]]]
-
Three approaches exist to construct synthetic promoters responsive to endogenous factors. First, the by structure of promoters is modeled by generating large data sets describing the relative spacing and coincidence of transcription factors (reviewed in [[Team:Heidelberg/Project_Synthetic_promoters#References|[4]]]). To our knowledge, such predictions have not been tested ''in vivo''. Second, promoters are generated by randomly or repeatedly cloning response elements upstream of a core promoter. To our knowledge, repeated cloning of response elements works well [[Team:Heidelberg/Project_Synthetic_promoters#References|[5]]] and is frequently applied, but no suggestions exist on how to apply this strategy to the generation of more complexly regulated promoters. The random creation of promoters works well to generate constitutive promoters [[Team:Heidelberg/Project_Synthetic_promoters#References|[6]]] and was even applied to broadly identify activating elements  [[Team:Heidelberg/Project_Synthetic_promoters#References|[2]]], but no promoters of specific regulation have been described for this approach. A third approach is the randomization of spacer elements between transcription factor binding sites, which is applied to generate libraries of promoters of varying strength [[Team:Heidelberg/Project_Synthetic_promoters#References|[3]]], [[Team:Heidelberg/Project_Synthetic_promoters#References|[8]]].
+
== Results ==
-
In order to be able to design synthetic promoters, an understanding of natural promoters is required. Mammalian promoters can be subdivided into several "domains". The ''core promoter'' is the binding site of the basal transcription machinery, i.e. RNA polymerase and associated factors. Core promoters differ in composition, but are more or less similar for most genes (reviewed in [[Team:Heidelberg/Project_Synthetic_promoters#References|[9]]]). The main regulatory domain is the proximal promoter, which is where regulatory elements bind. It can be very large (4kb), meaning that some transcription factors regulate transcription despite being very far away from the RNA polymerase. This is mainly possible because of the three-dimensional structure the DNA adopts. In addition to this, there are even more distal elements that are referred to as "enhancers" and "silencers". A further challenge is that some transcription factors are not able to initiate transcription on their own, but rather they require other transcription factors for their activity.
+
=== Identification of challenges unique to higher eukaryotes ===
-
== Results ==
+
In this chapter, we start by identifying critical points of promoter characterization in eukaryotes and specify how we proceeded to take on these challenges:<br>
-
=== RA-PCR, a method for the generation of randomized promoter libraries ===
+
==== Variance of transfection efficiencies ====
-
[[Image:HD09_rapcr.jpg|none|thumb|650px|'''Figure 1: The method of RA-PCR''']]
+
[[Image:HD09_tfeff.png|thumb|left|300px|<div style="text-align:justify;">'''Fig. 1: Transfection efficiency for vectors containing different inserts varies'''. By flow cytometry, we identified transfected cells and found transfection efficiencies to vary. Here, the transfection efficiency is shown for HeLa and e standard error of the mean (SEM) is also displayed.</div>]] <div style="text-align:justify;">In prokaryotes and yeast, plasmid copy number is a property of the plasmid as determined by the origin of replication. Higher eukaryotes are unable to stably propagate plasmids, unless stable integration occurs (which does not happen spontaneously). Even plasmids with minor differences show entirely different transfection efficiencies and therefore will result in large variations of GFP expression per well. By flow cytometry, we analyzed transfection efficiency for HeLa cells and calculated plasmid number to vary greatly. We transfected cells with plasmids containing the same backbone, but different (promoter) inserts and found transfection efficiency to vary as much as 20% (Fig. 1).  Also, as expected, a significant percentage of cells remains untransfected in a  experiment (Fig. 1).</div>
 +
Dual assay measurements using co-transfection as a control for transfection efficiency are commonly used[[Team:Heidelberg/Project_Measurement#References|[4]]]. Such assays can be utilized for relative characterizations, but do not provide accurate normalization. In order to resolve this issue, we suggest a method which not only allows relative characterizations but is also able to distinguish between transfected and untransfected cells (flow cytometry or image analysis). For experiments aiming at absolute numbers, stable integration of the measurement plasmid into the mammalian genome containing exactly one integration site is required. In this case the copy number per cell is one.<br>
-
We have developed a standard method (termed "Random Assembly PCR / RA-PCR") for the construction of randomized promoter libraries. We modified Assembly PCR [[Team:Heidelberg/Project_Synthetic_promoters#References|[7]]] to create randomized promoters instead of ordered genes by using different oligos containing a transcription factor binding site (or random DNA) plus two annealing sequences (see Figure 1 for a comprehensive explanation of the method).  We use two sets of oligos, one for the top strand, one for the bottom strand. The oligos for each strand have the same annealing sequences (which are complementary to the annealing sequences of the other strand). If these oligos are pooled, they will randomly anneal to each other, thus generating randomized repeats of the transcription factor binding sites of interest at varying spacing. In order to be able to clone the construct, we also add two stop oligos (termed stop 5' and top 3') which contain only one annealing sequence, plus a cutsite (SpeI 5', HindIII 3'). Double-stranded DNA is created by running a seven-cycle PCR, and amplified by a  25-cycle PCR. Then, the resulting (proximal) promoter is cloned 5' of a core promoter (we used the core promoter of JeT [[Team:Heidelberg/Project_Synthetic_promoters#References|[8]]]) by inserting it into [[Team:Heidelberg/Project_Measurement#A_promoter_measurement_kit_for_use_in_mammalian_systems|pSMB_MEASURE]], the promoter measurement plasmid we developed (from there, it can be excised like any standard biological part in a submission plasmid). Thus, a mixture of different promoters in the same plasmid backbone is generated. These can then be transformed into bacteria. Each colony represents a single putative promoter, which can the be transfected into mammalian cell under the conditions of interest, plus control conditions. Promoters active under the desired conditions, but not under control conditions, are selected for further characterization. <br>
+
==== Regulation of promoter activity by chromatin structure ====
-
Please see a detailled protocol for RA-PCR [[Team:Heidelberg/Project_Synthetic_promoters#RA-PCR_protocol|below]].
+
Considering stable integration of a promoter into the mammalian genome, another level of complexity is added: Chromatin structure affects transcription, though mostly not affected by the promoter[[Team:Heidelberg/Project_Measurement#References|[5]]]. Therefore, integration at different sites of the genome will not result in the same expression strength.<br>
-
=== Generation of a library of constitutive promoters ===
+
A cell line having a FRT site stably integrated into the genome is required. Such a cell line involves the recombination of sequences between short Flippase Recognition Target (FRT) sites by the Flippase recombination enzyme (FLP or Flp) derived from the 2µ plasmid of the baker's yeast Saccharomyces cerevisiae.[[Team:Heidelberg/Project_Measurement#References|[6]]]. We started the development of [[Team:Heidelberg/Project_Measurement#A_stable_cell_line_for_promoter_measurement|such a cell line]].
-
[[Image:HD09_constitutive.png|left|thumb|350px|'''Figure 2: A library of constitutive promoters created by RA-PCR''' Promoters were analyzed by the standards developed in the [[Team:Heidelberg/Project_Measurement#Measuring REU_by_flow_cytometry_and_image_analysis|Measurement]] part of our project in HeLa cells.]]
+
[[Team:Heidelberg/Project_Measurement#Measurement|[TOP]]]
-
As a first application of RA-PCR, we have created a library of constitutive promoters. We performed RA-PCR on oligos containing binding sites for some well known generally activating transcription factors (Sp1, Ap1, CREB, NF-Y)  which we identified from literature search [[Team:Heidelberg/Project_Synthetic_promoters#References|[2]]],[[Team:Heidelberg/Project_Synthetic_promoters#References|[6]]],[[Team:Heidelberg/Project_Synthetic_promoters#References|[8]]]. We also added NF-&kappa;B responsive oligos as NF-&kappa;B has non-specific activity and is therefore used by a variety of viral constitutive promoters, e.g. the HIV promoter [[Team:Heidelberg/Project_Synthetic_promoters#References|[13]]]. We picked 24 colonies, two of which we dismissed after a test digest (not shown). Figure 3 shows the sequence analysis of some randomly selected clones and demonstrates that RA-PCR is able to generate randomized repeats of Oligos. We then measured the activity of the clones we picked by applying the [[Team:Heidelberg/Project_Measurement|Concept of Relative Expression Units (REU)]] we developed. Figure 2 shows that we have been able to create a library of promoters of varying strength, some of which have an expression strength higher than JeT (which was not accomplished by JeT's developers, although attempted [[Team:Heidelberg/Project_Synthetic_promoters#References|[8]]]). Such a library is of great value for fine-tuning gene expression levels.
+
==== RNA processing ====
-
[[Image:HD09_0109_const.jpg|left|thumb|350px|'''Figure 3: RA-PCR generates randomized repeats of transcription factor binding sites.''' Sequence analysis of clones of constitutive promoters generated by RA-PCR. Transcription factor binding sites are marked in color, random sequences in light grey.]]
+
-
=== Generation and screening of a library of promoters putatively responsive to NF-&kappa;B ===
+
Many RNAs are spliced after transcription. Also, RNAs are processed before being exported from the nucleus, and can be retained in the nucleus both dependent and independently of processing [[Team:Heidelberg/Project_Measurement#References|[3]]], [[Team:Heidelberg/Project_Measurement#References|[7]]]. Therefore, several mRNA populations exist: Unprocessed, processed and retained in the nucleus, and functional mRNA outside of the nucleus. These effects are a current research focus of molecular cell biologists and barely understood even in model organisms as simple as yeast.<br>
-
[[Image:HD09_nfkb.png|left|thumb|350px|'''Figure 4: A library of putative NF-&kappa;B responsive promoters created by RA-PCR''' Promoters were induced by TNF-&alpha; in U2OS cells and screened by TECAN (automated plate reader).]]
+
Splicing does not present a problem for reporter gene constructs, as these are cDNAs which are not spliced. Nevertheless, this means that the ratios between RNA levels and protein levels will change for a single promoter if a different output gene is linked to the promoter. We therefore suggest to assume a black box between RNA and protein, and the introduction of two independent units.
 +
 
