Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
NUCLEOTIDE SEQUENCES, NUCLEIC ACID SENSORS AND METHODS THEREOF
Document Type and Number:
WIPO Patent Application WO/2015/033237
Kind Code:
A1
Abstract:
The present disclosure relates to particular nucleotide sequences comprising of at least one stem and/or loop structure and which optionally bind fluorescent labels, for example DFHBI and a process for obtaining said sequences. The present disclosure also provides nucleic acid sensor for cAMP comprising reporter domain, communication module and target recognition domain. Further, the present disclosure provides for ratiometric sensors 10 for quantifying cAMP using the nucleic sensors of the instant invention. The instant disclosure further provides method for obtaining the sensors, method for detecting and measuring small molecules, such as cAMP using the sensors of the instant disclosure and kits thereof.

Inventors:
KRISHNAN YAMUNA (IN)
SHARMA SURUCHI (IN)
Application Number:
PCT/IB2014/060734
Publication Date:
March 12, 2015
Filing Date:
April 15, 2014
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
NAT CT FOR BIOLOG SCIENCES NCBS TIFR (IN)
International Classes:
C07H21/04; C07H21/00
Domestic Patent References:
WO2013016694A22013-01-31
Other References:
KOIZUMI ET AL.: "Molecular recognition of cAMP by an RNA aptamer.", BIOCHEMISTRY, vol. 39, no. 30, 1 August 2000 (2000-08-01), pages 8983 - 8992
Attorney, Agent or Firm:
RAJAGOPAL, Lakshmi et al. (4121/B 6th Cross, 19A Main, HAL II Stage, Bangalore 8, IN)
Download PDF:
Claims:
We Claim:

1. A nucleotide sequence comprising sequence set forth as SEQ ID No. 1, wherein N is base selected from group comprising A, G, U, C and modified base or any combinations thereof and number of bases vary from about 4 to about 9 bases; and the nucleotide sequence optionally binds to fluorescent dye.

2. A method of obtaining nucleotide sequence as claimed in claim 1, said method comprising acts of:

(c) creating a nick in stem-loop 2 region of SEQ ID No. 24 to obtain stem-2 region with free 5' end and optionally varying nucleotide sequence of the stem-2 region to obtain the nucleotide sequence set forth as SEQ ID No. 1; and

(d) optionally adding the fluorescent dye to the nucleotide sequence of step (a).

3. The nucleotide sequence as claimed in claim 1 and the method of obtaining nucleotide sequence as claimed in claim 2, wherein the modified base is selected from group comprising m5G, m7G, inosine and xanthosine or any combinations thereof.

4. The nucleotide sequence as claimed in claim 1 and the method of obtaining nucleotide sequence as claimed in claim 2, wherein said sequence comprises atleast one stem and/or loop structure and said nucleotide sequence generates an optical signal on optionally binding with the fluorescent dye, preferably 3, 5-difluoro-4- hydroxybenzylidene imidazolinone (DFHBI).

5. The nucleotide sequence as claimed in claim 1 and the method of obtaining nucleotide sequence as claimed in claim 2, wherein the nucleotide sequence has three stem loop structures and a stem structure with free 5' end.

6. The nucleotide sequence as claimed in claim 1 and the method of obtaining nucleotide sequence as claimed in claim 2, wherein said nucleotide sequence is selected from group comprising SEQ ID No. 3, SEQ ID No. 4, SEQ ID No. 5, SEQ ID No. 6, SEQ ID No. 7 or SEQ ID No. 8.

7. A nucleic acid sensor for detecting target molecule, said sensor comprising:

a. target recognition domain; and b. reporter domain including communication module; comprises of nucleotide sequence as claimed in claim 1; and optionally hybridizes to fluorescently labelled DNA.

8. A method of obtaining nucleic acid sensor as claimed in claim 7, said method comprising acts of :

a. creating nick in stem-loop 2 region of SEQ ID No. 24 to obtain stem-2 region of reporter domain with free 5' end; and optionally varying nucleotide sequence of the stem-2 region, wherein the stem-2 region is communication module;

b. optionally extending, or splitting and extending the reporter domain;

c. obtaining target recognition domain; and

d. joining the target recognition domain to the free 5' end of the communication module to obtain the nucleic acid sensor.

9. The nucleic acid sensor as claimed in claim 7 and the method of obtaining nucleic acid sensor as claimed in claim 8, wherein the target recognition domain has sequence set forth in SEQ ID No. 23 and binds to target molecule selected from group comprising small molecules, preferably cAMP.

10. The nucleic acid sensor as claimed in claim 7 and the method of obtaining nucleic acid sensor as claimed in claim 8, wherein the communication module is variable part of the reporter domain and is a linker between the recognition domain and the reporter domain; and sequence of the sensor is DNA, RNA or a combination thereof.

11. The nucleic acid sensor as claimed in claim 7 and the method of obtaining nucleic acid sensor as claimed in claim 8, wherein said sensor has nucleotide sequence set forth as SEQ ID No. 2; wherein N is base selected from group comprising A, G, U, C and modified base or any combinations thereof, and number of bases vary from about 4 to about 9 bases.

12. The nucleic acid sensor as claimed in claim 11 and the method of obtaining nucleic acid sensor as claimed in claim 8, wherein said sensor has nucleotide sequence selected from group comprising SEQ ID No. 9, SEQ ID No. 16, SEQ ID No. 22 and SEQ ID No. 15.

13. The nucleic acid sensor as claimed in claim 7, wherein 3' end of the reporter domain of the sensor is extended and thereafter hybridized to the fluorescently labelled complementary DNA.

14. The nucleic acid sensor as claimed in claim 7, wherein the reporter domain is split into two halves-Split A and Split B, 3' end of Split A and 5 'end of Split B are extended and the extended Split B is hybridized to the fluorescently labelled complementary DNA.

15. The nucleic acid sensor as claimed in claim 13, wherein said sensor has sequence set forth in SEQ ID No. 18; and the labelled complementary DNA has sequence set forth in SEQ ID No. 19.

16. The nucleic acid sensor as claimed in claim 14, wherein said Split A has sequence set forth in SEQ ID No. 20 and said Split B has sequence set forth in SEQ ID No. 21; and the labelled complementary DNA has sequence set forth in SEQ ID NO. 19.

17. The method of obtaining nucleic acid sensor as claimed in claim 8, wherein 3' end of the reporter domain is extended from about 15 to about 40 nucleotides, selected from group comprising A, C, U, T and G, or any combinations thereof to obtain extended reporter domain and hybridizing the extended reporter domain with the fluorescently labelled complementary DNA sequence.

18. The method of obtaining nucleic acid sensor as claimed in claim 8, wherein the reporter domain is split into two halves - Split A and Split B; 3' end of the split A and 5' end of the split B are extended from about 15 to about 45 nucleotides selected from group comprising A, C, T, U, and G, or any combinations thereof and the extended Split B is hybridized with fluorescently labelled complementary DNA.

19. The nucleic acid sensor as claimed in claims 7, 13 and 14 and the method of obtaining nucleic acid sensor as claimed in claims 8, 17 and 18, wherein the fluorescent label is selected from group comprising Alexa dye, bodipy dye, Cy3, Cy5, FITC and TAMRA, preferably Alexa dye.

20. A method of identifying and optionally quantifying target molecule in a sample, said method comprising acts of:

a. contacting the sample with fluorescent dye, nucleic acid sensor as claimed in claim 7, and optionally along with fluorescent label;

b. identifying the target molecule by determining the optical signal generated; and c. optionally quantifying the target molecule by determining ratio of optical signal generated by fluorescent dye to fluorescent label.

21. The method as claimed in claim 20, wherein the optical signal of step b) is generated by fluorescent dye bound to reporter domain of the sensor; and the target molecule is selected from group comprising small molecules, preferably cAMP.

22. The method as claimed in claim 20, wherein the fluorescent dye is DFHBI; the fluorescent label in step (c) is attached to complementary DNA hybridized to reporter domain of the sensor; and the fluorescent label is selected from group comprising Alexa dye, bodipy dye, Cy3, Cy5, FITC and TAMRA, preferably Alexa dye.

23. The method as claimed in claim 20, wherein the sample is biological sample selected or derived from group comprising cells, cell extracts, cell lysates, tissues, tissue extracts, bodily fluids, serum, blood and blood product, cAMP secreted in growth media.

24. A kit for obtaining nucleotide sequence set forth as SEQ ID No. 1 or for obtaining nucleic acid sensor or for identifying and optionally quantifying target molecule in a sample, said kit comprising components selected from group of nucleotide sequence as claimed in claim 1 or reporter domain, sequence set forth as SEQ ID NO. 24, nucleic acid sensor as claimed in claim 7, target recognition domain set forth as SEQ ID No. 23, fluorescently labelled DNA set forth as SEQ ID No. 19, free bases, fluorescent dye, fluorescent label, buffers, and instruction manual or any combinations thereof.

25. A method of assembling kit as claimed in claim 24, said method comprising acts of combining components selected from group of nucleotide sequence as claimed in claim 1 or reporter domain, sequence set forth as SEQ ID NO. 24, nucleic acid sensor as claimed in claim 7, target recognition domain set forth as SEQ ID No. 23, fluorescently labelled DNA set forth as SEQ ID No. 19, free bases, fluorescent dye, fluorescent label, buffers, and instruction manual or any combinations thereof.

Description:
"NUCLEOTIDE SEQUENCES, NUCLEIC ACID SENSORS AND METHODS

THEREOF"

TECHNICAL FIELD

The instant disclosure relates to nucleotide sequences which comprise of atleast one stem and/or loop structure and which optionally bind fluorescent labels, for example, 3, 5-difluoro-4-hydroxybenzylidene imidazolinone (DFHBI) and methods thereof. The instant disclosure also relates to the field of sensors. More particularly, the instant disclosure relates to a nucleic acid sensor for detecting and/or quantifying cyclic 3'-5' adenosine monophosphate (cAMP) and methods thereof. The instant disclosure further relates to a kit comprising the nucleotide sequence and nucleic acid sensor.

BACKGROUND OF THE DISCLOSURE

Genetically encoded sensors are very useful for imaging small molecules in living cells in order to detect them. The modular and programmable nature of nucleic acids, coupled with their capacity for molecular recognition, positions nucleic acids as a uniquely versatile scaffold for molecular sensing within the cellular milieu.

Cyclic adenosine monophosphate (3'-5'-cyclic adenosine monophosphate, cAMP) is a secondary messenger and is important in many biological processes. Thus, it becomes very important to detect and quantify cAMP in biological systems. Presently radioimmunoassays are being used for measuring cAMP in in vitro studies which are variable, costly and highly time consuming and the existing sensors for live cell imaging of cAMP are based on proteins which are natural intracellular targets of cAMP and hence these sensors may interfere with cellular physiology.

