throbber
Molecular Biology Problem Solver: A Laboratory Guide. Edited by Alan S. Gerstein
`Copyright © 2001 by Wiley-Liss, Inc.
`ISBNs: 0-471-37972-7 (Paper); 0-471-22390-5 (Electronic)
`
`14
`
`Nucleic Acid Hybridization
`
`Sibylle Herzer and David F. Englert
`
`Planning a Hybridization Experiment . . . . . . . . . . . . . . . . . . . . . 401
`The Importance of Patience . . . . . . . . . . . . . . . . . . . . . . . . . . 401
`What Are Your Most Essential Needs?
`. . . . . . . . . . . . . . . . . 401
`Visualize Your Particular Hybridization Event . . . . . . . . . . . . 401
`Is a More Sensitive Detection System Always Better? . . . . . 403
`What Can You Conclude from Commercial
`Sensitivity Data?
`. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 403
`Labeling Issues . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 403
`Which Labeling Strategy Is Most Appropriate for
`Your Situation? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 403
`What Criteria Could You Consider When Selecting
`a Label? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 405
`Radioactive and Nonradioactive Labeling Strategies
`Compared . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 409
`What Are the Criteria for Considering Direct over
`Indirect Nonradioactive Labeling Strategies? . . . . . . . . . . . 410
`What Is the Storage Stability of Labeled Probes? . . . . . . . . . 411
`Should the Probe Previously Used within the
`Hybridization Solution of an Earlier Experiment Be
`Applied in a New Experiment? . . . . . . . . . . . . . . . . . . . . . . 412
`How Should a Probe Be Denatured for Reuse? . . . . . . . . . . 412
`Is It Essential to Determine the Incorporation Efficiency
`of Every Labeling Reaction? . . . . . . . . . . . . . . . . . . . . . . . . . 412
`Is It Necessary to Purify Every Probe? . . . . . . . . . . . . . . . . . 413
`
`399
`
`Enzo Exhibit 2111
`BD v. Enzo
`Case IPR2017-00181
`
`Exhibit 2111 Page 1
`
`

`

`Hybridization Membranes and Supports . . . . . . . . . . . . . . . . . . 413
`What Are the Criteria for Selecting a Support for Your
`Hybridization Experiment? . . . . . . . . . . . . . . . . . . . . . . . . . . 413
`Which Membrane Is Most Appropriate for Quantitative
`Experiments? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 417
`What Are the Indicators of a Functional Membrane? . . . . . 417
`Can Nylon and Nitrocellulose Membranes Be
`Sterilized? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 417
`Nucleic Acid Transfer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 418
`What Issues Affect the Transfer of Nucleic Acid from
`Agarose Gels? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 418
`Should Membranes Be Wet or Dry Prior to Use? . . . . . . . . 420
`What Can You Do to Optimize the Performance of
`Colony and Plaque Transfers? . . . . . . . . . . . . . . . . . . . . . . . 421
`Crosslinking Nucleic Acids . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 422
`What Are the Strengths and Limitations of Common
`Crosslinking Strategies? . . . . . . . . . . . . . . . . . . . . . . . . . . . . 422
`What Are the Main Problems of Crosslinking? . . . . . . . . . . . 423
`What’s the Shelf Life of a Membrane Whose Target DNA
`Has Been Crosslinked? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 423
`The Hybridization Reaction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 424
`How Do You Determine an Optimal Hybridization
`Temperature? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 424
`What Range of Probe Concentration Is Acceptable? . . . . . . 425
`What Are Appropriate Pre-hybridization Times? . . . . . . . . . 426
`How Do You Determine Suitable Hybridization Times? . . . 426
`What Are the Functions of the Components of a Typical
`Hybridization Buffer? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 427
`What to Do before You Develop a New Hybridization
`Buffer Formulation? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 430
`What Is the Shelf Life of Hybridization Buffers and
`Components? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 431
`What Is the Best Strategy for Hybridization of Multiple
`Membranes? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 432
`Is Stripping Always Required Prior to Reprobing? . . . . . . . . 432
`What Are the Main Points to Consider When Reprobing
`Blots?
`. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 433
`How Do You Optimize Wash Steps? . . . . . . . . . . . . . . . . . . . 434
`How Do You Select the Proper Hybridization
`Equipment? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 435
`Detection by Autoradiography Film . . . . . . . . . . . . . . . . . . . . . . 436
`How Does an Autoradiography Film Function? . . . . . . . . . . . 436
`What Are the Criteria for Selecting Autoradiogaphy
`Film? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 438
`Why Expose Film to a Blot at -70°C?
`. . . . . . . . . . . . . . . . . 440
`
`400
`
`Herzer and Englert
`
`
`Exhibit 2111 Page 2
`
`