 +
[[Team:Heidelberg/Project_Measurement#Measurement|[TOP]]]
 +
 
 +
 
 +
==== Variety of cell lines ====
 +
 
 +
<div style="text-align:justify;"> TOP10 or DH5&alpha; cells have been widely accepted as chassis systems by synthetic biologists working in bacteria. For the work with mammalian cells, no such consensus exists; also, it would not be sensible to limit synthetic mammalian biology to a small number of cell lines, as every cell line is suited for a special application. Scientists working on breast cancer virotherapy by synthetic promoters would choose a breast cancer cell line, whereas scientists working on [https://2008.igem.org/Team:Bay_Area_RSI stem cell therapies to myocardial infarction] would choose a cardiomyocyte cell line. Cell lines differ greatly, even in expression strength of constitutive promoters (shown [[Team:Heidelberg/Project_Measurement#Measuring_REU_by_flow_cytometry_and_image_analysis|below]]).
 +
<div style="text-align:justify;">Each part must be characterized in every cell line. We worked with three cancer cell lines, [[Team:Heidelberg/Eukaryopedia#HeLa|HeLa]] (cervical cancer), [[Team:Heidelberg/Eukaryopedia#MCF-7|MCF-7]] (breast cancer) and [[Team:Heidelberg/Eukaryopedia#U2-OS|U2-OS]] (osteosarcoma). We suggest that the synthetic biology community should pick a small number of cell lines as model systems. We argue that HeLa (well known, widely used, easy to transfect) and/or MCF-7 (very robust to apoptosis) should be part of these cell lines. </div>
<br>
<br>
-
RA-PCR was conducted with Oligos containing a NF-&kappa;B binding site, plus a small number of "general activators" (NF-Y, Sp1, Ap1, CREB) . Box 1 demonstrates how the oligos were designed from a frequency matrix.  33 clones were picked, miniprepped and transfected. NF-&kappa;B was then induced by the addition of TNF-&alpha (2.5µM) for 10 hours, and left uninduced as a control. The plate was then scanned by TECAN, an automated fluorescence plate reader. TECAN is very imprecise on eukaryotic cells, and the arbitrary fluorescence we meausred is not proportional to [[Team:Heidelberg/Project_Measurement|REU]] or another precise measure of mammalian promoter activity, but it can serve as a rough indicator of promoter induction. The result (Fig.4) shows that most clones appear not to be induced by NF-&kappa;B, whereas others are induced at varying levels of strength. Considering the sequence analysis of some randomly selected clones (Fig.5), this result is not intuitive, as most sequences contain a NF-&kappa;B binding site, but it demonstrates that simply cloning repeats of a Transcription Factor Binding Site in front of a core promoter will not necessarily work.<br>
+
==== Lack of truly constitutive promoters in mammalian cells ====
-
We picked clone 31 for further characterization in REU.
+
[[Image:HD09_FACS_cmv_medium_with_time.PNG|thumb|left|300px| <div style="text-align:justify;">'''Fig. 2: CMV and JeT strength changes depending on conditions''' We characterize GFP expression from [http://partsregistry.org/wiki/index.php?title=Part:BBa_K203112 JeT] and [http://partsregistry.org/Part:BBa_I712004 CMV] fluctuate dependent on condition (Everolimus induces extreme starvation). Measured by flow cytometry 20 hours after transfection (unless specified otherwise) in MCF-7. The standard deviation is represented by the error bars. </div>]]
-
[[Image:HD09_nfkbseq.jpg|left|thumb|350px|'''Figure 5: Sequence analysis of putative NF-&kappa;B responsive promoters]]
+
Figure 2 shows measurements of GFP expression from [http://partsregistry.org/Part:BBa_I712004 CMV] and  [http://partsregistry.org/wiki/index.php?title=Part:BBa_K203112 JeT] under varying conditions. This result demonstrates that every promoter in mammalian cells underlies regulation, and therefore, is not truly constitutive. We analyzed the sequence of CMV by [http://www.gene-regulation.com/pub/databases.html TRANSFAC Professional] and found it to contain two [[Team:Heidelberg/Eukaryopedia#Transcription_factors|NF-&kappa;B]] binding sites, two [[Team:Heidelberg/Eukaryopedia#CREB|CREB]]-binding sites, and single [[Team:Heidelberg/Eukaryopedia#Ap1|Ap1]], RFX1 and SRF binding sites. Of NF-&kappa;B, CREB and Ap1, we know that they have a high constitutive activity ([[Team:Heidelberg/Project_Synthetic_promoters#Generation_of_a_library_of_constitutive_promoters|compare to Synthetic promoters]]), but nevertheless, they underlie regulation. For example, NF-&kappa;B is induced by inflammation conditions, whereas CREB is activated by the second messenger cAMP[[Team:Heidelberg/Project_Measurement#References|[8]]] and thus responds to many hormones, starvation conditions etc. This impedes comparison of promoters in different conditions. We next [[Team:Heidelberg/Project_Measurement#Characterization_of_promoters_under_different_conditions|discuss]] how this can be achieved.
 +
<br>
 +
<br>
 +
<br>
 +
<br>
 +
<br>
 +
[[Team:Heidelberg/Project_Measurement#Measurement|[TOP]]]
-
[[Image:HD09_nfkbmatrix.jpg|left|thumb|350px|'''Box 1: RA-PCR allows for synthesis of promoters responsive of imprecisely described transcription  factors.''' Considering the graphical representation of NF-kappaB's frequency matrix shown above (source: [http://jaspar.cgb.ki.se/ JASPAR]), the oligo can be designed in order to represent this matrix, instead of a static NF-kappa B binding site. A sensible representation of this matrix would be GGGRHTTYCC (for the IUPAC nucleotide code, refer to [http://www.bioinformatics.org/sms/iupac.html bioinformatics.org]). Most oligonucleotide manufactures provide the option to synthesize such mixtures of individual oliogs without further cost. As our method is PCR-based (unlike other methods such as [[Team:Heidelberg/Project_Synthetic_promoters#References|[5]]] and [[Team:Heidelberg/Project_Synthetic_promoters#References|[6]]]), we are able to synthesize even promoters responsive to badly-described transcription factors ]]
 
-
=== Characterization of a NF-&kappa;B responsive promoter ===
+
=== A promoter measurement kit for use in mammalian systems ===
-
'''Hannah and Corinna, I need you data. Possibly also ibidi cotransfection, video etc.'''
+
[[Image:HD09_p31.png|thumb|left|385px|<div style="text-align:justify;">'''Fig. 3: Plasmid map of pSMB_MEASURE.''' BBb sites are shown in red, core promoter in light blue and proximal promoter in dark blue. The length of this plasmid is 5162 bp.</div>]]<div style="text-align:justify;">We created a plasmid , pSMB_MEASURE (SMB is for Synthetic Mammalian Biology; [http://partsregistry.org/wiki/index.php?title=Part:BBa_K203100 Part:BBa_K203100]), which should be used for promoter characterization in mammalian cells. pSMB_MEASURE (see Fig. 3) contains a reference promoter, JeT[[Team:Heidelberg/Project_Measurement#References|[9]]] ([http://partsregistry.org/wiki/index.php?title=Part:BBa_K203112 Part:BBa_K203112]), which is flanked by BBb_2 (Tom Knight) sites and can therefore be replaced by the promoter to be measured. JeT is ideal as a reference promoter for a variety of reasons. First, it has an intermediate expression strength; second, it is regulated by a wide variety of transcription factors and low levels of change in fluorescence among different conditions (compare Fig. 2)[[Team:Heidelberg/Project_Measurement#References|[9]]]. Third, we want to pay tribute to its creators as pioneers in synthetic promoter research.</div>
-
=== RA-PCR can generate promoters responsive to a variety of pathways ===
+
We separated JeT's core promoter from its proximal promoter by a HindIII site; it can therefore be used for [[Team:Heidelberg/Project_Synthetic_promoters#Results|the ''de novo'' creation of synthetic promoters]] containing the JeT core promoter or for modifying the strength of a promoter by [[Team:Heidelberg/Project_Measurement#Different_core_promoters_result_in_different_expression_strength|core promoter swapping]]. In addition, it contains a FRT site which will allow for stable integration into mammalian cells also containing a FRT site. Thus, it provides the possibility to characterize the promoter in a defined genome and in this way helps to avoid some of the [[Team:Heidelberg/Project_Measurement#Identification_of_challenges_unique_to_higher_eukaryotes|challenges outlined above]]. For the same reason, it also contains a mammalian selection marker (hygromycine). For the generation of the plasmid, please see  [http://partsregistry.org/Part:BBa_K203100:Design part design]. As a reporter gene, it contains GFP, which is followed by a SV40 mammalian terminator. We generated another plasmid pSMB_REFERENCE, which contains mCherry instead of GFP. It can be used for normalizations of transfection efficiency in flow cytometry and image analysis.
-
We performed RA-PCR to construct promoters putatively responsive to Transcription factors as diverse as p53 (DNA damage sensor), pPAR&gamma; (metabolism & diabtetis), SREBP (Sterol nutrition), HIF (hypoxia) and Estrogen receptor . While screening these promoters we found the following:
+
[[Team:Heidelberg/Project_Measurement#Measurement|[TOP]]]
-
[[Image:HD09_ppary_data.png|left|thumb|220px|'''Figure 6: pPAR&gamma; responsive promoters induced by Thiazolidinedione in U2OS cells.]]
+
-
* For pPAR&gamma;, we, by screening, identified two clones which appear to be responsive to the anti-diabetis drug [http://en.wikipedia.org/wiki/Thiazolidinedione Thiazolidinedione]. We roughly characterized these promoters by a triple TECAN read reltaive to JeT (Fig. 6)
+
-
* For p53, induction of the pathway by the Topoisomerase inhibitor [http://en.wikipedia.org/wiki/Camptothecin Camptothecin] (a anti-cancer drug) turned out to be difficult as is severly harms the cells and makes promoter indcution levels difficult. We therefore attempted to normalize screening conditions to number of living cells by Hoechst-Staining. We found that some promoters appeared to be strongly dowregulated by Camptothecin and therefore experimented with a variety of conditions inducing by p53 by different pathways, at different phosphorylation sites, but where unable to obtain a conclusive picture.
+
-
* For HIF, we failed to induced the condtions sufficiently to achieve promoter activation. We below [[Team:Heidelberg/Project_Synthetic_promoters#Improving_RA-PCR|discuss]] how screening can be improved.
+
-
* For SREBP and Estrogen, we encoutered technical problems during promoter synthesis (probably damaged HinDIII enzyme) and therefore were unable to produce enough clones for a sufficient screening. For SREBP, we therefore cloned two natural, SREBP-upregulated promoters we had at hand and submitted them to the registry (where a characterization can be found).
+
-
=== HEARTBEAT, a model describing promoter structure ===
+
=== Two units for promoter activity in mammalian cells ===
-
[[Project_heartbeat|Main article: HEARTBEAT]]<br>
+
-
[[Image:HD09_VDRspatial.png|left|thumb|220px|'''Figure 7: Probability density function for the distribution of VDR-Binding sites along an ideal promoter as modelled by HERATBEAT''']]Based on the assumption that transcription factors (TFs) have a spatial preference for binding to the natural promoters' sequence concerning the distance to the transcriptional start site (TSS) [[Team:Heidelberg/Project_Synthetic_promoters#References|[14]]], we developed HEARTBEAT (Heidelberg Artificial Transcription Factor Binding Site Engineering and Assembly Tool). In a first step 4395 human promoter sequences 1000 bp upstream from the TSS obtained from the UCSC genome browser were analysed by the program “Promotersweep” [[Team:Heidelberg/Project_Synthetic_promoters#References|[15]]]. Promotersweep is able to assign transcription factor binding sites (TFBS) to a given sequence by retrieving and combining information from three homology databases (EnsEMBL Compara, NCBI HomoloGene, DoOP database), five promoter databases (EPD, DBTSS), six sequence motive identification tools (e.g. Meme, Gibbs Motif Sampler) and two matrix profile databases (Jaspar Core Library, Transfac Professional Library). Each TFBS motive is further classified into weak, conserved and reliable according to the quality of the assignment. The final result of Promotersweep can be divided into general spatial information about the TFBS and the consensus sequence on the one hand and further detailed facts about the associated gene on the other.<br>
+
We define two units: 1 relative mammalian promoter unit (RMPU) is defined in analogy to[[Team:Heidelberg/Project_Measurement#References|[1]]], but mRNA-based. It is  the amount of total mRNA (that is, processed and unprocessed mRNA inside and outside of the nucleus) generated by a promoter x, relative to the amount of total mRNA generated by the JeT promoter j ([http://partsregistry.org/wiki/index.php?title=Part:BBa_K203112 BBa_K203112])[[Team:Heidelberg/Project_Measurement#References|[9]]] in steady state. It is directly proportional to PoPs, as we show. In steady state, change of mRNA levels is 0, thus:
-
[[Image:HD09_VDRcoin.png|left|thumb|220px|'''Figure 8: Frequency of other transcription factors occuring together with VDR. 680 transcription factors were examined, of which the displayed 340 show coincidence at least once.''']] In figure 7 the spatial distribution of VDR (Vitamin D receptor) binding sites within 140 natural promoter sequences is shown as an example. The size of each bin equals the number of VDR-TFBS within a range of 20 bps. The solid line represents the probability density function (pdf). Here, the maximum of the pdf is located 54 bps upstream to the TSS indicated by the vertical line. Natural promoter sequences usually exhibit multiple TFBS which implies dependencies between different TFs according to their binding behaviour to the DNA. Figure 8 shows the frequency distribution of coincidental appearing TFBSs if VDR is present. The highest peak represents VDR itself. The next three highest peaks are Kid3 (inhibitory), WT1 and AP-2 (stimulatory). In total, together with VDR, there are over 300 different TFBS coincidentally present. Both plots represent data deduced from the Heartbeat-database which enable a well-defined synthesis of promoter sequences.
+
[[Image:HD09_eq1.png]]
-
=== An ''in vivo'' test of predicted promoter sequences ===
+
Where M' is the change in total mRNA level, &gamma;M is the mRNA degradation rate and n is the number of promoters per cell (adapted from [[Team:Heidelberg/Project_Measurement#References|[1]]]. Therefore:
-
''ongoing work''
+
[[Image:HD09_eq2.png]]
-
== Discussion ==
+
Since both promoters are cloned seperatly into the same backbone, they generate identical mRNA. As the most important determinant for mRNA stability is the 3' untranslated region, and mRNA stability is generally not affected by promoter structure [[Team:Heidelberg/Project_Measurement#References|[10]]], we are able to assume the same mRNA degradation rate &gamma;M for both promoters.
-
The results shown above demonstrate the potential of RA-PCR towards the synthesis of any promoter. Even by analyzing modest amounts of clones for each individual pathway, we were able to obtain promoters of a wide variety of strength and inducibility. Also, we were able to obtain constitutive promoters of greater strength than JeT, which has not been possible before[8].<br>
+
[[Image:HD09_eq3.png]]
-
Many insights about promoter regulation are possible by analyzing different promoters created by RA-PCR. For example, clone 3 and clone 11 (see figure 4 and 5) differ only in the positioning of the single response element (RE), but still, induction strength differs threefold. This gives hints about Nf-&kappa;B's binding preference. A systematic study of promoters generated by RA-PCR and their strength could therefore be used to develop a comprehensive model of transcriptional regulation.  '''Nao, kann man hier dein model verlinken und mit einem oder zwei stzen elegant beschrieben?'''
+
We also expect the same n for both promoters as this is normalized for during the measurements (as shown below).
 +
We then define RMPU, and achieve a cancelation of terms:
-
=== Improving RA-PCR ===
+
[[Image:HD09_eq4.png]]
-
''Screening conditions and induction strength:'' <br>
+
On the other hand, we define Relative Expression Units (REU). 1 relative expression unit (REU) is the amount of total folded protein generated by a promoter, relative to the amount of folded protein generated by the JeT promoter ([http://partsregistry.org/wiki/index.php?title=Part:BBa_K203112 BBa_K203112]) under the same cellular condition. It is not directly proportional to PoPS as too many levels of regulation, such as post-transcriptional modifications, enhanced splicing and nuclear shuttling/transport lie between transcription and protein [[Team:Heidelberg/Project_Measurement#References|[3]]], and REUs would strongly depend on conditions which affect RNA. Still, it is a very useful measure, as for most applications such as metabolic pathway engineering, protein level, not mRNA levels, are of importance. Also, it can be measured with a much easier experimental setup than RMPU. See [[Team:Heidelberg/Project_Measurement#Characterization_of_promoters_under_different_condtions|discussion]] for ideas on how to compare REU measurements taken at different conditions.
-
As noted [[Team:Heidelberg/Project_Synthetic_promoters#RA-PCR_can_generate_promoters_responsive_to_a_variety_of_pathways|above]], we experienced difficulties inducing some of the pathways (namely, HIF and p53). From our cell culture work, we learned that finding the ideal timepoint of induction for a certain pathway and the ideal conditions is very difficult even with literature at hand. Also, one would expect a much higher induction than the one observed for the NF-&kappa;B responsive clone we describe. Our induction levels might be low because NF-&kappa;B has a high constant actvity '''link to nfkb review here''', especially if the cells encounter rough cell culture conditions. Therefore we suggest that for future screening, a library of siRNAs for the transcription factors of interest should be compiled. Also, a library of transcription factors mutated to be constantly active is is required. With these libraries at hand, individual transcription factors can be knocked down, and activated specifically at 100% efficiency. This will greaty facilitate screening and parts characterization.<br>
+
[[Team:Heidelberg/Project_Measurement#Measurement|[TOP]]]
-
''Generation of down-regulated promoters''<br>
 