Modular RNA aptamer sensors generally known in the art have three domains: target recognition domain, communication module and reporter domain. Reporter domain comprises molecules that bind a fluorescent label, thus emitting a fluorescent signal. Target recognition domain binds the target to be detected and communication module transfers the information from target recognition domain to reporter domain such that whenever recognition domain binds to target, the reporter domain will give fluorescence signal. Spinach is an RNA aptamer that binds the chromophore DFHBI. DFHBI, in general, is a chromophore with low fluorescence yields, i.e., it is very less fluorescent. However, when this chromophore is bound to Spinach, the fluorescence quantum yield increases by a factor of 80. This aptamer has a stem-loop called stem-loop-2, whose structure is important in order for binding to DFHBI but its sequence is not important. Stem loop-2 has been used as communication module for designing sensors for target analytes whose aptamers are known by attaching aptamer for the target analyte (which is target recognition domain) at stem loop-2.

However, RNA aptamers for various analytes have 5' ends that need to be free for their binding action e.g. cAMP aptamer. Spinach cannot be used as reporter domain for making sensors for such analytes as attaching aptamer to loop of stem-loop-2 will make analyte aptamer lose its 5' end. So, there is no Spinach based sensor for cAMP. Further, the available Spinach based aptameric sensors for cdiGMP, Adenosine, ADP SAM etc. are not ratiometric.

Therefore, in order to make Spinach aptamer amenable for such analytes, it is required to carry out a rearrangement in Spinach so as to create a free 5' end near stem-2. However, it is observed in the art that rearranging of nucleic acid molecules can affect their activity.

STATEMENT OF THE DISCLOSURE

Accordingly, the present disclosure relates to a nucleotide sequence comprising sequence set forth as SEQ ID No. 1, wherein N is base selected from group comprising A, G, U, C and modified base or any combinations thereof and number of bases vary from about 4 to about 9 bases, and the nucleotide sequence optionally binds to fluorescent dye; a method of obtaining nucleotide sequence as above, said method comprising acts of: (a) creating a nick in stem-loop 2 region of SEQ ID No. 24 to obtain stem-2 region with free 5' end and optionally varying nucleotide sequence of the stem-2 region to obtain the nucleotide sequence set forth as SEQ ID No. 1, and (b) optionally adding the fluorescent dye to the nucleotide sequence of step (a); a nucleic acid sensor for detecting target molecule, said sensor comprising (a) target recognition domain, and (b) reporter domain including communication module, comprises of nucleotide sequence as above and optionally hybridizes to fluorescently labelled DNA; a method of obtaining nucleic acid sensor as above, said method comprising acts of : (a) creating nick in stem-loop 2 region of SEQ ID No. 24 to obtain stem-2 region of reporter domain with free 5' end, and optionally varying nucleotide sequence of the stem-2 region, wherein the stem-2 region is communication module, (b) optionally extending, or splitting and extending the reporter domain, (c) obtaining target recognition domain, and (d) joining the target recognition domain to the free 5' end of the communication module to obtain the nucleic acid sensor; a method of identifying and optionally quantifying target molecule in a sample, said method comprising acts of: (a) contacting the sample with fluorescent dye, nucleic acid sensor as above, and optionally along with fluorescent label, (b) identifying the target molecule by determining the optical signal generated, and (c) optionally quantifying the target molecule by determining ratio of optical signal generated by fluorescent dye to fluorescent label; a kit for obtaining nucleotide sequence set forth as SEQ ID No. 1 or for obtaining nucleic acid sensor or for identifying and optionally quantifying target molecule in a sample, said kit comprising components selected from group of nucleotide sequence as above or reporter domain, sequence set forth as SEQ ID NO. 24, nucleic acid sensor as above, target recognition domain set forth as SEQ ID No. 23, fluorescently labelled DNA set forth as SEQ ID No. 19, free bases, fluorescent dye, fluorescent label, buffers, and instruction manual or any combinations thereof; and a method of assembling kit as above, said method comprising acts of combining components selected from group of nucleotide sequence as above or reporter domain, sequence set forth as SEQ ID NO. 24, nucleic acid sensor as above, target recognition domain set forth as SEQ ID No. 23, fluorescently labelled DNA set forth as SEQ ID No. 19, free bases, fluorescent dye, fluorescent label, buffers, and instruction manual or any combinations thereof.

BRIEF DESCRIPTION OF THE ACCOMPANYING FIGURES

In order that the disclosure may be readily understood and put into practical effect, reference will now be made to exemplary embodiments as illustrated with reference to the accompanying figures. The figures together with a detailed description below, are incorporated in and form part of the specification, and serve to further illustrate the embodiments and explain various principles and advantages, in accordance with the present disclosure where:

Figure 1 depicts the basic design of the sensor of the instant disclosure, wherein (I) represents Spinach, (II) represents Class II cAMP RNA Aptamer, (III) represents nucleotide sequence of the instant disclosure and (IV) represents nucleic acid sensor of the instant disclosure.

Figure 2 depicts a structural representation of Spinach (2A) and the nucleotide sequence of the instant disclosure (2B).

Figure 3- 'A' depicts a graph showing increase in fluorescence of DFHBI upon binding to the nucleotide sequences of the instant invention and 'B' is a bar graph showing the fold change in intensity of DFHBI upon binding to the nucleotide sequences of the instant disclosure.

Figure 4 (a) depicts the process of obtaining RNA sequence of the nucleic acid sensor. Figures 4(b)-4(e) depicts the response of sensors (caSpn) of the instant invention to cAMP, wherein in (c), red line CaSp4, black line is CaSp7m and blue line is CaSp5b and in (e), black represents sensor CaSp7NE in the absence of cAMP and red represents CaSp7NE in presence of 2mM cAMP.

Figure 5 depicts a schematic diagram showing the mechanism of sensing of the nucleic acid sensors of the instant disclosure. Figure 6- (a) depicts Strategy- 1 for designing the ratiometric nucleic acid sensors of the instant disclosure (caSpn A ) and (b) depicts Strategy-2 for designing the ratiometric nucleic acid sensors (cAMPhor) of the instant disclosure.

Figure 7- (a) depicts the detection of cAMP using the ratiometric sensors obtained by Strategy I of the instant disclosure, wherein Red line is CaSp7m A +2mM cAMP, Black line is CaSp7m A + no cAMP, Green line is CaSp4 A +2mM cAMP and Blue line is CaSp4 A + no cAMP; and (b) depicts graph showing the cAMP response of the sensors obtained by Strategy I (black line) and Strategy II (Red line). Figure 8 depicts a graph showing the cAMP sensitivity of the ratiometric sensor of the instant disclosure under different conditions wherein 'a' represents cAMP sensitivity curve at 25°C in about 5 minutes, 'b' represents cAMP sensitivity curve at 25° C in presence of ImM Mn 2+ in about 5 minutes, 'c' represents cAMP sensitivity curve at 5° C in about 2 hours, 'd' represents cAMP sensitivity curve at 25° C (black), at 25° C in presence of ImM Mn 2+ (red) and at 5° C (blue) for comparison, 'e' represents cAMP sensitivity curve at 25° C in about 2 hours, and 'f represents cAMP sensitivity curve in presence of 5mMATPMg at 25° C in about 2 hours. Figure 9 depicts a bar graph showing the comparison of secreted cAMP measured using a radioimmunoassay and ratiometric sensor of the instant disclosure, wherein the values plotted are the mean±SEM of five different biological replicates.

Figure 10 depicts a bar graph showing the specificity of the ratiometric sensors of the instant disclosure wherein Normalized G/R values of cAMPhor is shown in red and of CaSp7m A is shown in black.

Figure 11 depicts imaging of cAMP inside membrane bound compartments; (a) shows the bright field image (BF), images in Alexa 647 channel (R) and in DFHBI (G) channel of a given GUV containing 1 μΜ cAMPhor and indicated cAMP concentration as well as pseudocolored R/G images showing the ratio of R/G intensities of the above images. Scale bar: 5 μτη. (b) depicts a histogram showing spread of normalized G/R values of a collection of GUVs at the indicated cAMP concentrations (n=at least 25). Red dotted line indicates the mean of each histogram; (c) shows the comparison of normalized G/R values obtained at various cAMP concentrations in solution and inside GUVs. The normalized G/R values reported for GUV is mean±s.e.m. of mean normalized G/R of at least three independent GUV preparations.

DETAILED DESCRIPTION OF THE DISCLOSURE

The present disclosure relates to a nucleotide sequence comprising sequence set forth as SEQ ID No. 1, wherein N is base selected from group comprising A, G, U, C and modified base or any combinations thereof and number of bases vary from about 4 to about 9 bases; and the nucleotide sequence optionally binds to fluorescent dye.

The present disclosure also relates to a method of obtaining nucleotide sequence as above, said method comprising acts of:

(a) creating a nick in stem-loop 2 region of SEQ ID No. 24 to obtain stem-2 region with free 5' end and optionally varying nucleotide sequence of the stem-2 region to obtain the nucleotide sequence set forth as SEQ ID No. 1; and

(b) optionally adding the fluorescent dye to the nucleotide sequence of step (a).

In an embodiment of the present disclosure, the modified base is selected from group comprising m 5 G, m 7 G, inosine and xanthosine or any combinations thereof.

In another embodiment of the present disclosure, said sequence comprises atleast one stem and/or loop structure and said nucleotide sequence generates an optical signal on optionally binding with the fluorescent dye, preferably 3, 5-difluoro-4- hydroxybenzylidene imidazolinone (DFHBI).

In yet another embodiment of the present disclosure, the nucleotide sequence has three stem loop structures and a stem structure with free 5' end.

In still another embodiment of the present disclosure, said nucleotide sequence is selected from group comprising SEQ ID No. 3, SEQ ID No. 4, SEQ ID No. 5, SEQ ID No. 6, SEQ ID No. 7 or SEQ ID No. 8.

The present disclosure further relates to a nucleic acid sensor for detecting target molecule, said sensor comprising:

(a) target recognition domain; and

(b) reporter domain including communication module; comprises of nucleotide sequence as above; and optionally hybridizes to fluorescently labelled DNA. The present disclosure further relates to a method of obtaining nucleic acid sensor as above, said method comprising acts of:

(a) creating nick in stem-loop 2 region of SEQ ID No. 24 to obtain stem-2 region of reporter domain with free 5' end; and optionally varying nucleotide sequence of the stem-2 region, wherein the stem-2 region is communication module;

(b) optionally extending, or splitting and extending the reporter domain;

(c) obtaining target recognition domain; and

(d) joining the target recognition domain to the free 5' end of the communication module to obtain the nucleic acid sensor.

In an embodiment of the present disclosure, the target recognition domain has sequence set forth in SEQ ID No. 23 and binds to target molecule selected from group comprising small molecules, preferably cAMP.

In another embodiment of the present disclosure, the communication module is variable part of the reporter domain and is a linker between the recognition domain and the reporter domain; and sequence of the sensor is DNA, RNA or a combination thereof.