`

`Helpful Hints When Working With Autoradiography
`Film . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 441
`Detection by Storage Phosphor Imagers . . . . . . . . . . . . . . . . . . 441
`How Do Phosphor Imagers Work? . . . . . . . . . . . . . . . . . . . . . 441
`Is a Storage Phosphor Imager Appropriate for Your
`Research Situation? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 441
`What Affects Quantitation? . . . . . . . . . . . . . . . . . . . . . . . . . . . 443
`What Should You Consider When Using Screens? . . . . . . . . 445
`How Can Problems Be Prevented? . . . . . . . . . . . . . . . . . . . . . 447
`Troubleshooting . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 448
`What Can Cause the Failure of a Hybridization
`Experiment? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 448
`Bibliography . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 453
`
`PLANNING A HYBRIDIZATION EXPERIMENT
`Hybridization experiments usually require a considerable
`investment in time and labor, with several days passing before you
`obtain results. An analysis of your needs and an appreciation for
`the nuances of your hybridization event will help you select the
`most efficient strategies and appropriate controls.
`
`The Importance of Patience
`Hybridization data are the culmination of many events, each
`with several effectors. Modification of any one effector (salt con-
`centration, temperature, probe concentration) usually impacts
`several others. Because of this complex interplay of cause and
`effect, consider an approach where every step in a hybridization
`procedure is an experiment in need of optimization. Manufac-
`turers of hybridization equipment and reagents can often pro-
`vide strategies to optimize the performance of their products.
`
`What Are Your Most Essential Needs?
`Consider your needs before you delve into the many hybridiza-
`tion options. What criteria are most crucial for your research—
`speed, cost, sensitivity, reproducibility or robustness, and qualita-
`tive or quantitative data?
`
`Visualize Your Particular Hybridization Event
`Consider the possible structures of your labeled probes and
`compare them to your target(s). Be prepared to change your label-
`ing and hybridization strategies based on your experiments.
`
`Nucleic Acid Hybridization
`
`401
`
`
`Exhibit 2111 Page 3
`
`

`

`results are unsatisfactory, a point at which it might be too late to
`determine incorporation efficiency.
`Before skipping any control steps, consider the implications.
`Minimally, measure incorporation efficiency when working with a
`new technique, a new probe, a new protocol, or a new kit. Radio-
`labeled probes need to be purified or at least Trichloroacetic acid
`(TCA) precipitated to determine labeling efficiency, as discussed
`in Chapter 7, “DNA Purification.” Determining the efficiency of
`nonradioactive labeling reactions can be more time-consuming,
`often involving dot blots and/or scanning of probe spots. Follow
`manufacturer recommendations to determine labeling efficiency
`of nonradioactive probes.
`
`Is It Necessary to Purify Every Probe?
`Unincorporated nucleotides, enzyme, crosslinking reagents,
`buffer components, and the like, may cause high backgrounds
`or interfere with downstream experiments. Hybridization experi-
`ments where the volume of the probe labeling reaction is negli-
`gible in comparison to the hybridization buffer volume do not
`always require probe cleanup. If you prefer to minimize these
`risks, purify the probe away from the reaction components.
`While there are some labeling procedures (i.e., probes gener-
`ated by random primer labeling with 32P-dCTP), where unpurified
`probe can produce little or no background (Amersham Pharma-
`cia Biotech, unpublished observations), such ideal results can’t be
`guaranteed for every probe. When background is problematic,
`researchers have the option to repurify the probe preparation.
`Admittedly, this approach wouldn’t be of much use if the experi-
`ment producing the background problem required a five day
`exposure. (Purification options are discussed in Chapter 7, “DNA
`Purification.”)
`
`HYBRIDIZATION MEMBRANES AND SUPPORTS
`What Are the Criteria for Selecting a Support for Your
`Hybridization Experiment?
`Beyond the information listed below and your personal experi-
`ence, the most reliable approach to determine if a membrane can
`be used in your application is to ask the manufacturer for appli-
`cation and or quality control data. Whether a new membrane for-
`mulation will provide you with superior results is a matter that can
`usually be decided only at the bench, and the results can vary for
`different sets of targets, probes, and detection strategies.
`
`Nucleic Acid Hybridization
`
`413
`
`
`Exhibit 2111 Page 4
`
`