-
As shown, we were able to generate a set of promoters upregulated by certain factors. For several applications, promoters of a high constant strength, which become down-regulated by a signal, are required. We think it might be possible to construct such promoters by performing a RA-PCR with oligos containing weak binding sites for generally activating transcription factors (that is, binding sites which deviate from the consensus sequence), and to add some oligos containing very strong binding sites for the transcription factor of interest (say, NF-&kappaB). If this factor is not active, the general activators will be able to bind to the DNA and activate transcription. Upon factor activation, the general activator will be replaced. If the binding site is then in a position where it does not initiate transcription (as for some of the clones (32 etc.) shown in Figure 4 and 5), the promoter will be downregulated, instead of upregulated. This hypothesis remains to be tested.
+
=== Measuring RMPU by real-time RT-PCR ===
-
=== M-RA-PCR, a model-guided biochemical method for synthesis of complex promoters ===
+
To measure the RMPU (Relative Mammalian Promoter Units), HeLa cells were transfected with plasmids containing the promoter of interest. HeLa cells transfected with plasmid containing JeT promoter were used as reference. At two different time-points (20 h and 50 h after transfection), total RNA (> 200 bp) was isolated. This step was followed by  real-time RT-PCR, where the mRNA amount of GFP was quantified. For each promoter at each time point, 12 replicates were taken to obtain reliable results and reduce the variance. To get comparable results of different samples, we used multiple housekeeping genes as internal controls, while using non-transfected HeLa mRNA as plate-to-plate correction. <!--The program was performed with ABI StepOnePlus Real-time PCR System. Data was collected from the associated software. GFP expression was normalized against housekeeping genes using own written MatLab-script.--> The CMV promoter was calculated about 2.89 times stronger as JeT after 20 h and 2.04 times stronger after 50 h (Fig. 4 and 5).
-
[[Image:HD09_dual.jpg|thumb|left|250px|'''Figure 9: RA-PCR can be modified to refelct probability densitiy curves in vitro''']] RA-PCR can be modified to reflect modeled probability density curves. If a promoter regulated by multiple pathways, for example VDR (Vitamin D receptor) and SREBP (Sterol regulated element binding protein), is to be constructed, considering the density curves as obtained from the model (Figure 9) can give clues about its construction. A working VDR/SREBP promoter requires VDR and/or SREBP Response Elements (REs) in the close vicinity of the TSS (at approx. 850). It might require SREBP REs between 300 and 700, and VDR REs between 0 and 300. This distribution can be refelected by setting up 3 RA-PCRs with varying concentations of VDR-responsive, SREBP-responsive and spacer-oligos (compare figure B2.1). If a 3'Stop oligo containing a NheI cutsite, and a 5'Stop oligo containing a SpeI cutsite (or any combination of cutsites yielding compatible ends) is used, an infinite number of RA-PCR products can be assembled and cloned in front of a core promoter (having a SpeI cutsite 5').<br>
+
{|
-
We believe that this technique, termed Model-guided Random Assembly PCR, or M-RA-PCR, is the way forward to constructing the promoters of complex regulation described in the [[Team:Heidelberg/Project_Synthetic_promoters#Introduction|Introduction]].
+
|-valign="top" border="0"
 +
|[[Image:QPCR_result_matlab.png| thumb |left|300px|<div style="text-align:justify;">'''Figure 4: Real-time RT-PCR data of CMV and JeT promoters.''' One group of HeLa cells were transfected with plasmid containing CMV promoter coupled to GFP. Another group with JeT promoter coupled to GFP was used as reference. RNA was extracted after 20 h and 50 h, followed by real-time RT-PCR. The Ct values were collected with a threshold of 0.05. The CMV activity compared to JeT at the same time point was calculated in MatLab as "arbitrary units" which correspond to amount of mRNA.</div> ]]
 +
|[[Image:qpcr_result.png| thumb |left|300px|<div style="text-align:justify;">'''Figure 5: Real-time RT-PCR data of CMV promoter'''. Arbitrary units of CMV divided by that of JeT is the [https://2009.igem.org/Team:Heidelberg/Project_Measurement RMPU]. </div>]]
 +
|-
 +
|}
-
=== Final remarks ===
+
[[Team:Heidelberg/Project_Measurement#Measurement|[TOP]]]
-
We have developed two independent methods for the generation of truly synthetic promoter for use in mammalian cells and discussed possibilites for their combination and improvement.  We are looking forward to continuing this work and generating promoters which can be used in medical or biotechnological applications, such as transcriptional targeting in virotherapy or [[Team:Heidelberg/Project_SaO|a reporter cell line]].
+
=== Measuring REU by flow cytometry and image analysis===
-
== Methods ==
+
The GFP reporter expression from [http://partsregistry.org/Part:BBa_I712004 Part:BBa_I712004  (CMV)] is examined by two-color flow cytometry[[Team:Heidelberg/Project_Measurement#References| [11]]] as well as fluorescence microscopy with subsequent image analysis. To account for potential differences in transfection efficiency, we have used the fluorescent protein mCherry coupled to the reference promoter JeT, which is co-transfected together with GFP coupled to the promoter of interest. By comparing transfection/expression levels of mCherry, we could exclude samples with low cell numbers or low transfection efficiencies. The relative fluorescence of the CMV promoter coupled to GFP was measured in three different cell lines: [[Team:Heidelberg/Eukaryopedia#HeLa|HeLa]], [[Team:Heidelberg/Eukaryopedia#MCF-7|MCF-7]], and [[Team:Heidelberg/Eukaryopedia#U2-OS|U2-OS]]. The HeLa cell line was measured five times, MCF-7 four times and U2-OS also four times by flow cytometry. The microscopy measurement was performed once for the different cell lines. As one would expect, the Relative Expression Units (REU) varied slightly between the different cell lines. Overall, the CMV promoter is very strong relative to our reference promoter JeT with values of 5.52, 6.76 and 9.73 in HeLa, MCF-7 and U2-OS respectively (Fig. 6).
-
=== RA-PCR protocol ===
+
[[Team:Heidelberg/Project_Measurement#Measurement|[TOP]]]
-
* '''All Oligos we used can be found in [[Notebook/material#Oligos_used_for_RA-PCR|Material and Methods]]
+
-
* Obtain density curves about the distribution of your TF of interest from our model. If this densitiy curve shows a decisive peak at distance >250Bp from the Transcriptional Start Site (TSS), continue with Box 2 (M-RA-PCR). If a peak is present close to the TSS, or if data is insufficient, continue here.<br>How our model was developed is detailled on the model page.<br>
+
-
* Check our model for transcription factors coinceding with your transcription factor of interest<br>
+
-
* Design two annealing sites, each 15-18 base pairs long. Annealing sites should be void of transcription factor binding sites. Calculate the reverse complement of both sequences. We used the following sequences:
+
 
-
{| class="wikitable centered" border="2" rules="rows" style="border-color:white;"
+
=== Different core promoters result in different expression strength ===
-
|-
+
 
-
!
+
We cloned the CMV core promoter in front of the JeT proximal promoter to obtain JeT_CMV. We characterized this construct in three different cell lines, HeLa, MCF-7, and U2-OS and found it to have 50% - 60%  percent of JeT's activtiy depending on the cell line (Fig. 7). Thereby, the HeLa cell line was measured five times and the MCF-7 and U2-OS cell line measurements were performed four times. The fact that variations in the core promoter can be used to vary expression strength of a certain promoter of interest comes in useful if the transfer function of an existing promoter is to be altered, and it can be used to further diversify the synthetic promoters we created. We characterized these promoters by the same methods and accuracy as CMV and JeT/CMV (see [[Team:Heidelberg/Project_Synthetic_promoters|Synthetic Promoter project]])
-
! Forward (F)
+
 
-
! Reverse Complement (RC)
+
{|
-
|-
+
|-valign="top" border="0"
-
|Annealing Sequence 1 (AS1)
+
|
-
|GGGTGACGGGTTCA
+
[[Image:HD09_CMV_standard3.png|thumb|left|300px|<div style="text-align:justify;">'''Figure 6: Flow cytometry and microscopy measurement data of CMV (REU) in different cell lines.''' The three cell lines MCF-7, U2-OS and HeLa were cotransfected with the CMV promoter coupled to GFP and a reference plasmid including the promoter JeT coupled to mCherry. The relative fluorescence (REU) of GFP was measured 20 hours after transfection. All cell lines were measured once for microscopy. The HeLa cell line was measured five times by flow cytometry and MCF-7 and U2-OS were measured four times. In the flow cytometry measurement the standard error of the mean (SEM) is indicated by the error bars.</div> ]]
-
|AGTGAACCCGTCACCC
+
|
-
|-
+
[[Image:JeT_CMV_standard2.png|thumb|left|300px|<div style="text-align:justify;">'''Figure 7: Flow cytometry measurement data of JeT_CMV (REU) in different cell lines.''' The three cell lines MCF-7, U2-OS and HeLa were cotransfected with the JeT_CMV promoter coupled to GFP and a reference plasmid including the promoter JeT coupled to mCherry. The relative fluorescence (REU) of GFP was measured 20 hours after transfection. The MCF-7 and the U2-OS cell line were measured four times and the HeLa cell line five times. The standard error of the mean (SEM) is indicated by the error bars. </div>]]
-
|Annealing Sequence 2 (AS2)
+
-
|GCGATCGGCAGATCA
+
-
|TGATCTGCCGATCGC
+
|-
|-
|}
|}
-
* Design a 5' stop oligos containing a cutsite (SpeI) and AS1_F.
+
<br>
-
* Design a 3' stop oligos containing a cutsite (HindIII) and AS1_RC.
+
[[Team:Heidelberg/Project_Measurement#Measurement|[TOP]]]
-
* Design forward and reverse Oligos for each transcription factor of your interest. Forward oligos contain AS2_F, the transcription factor binding site and AS1_F. Reverse oligos contain AS2_RC, the TFBS and AS1_RC. TFBS should be designed to represent the matrix describing the factor's binding preferences (Box 1).
+
 