In yet another embodiment of the present disclosure, said sensor has nucleotide sequence set forth as SEQ ID No. 2; wherein N is base selected from group comprising A, G, U, C and modified base or any combinations thereof, and number of bases vary from about 4 to about 9 bases.

In yet another embodiment of the present disclosure, said sensor has nucleotide sequence selected from group comprising SEQ ID No. 9, SEQ ID No. 16, SEQ ID No. 22 and SEQ ID No. 15.

In yet another embodiment of the present disclosure, 3' end of the reporter domain of the sensor is extended and thereafter hybridized to the fluorescently labelled complementary DNA. In yet another embodiment of the present disclosure, the reporter domain is split into two halves-Split A and Split B, 3' end of Split A and 5 'end of Split B are extended and the extended Split B is hybridized to the fluorescently labelled complementary DNA.

In yet another embodiment of the present disclosure, said sensor has sequence set forth in SEQ ID No. 18; and the labelled complementary DNA has sequence set forth in SEQ ID No. 19.

In yet another embodiment of the present disclosure, said Split A has sequence set forth in SEQ ID No. 20 and said Split B has sequence set forth in SEQ ID No. 21; and the labelled complementary DNA has sequence set forth in SEQ ID NO. 19. In yet another embodiment of the present disclosure, 3' end of the reporter domain is extended from about 15 to about 40 nucleotides, selected from group comprising A, C, U, T and G, or any combinations thereof to obtain extended reporter domain and hybridizing the extended reporter domain with the fluorescently labelled complementary DNA sequence.

In yet another embodiment of the present disclosure, the reporter domain is split into two halves - Split A and Split B; 3' end of the split A and 5' end of the split B are extended from about 15 to about 45 nucleotides selected from group comprising A, C, T, U, and G, or any combinations thereof and the extended Split B is hybridized with fluorescently labelled complementary DNA.

In still another embodiment of the present disclosure, the fluorescent label is selected from group comprising Alexa dye, bodipy dye, Cy3, Cy5, FITC and TAMRA, preferably Alexa dye.

The present disclosure further relates to a method of identifying and optionally quantifying target molecule in a sample, said method comprising acts of: (a) contacting the sample with fluorescent dye, nucleic acid sensor as above, and optionally along with fluorescent label;

(b) identifying the target molecule by determining the optical signal generated; and

(c) optionally quantifying the target molecule by determining ratio of optical signal generated by fluorescent dye to fluorescent label.

In an embodiment of the present disclosure, the optical signal of step b) is generated by fluorescent dye bound to reporter domain of the sensor; and the target molecule is selected from group comprising small molecules, preferably cAMP.

In another embodiment of the present disclosure, the fluorescent dye is DFHBI; the fluorescent label in step (c) is attached to complementary DNA hybridized to reporter domain of the sensor; and the fluorescent label is selected from group comprising Alexa dye, bodipy dye, Cy3, Cy5, FITC and TAMRA, preferably Alexa dye.

In yet another embodiment of the present disclosure, the sample is biological sample selected or derived from group comprising cells, cell extracts, cell lysates, tissues, tissue extracts, bodily fluids, serum, blood and blood product, cAMP secreted in growth media.

The present disclosure further relates to a kit for obtaining nucleotide sequence set forth as SEQ ID No. 1 or for obtaining nucleic acid sensor or for identifying and optionally quantifying target molecule in a sample, said kit comprising components selected from group of nucleotide sequence as above or reporter domain, sequence set forth as SEQ ID NO. 24, nucleic acid sensor as above, target recognition domain set forth as SEQ ID No. 23, fluorescently labelled DNA set forth as SEQ ID No. 19, free bases, fluorescent dye, fluorescent label, buffers, and instruction manual or any combinations thereof.

The present disclosure further relates to a method of assembling kit as above, said method comprising acts of combining components selected from group of nucleotide sequence as above or reporter domain, sequence set forth as SEQ ID NO. 24, nucleic acid sensor as above, target recognition domain set forth as SEQ ID No. 23, fluorescently labelled DNA set forth as SEQ ID No. 19, free bases, fluorescent dye, fluorescent label, buffers, and instruction manual or any combinations thereof.

The present disclosure relates to a nucleotide sequence which comprises of atleast one stem and/or loop structure, optionally capable of binding to fluorescent dyes, such as DFHBI having low fluorescence quantum which upon binding, increases its fluorescence quantum yield. In an embodiment of the present disclosure, the nucleotide sequence comprises three stem-loop structures. The said nucleotide sequences may be used for designing sensors for small molecules whose aptamers are sensitive to manipulations at 5' ends, wherein the nucleotide sequences function as the reporter domain by binding fluorescent dye and emitting fluorescence.

In another embodiment, the present invention relates to nucleic acid sensors for detecting and quantifying small molecules, such as cAMP. The nucleic acid sensors comprise a target recognition domain which binds small molecules such as cAMP, a reporter domain for binding fluorescent label and a communication module which is part of the reporter domain whose sequence can be varied provided it forms a duplex. In an embodiment, the reporter domain of the nucleic acid sensor is the nucleotide sequence which comprises of atleast one stem and/or loop structure and binds to fluorescent dye, such as DFHBI. The target recognition domain is the RNA aptamer for cAMP, which is known in art (SEQ ID No.23) and the communication module is the stem region of the reporter domain with free 5' end; present within the reporter domain comprising the nucleotide sequence which is designed in such a way that it allows fluorescent dye such as DFHBI to bind the reporter domain only in the presence of cAMP. As a result, there is low fluorescence in absence and high fluorescence in the presence of cAMP.

In an embodiment of the present disclosure, nucleotide sequence with atleast one stem and/or loop structure for binding to fluorescent dyes such as DFHBI are provided. This nucleotide sequence is modified form of the Spinach aptamer, particularly the nucleotide sequences are circular permuted versions of Spinach wherein a nick is created in loop-2 of Spinach (SEQ ID No. 24) to create a free 5 'end and connecting the 5' and 3' end of the sequence by a 'UUCG' loop in order that it remains single stranded. Figure 2(A) of the instant disclosure depicts the structure of Spinach while Figure 2(B) depicts the structure of one of the nucleotide sequence of the instant invention which clearly shows the nick created in stem-loop 2 thereby creating a free 5' end in the sequence. The nucleotide sequence of the instant invention may be used to design sensors for molecules whose aptamers require free 5 'end for binding to the molecules.

In an embodiment of the instant invention, on testing for the ability to enhance the fluorescence of DFUBI, it is found that molecules with shorter stem sequences are less effective in increasing the fluorescence of DFUBI, as compared to longer stem sequences thereby showing that shorter sequences which are less stable might result in loss of activity. Therefore, stability of Stem-2 is very crucial for the activity of the nucleotide sequence of the instant invention and designing of various such sequences is one of the aspects of the instant disclosure.

The present disclosure also relates to nucleic acid sensors for detecting small molecules, such as cAMP. The nucleic acid sensors of the present disclosure are designed by linking/integrating/fusing/joining the 3' end of target recognition domain which is the RNA aptamer for cAMP (SEQ ID No. 23) to the 5' end of the reporter domain i.e., the nucleotide sequence of the instant invention. Figure 1 depicts the designing of the nucleic acid sensor of the instant disclosure, wherein (I) depicts the basic structure of Spinach, (II) depicts Class II cAMP specific aptamer which functions as the target recognition domain, (III) depicts the nucleotide sequence of the instant invention which functions as the reporter domain and binds DFUBI and (IV) depicts the nucleic acid sensor of the instant disclosure wherein the cAMP aptamer is joined to the free 5' end of stem- 2 region of the nucleotide sequence of the instant disclosure.

In another embodiment, many sequences were obtained by joining reporter domain and target recognition domain differing in communication module sequence. The communication module sequences vary in length from about 4 to about 9 base pairs, further mismatches are also created in them creating various communication module sequences. Of these, some of the sequences function as sensors by showing increase in fluorescence intensity of DFHBI only in the presence of cAMP and some do not show the functionality as sensors. Further, it is observed that those sequences that did not function as sensors for cAMP did not fold into proper secondary structures when entered into mfold as a functional sensor should as shown in Figure 5 right conformation. Also these sequences have at least one secondary structure that did not change the structure of the reporter domain at any other place other than at communication module (e.g. figure 5 left conformation). Among those which are able to function as sensors for cAMP, the sensitivity, Signal/Noise and percentage change in signal (%SC) differ depending on the communication module sequences.

The mechanism of detection of cAMP by the nucleic acid sensor of the instant disclosure is depicted in Figure 5 and is explained in detail below.

The nucleic acid sensor of the present disclosure is present in two conformations which exist in equilibrium with each other as shown in Figure 5. As captured in the figure, the conformation on the left cannot bind to the fluorescence dye, such as DFHBI in the absence of cAMP, as half of the communication module, i.e., stem-2 of the reporter domain pairs with the target recognition module and a proper duplex is not formed. Thus, fluorescence exhibited by this molecule is less or negligible. However, in the presence of cAMP, the target recognition domain binds to cAMP, as a result of which a stable duplex of communication module is formed which in turn enables the binding of DFHBI to the reporter domain/nucleotide sequence (right conformation of figure 5). This molecule thus exhibits enhanced fluorescence. In a further embodiment, the present disclosure provides ratiometric nucleic acid sensors that are capable of quantifying cAMP. Two different strategies are employed for designing such sensors as shown in Figure 6.

Figure 6(a) depicts Strategy I, wherein the 3' end of the nucleic acid sensor is extended by about 15-40 nucleotide bases, preferably 38 nucleotide bases, and this extended end is hybridized with a complementary DNA oligonucleotide labelled with Alexa 647. Other dyes may also be used selected from group comprising FITC, TAMRA, Alexa 488, or any flurophore that can be conjugated to DNA. Figure 6(b) depicts Strategy II, wherein the RNA of the nucleic acid sensor is split into two halves: Split A and Split B. About 15-45 nucleotide bases, preferably about 20 nucleotide bases are added to 3' end of Split A and complementary about 15-45 nucleotide bases, preferably about 20 nucleotide bases, are added to 5' end of Split B. The purpose of these extra nucleotide bases is to increase the strength of association of Split A and Split B. Further, 15-45 nucleotide bases, preferably about 38 more nucleotide bases complementary to Alexa 647 labelled DNA oligonucleotide are added to 5' end of Split B so as to facilitate the hybridization of Alexa 647 labelled DNA oligonucleotide to form a new trimeric assembly which can quantify cAMP in samples.