`

`Physical Strength
`Nitrocellulose remains popular for low to medium sensitivity
`(i.e., screening libraries) applications and for situations that
`require minimal handling. The greater mechanical strength of
`nylon makes it superior for situations that require repeated
`manipulation of your blot. Nylon filters may be probed 10 times
`or more (Krueger and Williams, 1995; Li, Parker, and Kowalik,
`1987). Even though nitrocellulose may be used more than once,
`brittleness, loss of noncovalently bound target during stripping,
`and decreased stability in harsh stripping solutions make nitro-
`cellulose a lesser choice for reusable blots. Glass supports and
`chips can be stripped, but stripping efficiency and aging of target
`on these supports may impair reuse of more than two to three
`cycles of stripping and reprobing. Supported nitrocellulose is stur-
`dier and easier to handle than pure nitrocellulose, but remember
`that it needs to be used in the proper orientation.
`
`Binding Capacity
`Nylon and PVDF (polyvinylidene difluoride) membranes bind
`significantly more nucleic acid than nitrocelluose; hence they can
`generate stronger signals after hybridization. Nucleic acids can be
`covalently attached to nylon but not to nitrocellulose, as discussed
`below. Positively charged nylon offers the highest binding capac-
`ities. As is the case with detection systems of greater sensitivity,
`the greater binding capacity of positively charged membranes
`could increase the risk of background signal. However, optimiza-
`tion of hybridization conditions, such as probe concentration and
`hybridization buffer composition, will usually prevent background
`problems. If such optimization steps do not prevent background,
`a switch to another membrane type, such as to a neutral nylon
`membrane, might be required. If your signal is too low, try a pos-
`itively charged nylon membrane. Positively charged nylon is often
`chosen for nonradioactive applications to ensure maximum signal
`strength. The quantity of positive charges (and potential for back-
`ground) can vary by 10-fold between manufacturers. The lower
`binding capacity of nitrocellulose decreases the likelihood of
`background problems under conditions that generate a detectable
`signal.
`
`Thickness
`Most membranes are approximately 100 to 150mm thick. Thick-
`ness influences the amount of buffer required per square
`
`414
`
`Herzer and Englert
`
`
`Exhibit 2111 Page 5
`
`