-
* Design forward and/or reverse oligos for coinceding transcription factors identified in step 2 in the same way as described in step 6.<br>
+
=== A stable cell line for promoter measurement ===
-
* Design forward and/or reverse oligos for general activators.
+
[[Image:Gel_LAM-PCR1.png‎|thumb|left|300px|<div style="text-align:justify;">'''Fig. 8:  Results of second exponential PCR.''' In all lanes are  several bands visible, indicating that there is more than one FRT-site integrated in each cell line. Negative controls are: 1. untransfected genomic DNA 2. H<sub>2</sub>O negative control LAM-PCR, 3. H<sub>2</sub>O negative control first exponential PCR, 4. H<sub>2</sub>O negative control second exponential PCR. Electrophoresis was carried out on 2% agarose; [http://tools.invitrogen.com/content/sfs/manuals/15628019.pdf 100 bp DNA ladder] was used.</div>]]
-
* Design forward and reverse spacer oligos, which contain 10-15*N (random nucleotide) instead of a TFBS.
+
'''[[Team:Heidelberg/stables|Main Article: Stable cell line]]'''
-
* Order oligos at 100µM. Pool the oligos. As a general rule, use 0,8µL oft Stop5' and Stop 3'; ~4µL of the transcription factor (forward), ~4µL of the transcription factor (reverse), 1-2µL each of the forward and reverse spacer oligo, ~1µL of coinceding transcription factors and a total of 0,5µL of general activators. For the examples shown below, we used the following mixtutres of oligos:<br>
+
 
-
{| class="wikitable centered" width="800px" border="2" rules="rows" style="border-color:white;"
+
We were able to generate HeLa, MCF-7 and U2-OS cells that stably integrated the FRT-site into their genome. Fig. 8 shows the PCR products of the second exponential PCR. For each cell line there are several bands visible. Since the number of bands correlates with the number of unique integration sites [[Team:Heidelberg/Project_Measurement#References| [2]]] there must have been more than one integration of the FRT-vector into the genome of the cells.
-
|- 
+
<br>
-
! p53
+
<br>
-
! NFkB II
+
<br>
-
! HIF
+
<br>
-
! Activator Mix
+
<br>
-
|-
+
<br>
-
|width="200px"|6µL  p53 (O.91)<br>
+
<br>
-
5µL p53 reverse (O.188)<br>
+
<br>
-
1µL random (O.56)<br>
+
<br>
-
1µL Activator Mix<br>
+
<br>
-
0.8µL Stop 5 new (O.187)<br>
+
<br>
-
0.8µL Stop 3 (O.58)<br>
+
<br>
-
|width="200px"|3µL NFkB-1 (O.93)<br>
+
<br>
-
3µL NFkB-2 (O.94)<br>
+
 
-
4µL NFkB-rev (O.194)<br>
+
[[Team:Heidelberg/Project_Measurement#Measurement|[TOP]]]
-
3µL Random (O.56)<br>
+
 
-
2µL Activator Mix<br>
+
 
-
0.8µL Stop 5 new (O.187)<br>
+
== Discussion ==
-
0.8µL Stop 3 (O.58)<br>
+
 
-
|width="200px"|2,5µL  HIF-1 (O.53)<br>
+
=== Flow Cytometry/Fluorescence Microscopy ===
-
2,5µL HIF-2 (O.54)<br>
+
 
-
1µL CREB(O.89)<br>
+
Our flow cytometer measurements of CMV and JeT-CMV in different cell lines (Fig.3 and 4) show that the strength of these promoters varies in different cell lines. Furthermore, the ratio between the two promoters varies slightly amongst different cell lines, but overall CMV is considerably stronger than JeT, while JeT-CMV is consistently lower than JeT. Our measurements were reproducible considering the strength ratios between our standard promoters.  
-
3µL HIF-rev (O.189)<br>
+
Between different measurements we detected minimal fluctuations (represented by the standard error of the mean). However, as transfection efficiencies also slightly vary between experiments, this is one source of error when comparing experiments. Additionally, due to the nature of the iGEM competition, experiments have been carried out by several persons, introducing another source for errors.<br>
-
3µL Random (O.56)<br>
+
We have applied microscopy measurements to support the data obtained by flow cytometry. The initial microscopy measurements so far confirm the observations made by flow cytometry, although some inconsistencies appeared. This might partially be due to the different cell preparation for flow cytometry and microscopy. While living cells have been used for the flow cytometric measurmentes, cells were fixed prior to analysis by microscopy. So far we could only carry out one set of microscopic measurements, therefore it will be necessary to repeat the microscopic measurements to obtain reliable data.<br>
-
1µL Stop 5 new (O.187)<br>
+
Apart from supporting the flow cytometry results, fluorescence microscopy will also gain importance in the future as we are working on a stable cell line which simultaneous expresses differently coloured fluorescent proteins. These fluorescent proteins will be directed to various compartments of the cell and thereby, allow to visualize the activation of several promoters at the same time in one single cell. Read more about our future plans in the [[Team:Heidelberg/Project_SaO|outlook section]].
-
1µL Stop 3 (O.58)
+
 
-
.2µL each Ap1, Sp1 (O.55, O.57)
+
[[Team:Heidelberg/Project_Measurement#Measurement|[TOP]]]
-
|width="200px"|2µL each Ap1, Sp1, CREB (O.55, O.57, O.89)<br>
+
 
-
1µL each NFY, Empty (O.90, O.95)
+
=== Real-time RT-PCR ===
-
Water to 30µL
+
The [[Team:Heidelberg/Project_Measurement#Measuring_RMPU_by_real-time_RT-PCR|results of the real-time RT-PCR]] measurement show that the promoter strength varied significantly over time. Reasons for the variations could be due to the fact, that the activity was measured under transient transfection where the plasmid containing the promoter is not replicated when the cells proliferate. This results in a decrease of amount of plasmids per cell, thus reduced copy of promoter present in the cells. This effect will be increase within each replication. Therefore, long time intervals between measurement and transfection will be more error-prone than shorter intervals. That is why the proportion of error to data is much higher 50 h after transfection compared to the 20 h measurement, although the standard deviations of the two time points are in the same range. It can also be possible that the cells containing the plasmid with promoter construct die faster due to changes in cell physiology. Furthermore, the fluctuation of the results could be from both systematic and random error where all the following reasons could be responsible: pipetting errors during plate preparation, influence of the freeze-thaw process on enzymes, differing contents of mastermix and random error caused by the machines. This could be caused by physiological changes the plasmids and consequently, the promoters put on the cells. Over the course of 20 h the effects those changes evoked were not that distinctiv but over 50 h of interaction enlarged the difference  greatly.
-
|
+
 
-
! SREBP
+
[[Team:Heidelberg/Project_Measurement#Measurement|[TOP]]]
-
! AHR
+
 
-
! pPAR&gamma;
+
=== Characterization of promoters under different condtions ===
-
! Estrogen receptor
+
 
-
|-  
+
Considering what we learned about [[Team:Heidelberg/Project_Measurement#Identification_of_challenges_unique_to_higher_eukaryotes|the lack of absolute, non-changing reference standards]], characterizing promoters under different conditions becomes inherently difficult. All parameters affecting REU and RPMU are altered by a change in conditions. For the assumptions on which REU and RMPU are based to remain valid, promoters must only be compared in one set of conditions. In most scientific work to this date, transcriptional activity is assessed by comparing protein levels between unrelated conditions (see[[Team:Heidelberg/Project_Measurement#References| [12]]] for a typical example). Although giving sufficient information as to whether a promoter is up- or downregulated, this standard cannot be used for an absolute characterization, as if a condition induces, for example, a system-wide increase in translation, it will result in a ratio between the promoters which is not proportional to the change in transcription. If, on the other hand, measurements are related to a reference promoter at each condition individually, this does not represent true levels of change if the reference promoter itself is up- or downregulated by that condition. Providing absolute measurements to compare between different conditions will be a major challenge for synthetic mammalian biology in the next years. We suggest, for now, always to give promoter strength relative to the strength of JeT under the condition regarded.
-
|width="200px"|5µL  SREBP (O.208)<br>
+
 
-
4µL SREBP reverse (O.209)<br>
+
[[Team:Heidelberg/Project_Measurement#Measurement|[TOP]]]
-
1µL Sp1 (O.57)
+
 
-
2µL random (O.56)<br>
+
=== Towards PoPs ===
-
1µL Activator Mix<br>
+
 
-
0.8µL Stop 5 new (O.187)<br>
+
The concept of RMPU can easily be converted into PoPs, at least for constitutive promoters. RNA degradation equals RNA synthesis in steady state. Therefore, we suggest to block RNA synthesis by applying Actinomycin D which works in minutes[[Team:Heidelberg/Project_Measurement#References| [13]]], and then measuring RNA degradation. According to [[Team:Heidelberg/Project_Measurement#Two_units_for_promoter_activity_in_mammalian_cells|equation 2]], PoPS will then equal mRNA degradation, divided by copy number (which is 1, if the experiment is conducted in a stable cell line). Of course, Actinomycin is toxic to cells and might affect mRNA turnover rates system-wide. Accounting for this effect will present a major challenge.
-
0.8µL Stop 3 (O.58)<br>
+
 
-
|width="200px"|5µL  AHR (O.212)<br>
+
[[Team:Heidelberg/Project_Measurement#Measurement|[TOP]]]
-
4µL AHR reverse (O.213)<br>
+
 
-
2µL random (O.56)<br>
+
=== Concluding remarks ===
-
1,5µL Activator Mix<br>
+
 
-
0.8µL Stop 5 new (O.187)<br>
+
We reported two new units for promoter characterization in mammalian cells, one of which being directly proportional to PoPS. We showed the advantages and limitations of the concept. Future work will have to focus on improving comparisons between different conditions.
-
0.8µL Stop 3 (O.58)<br>
+
 
-
|width="200px"|5µL  pPAR&gamma; (O.210)<br>
+
[[Team:Heidelberg/Project_Measurement#Measurement|[TOP]]]
-
4µL pPAR&gamma; reverse (O.211)<br>
+
 
-
2µL random (O.56)<br>
+
== Method details ==
-
1,5µL Activator Mix<br>
+
 
-
0.8µL Stop 5 new (O.187)<br>
+
=== Flow Cytometry ===
-
0.8µL Stop 3 (O.58)<br>
+
 
-
|width="200px"|5µL Estrogen receptor (O.210)<br>
+
Flow cytometry is a commonly used method for the measurement of fluorescence intensity levels. It uses the principles of light scattering, light excitation, and emission of fluorochrome molecules to generate specific multi-parameter data from particles and cells in the size range of 0.5 µm to 40 µm diameter . One unique feature of flow cytometry is that it measures fluorescence per cell or particle. In contrast to high-resolution microscopy, flow cytometric measurements do not reveal intracellular distribution of the measured signal, but give an integrated signal of each cell. However, flow cytometry allows fast, multi-parametric measurements of high numbers of cells (>1000 cells/sample) in a quantitative manner [[Team:Heidelberg/Project_Measurement#References|[14]]], [[Team:Heidelberg/Project_Measurement#References|[19]]].<br>
-
4µL Estrogen receptor reverse (O.211)<br>
+
A flow cytometer consists of the fluidics system where cells are hydro-dynamically focused in a sheath of fluid; one or several laser which is the source of focused light for fluorescence and scatter; the optics which gather and direct the light emitted from the cells; up to 8 detectors which receive the light and convert it into an electrical signal, which is finally delivered to a computer system to analyze the signals of each cell. <br>
-
2µL random (O.56)<br>
+
The first detector (Forward Scatter; FSC) is in line with the light beam; its signal is proportional to the size of the cell. The larger the cell, the more light is scattered and the higher the electric signal in the detector. Perpendicular to the light beam are several other detectors, including the Side Scatter (SSC). The Side Scatter represents the granularity of the cell and this can be used to further distinguish different cell populations and exclude apoptotic cells. A two-dimensional scatter plot of Forward versus Side Scatter can give a more detailed overview over the nature of the cells in the sample. This plot is used to separate dead cells and cell debris from healthy cells. [[Team:Heidelberg/Project_Measurement#References|[14]]], [[Team:Heidelberg/Project_Measurement#References|[19]]]<br>  
-
1,5µL Activator Mix<br>
+
Following the Side Scatter, the light is directed through a series of mirrors and filters, so that particular wavelengths are delivered to the correct detector. The fluorescence signal is then converted to a voltage signal and can be visualized as a histogram, showing frequencies of cells with different fluorescence levels (usually distributed over 1024 channels) [[Team:Heidelberg/Project_Measurement#References|[14]]], [[Team:Heidelberg/Project_Measurement#References|[19]]]. <br>
-
0.8µL Stop 5 new (O.187)<br>
+
We have used a Beckman Coulter FC500 MPL, equipped with a 488 nm and 561 nm laser, thereby allowing simultaneous and quantitative measurement of GFP and mCherry fluorescence. <br>
-
0.8µL Stop 3 (O.58)<br>
+
Cells were prepared in 96-well format with 10<sup>4</sup> cells/well and transfected with the promoter of interest and when necessary, induction drug was added for inducible promoters. Before measurement, the medium was removed, the cells were washed with 1xPBS and trypsinized with 60 µl of trypsin per well. After 10 minutes incubation at 37°C, 1xPBS + 1% BSA were added up to a volume of 200 μl per well. When starting the flow cytometry measurement we first adjusted the gates and the background signal to the negative control (Fig. 9). Fig. 10 shows an example of a gated positive control (JeT), where we can see a second normal distribution peak indicating the gated GFP positive cells.
 +
 