The principle of the functioning of the ratiometric sensors of the present disclosure is provided below:

In the normal sensors for detecting cAMP, the intensity at 501 nm obtained by binding cAMP depends on cAMP concentration as well as concentration of sensor RNA molecule. Therefore, this would not give an accurate estimation of the concentration of cAMP alone. Therefore, the ratiometric sensors of the present invention are designed with the objective of making it independent of RNA molecule concentration. For this purpose, Alexa 647 is attached to the sequence which is insensitive to cAMP concentration and whose intensity will thus be proportional to the sensor concentration alone. Thus, the ratio of fluorescence of DFHB I/fluorescence of Alexa 647 gives an accurate estimation of the concentration of cAMP. Said calculation is provided below:

Fluorescence of DFHBI a [cAMP] [Sensor]

Fluorescence of Alexa 647 a [Sensor]

DFHBI Fluorescence/ Alexa 647 fluorescence a [cAMP]

It is essential to note here that although the examples provide specific sequences for extension of the nucleotide sequences of the ratiometric sensors for hybridizing with Alexa 647 labelled DNA, ratiometric sensors using other sequences of varying lengths for hybridizing with complementary DNA labelled with Alexa 647 also fall within the purview of the instant disclosure provided, the strength of association is strong enough under experimental conditions.

Table 1 below shows some the nucleotide sequences used in the present disclosure. The portions of the sequences in blue relate to the cAMP aptamer, portions of the sequences in red relate to the communication module and the portions of the sequences in green relate to the extended region of the reporter domain. However, it is important to note that other sequences formed by varying stem -2 and/or varying length of the extended region of the ratiometric sensors and/or any other variation in the sequence of the nucleotide sequence and the nucleic acid sensors will also fall within the scope of this invention as long as they retain the required activity.



Name SEQ Sequence (5'-3')*

ID No.

D2 Alexa 647 19 TATATATAGGATCTTGCTGTCTGGTGTGCAGTGTTGAT caSp4SplitA 20 GGAAGAGAUGGCGACUAAAAOGACUUGUGGGCAGGUUGUUGAGUAGA

GUGUGAGCUCCGUAACUAGUCGCGUCCGGCGUACCGUACCGUACCCU

G

caSp4SplitB 21 ·-· CAGGGUACGG

UACGGUACGCCGGACGCGACUGAAUGAAAUGGUGAAGGACGGGUCCA GCUG

caSp6b 22

GAGUGUGAGCUCCGUAACUAGUCGCGUC

UUCGGACGCGACUGAAUGAAAUGGUGAAGGACGGGUCCAGAGCUG

cAMP aptamer 23 GGAAGAGAUGGCGACUAAAACGACUUGUCGC

Spinach 24 GACGCGACUGAAUGAAAUGGUGAAGGACGGGUCCAGCUGCUUCGGCA

GCUUGUUGAGUAGAGUGUGAGCUCCGUAACUAGUCGCGUC

caSp4DNA-1 25 TAATACGACTCACTATAGGAAGAGATGGCGACTAAAACGACTTGTCGCCA

GCTTGTTGAGTAGAGTGTGAGCTCCG

caSp4DNA-2 26 CAGCTGGACCCGTCCTTCACCATTTCATTCAGTCGCGTCCGAAGACGCG

ACTAGTTACGGAGCTCACACTCTACTC

caSp4SplitBDNA1 27 TAATACGACTCACTATAGGGATCAACACTGCACACCAGACAGCAAGATCC

TATATATACAGGGTACGGTACGG

caSp4SplitBDNA2 28 CAGCTGGACCCGTCCTTCACCATTTCATTCAGTCGCGTCCGGCGTACCG

TACCGTACCCTGTATATATAGGATC

FPcaSp4 29 TAATACGACTCACTATAGGAAGAGA

RPcaSp4 30 CAGCTGGACCCGTCCTTCACCATTTCATTC

FPIVTcaSpn 31 ATTCGCCCTTTAATACGACTCACTATAGGAAGAGA RPcaSp5aPartA 32 TCTACTCAACAAGCTGCGCGACAAGTCG

FPcaSp5aPartB 33 CGACTTGTCGCGCAGCTTGTTGAGTAG

RPcaSp5a 34 GCAGCTGGACCCGTCCTTCACCATTTC

RPcaSp5bPartA 35 CTACTCAACAAAGCTGGCGACAAGTCG

FPcaSp5bPartB 36 CGACTTGTCGCCAGCTTTGTTGAGTAG

RPcaSp5b 37 CAGCTTGGACCCGTCCTTCACCATTTC

RPcaSp6aPartA 38 TCTACTCAACAAGCTGCCGGCGACAAGTCG

FPcaSp6aPartB 39 CGACTTGTCGCCGGCAGCTTGTTGAGTAG

RPcaSp6a 40 AAGCAGCTGGACCCGTCCTTCACCATTTC

RPcaSp6bPart A 41 CTACTCAACAAGAGCTGGCGACAAGTCG

FPcaSp6bPartB 42 CGACTTGTCGCCAGCTCTTGTTGAGTAG

RPcaSp6b 43 CAGCTCTGGACCCGTCCTTCACCATTTC

RPcaSp7PartA 44 TCTACTCAACAAGTGCCTGGCGACAAGTCG

FPcaSp7PartB 45 ACGACTTGTCGCCAGGCACTTGTTGAGTAG

RPcaSp7 46 CAGACACTGGACCCGTCCTTCACCATTTC

RPcaSp7m 47 CAGTCACTGGACCCGTCCTTCACCTTTC

RPcaSp9PartA 48 CTCTACTCAACAAGTGCCTGCCGCGACAAGTCG FPcaSp9PartB 49 AACGACTTGTCGCGGCAGGCACTTGTTGAGTAGAG

RPcaSp9 50 AGCAGACACTGGACCCGTCCTTCACC

RPcaSp9m 51 AGCACACACTGGACCCGTCCTTCACCTTTC

RPcaSp9m' 52 AGCACACACTGGACCCGTCCTTCACCTTTC

RPcaSp9m" 53 AGCCGACACTGGACCCGTCCTTCACCTTTC

FP caSp4SplitB 54 TAATACGACTCACTATAGGGATCAACACTG

RP caSp4SplitB 55 ATTCGCCCTTTAATACGACTCACTATAGGG

RP caSp4SplitA 56 CAGGGTACGGTACGGTACGCCGGACGCGACTAGTTACGG

RP caSp4A 57 TATATATAGGATCTTGCTGTCTGGTGTGCAGTGTTGATCAGCTGGACCCG

TCCTTCACC

RP caSp7mA 58 TATATATAGGATCTTGCTGTCTGGTGTGCAGTGTTGATAGTCACTGGACC

CGTCCTTCACC

RPcaSp7nePartA 59 TCTACTCAACAAGTCCCTGGCGACAAGTCG

FPcaSp7nePartB 60 ACGACTTGTCGCCAGGGACTTGTTGAGTAG

RPcaSp7ne 61 AAGAGACTGGACCCGTCCTTCACCATTTC caSp7ne 62 GGAAGAGAUGGCGACUAAAACGACUUGUCGCCAGGG/¾CUUGUUGAGU

AGAGUGUGAGCUCCGUAACUAGUCGCGUCUUCGGACGCGACUGAAUG A A A U G G U G A A G G A C G G G U C C A G l ; C 'C U U

Duplex IVT DNA 63

ATTCGCCCTTTAATACGACTCACTATAGGAAGAGATGGCGACTAAAACGAC " positive strand

TGTCGCCAGCTTGTTGAGTAGAGTGTGAGCTCCGTAACTAGTCGCGTCTTC GGACGCGACTGAATGAAATGGTGAAGGACGGGTCCAGCTG Duplex IVT DNA 64 TAAGCGGGAAATTATGCTGAGTGATATCCTTCTCTACCGCTGATTTTGCTG

ACAGCGGTCGAACAACTCATCTCACACTCGAGGCATTGATCAGCGCAGAAC

negative strand

CCTGCGCTGACTTACTTTACCACTTCCTGCCCAGGTCGAC

TABLE 1- Sequences of the instant invention

*Duplex IVT DNA negative strand is in 3 '-5' direction; remaining strands have been given in 5 '-3' direction following the convention.

RP in the table denotes Reverse Primer and FP in table denotes Forward Primer. (N)n means a tract having any base selected from group comprising A, G, U,C and modified base or any combinations thereof and n indicates the number of bases that can vary from about 4-9 bases.

In D2 Alexa 647 (SEQ ID No. 19), the Alexa 647 is attached to the T shown in bold and italic and red. Additionally, '(N)n' may alternately be represented as 'N' wherein N is base selected from group comprising A, G, U, C and modified base or any combinations thereof and number of bases vary from about 4 to about 9 bases.

Further the Duplex IVT DNA positive strand set forth as SEQ ID No. 63 and the Duplex IVT DNA negative strand set forth as SEQ ID No. 64 together form the Duplex IVT DNA template having the following sequence:

5)'ATTCGCCCTTTAATACGACTCACTATAGGAAGAGATGGCGACTAAAACGACTTG TCGCCAGCTTGTTGAGTAGAGTGTGAGCTCCGTAACTAGTCGCGTCTTCGGACGCGACTG AATGAAATGGTGAAGGACGGGTCCAGCTG TAAGCGGGAAATTATGCTGAGTGATATCCTTCTCTACCGCTGATTTTGCTGAACAGCGGT CGAACAACTCATCTCACACTCGAGGCATTGATCAGCGCAGAAGCCTGCGCTGACTTACTT TACCAC CCTGCCCAGGTCGAC5 '

In another embodiment of the instant disclosure, the nucleic acid sensors of the present disclosure can be delivered into cytoplasm by electroporation. They can also be used in genetically encodable formats which involves making cells express the sensors of the invention, followed by incubation of the cell in DFHBI to measure the cAMP concentration inside cytoplasm of the cell expressing the sensor of invention. Further, the sensors of the present disclosure can be targeted to various sub-cellular compartments for determining the concentration of cAMP in those compartments. This can be done by using a RNA-DNA hybrid in place of Split B RNA, wherein in the sensor designed by Strategy 2 described above, the portion complementary to D2 Alexa 647 is DNA instead of RNA. This would result in duplex DNA at the end of sensor which helps in targeting this sensor to various sub-cellular compartments.

The present disclosure is further elaborated with the help of following examples and associated figures. However, these examples should not be construed to limit the scope of the present disclosure.