`

`centimeter. Thicker membranes soak up more buffer, wet more
`slowly, and dry application of thicker filters to the surface of a gel
`can be more difficult.
`
`Pore Size
`Pore size limits the size of the smallest fragment that can be
`bound and fixed onto a membrane, but bear in mind that pore size
`is an average value. In general, 0.45mm micron pore sizes can bind
`oligonucleotides down to 50 bases in length, but the more common
`working limit is 100 to 150 nucleotides or base pairs. Membranes
`comprised of 0.22mm micron pores are recommended for work
`with the smallest single- and double-stranded DNA fragments.
`Custom manufacturing of membranes with 0.1mm pore size is also
`available. Table 14.1 compares membrane characteristics.
`
`Specialized Application
`Microarrays
`Glass slides stand apart from membrane supports because glass
`allows for covalent attachment of oriented nucleic acid, is non-
`porous and offers low autofluorescence. On a nonporous support,
`buffer volumes can be kept low, which decreases cost and allows
`increased probe concentration. Unlike nylon and nitrocellulose
`membranes, background isn’t problematic under these aggressive
`hybridization conditions. Probes are labeled with different dyes
`and allow detection of multiple targets in a single hybridization
`experiment; nylon arrays are often restricted to serial or parallel
`hybridization, although examples of simultaneous detection on
`nylon membranes can be found in the literature. (Some references
`for multiple probes on nylon are Kondo et al. (1998), Holtke et al.
`(1992), and Bertucci et al. (1999).) These features make glass slides
`ideal for nonradioactive detection in micro arrays.
`
`Macroarrays
`Background problems, high buffer volumes, and hence cost,
`limit the usefulness of nonradioactive labels for macroarrays
`on nylon filters. Macroarrays employ thin charged or uncharged
`nylon membranes to reduce buffer consumption but suffer from
`low sensitivity due to the high autofluorescence of nylon. Stronger
`signals derived from enzyme-substrate driven signal amplification
`compromise resolution and quantitation. Radioactive labels such
`as 33P are preferred for macroarrays (Moichi et al., 1999; Yano
`et al., 2000; Eickhoff et al., 2000).
`
`Nucleic Acid Hybridization
`
`415
`
`
`Exhibit 2111 Page 6
`
`

`

`optimzed properly
`backgrounds if not
`
`Can give high
`
`detection systems
`nonradioactive
`interfere with
`membranes can
`supercharged
`systems,but some
`most nonradioactive
`usually optimal for
`
`Highest sensitivity;
`
`As for uncharged
`
`nylon
`
`signal/retention
`Poor specificity of
`
`all nylon membranes
`Lowest background of
`
`As for uncharged
`
`nylon
`
`charged nylon
`problematic with
`background is
`inappropriate and
`Where nitrocelluose
`
`often lowers signal
`many stains,staining
`
`Irreversibly binds
`
`applications
`nucleic acid
`mixed protein/
`applications,some
`some nonradioactive
`
`difficult to optimize
`that might be more
`documented method
`not so well
`
`Fairly new and hence
`
`As for unsupported
`
`nitrocellulose
`
`As for unsupported
`
`nitrocellulose
`
`applications
`sensitivity
`screens,low
`plaque lifts,library
`Radioactive colony/
`Recommended Use
`
`for RNA
`binds proteins,not
`systems because it
`not good for nonrad
`low binding capacity
`handle,flammable,
`
`More difficult to
`Limitations
`
`brittleness
`drawbacks of
`nitrocellulose
`sensitivity without
`capacity and hence
`Intermediate binding
`
`block
`background,easy to
`
`Low cost,low
`
`block
`background,easy to
`
`Low cost,low
`
`Benefits
`
`hydrolyze the target
`conditions that don’t
`Up to 10 times if under
`
`water,formamide
`Yes,strip with SDS,
`
`Possibly 2–6 times via
`
`gentle conditions
`
`Strip or Reprobe
`
`and physical strength
`solvent resistance
`membrane of good
`hygroscopic
`
`Hydrophilic,
`
`and physical strength
`solvent resistance
`membrane of good
`hygroscopic
`
`Hydrophilic,
`
`charged
`positively
`
`Nylon
`
`charged
`negatively
`
`Nylon
`
`and physical strength
`solvent resistance
`membrane of good
`hygroscopic
`
`Nylon neutralHydrophilic,
`
`stability <135C
`with a thermal
`physical strength
`solvent resistance/
`membrane of fair
`
`Hydrophobic
`
`PVDF
`
`Nitrocellulose
`
`supported
`
`NitrocelluloseHydrophilic membrane Not recommended
`Membrane
`
`physical strength
`resistance and
`of low solvent
`
`Characteristics
`
`Physical
`
`Table 14.1Characteristics of Membranes Used in Nucleic Acid Blotting Applications
`
`416
`
`Herzer and Englert
`
`
`Exhibit 2111 Page 7
`
`