 +
{|
 +
|-valign="top" border="0"
 +
|width="300px" style="padding: 0 20px 0 0;"|
 +
[[image:HD09_GFP_negative.png|center|300px|thumb|<div style="text-align:justify;">'''Figure 9 : Example of a negative control in Hela cells by flow cytometry.''' The number of events is plotted against the fluorescence (log) of GFP. Background signal is set under 10 and the area under the curve is colored in green.</div>]]
 +
|width="30px"|
 +
[[image:HD09_GFP_positive_HeLa_Jet.png|center|300px|thumb| <div style="text-align:justify;">'''Figure 10 : Example of a positive control (JeT) in Hela cells by flow cytometry.''' The number of events is plotted against the fluorescence (log) of GFP. The area under the curve is colored in green.</div>]]
|}
|}
-
* Introduce the oligos thus pooled into a PCR reactions at a final dilutionof 1:200-1:500. We used Phusion MasterMix 2x (Finnzymes) as PCR reagent. Do that PCR reaction twice in order to achieve greater heterogenety.<br>
 
-
* Run the PCR, 7-10 cycles, with the following setup:<br>
 
-
**1 cycle Initial dentaturing, 5 minute 95°C<br>
 
-
**7-10  cycles assembly: 30 seconds 95°C, 45 seconds 58°C, 45 seconds 72°C<br>
 
-
**Terminal hold, 4°C, forever<br>
 
-
* Remove oligonucleotides by performing a PCR purification using PCR purification kit (QIAGEN) or a gel extraction using Gel extraction kit (QIAGEN)<br>
 
-
* Add PCR reagent (Phusion MasterMix 2x) again. Add 5' stop oligo and 3' Stop oligo, 25pmol (1µL of 1:4 diluted stock). <br>
 
-
* Run the PCR, 25 cycles, with the following setup:<br>
 
-
**1 cycle Initial dentaturing, 5 minute 95°C<br>
 
-
**25 cycles amplification 30 seconds 95°C, 45 seconds 68°C, 60 seconds 72°C<br>
 
-
**Terminal hold, 4°C, forever<br>
 
-
* Gel purify PCR products to exclude everything <200Bp. Use a 1% agarose gel, 50V for at least 2h to achieve a good resolution<br>
 
-
* Digest with HindIII and SpeI (or whatever cutsites were included in step 4 and 5). Digest a reporter plasmid containing a core promoter and a reporter gene with the same enzymes. We used the plasmids (containing GFP as a reporter) for this task. Make sure to perform a thorough digest; in addition, digest the plasmid with shrimp alkaline phosphatase or calf intestine phosphatase afterwards. Gel purify the plasmid backbone, PCR purify the digested PCR products.<br>
 
-
* Ligate. Perform a thorough ligation to increase transformation efficiency. We used Fermentas T4 DNA Ligase for 5h, 19°C or overnight, 16°C.<br>
 
-
* Transform into comptetent E. Coli cells and plate out. Pick no more than 20 colonies per individual PCR reaction. If more putative promoters are desired, set up several PCR reactions<br>
 
-
* Isolate plasmid DNA from the selected colonies. We used a QIAGEN Miniprep kit for this tasked.<br>
 
-
* Recommended step: Test-digest miniprep DNA with the same enzymes used in step 17 to make sure you get plasmid with synthesized promoters of varying length. Length of the inserts (that is, synthetic promoters) should be between 100 and 600 basepairs. If this is not the case, vary stop oligo concentration in step 10, improve gel purification setup in step 16 or alter PCR conditions in step 12 and 15.<br>
 
-
* Perfom a screening to select functioning clones. For example, transfect clones in triplicates into eukaryotic cells on a 96 well palte by using transfection agents such as EFFECTENE or Lipofectamine. Then, induce the conditions of interest in one replicate, shut them off in a second replicate, and leave control medium on the third replicate. When the pathway is fully active,  read flourescence (or luminescence, if a luciferase reporter is used) by a plate reader (TECAN) or other automated methods. We used the following conditions for promoter screening:<br>
 
-
'' to be added ''
 
-
=== References ===
+
[[Team:Heidelberg/Project_Measurement#Measurement|[TOP]]]
-
[1] Alberts, B. et al. Molecular Biology of the, Cell (5th edition). New York: Garland Science, p. 432-453
+
 
 +
=== Microscopy and Image Analysis ===
 +
 
 +
For further characterization of our promoters we decided to use fluorescence microscopy. Our cells are co-transfected (1:2) with a reference plasmid, which contains JeT in front of mCherry. Analysis of reporter gene expression can be restricted to cells that express the reference plasmid and are therefore likely to have been successfully co-transfected with the reporter plasmid of interest. This is particularly useful when transfection efficiency is low. We applied this approach to microscopy measurement to minimize the experimental error. The promoter strength was evaluated in relative expression units (REU) relative to the JeT promoter which was set to 1. Microscopy images of our samples fixed with 4% formaldehyde were taken using the Nikon Eclipse 90i upright automated widefield microscope in the Nikon Imaging Center at the University of Heidelberg. Each image was taken in the GFP and mCherry channel and the exposure time had to be adjusted for over- or underexposed images. The conversion factor was experimentally determined by imaging a fluorescent plate with exposure time varying from 10 to 70 ms (the estimated range of our promoters) and plotting the mean grey values of these images versus time. A linear relationship between the exposure times and grey values was observed for this time frame and the conversion factor was determined to be 0.998. For image analysis we used ImageJ (Image Processing and Analysis in Java) software tools for obtaining the mean grey values of each cell containing the promoter of interest. ImageJ is an open source software developed at the National Institutes of Health (NIH (http://rsbweb.nih.gov/ij/).
 +
The measurement is performed by setting the boundaries around the cells using the threshold in the mCherry channel image and generating a mask that is then applied to the image of the GFP channel. For a step by step description on ImageJ analysis see: [[Team:Heidelberg/Notebook_MaM| Materials and Methods]].
 +
 
 +
[[Team:Heidelberg/Project_Measurement#Measurement|[TOP]]]
 +
 
 +
=== Real-time RT-PCR ===
 +
Real-time RT-PCR, which has been developed tremendously during the last 20 years, is a specific and sensitive technique for detecting and quantification of gene expression on RNA level. In order to obtain reliable results and reduce the variance during the extraction, reverse transcription and PCR, we did the measurement for each promoter with 12 replicates from an identical sample. Prior to PCR, we extracted the total RNA using Qiagen RNeasy Plant Mini Kit following the protocol from the handbook [[Team:Heidelberg/Project_Measurement#References|[15]]]. The extraction was performed by the QIAcube<sup>TM</sup> [[Team:Heidelberg/Project_Measurement#References|[16]]], the automated spin-column kits preparation robot, to avoid possible operator error. In order to perform the real-time PCR, primers and probes were designed according to QuantiFast Probe RT-PCR Handbook [[Team:Heidelberg/Project_Measurement#References|[17]]]: The target PCR product length is set between 70-200 bp. The target spans over exon-exon boundary to exclude amplification of genome DNA which may be presented in the RNA extraction as contamination. The probes have reporter fluorophore and quencher fluorophore at 5' and 3' end respectively. During amplification, probes bind specifically to the target gene. The polymerase hydrolyzes the probe during elongation due to its exonuclease activity. The signal of the released fluorophore can then be detected and correlates therefore to the amount of PCR product. The real-time RT-PCR was performed on StepOnePlus<sup>TM</sup> Real-time PCR System from Applied Biosystems [[Team:Heidelberg/Project_Measurement#References|[18]]]. We quantified the expression of GFP, the reporter gene, together with other 5 housekeeping genes (ß-actin, glycerol-aldehyde-3-phosphate de-hydrogenase, glucose-6-phosphate dehydrogenase, gamma-tubulin, 18S rRNA) for normalization. At the end of each cycle, the fluorophore of each well is read and recorded. Ct (threshold cycle) is defined as the cycle number at which the fluorescent strength (the curve) crosses a certain threshold (Fig. 11). The more amount of starting mRNA, the sooner will the machine detect the fluorescent signals from the PCR reaction, which means a smaller Ct under the same threshold. The threshold should be a careful decision. With a too small threshold, the signal may be too weak for reliable detection. If it is too big, the reaction might be restricted by the enzymes and other materials. Usually, we set the threshold at 0.05 for all plates (Fig. 12.1 and Fig. 12.2). Calculating the difference between Ct values, with normalization of the multiple housekeeping genes, we get the ratio of the activity of the promoter of interest and our reference.
 +
 
 +
{|
 +
|-valign="top" border="0"
 +
|width="300px" style="padding: 0 20px 0 0;"|
 +
[[image:qPCR_Ct_18s_CMV.png|center|210px|thumb|<div style="text-align:justify;">'''Figure 11 : Ct values of 18s rRNA with CMV promoter 20h after transfection''' Threshold is set at 0.05, the cycle number at when the amplification plot crosses the threshold is the Ct value.</div>]]
 +
|width="30px"|
 +
[[image:qPCR_Ct_eGFP_CMV.png|center|210px|thumb| <div style="text-align:justify;">'''Figure 12.1 : Ct values of eGFP with CMV promoter 20h after transfection''' Threshold is set at 0.05. 12 dots represent 12 RNA extractions from identical sample.</div>]]
 +
|width="30px"|
 +
[[image:qPCR_Ct_eGFP_JeT.png|center|210px|thumb|<div style="text-align:justify;"> '''Figure 12.2 : Ct values of eGFP with JeT promoter 20h after transfection''' Threshold is set at 0.05. 12 dots represent 12 RNA extractions from identical sample.</div>]]
 +
|}
 +
 
 +
[[Team:Heidelberg/Project_Measurement#Measurement|[TOP]]]
 +
 
 +
== References ==
 +
<div style="text-align:justify;">
 +
[1] Kelly J. R., Rubin A. J., Davis J. H., Ajo-Franklin C. M., Cumbers J., Czar M. J., de Mora K., Glieberman A. L., Monie D. D. & Endy D. Measuring the activity of BioBrick promoters using an in vivo reference standard. ''Journal of Biological Engineering'' 3 (2009).<br>
 +
 
 +
[2] Endy D. & Deese I. Adventures in synthetic biology (comic) ''Nature'' 438: 449-453 (2005). Available online at [http://www.nature.com/nature/comics/syntheticbiologycomic/index.html nature.com]<br>
 +
 
 +
[3] Alberts B., Johnson A., Walter P. & Lewis J. ''Molecular Biology of the Cell.'' 5th edition, 2008. Garland Science) Chapter 6<br>
 +
 
 +
[4] http://www.promega.com/tbs/tm058/tm058.html<br>
 +
 
 +
[5]  Alberts B., Johnson A., Walter P. & Lewis J. ''Molecular Biology of the Cell''. 5th edition, 2008. Garland Science) Chapter 7, p. 467-476<br>
 +
 