EXAMPLES

EXAMPLE 1: EFFECT OF STEM-2 ON THE DFHBI BINDING ACTIVITY OF THE NUCLEOTIDE SEQUENCE

Various nucleotide sequences for binding DFHBI are designed by varying the length of the stem-2 region of the nucleotide sequence of the instant invention in between 4-9 base pairs. The nucleotide sequences of interest (aSp9/ aSp6/ aSp4) and Sp at a concentration of about 2.5μΜ in water is heated to 60° C for 10 min followed by incubation in ice for 5 min. About 14.4 μΙ_ of this RNA is then added to about 345 L solution of about IX sensing buffer containing about 10 μΜ DFFIBI. DFFIBI emission spectra of this solution are acquired using A ex= 469 nm. Fluorescence emission spectra shown in figure 3 A shows that fluorescence emission of DFFIBI increases upon binding to Sp (Spinach) 5 bases (5 bases are present in the stem of stemloop-2 of Sp), aSp9 (SEQ ID No. 4), aSp6 (SEQ ID No. 3), and aSp4 (SEQ ID No. 5) (named according to the number of base pairs in stem-2 region of the nucleotide sequence). In order to calculate the fold change in intensity of DFFIBI upon binding to Sp, aSp9, aSp6 and aSp4 , 100 nM of the RNA (Sp, aSp9/ aSp6/ aSp4) in water is heated to 60° C for 10 min followed by incubation in ice for 5 min. About 36 μΙ_ of this RNA is then added to about 324 L solution of about IX sensing buffer containing about 10 μΜ DFFIBI. DFFIBI emission spectra of this solution are acquired using A ex= 469 nm. DFFIBI emission intensity at 501 nm of this solution is divided with the DFFIBI emission intensity at 501 nm of the similar solution without nucleotide sequence to get the fold change as shown in Figure 3B. From the figures it is clear that sequences with shorter stem sequences are less effective in increasing the fluorescence of DFFIBI as compared to longer stem. Additionally sequences aSp7m (SEQ ID NO. 6), aSp5b (SEQ ID No. 7) and aSp6b (SEQ ID No. 8) also enable increase in fluorescence of DFFIBI upon binding. This indicates that for the activity of the nucleotide sequences of the instant disclosure, the stability of Stem-2 is very crucial which is conferred upon by 4 to 9 number of base pairs in the stem, and not any random stem sequence and length will work. EXAMPLE 2: PREPARATION OF RNA

1. Preparation of template for IVT (In Vitro Transcription) As depicted in Figure 4(a), a duplex DNA sequence corresponding to the nucleic acid RNA sensors and T7 promoter at its 5 ' end is used as template for IVT to get the RNA. The IVT template for caSp4 is made by overlap extension PCR using two overlapping ss DNA oligonucleotides obtained from Sigma. These overlapping sequences are caSp4DNA- 1 and caSp4DNA-2 shown in Table 1 (SEQ ID No. 25 and 26). This duplex DNA template is cloned in TopoTA vector using commercially available kit from Life Technologies, USA, i.e., TOPO® TA Cloning® Kit according to the manufacturer's instructions. The TopoTA vector carrying the IVT template for caSp4 as insert is transformed into E. coli DH5a cells. These cells are obtained from Life Technologies, USA (One Shot® MAX Efficiency™ DH5α-T1R). These transformed E. Coli are grown in media such as Luria Broth and plasmid is isolated from them. The sequence of the inserted template for IVT is confirmed by sequencing of the isolated plasmid. The inserted DNA is PCR amplified from the plasmid carrying the correct sequence using primers FPIVTcaSpn and RPcaSp4 shown in Table 1 (SEQ ID No. 31 and 30). This PCR product is ethanol precipitated and used as IVT template for getting RNA corresponding to caSp4. PCR reactions are carried out using Ι μΜ Forward Primer and Reverse Primer each using 0.3U Pfu DNA polymerase, 0.2 mM dNTP and 5 ng of template DNA of interest, using 30 cycle PCR reaction involving the following steps:

1. Initial denaturation at 94° C for 5 minutes

2. Denaturation at 94° C for 1 minute

3. Annealing at 60° C for 1 minute

4. Elongation at 72° C for 1 minute

5. Steps (2), (3) and (4) are repeated 30 times

6. Final elongation is carried out at 72°C for 5 minutes

7. Hold at 4°C.

IVT template for other constructs is made by modifying the IVT template for caSpn obtained as described ahead. For the constructs which need changes only at 3' end, PCR amplification is done using caSpn IVT template as PCR template using reverse primer RPcaSpn shown in Table 1 having desired change in the sequence. For those constructs that need changes in the middle, the entire construct is divided into two overlapping fragments namely, part A and part B, such that the changes needed in the sequence are at the ends of part A and part B. Part A is PCR amplified from IVT template of caSp4 using FPIVTcaSpn (SEQ ID No. 31) and RPcaSpnPartA and Part B is PCR amplified from IVT template caSpn using FPcaSpnPartB and RPcaSpn. Sequences of these primers are shown in Table 1. Overlap extension PCR is then done using about 5 ng of Part A and Part B each using FPIVTcaSpn and RPcaSpn. The PCR product obtained is cloned into TopoTA vector and transformed into E. coli DH5a cells as described earlier. E. Coli carrying plasmid is grown in media and plasmid is isolated from the bacteria and sequenced. From the plasmid carrying the correct sequence, template for IVT is made my PCR amplification using suitable forward primer (FPIVTcaSpn, SEQ ID No. 31), and reverse primer (RPcaSpn) (Table 1).

2. Preparation of RNA using IVT (In Vitro Transcription)

About 100 ng of template for IVT for a given of caSpn is used for IVT using MEGAscript® T7 Transcription Kit from life technologies, USA according to manufacturer's instructions. Formation of intact RNA is confirmed by running on 10 % denaturing PAGE in 0.5 X TBE. If only single band of RNA is present, the IVT product is purified using NucAway™ Spin Column purchased from life technologies, USA according to manufacturer's instructions to remove unused NTPs and salt. NucAway purification is followed by ethanol precipitation. The precipitated RNA is resuspended in RNAase free water. RNA obtained now is quantified using OD leO nm his RNA is used to study the cAMP sensing ability.

EXAMPLE 3: SCREENING FOR RNA CONSTRUCTS FOR cAMP SENSING ABILITY

Various nucleic acid sensor sequences obtained by joining cAMP aptamer and modified DFUBI aptamer having varying length and sequence of stem-2 are synthesized and screened for their ability to enhance DFUBI fluorescence in the presence of cAMP. In this regard, various nucleic acid sensors are prepared by varying nucleotide sequences and length of stem-2 from 4 to 9 bp. Fluorescence emission spectra of about 360 μΐ. solution of about 10 μΜ of DFUBI in about IX sensing buffer with and without about 2mM cAMP are acquired using ^ χ =469 nm and used as blank spectra. About 100 nM of the nucleic acid sensors (about 36 L from about 1 μΜ stock in water) and about 10 μΜ of DFUBI (about 36 L from about 100 μΜ stock) in about IX sensing buffer (about 40 mM HEPES pH about 7.4, about 125 mM KC1 and about ImM MgCl 2 ) are mixed in presence and absence of about 2 mM cAMP (about 36 L from about 20 mM stock) in a total volume of about 360 L. After 2 hours of incubation at about 25°C, fluorescence emission spectra are acquired using λ εχ 469 nm. The corresponding blank spectra are subtracted from these spectra. After blank subtraction, emission intensity at 501 nm in presence of about 2 mM cAMP is divided by emission intensity at 501 nm in absence of cAMP and is reported as fold change. As seen in figure 4(b), few of the sequences did not respond to cAMP at all e.g. 5a (SEQ ID. No. 10), 6a (SEQ ID No. 1 1), 7 (SEQ ID No. 12), 9m (SEQ ID No.13) and 9m' (SEQ ID No. 14) (named so based on the length of the stem-2). Additionally, as can be seen from the figure 4(d), sensors with stem-2 of 6, 7 and 9 are highly fluorescent even in the absence of cAMP and thus not used as sensors. Further, as can be seen from figure 4(e), 7NE (SEQ ID No. 62) does not show any detectable change in signal in presence and absence of cAMP. Further, from figures 4(b) and 4(d), it is clear that certain constructs, e.g., 4 (SEQ ID No. 9), 5b (SEQ ID No. 16), 7m (SEQ ID No. 15) and 6b (SEQ ID No. 22) respond to cAMP.

EXAMPLE 4: cAMP RESPONSE CHARACTERISTICS OF NON-RATIO METRIC SENSORS

The response characteristics of the RNA constructs that did respond to cAMP are studied in detail as described ahead. A 2.5μΜ solution of nucleic acid sensor of interest (caSp4 (SEQ ID No. 9)1 caSp5b (SEQ ID No. 16) / caSp7m (SEQ ID No. 15),) in about IX annealing buffer (about 8 mM HEPES pH of about 7.4, about 20 mM NaCl and about lmM EDTA) with about 2.5 μΜ free Alexa 647 in a total volume of about 500 L-l mL is heated for about 10 minutes, followed by about 4 hour incubation at about 5° C.

DFHBI and Alexa 647 fluorescence emission spectra of about 345 SL solution containing about 10 μΜ DFHBI, in about IX sensing buffer, about 4mM MgCl 2 and varying cAMP concentrations are acquired at 25° C and taken as time t=0 spectra. Fluorescence emission spectra are taken using λ €χ =469 nm for DFHBI and for Alexa 647 using λς Χ =647nm.Then, about 14.4 μΙ_, of about 2.5μΜ stock solution of RNA sensor prepared as described above is added to this solution. DFHBI and Alexa 647 emission spectra are acquired again as described after t=5, 10, 15 and 30 minute of addition of sensor. Time t=0 spectra are used as blank and subtracted from all other time point spectra. After subtraction, emission intensity of DFHBI at 501 nm is divided by emission intensity of Alexa 647 at 670 nm to obtain normalized intensity. Normalized intensity (at t=5 minutes) is plotted as against respective cAMP concentrations for these RNA as shown in Figure 4(c).

The response characteristics of these sensors vary in terms of percentage change in signal (%SC) cAMP sensitive regime as well as Signal/noise as shown in figure 4(c) and Figure 4(d) and also shown in Table 2 below. This therefore confirms that the sensors of the present disclosure are able to detect cAMP and also shows the importance of the communication module in stabilizing the sensor and determining its activity.

Table 2: cAMP response characteristics the cAMP sensors indicating the cAMP concentration where 10-90 % change in fluorescence emission of DFFIBI takes place, EC 50 relates to the cAMP concentration where half maximal change in DFFIBI fluorescence is observed and % change in signal (% SC) is percentage change in DFFIBI fluorescence emission between two extreme cAMP concentrations.

EXAMPLE 5: QUANTIFICATION OF cAMP USING THE RATIOMETRIC NUCLEIC ACID SENSORS

The various nucleotide sequences of the present disclosure are taken and further modified as per Strategy I to produce ratiometric sensors. Two of these sensors (SEQ ID No. 17 and SEQ ID No. 18) are then tested for their ability to detect cAMP as shown in Figure 7(a) and tested for fluorescence in the presence and absence of cAMP. As seen in figure 7(a), one of the sensors (SEQ ID No. 18 or CaSp7m A ) is able to sense cAMP, whereas the other sensor (CaSp4 A , SEQ ID No. 17) lost the DFFIBI binding activity. This loss of activity of caSp4 A is possibly because the extended 3' end creates a steric hindrance near sensing and communication module. To overcome the above drawback, ratiometric sensors are designed as per Strategy II.