`

`Which Membrane Is Most Appropriate for
`Quantitative Experiments?
`The size of the nucleic acid being transferred, the physical
`characteristics of the membrane, and the composition of transfer
`buffer affect the transfer efficiency. There is no magic formula
`guaranteeing linear transfer of all nucleic acids at all times.
`Linearity of transfer needs to be tested empirically with dilution
`series of nucleic acid molecular weight markers.
`
`What Are the Indicators of a Functional Membrane?
`Membranes will record every fingerprint, drop of powder,
`knick, and crease. Always handle membranes with plastic forceps
`and powder-free gloves.
`Membranes should be dry and uniform in appearance. They
`should be wrinkle- and scratch-free since mechanical damage
`may lead to background problems in these affected areas. Mem-
`branes should wet evenly and quickly. If membranes do appear
`blotchy or spotty, or seem to have different colors, it is best not to
`use them. Membranes are hygroscopic, light sensitive, and easily
`damaged, but as long as membranes are properly stored, may
`remain functional for years. Please note that manufacturers only
`guarantee potency for shorter time periods, usually six to twelve
`months. If the vitality of the membrane is in doubt, a quick dot
`blot or test of the binding capacity may help. Manufacturers can
`provide guidelines for assessing binding capacity. Including an
`untreated, target-free piece of membrane to evaluate background
`in a given hybridization buffer or wash system can help to
`troubleshoot background problems.
`
`Can Nylon and Nitrocellulose Membranes Be Sterilized?
`Researchers performing colony hyrbidizations often ask about
`membrane sterilization. While membranes might not be supplied
`guaranteed to be sterile, they are typically produced and packaged
`with extreme care, minimizing the likelihood of contamination.
`Theoretically it is possible to autoclave membranes, but cycles
`should be very short (two minutes at 121°C in liquid cycle). Note
`that such short durations cannot guarantee sterility. Membranes
`should be removed as soon as the autoclave comes down to a safe
`temperature, and dried at room temperature. Multiple membranes
`should be separated by single sheets of Whatman paper. Note that
`filters may turn brown, become brittle, may shrink and warp and
`become difficult to align with plates, but this does not interfere
`with probe hybridization.
`
`Nucleic Acid Hybridization
`
`417
`
`
`Exhibit 2111 Page 8
`
`

`

`One reference cited decreased shelf lives for storage at room
`temperature (Giusti and Budowle, 1992). Blots maintained
`dry (desiccant for long-term storage), dark, and protected from
`mechanical damage may be stored safely for 6 to 12 months.
`
`THE HYBRIDIZATION REACTION
`The hybridization step is central to any nucleic acid detection
`technique. Choices of buffer, temperature, and time are never
`trivial because these effectors in combination with membrane,
`probe, label, and target form a complex network of cause and
`effect. Determining the best conditions for your experiment will
`always require a series of optimization experiments; there is no
`magic formula. The role of the effectors of hybridization, recom-
`mended starting levels, and strategies to optimize them will be the
`focus of this section. Readers interested in greater detail on the
`intricacies and interplay of events within hybridization reactions
`are directed to Anderson (1999), Gilmartin (1996), Thomou and
`Katsanos (1976), Ivanov et al. (1978), and Pearson, Davidson, and
`Britten (1977).
`
`How Do You Determine an Optimal Hybridization
`Temperature?
`Hybridization temperature depends on melting temper-
`ature (Tm) of the probe, buffer composition, and the nature of the
`target:hybrid complex. Formulas to calculate the Tm of oligos,
`RNA, DNA, RNA-DNA, and PNA-DNA hybrids have been de-
`scribed (Breslauer et al., 1986; Schwarz, Robinson, and Butler,
`1999; Marathias et al., 2000). Software that calculates Tm is
`described by Dieffenbach and Dveksler (1995).
`The effects of labels on melting temperatures should be taken
`into consideration. While some claim little effect of tags as large
`as horseradish peroxidase on hybrid stability/Tm (Pollard-Knight
`et al., 1990a), others observed Tm changes with smaller base mod-
`ifications (Pearlman and Kollman, 1990). It will have to suffice that
`nonradioactive tags may alter the hybridization characteristics of
`probes and that empiric determination of Tm may be quicker than
`developing a formula to accurately predict hybridization behav-
`ior of tagged probes. Hybridization temperatures should also take
`into account the impact of hybridization temperature on label sta-
`bility. Alkaline phosphatase is more stable at elevated tempera-
`tures than horseradish peroxidase. Thermostable versions of
`enzymes or addition of thermal stabilizer such as trehalose
`
`424
`
`Herzer and Englert
`
`
`Exhibit 2111 Page 9
`
`