 +
[6] Zhu X.D. & Sadowski P. D. Cleavage-dependent Ligation by the FLP Recombinase. ''J Biol Chem'' 270: 23044-23054 (1995).<br>
-
[2] Edelmann, G.M. et al. Synthetic promoter elements obtained by nucleotide sequence variation and selection for activity. PNAS 97, 3038-43 (2000).
+
[7] York J. D., Odom A. R., Murphy R., Ives E. B. & Wente S. R. A phospholipase C-dependent inositol polyphosphate kinase pathway required for efficient messenger RNA export. ''Science'' 285: 96-100 (1999).<br>
-
[3] Ellis, T. et al. Diversity-based, model-guided construction of synthetic gene networks with predicted functions. Nature Biotechnology 27, 465-471 (2009).
+
[8] Alberts B., Johnson A., Walter P. & Lewis J. ''Molecular Biology of the Cell.'' 5th edition, 2008. Garland Science) Chapter 7, p. 905-908<br>
-
[4] Venter, M. Synthetic promoters: genetic control through cis engineering. Trends in Plant Science 12, 118-124 (2007). (and the references cited therein)
+
[9] Tornoe J. Generation of a synthetic mammalian promoter library by modification of sequences spacing transcription factor binding sites. ''Gene'' 297: 21-32 (2002) <br>
-
[5] Rushton, P.J. et al. Synthetic plant promoters containing defined regulatory elements provide novel insights into pathogen- and wound-induced signalling. in Plant Cell 14, 749–762 (2002).
+
[10] Alberts B., Johnson A., Walter P. & Lewis J. ''Molecular Biology of the Cell.'' 5th edition, 2008. Garland Science) Chapter 7, p. 492<br>
-
[6] Ogawa, R. Construction of strong mammalian promoters by random cis-acting element elongation. Biotechniques 42, 628-632 (2007).  
+
[11] Ducrest A. L., Amacker M., Lingner J. & Nabholz M. Detection of promoter activity by flow cytometric analysis of GFP reporter expression. ''Nucleic Acids Res.'' 30: e65 (2002). <br>
-
[7] Stemmer, W.P.C. et al. Single-step assembly of a gene and entire plasmid from large numbers of oligodeoxyribonucleotides. Gene 164, 49-53 (1995).
+
[12] Rushton P. J., Reinstädler A., Lipka V., Lippok B. & Somssich I. E. Synthetic plant promoters containing defined regulatory elements provide novel insights into pathogen- and wound-induced signalling. ''Plant Cell'' 14: 749-762 (2002).<br>
-
[8] Tornoe, J. Generation of a synthetic mammalian promoter library by modification of sequences spacing transcription factor binding sites. Gene 297, 21-32 (2002).
+
[13] Degenhardt T., Rybakova K. N., Tomaszewska A., Moné M. J., Westerhoff H. V., Bruggeman F. J. & Carlberg C. Population-Level Transcription Cycles Derive from Stochastic Timing of Single-Cell Transcription. ''Cell'' 138: 489-501 (2009).<br>
-
[9] Heintzman ND, Ren B. The gateway to transcription: identifying, characterizing and understanding promoters in the eukaryotic genome. Cellular and Molecular Life Science 64, 386-400 (2007).
+
[14] Lottspeich F. & Engels J. W.: ''Bioanalytik'' (Book. 2nd edition, 2006. Spektrum Akademischer Verlag) Chapter 5, p. 95-96.
-
[10] Fussenegger, M., Weber, W. Engineering of Synthetic Mammalian Gene Networks. Chemistry and Biology 16, 287-297 (2009).
+
[15] http://www1.qiagen.com/Products/RnaStabilizationPurification/RNeasySystem/RNeasyPlantMini.aspx
-
[11] Gossen, M., Bujard, L. Tight control of gene expression in mammalian cells by tetracycline-responsive promoters. PNAS 89, 5547-5551 (1992).
+
[16] http://www1.qiagen.com/products/automation/qiacube.aspx
-
[12] Dorer, D.E., Nettelbeck, D. Targeting cancer by transcriptional control in cancer gene therapy and viral oncolysis. Advanced Drug Delivery Reviews 61, 554-557 (2009).
+
[17] http://www1.qiagen.com/literature/render.aspx?id=398
-
[13] Rattner, A. NF-kappa B activates the HIV promoter in neurons. EMBO 12, 4261–4267 (1993).
+
[18] https://products.appliedbiosystems.com/ab/en/US/adirect/ab?cmd=catNavigate2&catID=604109&tab<br>=DetailInfo<
-
[14] Yokoyama KD et al. Measuring spatial preferences at fine-scale resolution identifies known and novel cis-regulatory element candidates and functional motif-pair relationships. Nuc Acids Res, 1-21 (2009).
+
[19] Ormerod M.G.: ''Flow cytometry: a practical approach'' (Book. 3rd edition, 2000. Oxford University Press) Chapter 1, p. 7-21.
-
[15] del Val C. et al.  PromoterSweep: a tool for identification of transcription factor binding site. Theor Chem Acc (in press)
+
[20] Zhou H. Development of site-specific integration system to high-level expression recombinant proteins in CHO cells. ''Chinese journal of biotechnology'' 23: 756-62 (2007).
 +
[[Team:Heidelberg/Project_Measurement#Measurement|[TOP]]]
 +
</div>
|width="250px" style="padding: 0 20px 15px 15px; background-color:#d8d5d0"|
|width="250px" style="padding: 0 20px 15px 15px; background-color:#d8d5d0"|
|}
|}

Latest revision as of 23:22, 21 October 2009

Measurement

Abstract

In order to achieve defined protein levels in a cell, promoters of defined strength are an obvious requirement. Such promoters can only be valuable to synthetic biology if they are well characterized. For future eukaryotic devices that require PoPS (Polymerase per second) as an input, our promoters will be very suitable since they deliver PoPS as an output. PoPS is the standard unit of synthetic biology, but it is very difficult to measure directly. For bacteria, relative measurements (relative promoter units, RPU) are most commonly used and it has been shown how to convert them to PoPS [1]. In this project, we identify and deal with several challenges to achieve the same in mammalian cells, suggest solutions and provide easy-to-use relative measures for application in mammalian cells - one based on RNA levels (Relative Mammalian Promoter Units, RMPU), the other based on folded protein levels (Relative Expression Units, REU). We apply those measurements on the characterization of CMV, an existing promoter from the registry. Finally, we discuss how to transform these units into PoPS.

Introduction

The need for standardized measurements of promoter activity in vivo has been widely accepted across the synthetic biology community [1]. Only if a part is well characterized initially, function of an engineered device or system can be predicted reliably. Most work of synthetic biology has focused on bacteria, especially Escherichia coli, as a model system. Novel tasks in synthetic biology, especially for medical applications, will require synthetic biology of mammalian cells. Mammalian systems are the most complex biological systems, and therefore, little work has been done in the field of mammalian synthetic biology, leaving a huge potential for future research.

Box 1: Calculation of PoPS, where γM is the mRNA degradation rate, a is the GFP maturation rate, γI is the degradation rate of immature GFP, ρ is the translation rate of immature GFP from mRNA and n is the number of copies of promoter per cell. [1]
The holy grail of synthetic biology measurements is to express device input and output in "Polymerase per Second" (PoPS), an absolute unit which describes the rate of RNA polymerase molecules passing by the final base pair of the promoter [2]. If PoPS is to be measured from protein expression (e.g. GFP expression), in bacteria PoPS can be calculated from GFP synthesis rate, mRNA degradation, GFP maturation rate, GFP translation rate, plasmid copy number and degradation rate of immature GFP (see Box 1). Only GFP levels are easy to measure by fluorescence readouts, the other factors require complex experiments. Therefore, it has become a standard procedure to characterize promoters in RPUs. One RPU is the activity equivalent to BBa_J23101. [1] This is possible because by cloning both the promoter and the reference separately but into the same plasmid backbone, promoters will create the same mRNA (as we always used GFP as a readout), thus having the same mRNA degradation and translation rate. In the same way, plasmid copy number, GFP maturation and degradation rates should be equal when using the one cell line at same conditions.

For mammalian cells, no such unit has been reported, nor is there a kit for measuring promoter activity. Mammalian cells have higher levels of complexity - the process from a gene to a functional protein involves transcription (regulated by the promoter itself and chromatin structure), RNA modification, RNA splicing, RNA export, RNA degradation, protein folding and protein degradation [3]. All of these processes are highly regulated, both generally and specifically for the individual gene.

A frequently used system is the dual luciferase assay. We dismissed the idea of using luciferase as it is an invasive technique - it requires killing the cells before measuring [4]. This makes time-course experiments impossible. Moreover, GFP expression can be detected non-invasively with a wide variety of methods. As outlined below, simple fluorescence measurements by fluorimeters cannot be the method of choice for measuring promoter activity in mammalian systems.

For the quantification of reporter gene expression in each transfected cell, we used a dual assay with GFP and mCherry. Our cells are co-transfected with a reference plasmid containing our reference promoter in front of mCherry. The fluorescence intensities were measured by flow cytometry and fluorescence microscopy followed by ImageJ processing.

On the other hand, we attempted to measure the promoter strength through quantification of gene expression on mRNA level. Hereby, we applied the novel technique real-time RT-PCR. The method consists of 2 steps, a reverse transcription, which synthesizes cDNA from extracted mRNA, and a real-time PCR, which is based on normal PCR, but collects the data throughout the PCR process, thus in real-time. Signal detection is achieved using fluorescent dye, which can be attached to a probe . The accumulation of PCR products over cycles results in the increase of fluorescence. By comparing of cycles required to achieve a certain amount of fluorescence the difference in mRNA expression profile can be illustrated. Through quantification of GFP mRNA expression driven by the test promoters, the difference in promoter strength can be easily illustrated. This approach measures directly on mRNA level, which means, translation and maturation of the GFP molecule do not have to be taken into account as in the flow cytometry approach.

However, a common problem when measuring any construct or device in mammalian cells is the fact that the transfection rate is not always constant. Thus, one never knows how many copies of a construct are actually in the cell. This is why we attempted to create a system that allows a controlled integration of our constructs into the genome. To accomplish this we used the FRT/Flp system which is based on homologous recombination by the enzyme flippase at a specific sequence – the FRT site [20]. Read more about our stable cell lines

[TOP]

Results

Identification of challenges unique to higher eukaryotes

In this chapter, we start by identifying critical points of promoter characterization in eukaryotes and specify how we proceeded to take on these challenges:

Variance of transfection efficiencies

Fig. 1: Transfection efficiency for vectors containing different inserts varies. By flow cytometry, we identified transfected cells and found transfection efficiencies to vary. Here, the transfection efficiency is shown for HeLa and e standard error of the mean (SEM) is also displayed.
In prokaryotes and yeast, plasmid copy number is a property of the plasmid as determined by the origin of replication. Higher eukaryotes are unable to stably propagate plasmids, unless stable integration occurs (which does not happen spontaneously). Even plasmids with minor differences show entirely different transfection efficiencies and therefore will result in large variations of GFP expression per well. By flow cytometry, we analyzed transfection efficiency for HeLa cells and calculated plasmid number to vary greatly. We transfected cells with plasmids containing the same backbone, but different (promoter) inserts and found transfection efficiency to vary as much as 20% (Fig. 1). Also, as expected, a significant percentage of cells remains untransfected in a experiment (Fig. 1).

Dual assay measurements using co-transfection as a control for transfection efficiency are commonly used[4]. Such assays can be utilized for relative characterizations, but do not provide accurate normalization. In order to resolve this issue, we suggest a method which not only allows relative characterizations but is also able to distinguish between transfected and untransfected cells (flow cytometry or image analysis). For experiments aiming at absolute numbers, stable integration of the measurement plasmid into the mammalian genome containing exactly one integration site is required. In this case the copy number per cell is one.

Regulation of promoter activity by chromatin structure

Considering stable integration of a promoter into the mammalian genome, another level of complexity is added: Chromatin structure affects transcription, though mostly not affected by the promoter[5]. Therefore, integration at different sites of the genome will not result in the same expression strength.

A cell line having a FRT site stably integrated into the genome is required. Such a cell line involves the recombination of sequences between short Flippase Recognition Target (FRT) sites by the Flippase recombination enzyme (FLP or Flp) derived from the 2µ plasmid of the baker's yeast Saccharomyces cerevisiae.[6]. We started the development of such a cell line.

[TOP]

RNA processing

Many RNAs are spliced after transcription. Also, RNAs are processed before being exported from the nucleus, and can be retained in the nucleus both dependent and independently of processing [3], [7]. Therefore, several mRNA populations exist: Unprocessed, processed and retained in the nucleus, and functional mRNA outside of the nucleus. These effects are a current research focus of molecular cell biologists and barely understood even in model organisms as simple as yeast.

Splicing does not present a problem for reporter gene constructs, as these are cDNAs which are not spliced. Nevertheless, this means that the ratios between RNA levels and protein levels will change for a single promoter if a different output gene is linked to the promoter. We therefore suggest to assume a black box between RNA and protein, and the introduction of two independent units.