Ratiometric sensor samples are prepared by heating at about 60° C about 2.5μΜ solution of RNA (or two RNAs caSp4SplitA and caSp4SplitB in case of strategy II) in about IX annealing buffer (about 8 mM HEPES pH of about 7.4, about 20 mM NaCl and about ImM EDTA) with about 2.5 μΜ D2Alexa 647 labelled DNA in a total volume of about 500 L-l mL for about 10 minutes, followed by about 4 hour incubation at about 5° C. DFHBI and Alexa 647 fluorescence emission spectra of about 345 L solution containing about 10 μΜ DFHBI, in about IX sensing buffer, about 4mM MgCl 2 and varying cAMP concentrations are acquired at 25° C and taken as time t=0 spectra. Fluorescence emission spectra are taken using ^ χ 469 nm for DFHBI and for Alexa 647 using ^ χ 647 nm. Then, about 14.4 μΙ_, of about 2.5μΜ stock solution of sensor prepared as described above is added to this solution. DFHBI and Alexa 647 emission spectra are acquired as described earlier after t=5, 10, 15 and 30 minute of addition of sensor. Time t=0 spectra are used as blank and subtracted from all other time point spectra. After subtraction, emission intensity of DFHBI at 501 nm (G) is divided by emission intensity of Alexa 647 at 670 nm (R) to obtain G/R values. G/R values against respective cAMP concentrations for both the sensors (caSp7m A (SEQ ID No. 18)) and (CaSp4split (SEQ ID No. 20-split A and SEQ ID NO. 21-splitB)) hybridized with Alexa 647 labelled DNA is plotted to get the cAMP response curve as shown in Figure 7(b). Figure shows cAMP response curve for ratiometric sensor prepared by strategy I and caSp4split/cAMPhor (SEQ ID No. 20 and SEQ ID No. 21) hybridized with Alexa 647 labelled DNA (ratiometric sensor prepared by strategy II).

G/R values for caSp7m A is at t= 30 minutes and for caSp4split/cAMPhor is at t=5 minutes. From the figure, it is clear that the ratiometric sensors can quantify cAMP. The detailed cAMP response characteristics have been summarized in Table 3 below.

Name 10 %-90% change EC so % Signal Change (%SC) cAMPhor (5 min, 25° C) 100 μΜ-4.5 mM 800 μΜ 690% (0 μΜ-9.7 mM) cAMPhor (10 min, 25° C) 70 μΜ-3.2 ιηΜ 524 μΜ 470% (0 μΜ-9.7 mM) cAMPhor (15 min, 25° C) 50 μΜ-2.7 mM 456 μΜ 390% (0 μΜ-9.7 mM) cAMPhor (2 h, 25° C) 40 μΜ-4.2 mM 403 μΜ 213% (0.25 μΜ-lO mM) cAMPhor (MnCl 2 5 min, 25° 45 μΜ-4 mM 400 μΜ 669 % (5 μΜ-9.7 mM)

C) cAMPhor (5 mMATP, 25°C) 100 μΜ -3.8 ιηΜ 25 % (100 μΜ-3.8 mM) cAMPhor (2 h, 5° C) 10 μΜ-1.5 mM 140 μΜ 510% (0.25 μΜ-lO mM) cAMPhor (12 h, 5° C) 10 μΜ-1.5 mM 260 μΜ 110% (0.25 μΜ-lO mM) caSp7m A647 (30 min, 25° C) 250 μΜ-6.3 mM 1.7 mM 767% (0 μΜ-lO mM)

Table 3: Summary of the response characteristics of ratio metric sensors.

Further, figure 8 shows the cAMP sensitivity of caSp4 split under different conditions. From the figure 8a , it a can be seen that at room temperature, in about IX sensing buffer, about 4 mM MgC12 and about 10 μΜ DFHBI at about 25° C in about 5 minutes, the sensors of the invention cAMPhor can measure cAMP from about 100 uM- to about 4.5 mM. From the figure 8b, it can be seen that in about IX sensing buffer, about 4 mM MgCl 2 , about 1 mM MnCl 2 and about 10 μΜ DFHBI at about 25° C in about 5 minutes, the sensors of the invention cAMPhor can measure cAMP from about 45 μΜ to about 4mM. From figure 8c, it can be seen that in about IX sensing buffer, about 4 mM MgCl 2 , and about 10 μΜ DFFIBI at about 5° C in about 2 hours, the sensors of the invention cAMPhor can measure cAMP from about 10 μΜ to about 1.5 mM. Figure 8d shows the comparative graph of all three conditions. From Figure 8e, it can be seen that in about IX sensing buffer, about 4 mM MgCl 2; and about 10 μΜ DFHBI at about 25° C in about 2 hours, cAMPhor can measure cAMP from about 40 μΜ to about 4.2 mM. Further, from Figure 8f, it can be seen that in about IX sensing buffer, about 4 mM MgCl 2; about 10 μΜ DFHBI and 5mMATPMg at about 25° C in about 2 hours, cAMPhor can measure cAMP from about 100 μΜ to about 3.8 mM. EXAMPLE 6: MEASUREMENT OF cAMP SECRETED BY BACTERIA INTO GROWTH MEDIA

M. smegmatis MC2155 obtained from USA is grown from glycerol stocks in 7H9 media supplemented with about 0.2% glycerol and about 0.05% Tween-80 at about 37° C with shaking at about 180 rpm, till the culture reaches saturation. Cells are centrifuged at about 13000 rcf and washed with TBST (about lOmM Tris-HCl (pH of about 7.5), about 0.89% NaCl, and about 0.05% Tween-80). Cells are re-suspended in original volume of fresh 7H9 media supplemented with about 0.2% glycerol and about 0.05% Tween-80. To this suspension, Sodium Dodecyl Sulphate (SDS) is added to achieve a final concentration of about 0.05%. This suspension is allowed to grow for about 1.5 hours at about 37°C. After about 1.5 hours, cells are centrifuged at about 13000 rcf and the cell free supernatant is recovered. An aliquot of the supernatant is acidified with HCl at a final concentration of about 0.1N, boiled for about 10 minutes and used for radioimmunoassay. The remaining supernatant is boiled and used for measurements with caSp4split. Measurements using about 100 nM caSp4split at about 5°C in about IX sensing buffer + about 10μΜ DFHBI + about 4mM MgC12 and G/R values are taken at t=2 hours as described earlier. Samples are spun at about 10000 rcf for about 4 minutes immediately before acquiring the fluorescence spectra. As can be observed from Figure 9, the cAMP concentration measured using caSp4Split matched well with values obtained using standard radioimmunoassay technique thus validating that caSP4Spilt can measure unknown cAMP concentrations in complex biological mixtures provided they lie in sensitive regime.

EXAMPLE 7: SPECIFICITY OF RATIOME TRIC SENSORS

Ratiometric sensor samples are prepared by heating about 2.5μΜ solution of RNA (or two RNAs caSp4SplitA and caSp4SplitB in case of strategy II) in about IX annealing buffer (about 8 mM HEPES pH of about 7.4, about 20 mM NaCl and about ImM EDTA) with about 2.5 μΜ D2Alexa 647 labelled DNA in a total volume of about 500 μΙ_-1 mL for about 10 minutes, followed by about 4 hour incubation at about 5° C. DFHBI and Alexa 647 fluorescence emission spectra of about 345 μΐ ^ solution containing about 10 μΜ DFHBI, in about IX sensing buffer, about 4mM MgCl 2 and structural variant of cAMP (ATP/ADP/AMP/cGMP) at about 25° C are taken as time t=0 spectra. Fluorescence emission spectra are taken using X, x = 469 nm for DFHBI and for Alexa 647 using X> x 647. Then, about 14.4 μΙ_, of about 2.5μΜ stock solution of sensor prepared as described above is added to this solution. DFHBI and Alexa 647 emission spectra are acquired as described earlier after t=5, and 30 minute of addition of sensor. Time t=0 spectra are used as blank and subtracted from all other time point spectra. After subtraction, emission intensity of DFHBI at 501 nm (G) is divided by emission intensity of Alexa 647 at 670 nm (R) to obtain G/R values. G/R values obtained in the presence of structural variants of cAMP are divided by the corresponding G/R values for equi-molar cAMP concentration to get normalized G/R value against respective sensors (Figure 10). Figure 10 shows normalized G/R values for structural variants of cAMP of the sensor prepared by strategy I (caSp7m A ) and caSp4split/cAMPhor (SEQ ID No. 20 and SEQ ID No. 21) hybridized with Alexa 647 labelled DNA (ratiometric sensor prepared by strategy II).

Normalized G/R values for caSp7m A is at t=30 minutes and for caSp4split/cAMPhor is at t=5 minutes. From the figure, it is clear that the ratiometric sensors are specific for cAMP.

EXAMPLE 8: cAMP IMAGING INSIDE MEMBRANE BOUND COMPARTMENTS

To determine the ability of cAMPhor for imaging cAMP in model membrane-bound vesicles, giant Unilamellar Vesicles (GUVs) are prepared from DMPC lipids (1,2- dimyristoyl-sn-glycero-3-phosphocholine) encapsulating about 1 μΜ cAMPhor, about 50 μΜ DFUBI and varying concentrations of cAMP in about IX sensing buffer pH of about 6.8, about 4 mM MgCl 2 and about 500 mM sucrose. The response curve for cAMPhor in bulk solution under identical conditions is constructed. cAMPhor retained its 100 μΜ-4.5 mM sensitive regime with an overall % SC of -100 % as shown in Figure 11(c) (open circles). GUVs containing cAMPhor, DFUBI and cAMP at the indicated concentrations are imaged under a fluorescence microscope in bright field (BF), DFUBI (G) and Alexa 647 (R) channels. The images acquired in R channel are divided by the corresponding images in G channel and pseudocolored into 16 colours to get R/G images (Figure 11(a)). These clearly revealed decreasing R/G values as the cAMP inside the GUVs increases. The frequency histogram of normalized G/R ratios of individual GUVs (n~25) at the indicated cAMP concentrations showed with high clarity, the expected shifts towards higher G/R values with increasing cAMP concentrations (Figure 11(b)). Moreover, the mean G/R from three different experiments correlated well with the expected G/R values in bulk solution for every concentration of elevated cAMP tested as shown in Figure 11(c). This confirms that cAMPhor can image cAMP quantitatively inside membrane bound compartments.

EXAMPLE 9: COMPARISON OF THE SENSORS OF THE INSTANT INVENTION WITH THE EXISTING SENSORS FOR cAMP. The % signal change of the sensor of the instant invention is very high and it measures above 100 uM levels of cAMP as can be observed from Figure 7(b) explained in Example 5. It is observed from the prior art, that the sensors presently known in art cannot measure this high cAMP concentrations. The cAMP sensor of present invention helps in measuring elevated cAMP levels that are present at various pathological conditions and infections. None of the existing sensors of the prior art have comparable %SC to the present sensor. Also the % signal change (% SC) shown by sensors is much higher than existing cAMP sensors for live cell imaging of cAMP as shown in table 4 below.