`

`(Carninci et al., 1998) may provide alternatives to hybridization
`at low temperatures.
`When switching from a DNA to an RNA probe, hybridization
`temperatures can be increased due to the increased Tm of RNA-
`DNA heteroduplexes. Because of concerns about instability of
`RNA at elevated temperatures, an alternative approach with
`RNA probes is the use of a denaturing formamide or urea buffer
`that allows hybridization at lower temperature.
`A good starting point for inorganic (nondenaturing) buffers are
`hybridization temperatures of 50 to 65°C for DNA applications
`and 55 to 70°C for RNA applications. Formamide buffers offer
`hybridization at temperatures as low as 30°C, but temperatures
`between 37 and 45°C are more common. Enzyme-linked probes
`should be used at the lowest possible temperature to guarantee
`enzyme stability.
`After hybridization and detection has been performed at the
`initially selected hybridization temperature, adjustments may be
`required to improve upon the results. A hybridization tempera-
`ture that is too low will manifest itself as a high nonspecific back-
`ground. The degree by which the temperature of subsequent
`hybridizations should be adjusted will depend on other criteria
`discussed throughout this chapter (GC content of the probe and
`template, RNA vs. DNA probe, etc.), and thus hybridization tem-
`perature can’t be exactly predicted. Most hybridization protocols
`employ temperatures of 37°C, 42°C, 50°C, 55°C, 60°C, 65°C, and
`68°C.
`Note that sometimes a clean, strong, specific signal that is totally
`free of nonspecific background cannot be obtained. Background
`reduction, especially through the use of increased hybridization
`temperatures, will result in the decrease of specific hybridization
`signal as well. There is often a trade-off between specific signal
`strength and background levels. You may need to define in each
`experiment what amount of background is acceptable to obtain
`the necessary level of specific hybridization signal. If the results
`are not acceptable, the experiment might have to be redesigned.
`
`What Range of Probe Concentration Is Acceptable?
`Probe concentration is application dependent. It will vary with
`buffer composition, anticipated amount of target, probe length
`and sequence, and the labeling technique used.
`Background and signal correlate directly to probe concentra-
`tion. If less probe than target is present, then the accuracy of band
`quantities is questionable.
`
`Nucleic Acid Hybridization
`
`425
`
`
`Exhibit 2111 Page 10
`
`