[TOP]


Variety of cell lines

TOP10 or DH5α cells have been widely accepted as chassis systems by synthetic biologists working in bacteria. For the work with mammalian cells, no such consensus exists; also, it would not be sensible to limit synthetic mammalian biology to a small number of cell lines, as every cell line is suited for a special application. Scientists working on breast cancer virotherapy by synthetic promoters would choose a breast cancer cell line, whereas scientists working on stem cell therapies to myocardial infarction would choose a cardiomyocyte cell line. Cell lines differ greatly, even in expression strength of constitutive promoters (shown below).
Each part must be characterized in every cell line. We worked with three cancer cell lines, HeLa (cervical cancer), MCF-7 (breast cancer) and U2-OS (osteosarcoma). We suggest that the synthetic biology community should pick a small number of cell lines as model systems. We argue that HeLa (well known, widely used, easy to transfect) and/or MCF-7 (very robust to apoptosis) should be part of these cell lines.


Lack of truly constitutive promoters in mammalian cells

Fig. 2: CMV and JeT strength changes depending on conditions We characterize GFP expression from JeT and CMV fluctuate dependent on condition (Everolimus induces extreme starvation). Measured by flow cytometry 20 hours after transfection (unless specified otherwise) in MCF-7. The standard deviation is represented by the error bars.

Figure 2 shows measurements of GFP expression from CMV and JeT under varying conditions. This result demonstrates that every promoter in mammalian cells underlies regulation, and therefore, is not truly constitutive. We analyzed the sequence of CMV by TRANSFAC Professional and found it to contain two NF-κB binding sites, two CREB-binding sites, and single Ap1, RFX1 and SRF binding sites. Of NF-κB, CREB and Ap1, we know that they have a high constitutive activity (compare to Synthetic promoters), but nevertheless, they underlie regulation. For example, NF-κB is induced by inflammation conditions, whereas CREB is activated by the second messenger cAMP[8] and thus responds to many hormones, starvation conditions etc. This impedes comparison of promoters in different conditions. We next discuss how this can be achieved.




[TOP]


A promoter measurement kit for use in mammalian systems

Fig. 3: Plasmid map of pSMB_MEASURE. BBb sites are shown in red, core promoter in light blue and proximal promoter in dark blue. The length of this plasmid is 5162 bp.
We created a plasmid , pSMB_MEASURE (SMB is for Synthetic Mammalian Biology; Part:BBa_K203100), which should be used for promoter characterization in mammalian cells. pSMB_MEASURE (see Fig. 3) contains a reference promoter, JeT[9] (Part:BBa_K203112), which is flanked by BBb_2 (Tom Knight) sites and can therefore be replaced by the promoter to be measured. JeT is ideal as a reference promoter for a variety of reasons. First, it has an intermediate expression strength; second, it is regulated by a wide variety of transcription factors and low levels of change in fluorescence among different conditions (compare Fig. 2)[9]. Third, we want to pay tribute to its creators as pioneers in synthetic promoter research.

We separated JeT's core promoter from its proximal promoter by a HindIII site; it can therefore be used for the de novo creation of synthetic promoters containing the JeT core promoter or for modifying the strength of a promoter by core promoter swapping. In addition, it contains a FRT site which will allow for stable integration into mammalian cells also containing a FRT site. Thus, it provides the possibility to characterize the promoter in a defined genome and in this way helps to avoid some of the challenges outlined above. For the same reason, it also contains a mammalian selection marker (hygromycine). For the generation of the plasmid, please see part design. As a reporter gene, it contains GFP, which is followed by a SV40 mammalian terminator. We generated another plasmid pSMB_REFERENCE, which contains mCherry instead of GFP. It can be used for normalizations of transfection efficiency in flow cytometry and image analysis.

[TOP]

Two units for promoter activity in mammalian cells

We define two units: 1 relative mammalian promoter unit (RMPU) is defined in analogy to[1], but mRNA-based. It is the amount of total mRNA (that is, processed and unprocessed mRNA inside and outside of the nucleus) generated by a promoter x, relative to the amount of total mRNA generated by the JeT promoter j (BBa_K203112)[9] in steady state. It is directly proportional to PoPs, as we show. In steady state, change of mRNA levels is 0, thus:

HD09 eq1.png

Where M' is the change in total mRNA level, γM is the mRNA degradation rate and n is the number of promoters per cell (adapted from [1]. Therefore:

HD09 eq2.png

Since both promoters are cloned seperatly into the same backbone, they generate identical mRNA. As the most important determinant for mRNA stability is the 3' untranslated region, and mRNA stability is generally not affected by promoter structure [10], we are able to assume the same mRNA degradation rate γM for both promoters.

HD09 eq3.png

We also expect the same n for both promoters as this is normalized for during the measurements (as shown below). We then define RMPU, and achieve a cancelation of terms:

HD09 eq4.png

On the other hand, we define Relative Expression Units (REU). 1 relative expression unit (REU) is the amount of total folded protein generated by a promoter, relative to the amount of folded protein generated by the JeT promoter (BBa_K203112) under the same cellular condition. It is not directly proportional to PoPS as too many levels of regulation, such as post-transcriptional modifications, enhanced splicing and nuclear shuttling/transport lie between transcription and protein [3], and REUs would strongly depend on conditions which affect RNA. Still, it is a very useful measure, as for most applications such as metabolic pathway engineering, protein level, not mRNA levels, are of importance. Also, it can be measured with a much easier experimental setup than RMPU. See discussion for ideas on how to compare REU measurements taken at different conditions.

[TOP]


Measuring RMPU by real-time RT-PCR

To measure the RMPU (Relative Mammalian Promoter Units), HeLa cells were transfected with plasmids containing the promoter of interest. HeLa cells transfected with plasmid containing JeT promoter were used as reference. At two different time-points (20 h and 50 h after transfection), total RNA (> 200 bp) was isolated. This step was followed by real-time RT-PCR, where the mRNA amount of GFP was quantified. For each promoter at each time point, 12 replicates were taken to obtain reliable results and reduce the variance. To get comparable results of different samples, we used multiple housekeeping genes as internal controls, while using non-transfected HeLa mRNA as plate-to-plate correction. The CMV promoter was calculated about 2.89 times stronger as JeT after 20 h and 2.04 times stronger after 50 h (Fig. 4 and 5).

Figure 4: Real-time RT-PCR data of CMV and JeT promoters. One group of HeLa cells were transfected with plasmid containing CMV promoter coupled to GFP. Another group with JeT promoter coupled to GFP was used as reference. RNA was extracted after 20 h and 50 h, followed by real-time RT-PCR. The Ct values were collected with a threshold of 0.05. The CMV activity compared to JeT at the same time point was calculated in MatLab as "arbitrary units" which correspond to amount of mRNA.
Figure 5: Real-time RT-PCR data of CMV promoter. Arbitrary units of CMV divided by that of JeT is the RMPU.

[TOP]

Measuring REU by flow cytometry and image analysis

The GFP reporter expression from Part:BBa_I712004 (CMV) is examined by two-color flow cytometry [11] as well as fluorescence microscopy with subsequent image analysis. To account for potential differences in transfection efficiency, we have used the fluorescent protein mCherry coupled to the reference promoter JeT, which is co-transfected together with GFP coupled to the promoter of interest. By comparing transfection/expression levels of mCherry, we could exclude samples with low cell numbers or low transfection efficiencies. The relative fluorescence of the CMV promoter coupled to GFP was measured in three different cell lines: HeLa, MCF-7, and U2-OS. The HeLa cell line was measured five times, MCF-7 four times and U2-OS also four times by flow cytometry. The microscopy measurement was performed once for the different cell lines. As one would expect, the Relative Expression Units (REU) varied slightly between the different cell lines. Overall, the CMV promoter is very strong relative to our reference promoter JeT with values of 5.52, 6.76 and 9.73 in HeLa, MCF-7 and U2-OS respectively (Fig. 6).

[TOP]


Different core promoters result in different expression strength

We cloned the CMV core promoter in front of the JeT proximal promoter to obtain JeT_CMV. We characterized this construct in three different cell lines, HeLa, MCF-7, and U2-OS and found it to have 50% - 60% percent of JeT's activtiy depending on the cell line (Fig. 7). Thereby, the HeLa cell line was measured five times and the MCF-7 and U2-OS cell line measurements were performed four times. The fact that variations in the core promoter can be used to vary expression strength of a certain promoter of interest comes in useful if the transfer function of an existing promoter is to be altered, and it can be used to further diversify the synthetic promoters we created. We characterized these promoters by the same methods and accuracy as CMV and JeT/CMV (see Synthetic Promoter project)

Figure 6: Flow cytometry and microscopy measurement data of CMV (REU) in different cell lines. The three cell lines MCF-7, U2-OS and HeLa were cotransfected with the CMV promoter coupled to GFP and a reference plasmid including the promoter JeT coupled to mCherry. The relative fluorescence (REU) of GFP was measured 20 hours after transfection. All cell lines were measured once for microscopy. The HeLa cell line was measured five times by flow cytometry and MCF-7 and U2-OS were measured four times. In the flow cytometry measurement the standard error of the mean (SEM) is indicated by the error bars.
Figure 7: Flow cytometry measurement data of JeT_CMV (REU) in different cell lines. The three cell lines MCF-7, U2-OS and HeLa were cotransfected with the JeT_CMV promoter coupled to GFP and a reference plasmid including the promoter JeT coupled to mCherry. The relative fluorescence (REU) of GFP was measured 20 hours after transfection. The MCF-7 and the U2-OS cell line were measured four times and the HeLa cell line five times. The standard error of the mean (SEM) is indicated by the error bars.


[TOP]

A stable cell line for promoter measurement

Fig. 8: Results of second exponential PCR. In all lanes are several bands visible, indicating that there is more than one FRT-site integrated in each cell line. Negative controls are: 1. untransfected genomic DNA 2. H2O negative control LAM-PCR, 3. H2O negative control first exponential PCR, 4. H2O negative control second exponential PCR. Electrophoresis was carried out on 2% agarose; 100 bp DNA ladder was used.

Main Article: Stable cell line

We were able to generate HeLa, MCF-7 and U2-OS cells that stably integrated the FRT-site into their genome. Fig. 8 shows the PCR products of the second exponential PCR. For each cell line there are several bands visible. Since the number of bands correlates with the number of unique integration sites [2] there must have been more than one integration of the FRT-vector into the genome of the cells.












[TOP]


Discussion

Flow Cytometry/Fluorescence Microscopy

Our flow cytometer measurements of CMV and JeT-CMV in different cell lines (Fig.3 and 4) show that the strength of these promoters varies in different cell lines. Furthermore, the ratio between the two promoters varies slightly amongst different cell lines, but overall CMV is considerably stronger than JeT, while JeT-CMV is consistently lower than JeT. Our measurements were reproducible considering the strength ratios between our standard promoters. Between different measurements we detected minimal fluctuations (represented by the standard error of the mean). However, as transfection efficiencies also slightly vary between experiments, this is one source of error when comparing experiments. Additionally, due to the nature of the iGEM competition, experiments have been carried out by several persons, introducing another source for errors.
We have applied microscopy measurements to support the data obtained by flow cytometry. The initial microscopy measurements so far confirm the observations made by flow cytometry, although some inconsistencies appeared. This might partially be due to the different cell preparation for flow cytometry and microscopy. While living cells have been used for the flow cytometric measurmentes, cells were fixed prior to analysis by microscopy. So far we could only carry out one set of microscopic measurements, therefore it will be necessary to repeat the microscopic measurements to obtain reliable data.
Apart from supporting the flow cytometry results, fluorescence microscopy will also gain importance in the future as we are working on a stable cell line which simultaneous expresses differently coloured fluorescent proteins. These fluorescent proteins will be directed to various compartments of the cell and thereby, allow to visualize the activation of several promoters at the same time in one single cell. Read more about our future plans in the outlook section.

[TOP]

Real-time RT-PCR

The results of the real-time RT-PCR measurement show that the promoter strength varied significantly over time. Reasons for the variations could be due to the fact, that the activity was measured under transient transfection where the plasmid containing the promoter is not replicated when the cells proliferate. This results in a decrease of amount of plasmids per cell, thus reduced copy of promoter present in the cells. This effect will be increase within each replication. Therefore, long time intervals between measurement and transfection will be more error-prone than shorter intervals. That is why the proportion of error to data is much higher 50 h after transfection compared to the 20 h measurement, although the standard deviations of the two time points are in the same range. It can also be possible that the cells containing the plasmid with promoter construct die faster due to changes in cell physiology. Furthermore, the fluctuation of the results could be from both systematic and random error where all the following reasons could be responsible: pipetting errors during plate preparation, influence of the freeze-thaw process on enzymes, differing contents of mastermix and random error caused by the machines. This could be caused by physiological changes the plasmids and consequently, the promoters put on the cells. Over the course of 20 h the effects those changes evoked were not that distinctiv but over 50 h of interaction enlarged the difference greatly.