Table 4: Summarising the cAMP sensors known in art and one the cAMP sensors present invention in order to facilitate the comparison. Thus, it is clear from the above examples and figures that the sensors of the instant disclosure are highly useful for detecting and quantifying cAMP. Further, the sensors of the instant disclosure may be used in various applications such as, but not limiting to:

• Measuring cAMP in in vitro assays, wherein one is interested in measuring the amount of either secreted or intracellular accumulated cAMP by a population of cells;

• Visualizing and measuring levels of cAMP in both prokaryotic and eukaryotic cells;

• Monitoring the elevation in cAMP levels (cAMP burst) that occurs during various pathogen infections;

• For studying the role of cAMP in compartmentalization of eukaryotic cells, where cAMP regulates diverse cellular processes in such cells;

• For studying the activity of adenylyl cyclase and hence for screening to identify inhibitors and activators of this enzyme. This has direct application in screening for drug molecules.

• Further, the sensors of the instant disclosure are targeted to various compartments inside cells including by interfacing them with DNA, proteins and other small molecules.

While a number of embodiments and examples of this invention are described herein, it is apparent that these embodiments and examples may be altered to provide additional embodiments and examples which use the processes of this invention.

SEQUENCE LISTING

<1 10> National Centre for Biological Sciences (NCBS-TIFR) <120> NUCLEOTIDE SEQUENCES, NUCLEIC ACID SENSORS AND METHODS THEREOF

<130> IP23185 <140> 3973/CHE/2013

<141> 2013-09-04

<160> 64 <170> Patentln version 3.5

<210> 1

<211> 79

<212> RNA

<213> Artificial Sequence

<220>

<223> nucleotide sequence

<220>

<221> misc_feature

<222> (1)..(79)

<223> n is base selected from group comprising A, G, U, C and modified

base or any combinations thereof and number of bases vary from

about 4 to about 9 bases

<400> 1

nuuguugagu agagugugag cuccguaacu agucgcgucu ucggacgcga cugaaugaaa 60 uggugaagga cggguccan 79

<210> 2

<211> 110

<212> RNA

<213> Artificial Sequence

<220>

<223> nucleic acid sensor

<220> <221> misc_feature

<222> (1) .(110)

<223> n is base selected from group comprising A, G, U, C and modified

base or any combinations thereof and number of bases vary from

about 4 to about 9 bases

<400> 2

ggaagagaug gcgacuaaaa cgacuugucg cnuuguugag uagaguguga gcuccguaac uagucgcguc uucggacgcg acugaaugaa auggugaagg acggguccan

<210> 3

<2\ \> 91

<212> RNA

<213> Artificial Sequence

<220>

<223> aSp6

<220>

<221> misc_feature

<222> (1)..(91)

<400> 3

cggcagcuug uugaguagag ugugagcucc guaacuaguc gcgucuucgg acgcgacuga 60 augaaauggu gaaggacggg uccagcugcu u 91

<210> 4

<211> 95

<212> RNA

<213> Artificial Sequence

<220>

<223> aSp9

<220>

<221> misc_feature

<222> (1)..(95) <400> 4

ggcaggcacu uguugaguag agugugagcu ccguaacuag ucgcgucuuc ggacgcgacu 60 gaaugaaaug gugaaggacg gguccagugu cugcu 95 <210> 5

<2\ \> 85

<212> RNA

<213> Artificial Sequence

<220>

<223> aSp4

<220>

<221> misc_feature

<222> (1)..(85)

<400> 5

cagcuuguug aguagagugu gagcuccgua acuagucgcg ucuucggacg cgacugaaug 60 aaauggugaa ggacgggucc agcug 85

<210> 6

<21 1> 91

<212> RNA

<213> Artificial Sequence

<220>

<223> aSp7m

<220>

<221> misc feature

<222> (1)..(91) <400> 6

caggcacuug uugaguagag ugugagcucc guaacuaguc gcgucuucgg acgcgacuga 60 augaaauggu gaaggacggg uccagugacu g 1

<210> 7

<21 1> 87

<212> RNA

<213> Artificial Sequence

<220>

<223> aSp5b <220>

<221> misc_feature

<222> (1)..(87)

<400> 7

cagcuuuguu gaguagagug ugagcuccgu aacuagucgc gucuucggac gcgacugaau 60 gaaaugguga aggacggguc caagcug 87

<210> 8

<211> 89

<212> RNA

<213> Artificial Sequence

<220>

<223> aSp6b

<220>

<221> misc_feature

<222> (1)..(89) <400> 8

cagcucuugu ugaguagagu gugagcuccg uaacuagucg cgucuucgga cgcgacugaa 60 ugaaauggug aaggacgggu ccagagcug 89

<210> 9

<211> 116

<212> RNA

<213> Artificial Sequence

<220>

<223> caSp4 <220>

<221> misc_feature

<222> (1)..(116)

<400> 9

ggaagagaug gcgacuaaaa cgacuugucg ccagcuuguu gaguagagug ugagcuccgu 60 aacuagucgc gucuucggac gcgacugaau gaaaugguga aggacggguc cagcug 116 <210> 10

<211> 118

<212> RNA

<213> Artificial Sequence

<220>

<223> caSp5a

<220>

<221> misc feature

<222> (1)..(1 18)

<400> 10

ggaagagaug gcgacuaaaa cgacuugucg cgcagcuugu ugaguagagu gugagcuccg uaacuagucg cgucuucgga cgcgacugaa ugaaauggug aaggacgggu ccagcugc 118

<210> 1 1

<211> 122

<212> RNA

<213> Artificial Sequence

<220>

<223> caSp6a <220>

<221> misc feature

<222> (1)..(122)

<400> 11

ggaagagaug gcgacuaaaa cgacuugucg ccggcagcuu guugaguaga gugugagcuc 60 cguaacuagu cgcgucuucg gacgcgacug aaugaaaugg ugaaggacgg guccagcugc 120 uu 122

<210> 12

<211> 122

<212> RNA

<213> Artificial Sequence

<220>

<223> caSp7 <220>

<221> misc feature

<222> (1)..(122)

<400> 12

ggaagagaug gcgacuaaaa cgacuugucg ccaggcacuu guugaguaga gugugagcuc 60 cguaacuagu cgcgucuucg gacgcgacug aaugaaaugg ugaaggacgg guccaguguc 120 ug 122

<210> 13

<2\ \> 126

<212> RNA

<213> Artificial Sequence

<220>

<223> caSp9m'

<220>

<221> misc_feature

<222> (1)..(126)

<400> 13

ggaagagaug gcgacuaaaa cgacuugucg cggcaggcac uuguugagua gagugugagc 60 uccguaacua gucgcgucuu cggacgcgac ugaaugaaau ggugaaggac ggguccagug 120 ugugcu 126

<210> 14

<2\ \> 126

<212> RNA

<213> Artificial Sequence

<220>

<223> caSp9m" <220>

<221> misc_feature

<222> (1)..(126) <400> 14

ggaagagaug gcgacuaaaa cgacuugucg cggcaggcac uuguugagua gagugugagc 60 uccguaacua gucgcgucuu cggacgcgac ugaaugaaau ggugaaggac ggguccagug 120 ucggcu 126

<210> 15

<2\ \> 122

<212> RNA

<213> Artificial Sequence

<220>

<223> caSp7m

<220>

<221> misc_feature

<222> (1)..(122)

<400> 15

ggaagagaug gcgacuaaaa cgacuugucg ccaggcacuu guugaguaga gugugagcuc 60 cguaacuagu cgcgucuucg gacgcgacug aaugaaaugg ugaaggacgg guccagugac 120 ug 122

<210> 16

<2\ \> 118

<212> RNA

<213> Artificial Sequence

<220>

<223> caSp5b

<220>

<221> misc_feature

<222> (1) .(118)

<400> 16

ggaagagaug gcgacuaaaa cgacuugucg ccagcuuugu ugaguagagu gugagcuccg uaacuagucg cgucuucgga cgcgacugaa ugaaauggug aaggacgggu ccaagcug 118 <210> 17

<211> 154

<212> RNA

<213> Artificial Sequence

<220>

<223> caSp4A <220>

<221> misc_feature

<222> (1)..(154)

<400> 17

ggaagagaug gcgacuaaaa cgacuugucg ccagcuuguu gaguagagug ugagcuccgu 60 aacuagucgc gucuucggac gcgacugaau gaaaugguga aggacggguc cagcugauca 120 acacugcaca ccagacagca agauccuaua uaua 154

<210> 18

<2\ \> 160

<212> RNA

<213> Artificial Sequence

<220>

<223> caSp7mA

<220>

<221> misc_feature

<222> (1)..(160) <400> 18

ggaagagaug gcgacuaaaa cgacuugucg ccaggcacuu guugaguaga gugugagcuc 60 cguaacuagu cgcgucuucg gacgcgacug aaugaaaugg ugaaggacgg guccagugac 120 ugaucaacac ugcacaccag acagcaagau ccuauauaua 160

<210> 19

<211> 38

<212> DNA

<213> Artificial Sequence

<220> <223> D2 Alexa 647

<220>

<221> misc_feature

<222> (1)..(38)

<400> 19

tatatatagg atcttgctgt ctggtgtgca gtgttgat 38

<210> 20

<211> 95

<212> RNA

<213> Artificial Sequence

<220>

<223> caSp4SplitA

<220>

<221> misc feature

<222> (1)..(95) <400> 20

ggaagagaug gcgacuaaaa cgacuugucg ccagcuuguu gaguagagug ugagcuccgu 60 aacuagucgc guccggcgua ccguaccgua cccug 95

<210> 21

<211> 99

<212> RNA

<213> Artificial Sequence

<220>

<223> caSp4SplitB <220>

<221> misc feature

<222> (1)..(99)

<400> 21

aucaacacug cacaccagac agcaagaucc uauauauaca ggguacggua cgguacgccg 60 gacgcgacug aaugaaaugg ugaaggacgg guccagcug 99 <210> 22

<2U> 120

<212> RNA

<213> Artificial Sequence

<220>

<223> caSp6b

<220>

<221> misc feature

<222> (1)..(120) <400> 22

ggaagagaug gcgacuaaaa cgacuugucg ccagcucuug uugaguagag ugugagcucc 60 guaacuaguc gcgucuucgg acgcgacuga augaaauggu gaaggacggg uccagagcug 120

<210> 23

<2\ \> 31

<212> RNA

<213> Artificial Sequence

<220>

<223> cAMP aptamer <220>

<221> misc_feature

<222> (1) .(31)

<400> 23

ggaagagaug gcgacuaaaa cgacuugucg c 31

<210> 24

<2\ \> 87

<212> RNA

<213> Artificial Sequence

<220>

<223> Spinach

<220>

<221> misc feature <222> (1)..(87) <400> 24

gacgcgacug aaugaaaugg ugaaggacgg guccagcugc uucggcagcu uguugaguag 60 agugugagcu ccguaacuag ucgcguc 87