`

`In the absence of rate-accelerating “fast” hybridization buffers,
`probe concentration is typically 5 to 10ng/ml of buffer. Another
`convention is to apply 2 to 5 million counts/ml of hybridization
`buffer, which may add up to more than 10ng/ml if the probe
`was end-labeled, as compared to a random primer-generated
`probe. The use of rate accelerators or “fast” hybridization buffers
`requires a reduction in probe concentration to levels of 0.1 to
`5ng/ml of hybridization buffer.
`Another approach to select probe concentration is based on the
`amount of target. A greater than 20¥ excess of probe over target
`is required in filter hybridization (Anderson, 1999). Solution
`hybridization may not require excess amounts for qualitative
`experiments. To determine if probe is actually present in excess
`over target, perform replicate dot or slot blots containing a dilu-
`tion series of immobilized target and varying amounts of input
`probe (Anderson, 1999). If probe is present in excess, the signal
`should reflect the relative ratios of the different concentrations of
`target. If you do not observe a proportional relationship between
`target concentration and specific hybridization signal at any of
`the probe concentrations used, you may need to increase your
`probe concentration even higher. Probe concentration cannot be
`increased indefinitely; a high background signal will eventually
`appear.
`
`What Are Appropriate Pre-hybridization Times?
`Prehybridization time is also affected by the variables of
`hybridization time. For buffers without rate accelerators, prehy-
`bridization times of at least 1 to 4 hours are a good starting point.
`Some applications may afford to skip prehybridization altogether
`(Budowle and Baechtel, 1990). Buffers containing rate accelera-
`tors or volume excluders usually do not benefit from prehy-
`bridization times greater than 30 minutes.
`
`How Do You Determine Suitable Hybridization Times?
`Hybridization time depends on the kinetics of two reactions or
`events: a slow nucleation process and a fast “zippering” up. Nucle-
`ation is rate-limiting and requires proper temperature settings
`(Anderson, 1999). Once a duplex has formed (after “zippering”),
`it is very stable at temperatures below melting, given that the
`duplex is longer >50bp. Hybridizing overnight works well for a
`wide range of target or probe scenarios. If this generates a dissat-
`isfactory signal, consider the following.
`There are several variables that affect hybridization time.
`Double-stranded probes (i.e., an end-labeled 300bp fragment)
`
`426
`
`Herzer and Englert
`
`
`Exhibit 2111 Page 11
`
`

`

`require longer hybridization times than single-stranded probes
`(end-labeled oligonucleotide), because reassociation of double-
`stranded probes in solution competes with annealing events of
`probes to target. At 50% to 75% reassociation, free probe con-
`centration has dwindled to amounts that make further incubation
`futile. Hybridization time for a double-stranded probe can there-
`fore be deduced from its reassociation rate (Anderson, 1999).
`Glimartin (1996) discusses methods to predict hybridization times
`for single-stranded probes, as does Anderson (1999). Other vari-
`ables of hybridization time include probe length and complexity,
`probe concentration, reaction volume, and buffer concentration.
`Buffer
`formulations
`containing higher
`concentrations
`(≥10ng/ml) of probe and/or rate accelerators or blots with high
`target concentrations may require as little as 1 hour for hybridiza-
`tion. Prolonged hybridization in systems of increased hybridiza-
`tion rate will lead to background problems. The shortest possible
`hybridization time can be tested for by dot blot analysis. Standard
`buffers usually require hybridization times between 6 and 24
`hours. Plateauing of signal sets the upper limit for hybridization
`time. Again, optimization of hybridization time by a series of dot
`blot experiments, removed and washed at different times, is rec-
`ommended. Plaque or colony lifts may benefit from extended
`hybridization times if large numbers of filters are simultaneously
`hybridized.
`
`What Are the Functions of the Components of a Typical
`Hybridization Buffer?
`Hybridization buffers could be classified as one of two types:
`denaturing buffers, which lower the melting temperatures (and
`thus hybridization
`temperatures) of nucleic acid hybrids
`(i.e., formamide buffers), and salt/detergent based buffers, which
`require higher hybridization temperatures, such as sodium phos-
`phate buffer (as per Church and Gilbert, 1984).
`
`Denaturants
`Denaturing buffers are preferred if membrane, probe, or label
`are known to be less stable at elevated temperatures. Examples
`are the use of formamide with RNA probes and nitrocellulose
`filters, and urea buffers for use with HRP-linked nucleic acid
`probes. Imperfectly matched target:probe hybrids are hybridized
`in formamide buffers as well.
`For denaturing, 30% to 80% formamide, 3 to 6M urea, ethyl-
`ene glycol, 2 to 4M sodium perchlorate, and tertiary alkylamine
`
`Nucleic Acid Hybridization
`
`427
`
`
`Exhibit 2111 Page 12
`
`