[TOP]

Characterization of promoters under different condtions

Considering what we learned about the lack of absolute, non-changing reference standards, characterizing promoters under different conditions becomes inherently difficult. All parameters affecting REU and RPMU are altered by a change in conditions. For the assumptions on which REU and RMPU are based to remain valid, promoters must only be compared in one set of conditions. In most scientific work to this date, transcriptional activity is assessed by comparing protein levels between unrelated conditions (see [12] for a typical example). Although giving sufficient information as to whether a promoter is up- or downregulated, this standard cannot be used for an absolute characterization, as if a condition induces, for example, a system-wide increase in translation, it will result in a ratio between the promoters which is not proportional to the change in transcription. If, on the other hand, measurements are related to a reference promoter at each condition individually, this does not represent true levels of change if the reference promoter itself is up- or downregulated by that condition. Providing absolute measurements to compare between different conditions will be a major challenge for synthetic mammalian biology in the next years. We suggest, for now, always to give promoter strength relative to the strength of JeT under the condition regarded.

[TOP]

Towards PoPs

The concept of RMPU can easily be converted into PoPs, at least for constitutive promoters. RNA degradation equals RNA synthesis in steady state. Therefore, we suggest to block RNA synthesis by applying Actinomycin D which works in minutes [13], and then measuring RNA degradation. According to equation 2, PoPS will then equal mRNA degradation, divided by copy number (which is 1, if the experiment is conducted in a stable cell line). Of course, Actinomycin is toxic to cells and might affect mRNA turnover rates system-wide. Accounting for this effect will present a major challenge.

[TOP]

Concluding remarks

We reported two new units for promoter characterization in mammalian cells, one of which being directly proportional to PoPS. We showed the advantages and limitations of the concept. Future work will have to focus on improving comparisons between different conditions.

[TOP]

Method details

Flow Cytometry

Flow cytometry is a commonly used method for the measurement of fluorescence intensity levels. It uses the principles of light scattering, light excitation, and emission of fluorochrome molecules to generate specific multi-parameter data from particles and cells in the size range of 0.5 µm to 40 µm diameter . One unique feature of flow cytometry is that it measures fluorescence per cell or particle. In contrast to high-resolution microscopy, flow cytometric measurements do not reveal intracellular distribution of the measured signal, but give an integrated signal of each cell. However, flow cytometry allows fast, multi-parametric measurements of high numbers of cells (>1000 cells/sample) in a quantitative manner [14], [19].
A flow cytometer consists of the fluidics system where cells are hydro-dynamically focused in a sheath of fluid; one or several laser which is the source of focused light for fluorescence and scatter; the optics which gather and direct the light emitted from the cells; up to 8 detectors which receive the light and convert it into an electrical signal, which is finally delivered to a computer system to analyze the signals of each cell.
The first detector (Forward Scatter; FSC) is in line with the light beam; its signal is proportional to the size of the cell. The larger the cell, the more light is scattered and the higher the electric signal in the detector. Perpendicular to the light beam are several other detectors, including the Side Scatter (SSC). The Side Scatter represents the granularity of the cell and this can be used to further distinguish different cell populations and exclude apoptotic cells. A two-dimensional scatter plot of Forward versus Side Scatter can give a more detailed overview over the nature of the cells in the sample. This plot is used to separate dead cells and cell debris from healthy cells. [14], [19]
Following the Side Scatter, the light is directed through a series of mirrors and filters, so that particular wavelengths are delivered to the correct detector. The fluorescence signal is then converted to a voltage signal and can be visualized as a histogram, showing frequencies of cells with different fluorescence levels (usually distributed over 1024 channels) [14], [19].
We have used a Beckman Coulter FC500 MPL, equipped with a 488 nm and 561 nm laser, thereby allowing simultaneous and quantitative measurement of GFP and mCherry fluorescence.
Cells were prepared in 96-well format with 104 cells/well and transfected with the promoter of interest and when necessary, induction drug was added for inducible promoters. Before measurement, the medium was removed, the cells were washed with 1xPBS and trypsinized with 60 µl of trypsin per well. After 10 minutes incubation at 37°C, 1xPBS + 1% BSA were added up to a volume of 200 μl per well. When starting the flow cytometry measurement we first adjusted the gates and the background signal to the negative control (Fig. 9). Fig. 10 shows an example of a gated positive control (JeT), where we can see a second normal distribution peak indicating the gated GFP positive cells.

Figure 9 : Example of a negative control in Hela cells by flow cytometry. The number of events is plotted against the fluorescence (log) of GFP. Background signal is set under 10 and the area under the curve is colored in green.
Figure 10 : Example of a positive control (JeT) in Hela cells by flow cytometry. The number of events is plotted against the fluorescence (log) of GFP. The area under the curve is colored in green.

[TOP]

Microscopy and Image Analysis

For further characterization of our promoters we decided to use fluorescence microscopy. Our cells are co-transfected (1:2) with a reference plasmid, which contains JeT in front of mCherry. Analysis of reporter gene expression can be restricted to cells that express the reference plasmid and are therefore likely to have been successfully co-transfected with the reporter plasmid of interest. This is particularly useful when transfection efficiency is low. We applied this approach to microscopy measurement to minimize the experimental error. The promoter strength was evaluated in relative expression units (REU) relative to the JeT promoter which was set to 1. Microscopy images of our samples fixed with 4% formaldehyde were taken using the Nikon Eclipse 90i upright automated widefield microscope in the Nikon Imaging Center at the University of Heidelberg. Each image was taken in the GFP and mCherry channel and the exposure time had to be adjusted for over- or underexposed images. The conversion factor was experimentally determined by imaging a fluorescent plate with exposure time varying from 10 to 70 ms (the estimated range of our promoters) and plotting the mean grey values of these images versus time. A linear relationship between the exposure times and grey values was observed for this time frame and the conversion factor was determined to be 0.998. For image analysis we used ImageJ (Image Processing and Analysis in Java) software tools for obtaining the mean grey values of each cell containing the promoter of interest. ImageJ is an open source software developed at the National Institutes of Health (NIH (http://rsbweb.nih.gov/ij/). The measurement is performed by setting the boundaries around the cells using the threshold in the mCherry channel image and generating a mask that is then applied to the image of the GFP channel. For a step by step description on ImageJ analysis see: Materials and Methods.

[TOP]

Real-time RT-PCR

Real-time RT-PCR, which has been developed tremendously during the last 20 years, is a specific and sensitive technique for detecting and quantification of gene expression on RNA level. In order to obtain reliable results and reduce the variance during the extraction, reverse transcription and PCR, we did the measurement for each promoter with 12 replicates from an identical sample. Prior to PCR, we extracted the total RNA using Qiagen RNeasy Plant Mini Kit following the protocol from the handbook [15]. The extraction was performed by the QIAcubeTM [16], the automated spin-column kits preparation robot, to avoid possible operator error. In order to perform the real-time PCR, primers and probes were designed according to QuantiFast Probe RT-PCR Handbook [17]: The target PCR product length is set between 70-200 bp. The target spans over exon-exon boundary to exclude amplification of genome DNA which may be presented in the RNA extraction as contamination. The probes have reporter fluorophore and quencher fluorophore at 5' and 3' end respectively. During amplification, probes bind specifically to the target gene. The polymerase hydrolyzes the probe during elongation due to its exonuclease activity. The signal of the released fluorophore can then be detected and correlates therefore to the amount of PCR product. The real-time RT-PCR was performed on StepOnePlusTM Real-time PCR System from Applied Biosystems [18]. We quantified the expression of GFP, the reporter gene, together with other 5 housekeeping genes (ß-actin, glycerol-aldehyde-3-phosphate de-hydrogenase, glucose-6-phosphate dehydrogenase, gamma-tubulin, 18S rRNA) for normalization. At the end of each cycle, the fluorophore of each well is read and recorded. Ct (threshold cycle) is defined as the cycle number at which the fluorescent strength (the curve) crosses a certain threshold (Fig. 11). The more amount of starting mRNA, the sooner will the machine detect the fluorescent signals from the PCR reaction, which means a smaller Ct under the same threshold. The threshold should be a careful decision. With a too small threshold, the signal may be too weak for reliable detection. If it is too big, the reaction might be restricted by the enzymes and other materials. Usually, we set the threshold at 0.05 for all plates (Fig. 12.1 and Fig. 12.2). Calculating the difference between Ct values, with normalization of the multiple housekeeping genes, we get the ratio of the activity of the promoter of interest and our reference.

Figure 11 : Ct values of 18s rRNA with CMV promoter 20h after transfection Threshold is set at 0.05, the cycle number at when the amplification plot crosses the threshold is the Ct value.
Figure 12.1 : Ct values of eGFP with CMV promoter 20h after transfection Threshold is set at 0.05. 12 dots represent 12 RNA extractions from identical sample.
Figure 12.2 : Ct values of eGFP with JeT promoter 20h after transfection Threshold is set at 0.05. 12 dots represent 12 RNA extractions from identical sample.

[TOP]

References

[1] Kelly J. R., Rubin A. J., Davis J. H., Ajo-Franklin C. M., Cumbers J., Czar M. J., de Mora K., Glieberman A. L., Monie D. D. & Endy D. Measuring the activity of BioBrick promoters using an in vivo reference standard. Journal of Biological Engineering 3 (2009).

[2] Endy D. & Deese I. Adventures in synthetic biology (comic) Nature 438: 449-453 (2005). Available online at nature.com

[3] Alberts B., Johnson A., Walter P. & Lewis J. Molecular Biology of the Cell. 5th edition, 2008. Garland Science) Chapter 6

[4] http://www.promega.com/tbs/tm058/tm058.html

[5] Alberts B., Johnson A., Walter P. & Lewis J. Molecular Biology of the Cell. 5th edition, 2008. Garland Science) Chapter 7, p. 467-476

[6] Zhu X.D. & Sadowski P. D. Cleavage-dependent Ligation by the FLP Recombinase. J Biol Chem 270: 23044-23054 (1995).

[7] York J. D., Odom A. R., Murphy R., Ives E. B. & Wente S. R. A phospholipase C-dependent inositol polyphosphate kinase pathway required for efficient messenger RNA export. Science 285: 96-100 (1999).

[8] Alberts B., Johnson A., Walter P. & Lewis J. Molecular Biology of the Cell. 5th edition, 2008. Garland Science) Chapter 7, p. 905-908

[9] Tornoe J. Generation of a synthetic mammalian promoter library by modification of sequences spacing transcription factor binding sites. Gene 297: 21-32 (2002)

[10] Alberts B., Johnson A., Walter P. & Lewis J. Molecular Biology of the Cell. 5th edition, 2008. Garland Science) Chapter 7, p. 492

[11] Ducrest A. L., Amacker M., Lingner J. & Nabholz M. Detection of promoter activity by flow cytometric analysis of GFP reporter expression. Nucleic Acids Res. 30: e65 (2002).

[12] Rushton P. J., Reinstädler A., Lipka V., Lippok B. & Somssich I. E. Synthetic plant promoters containing defined regulatory elements provide novel insights into pathogen- and wound-induced signalling. Plant Cell 14: 749-762 (2002).

[13] Degenhardt T., Rybakova K. N., Tomaszewska A., Moné M. J., Westerhoff H. V., Bruggeman F. J. & Carlberg C. Population-Level Transcription Cycles Derive from Stochastic Timing of Single-Cell Transcription. Cell 138: 489-501 (2009).

[14] Lottspeich F. & Engels J. W.: Bioanalytik (Book. 2nd edition, 2006. Spektrum Akademischer Verlag) Chapter 5, p. 95-96.

[15] http://www1.qiagen.com/Products/RnaStabilizationPurification/RNeasySystem/RNeasyPlantMini.aspx

[16] http://www1.qiagen.com/products/automation/qiacube.aspx

[17] http://www1.qiagen.com/literature/render.aspx?id=398

[18] https://products.appliedbiosystems.com/ab/en/US/adirect/ab?cmd=catNavigate2&catID=604109&tab
=DetailInfo<

[19] Ormerod M.G.: Flow cytometry: a practical approach (Book. 3rd edition, 2000. Oxford University Press) Chapter 1, p. 7-21.

[20] Zhou H. Development of site-specific integration system to high-level expression recombinant proteins in CHO cells. Chinese journal of biotechnology 23: 756-62 (2007).

[TOP]