<210> 25

<2\ \> 76

<212> DNA

<213> Artificial Sequence

<220>

<223> caSp4DNA-l

<220>

<221> misc_feature

<222> (1)..(76)

<400> 25

taatacgact cactatagga agagatggcg actaaaacga cttgtcgcca gcttgttgag 60 tagagtgtga gctccg 76

<210> 26

<2\ \> 76

<212> DNA

<213> Artificial Sequence

<220>

<223> caSp4DNA-2

<220>

<221> misc_feature

<222> (1)..(76)

<400> 26

cagctggacc cgtccttcac catttcattc agtcgcgtcc gaagacgcga ctagttacgg 60 agctcacact ctactc 76

<210> 27

<211> 73 <212> DNA

<213> Artificial Sequence

<220>

<223> caSp4SplitBDNAl

<220>

<221> misc feature

<222> (1)..(73)

<400> 27

taatacgact cactataggg atcaacactg cacaccagac agcaagatcc tatatataca 60 gggtacggta egg 73

<210> 28

<211> 74

<212> DNA

<213> Artificial Sequence

<220>

<223> caSp4SplitBDNA2

<220>

<221> misc_feature

<222> (1)..(74)

<400> 28

cagctggacc cgtccttcac catttcattc agtcgcgtcc ggcgtaccgt accgtaccct 60 gtatatatag gate 74

<210> 29

<211> 25

<212> DNA

<213> Artificial Sequence

<220>

<223> FPcaSp4

<220>

<221> misc_feature

<222> (1)..(25) <400> 29

taatacgact cactatagga agaga 25

<210> 30

<211> 30

<212> DNA

<213> Artificial Sequence

<220>

<223> RPcaSp4 <220>

<221> misc_feature

<222> (1)..(30)

<400> 30

cagctggacc cgtccttcac catttcattc

<210> 31

<211> 35

<212> DNA

<213> Artificial Sequence

<220>

<223> FPIVTcaSpn

<220>

<221> misc_feature

<222> (1)..(35)

<400> 31

attcgccctt taatacgact cactatagga agaga 35

<210> 32

<211> 28

<212> DNA

<213> Artificial Sequence <220>

<223> RPcaSp5aPartA <220>

<221> misc feature

<222> (1)..(28)

<400> 32

tctactcaac aagctgcgcg acaagtcg 28

<210> 33

<2\ \> 27

<212> DNA

<213> Artificial Sequence

<220>

<223> FPcaSp5aPartB

<220>

<221> misc_feature

<222> (1)..(27)

<400> 33

cgacttgtcg cgcagcttgt tgagtag

<210> 34

<211> 27

<212> DNA

<213> Artificial Sequence

<220>

<223> RPcaSp5a

<220>

<221> misc_feature

<222> (1)..(27)

<400> 34

gcagctggac ccgtccttca ccatttc

<210> 35

<211> 27

<212> DNA

<213> Artificial Sequence

<220> <223> RPcaSp5bPartA

<220>

<221> misc feature

<222> (1)..(27)

<400> 35

ctactcaaca aagctggcga caagtcg

<210> 36

<211> 27

<212> DNA

<213> Artificial Sequence

<220>

<223> FPcaSp5bPartB

<220>

<221> misc_feature

<222> (1)..(27)

<400> 36

cgacttgtcg ccagctttgt tgagtag

<210> 37

<21 1> 27

<212> DNA

<213> Artificial Seqi

<220>

<223> RPcaSp5b

<220>

<221> misc_feature

<222> (1)..(27)

<400> 37

cagcttggac ccgtccttca ccatttc 27

<210> 38

<211> 30

<212> DNA <213> Artificial Sequence

<220>

<223> RPcaSp6aPartA

<220>

<221> misc_feature

<222> (1)..(30)

<400> 38

tctactcaac aagctgccgg cgacaagtcg 30

<210> 39

<2\ \> 29

<212> DNA

<213> Artificial Sequence

<220>

<223> FPcaSp6aPartB

<220>

<221> misc_feature

<222> (1)..(29)

<400> 39

cgacttgtcg ccggcagctt gtt

<210> 40

<2\ \> 29

<212> DNA

<213> Artificial Seq

<220>

<223> RPcaSp6a

<220>

<221> misc feature

<222> (1)..(29)

<400> 40

aagcagctgg acccgtcctt caccatttc 29 <210> 41

<211> 28

<212> DNA

<213> Artificial Sequence

<220>

<223> RPcaSp6bPart A <220>

<221> misc_feature

<222> (1)..(28)

<400> 41

ctactcaaca agagctggcg acaagtcg 28

<210> 42

<211> 28

<212> DNA

<213> Artificial Sequence

<220>

<223> FPcaSp6bPartB

<220>

<221> misc feature

<222> (1)..(28)

<400> 42

cgacttgtcg ccagctcttg ttgagtag 28 <210> 43

<211> 28

<212> DNA

<213> Artificial Sequence <220>

<223> RPcaSp6b

<220>

<221> misc_feature

<222> (1)..(28)

<400> 43 Lgctctgga cccgtccttc accatttc

<210> 44

<211> 30

<212> DNA

<213> Artificial Sequence

<220>

<223> RPcaSp7PartA

<220>

<221> misc_feature

<222> (1)..(30)

<400> 44

tctactcaac aagtgcctgg cgacaagtcg 30

<210> 45

<211> 30

<212> DNA

<213> Artificial Sequence

<220>

<223> FPcaSp7PartB <220>

<221> misc_feature

<222> (1)..(30)

<400> 45

acgacttgtc gccaggcact tgttgagtag 30

<210> 46

<211> 29

<212> DNA

<213> Artificial Sequence

<220>

<223> RPcaSp7

<220>

<221> misc feature <222> (1)..(29)

<400> 46

cagacactgg acccgtcctt caccatttc 29

<210> 47

<2\ \> 28

<212> DNA

<213> Artificial Sequence

<220>

<223> RPcaSp7m

<220>

<221> misc feature

<222> (1)..(28)

<400> 47

cagtcactgg acccgtcctt cacctttc

<210> 48

<21 1> 33

<212> DNA

<213> Artificial Sequence

<220>

<223> RPcaSp9PartA

<220>

<221> misc_feature

<222> (1)..(33)

<400> 48

ctctactcaa caagtgcctg ccgcgacaag teg 33

<210> 49

<211> 35

<212> DNA

<213> Artificial Sequence

<220>

<223> FPcaSp9PartB <220>

<221> misc feature

<222> (1)..(35)

<400> 49

aacgacttgt cgcggcaggc acttgttgag tagag 35

<210> 50

<2\ \> 26

<212> DNA

<213> Artificial Sequence <220>

<223> RPcaSp9

<220>

<221> misc_feature

<222> (1)..(26)

<400> 50

agcagacact ggacccgtcc ttcacc

<210> 51

<211> 30

<212> DNA

<213> Artificial Seq

<220>

<223> RPcaSp9m

<220>

<221> misc feature

<222> (1)..(30)

<400> 51

agcacacact ggacccgtcc ttcacctttc 30

<210> 52

<21 1> 30

<212> DNA

<213> Artificial Sequence <220>

<223> RPcaSp9m' <220>

<221> misc_feature

<222> (1)..(30)

<400> 52

agcacacact ggacccgtcc ttcacctttc 30

<210> 53

<2\ \> 30

<212> DNA

<213> Artificial Sequence

<220>

<223> RPcaSp9m"

<220>

<221> misc feature

<222> (1)..(30)

<400> 53

agccgacact ggacccgtcc ttcacctttc 30 <210> 54

<211> 30

<212> DNA

<213> Artificial Sequence

<220>

<223> FPcaSp4SplitB

<220>

<221> misc_feature

<222> (1)..(30)

<400> 54

taatacgact cactataggg atcaacactg

<210> 55

<211> 30 <212> DNA

<213> Artificial Sequence

<220>

<223> RPcaSp4SplitB

<220>

<221> misc_feature

<222> (1)..(30)

<400> 55

attcgccctt taatacgact cactataggg 30

<210> 56

<21 1> 39

<212> DNA

<213> Artificial Sequence

<220>

<223> RPcaSp4SplitA <220>

<221> misc_feature

<222> (1)..(39)

<400> 56

cagggtacgg tacggtacgc cggacgcgac tagttacg 39

<210> 57

<211> 59

<212> DNA

<213> Artificial Sequence

<220>

<223> RPcaSp4A

<220>

<221> misc_feature

<222> (1)..(59)

<400> 57

tatatatagg atcttgctgt ctggtgtgca gtgttgatca gctggacccg tccttcacc 59 <210> 58

<211> 61

<212> DNA

<213> Artificial Sequence

<220>

<223> RPcaSp7mA

<220>

<221> misc_feature

<222> (1) .(61) <400> 58

tatatatagg atcttgctgt ctggtgtgca gtgttgatag tcactggacc cgtccttcac 60

<210> 59

<211> 30

<212> DNA

<213> Artificial Sequence

<220>

<223> RPcaSp7nePartA <220>

<221> misc_feature

<222> (1)..(30)

<400> 59

tctactcaac aagtccctgg cgacaagtcg 30

<210> 60

<2\ \> 30

<212> DNA

<213> Artificial Sequence

<220>

<223> FPcaSp7nePartB

<220>

<221> misc feature <222> (1)..(30)

<400> 60

acgacttgtc gccagggact tgttgagtag 30

<210> 61

<2\ \> 29

<212> DNA

<213> Artificial Sequence

<220>

<223> RPcaSp7ne

<220>

<221> misc_feature

<222> (1)..(29)

<400> 61

aagagactgg acccgtcctt caccatttc

<210> 62

<2\ \> 122

<212> RNA

<213> Artificial Sequence

<220>

<223> caSp7NE

<220>

<221> misc feature

<222> (1)..(122)

<400> 62

ggaagagaug gcgacuaaaa cgacuugucg ccagggacuu guugaguaga gugugagcuc 60 cguaacuagu cgcgucuucg gacgcgacug aaugaaaugg ugaaggacgg guccagucuc 120

<210> 63

<2\ \> 143

<212> DNA

<213> Artificial Sequence <220>

<223> Duplex IVT DNA positive strand

<220>

<221> misc feature

<222> (1)..(143)

<400> 63

attcgccctt taatacgact cactatagga agagatggcg actaaaacga cttgtcgcca gcttgttgag tagagtgtga gctccgtaac tagtcgcgtc ttcggacgcg actgaatgaa atggtgaagg acgggtccag ctg 143

<210> 64

<211> 143

<212> DNA

<213> Artificial Sequence

<220>

<223> Duplex IVT DNA negative strand

<220>

<221> misc_feature

<222> (1)..(143)

<400> 64

taagcgggaa attatgctga gtgatatcct tctctaccgc tgattttgct gaacagcggt 60 cgaacaactc atctcacact cgaggcattg atcagcgcag aagcctgcgc tgacttactt 120 taccacttcc tgcccaggtc gac 143