`

`chloride salts have been used. High-quality reagents, such as
`deionized formamide, sequencing grade or higher urea, and
`reagents that are DNAse- and/or RNAse-free are critical.
`Formamide concentration can be used to manipulate stringency,
`but needs to be >20%. Hybrid formation is impaired at 20% for-
`mamide but not at 30 or 50% (Howley et al., 1979). 50% to 80%
`formamide may be added to hybridization buffers. 50% is rou-
`tinely used for filter hybridization. 80% formamide formulations
`are mostly used for in situ hybridization (ISH) where temperature
`has the greatest influence on overall stability of the fixed tissue
`and probe, and in experiments where RNA:DNA hybrid forma-
`tion is desired rather than DNA:DNA hybridization. In 80% for-
`mamide, the rate of DNA:DNA hybridization is much lower than
`RNA:DNA hybrid formation (Casey and Johnson 1977). Phos-
`phate buffers are preferred over citrate buffers in formamide
`buffers because of superior buffering strength at physiological pH.
`In short oligos 3M tetramethylammonium chloride (TMAC)
`will alter their Tm by making it solely dependent on oligonu-
`cleotide length and independent of GC content (Bains, 1994;
`Honore, Madsen, and Leffers, 1993). This property has been
`exploited to normalize sequence effects of highly degenerate
`oligos, as are used in library screens. Note that some specificity
`may be lost.
`
`Salts
`Binding Effects
`Hybrid formation must overcome electrostatic repulsion forces
`between the negatively charged phosphate backbones of the
`probe and target. Salt cations, typically sodium or potassium, will
`counteract these repulsion effects. The appropriate salt concen-
`tration is an absolute requirement for nucleic acid hybrid
`formation.
`Hybrid stability and sodium chloride concentration correlate in
`a linear relation in a range of up to 1.2M. Stability may be
`increased by adding salt up to a final concentration of 1.2M, or
`decreased by lowering the amount of sodium chloride. It is the
`actual concentration of free cations, or sodium, that influences sta-
`bility (Nakano et al., 1999; Spink and Chaires, 1999). Final con-
`centrations of 5 to 6¥ SSC or 5 to 6¥ SSPE (Sambrook, Fritsch,
`and Maniatis, 1989), equivalent to approximately 0.8 to 0.9M
`sodium chloride and 80 to 90mM citrate buffer or 50mM sodium
`phosphate buffer, are common starting points for hybridization
`buffers. At 0.4 to 1.0M sodium chloride, the hybridization rate of
`
`428
`
`Herzer and Englert
`
`
`Exhibit 2111 Page 13
`
`

`

`DNA:DNA hybrids is increased twofold. Below 0.4M sodium
`chloride, hybridization rate drops dramatically (Wood et al., 1985).
`RNA:DNA and RNA:RNA hybrids require slightly lower salt
`concentrations of 0.18 to 1.0M to increase hybridization by
`twofold.
`
`pH Effects
`Incorrect pH may impair hybrid formation because the charge
`of the nucleic acid phosphate backbone is pH dependent. The pH
`is typically adjusted to 7.0 or from 7.2 to 7.4 for hybridization
`experiments. Increasing concentrations of buffer substances may
`also affect stringency. EDTA is sometimes added to 1 to 2mM to
`protect against nuclease degradation.
`
`Detergent
`Detergents prevent nonspecific binding caused by ionic or
`hydrophobic interaction with hydrophobic sites on the membrane
`and promote even wetting of membranes. 1% to 7% SDS, 0.05%
`to 0.1% Tween-20, 0.1% N-lauroylsarcosine, or Nonidet P-40 have
`been used in hybridization buffers. Higher concentrations of SDS
`(7%) seem to reduce background problems by acting

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket