10
BNL-113700-2017-JA Micro-differential scanning calorimeter for liquid biological samples Shuyu Wang, Shifeng Yu, Michael S. Siedler, Peter M. Ihnat, Dana I. Filoti, Ming Lu, Lei Zuo Submitted to the Review of Scientific Instruments October 2016 Center for Functional Nanomaterials Brookhaven National Laboratory U.S. Department of Energy USDOE Office of Science (SC), Basic Energy Sciences (SC-22) Notice: This manuscript has been authored by employees of Brookhaven Science Associates, LLC under Contract No. DE- SC0012704 with the U.S. Department of Energy. The publisher by accepting the manuscript for publication acknowledges that the United States Government retains a non-exclusive, paid-up, irrevocable, world-wide license to publish or reproduce the published form of this manuscript, or allow others to do so, for United States Government purposes.

Micro-differential scanning calorimeter for liquid ...for protein liquid sample and the other for reference buffer liquid sample. The chamber had a volume of 1µL (height 200µm, radius

  • Upload
    others

  • View
    5

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Micro-differential scanning calorimeter for liquid ...for protein liquid sample and the other for reference buffer liquid sample. The chamber had a volume of 1µL (height 200µm, radius

BNL-113700-2017-JA

Micro-differential scanning calorimeter

for liquid biological samples

Shuyu Wang, Shifeng Yu, Michael S. Siedler,

Peter M. Ihnat, Dana I. Filoti, Ming Lu, Lei Zuo

Submitted to the Review of Scientific Instruments

October 2016

Center for Functional Nanomaterials

Brookhaven National Laboratory

U.S. Department of Energy USDOE Office of Science (SC), Basic Energy Sciences (SC-22)

Notice: This manuscript has been authored by employees of Brookhaven Science Associates, LLC under

Contract No. DE- SC0012704 with the U.S. Department of Energy. The publisher by accepting the

manuscript for publication acknowledges that the United States Government retains a non-exclusive, paid-up,

irrevocable, world-wide license to publish or reproduce the published form of this manuscript, or allow others

to do so, for United States Government purposes.

Page 2: Micro-differential scanning calorimeter for liquid ...for protein liquid sample and the other for reference buffer liquid sample. The chamber had a volume of 1µL (height 200µm, radius

DISCLAIMER

This report was prepared as an account of work sponsored by an agency of the

United States Government. Neither the United States Government nor any

agency thereof, nor any of their employees, nor any of their contractors,

subcontractors, or their employees, makes any warranty, express or implied, or

assumes any legal liability or responsibility for the accuracy, completeness, or any

third party’s use or the results of such use of any information, apparatus, product,

or process disclosed, or represents that its use would not infringe privately owned

rights. Reference herein to any specific commercial product, process, or service

by trade name, trademark, manufacturer, or otherwise, does not necessarily

constitute or imply its endorsement, recommendation, or favoring by the United

States Government or any agency thereof or its contractors or subcontractors.

The views and opinions of authors expressed herein do not necessarily state or

reflect those of the United States Government or any agency thereof.

Page 3: Micro-differential scanning calorimeter for liquid ...for protein liquid sample and the other for reference buffer liquid sample. The chamber had a volume of 1µL (height 200µm, radius

Micro-differential scanning calorimeter for liquid biological

samples

Shuyu Wang1, Shifeng Yu2, Michael S. Siedler3, Peter M. Ihnat4, Dana I. Filoti4, Ming Lu5, Lei Zuo2*

1 Department of Mechanical Engineering, Stony Brook University, Stony Brook, NY 11794, USA

2 Department of Mechanical Engineering, Virginia Tech, Blacksburg, VA, 24061, USA

3AbbVie, Deutschland, 67061 Ludwigshafen, Germany

4 AbbVie Bioresearch Center, Worcester, MA 01605, USA

5Center for Functional Nanomaterials, Brookhaven National Laboratory, Upton, NY 11973, USA

*corresponding author: [email protected]

Abstract: We developed an ultrasensitive micro-DSC (differential scanning calorimeter) for liquid

protein samples characterization. This design integrated vanadium oxide thermistors and flexible polymer

substrates with microfluidics chambers to achieve high sensitivity (6V/W), low thermal conductivity

(0.7mW/K), high power resolutions (40nW) and well-defined liquid volume (1µL) calorimeter sensor in a

compact and cost-effective way. We further demonstrated the performance of the sensor with lysozyme

unfolding. The measured transition temperature and enthalpy change were in accordance with the previous

literature data. This micro-DSC could potentially raise the prospect of high-throughput biochemical

measurement by parallel operation with miniaturized sample consumption.

Keywords: microcalorimeter, differential scanning calorimeter, protein unfolding, flexible substrate,

microfluidics, MEMS (micro electro-mechanical systems)

Introduction:

Kinetic processes are playing a critical role in

physical transformations, such as glass transition,

and in macromolecule transformations, such as

protein denaturation. Understanding the effects of

external influences on the transformation may

provide valuable insights into the process[1]. The

first step to such understanding is to detect the

reaction accurately in the real time. Calorimetry

is the technique that has been widely used to

study such kinetic process and thermodynamic

properties of the materials or biological system[2].

Among several different operation modes of

calorimetry, differential scanning calorimetry

(DSC) is a well-known dynamic calorimetric

method. It imposes a temperature ramp

continuously and measures heat capacity change

of the material (both in liquid and solid forms).

For the past decades, the micro/nanofabrication

technology has been enabling calorimeters to be

smaller, faster and more accurate. As a result, the

conventional bench-top scale instruments were

miniaturized into microelectromechanical sensor

systems and some may integrate microfluidics

systems [3-5]. These miniaturized calorimeters

were often called micro/nanocalorimeter or chip

calorimeters. Normally, they were based on the

thin films, either made of silicon/silicon nitride [6,

7] or polymers [5, 8]. These chip calorimeters

were capable of measuring samples with very

small thermal mass due to significantly reduced

addenda[9] and may measure the energy on the

Page 4: Micro-differential scanning calorimeter for liquid ...for protein liquid sample and the other for reference buffer liquid sample. The chamber had a volume of 1µL (height 200µm, radius

order of nanojoule[4] accounted from the superb

thermal insulation. We have already seen chip

calorimeters’ application to study thin films [8],

polymer [10], cells [11],protein [12] and so on.

Such studies in biological systems can be very

crucial to the pharmaceutical industry as several

critical steps in the drug discovery process are

associated with calorimeters. Currently, the long

measurement time of the conventional scanning

calorimeters often requires years to finish

thousands of the chemical compounds[13] and

the liquid volume of protein sample required is

very large(500µL-2mL). Chip calorimeter might

be a potential solution since they use very small

amount of protein sample and can easily provide

parallel operations to achieve high-throughput

[12]. However, chip calorimeter as DSC to study

biological system still remains very scarce in

academics compare to micro isothermal titration

calorimeter (ITC) [4, 12, 14, 15] due to

technology difficulties to develop them[13].

Several different configurations of micro-DSC

for liquid sample appeared in recent years. Olson

[2] and Youssef[16] studied the scanning

calorimeter with liquid samples, yet their uses are

limited to measure latent heat of transformation

due to evaporation. Yao [17] built an AC

calorimeter with glass capillary tube that

consumed 10µL liquid. The device (heat capacity

resolution ±300nJ/K) successfully tested the

lysozyme solution’s heat capacity when

denaturing. Garden’s [18] scanning AC

calorimeter has also measured 5µL biological

liquid sample’s heat capacity with high

resolution(±150nJ/K). They used a gasket and

polyimide membranes (supported by micro-posts)

to construct the chamber. However, their design

suffered from large thermal conduction

(30mW/K) which limited the sensitivity. Lee[19]

constructed an enclosed parylene chamber (1µL)

to tested DNA and hydrocarbon yet the signal-to-

noise ratio is too low. Lin’s group successfully

integrated microfluidic systems in the DSC using

polydimethylsiloxane (PDMS) microfluidics

chamber (1.2µL) and SU8 diaphragm to and

reached high detection limit (30nW)[20]. They

further developed AC-DSC[5] and changed

substrates to flexible polyimide substrates. These

devices reached very high performance

(sensitivity 4-8V/W) when demonstrating with

protein samples[21]. Yet, the thermopiles

deposited on the flexible substrate could not

determine absolute temperature, and therefore the

transition temperature might not be accurately

measured.

In this paper, we reported the fabrication and

operation of a micro-DSC for liquid biological

samples which can address the above problems.

We showed how to make a flexible calorimeter

by microscale fabrication technology and

integrate it with PDMS microfluidics chamber

(1µL). Due to the highly temperature-sensitive

vanadium oxide thermistors and superb thermal

insulation of polyimide thin films, the device had

proven to be very sensitive to thermal events.

This enabled the sensor to be used for protein

interaction detection at low cost and disposable

way. This technology could readily scale up to

array format to apply in high-throughput drug

discovery.

Theory

Virtually all DSC measures the temperature

difference between the sample and reference

region since this temperature difference can be

converted proportionally to heat flow rate

difference based on a simple linear model. This

conversion is the zeroth approximation, and more

accurate conversions can use higher-order

approximations[22].

Within one chamber, the differential heat

equation can be described as Eq.1, where Cp is the

heat capacity, G is the heat conduction, and P is

power or heat flow.

0( ( ) ) ( )p

dTC G T t T P t

dt (1)

The time constant τ is determined by 𝐶𝑝

𝐺 . When

time constant is much smaller than the

biomolecular thermal event (time constant of

guanidine induced unfolding is on the order of

Page 5: Micro-differential scanning calorimeter for liquid ...for protein liquid sample and the other for reference buffer liquid sample. The chamber had a volume of 1µL (height 200µm, radius

20-50s[17]), the process can be considered as in

the steady state. Therefore, ∆T=Ts-Tr can be

related to ∆P=Ps-Pr with a linear relationship

∆P=G∆T. Using temperature sensors to generate

voltage signal ∆U proportional to the ∆T, we can

have

∆𝑈 = 𝑆∆𝑃 (2)

where S is the sensitivity of the calorimeter.

Consequently, the thermal insulation of the

chamber (G) and temperature sensor’s sensitivity

can determine the device’s sensitivity. Based on

the mathematical model, the heat capacity

difference between the sample and reference

solution ∆C=Cs-Cr can be obtained from Eq3

where is the scanning rate of the calorimeter.

∆𝐶 =∆𝑃

[20] (3)

Since the differentiation method used in the DSC

measurement, the symmetry of the sample and

reference can be crucial to the final results.

Device description and fabrication

Figure 1and Figure 2 showed the 3D schematic

view and the cross section view of the

microcalorimeter respectively. The device

consisted of two parts: the PDMS microfluidic

chamber and the flexible calorimeter sensor.

They were fabricated separately and later bonded

together to formulate the whole device. There

were two identical microfluidic chambers: one

for protein liquid sample and the other for

reference buffer liquid sample. The chamber had

a volume of 1µL (height 200µm, radius 1.25mm)

and was surrounded by air gaps to reduce the

thermal mass of the close chamber and increase

the thermal insulation. We cast the liquid PDMS

onto the master pattern made from SU8 100

photoresist and then solidified it by soft baking.

Last, we drilled the tiny holes for inlet and outlet

of the chambers. The microfluidic chamber could

serve to prevent liquid evaporation and provide

well-defined liquid volume.

The flexible calorimeter sensor used polyimide

substrate (7µm) and integrates the thermistors

and the microheaters for sensing and calibration.

ANSYS simulation was used to assist the pattern

design of the microheater: the widths of the heater

in each circle were finely tuned to reach the

maximum temperature uniformity while heating.

The Au traces (width and gap were 10µm) were

the metallization features and served for electrical

interconnection of the thermistor layer. This

design was amount to parallel connection of

multiple resistors and could significantly lower

the resistance of the thermistor.

We spun coat PI 2611(HD microsystem)

polyimide on a silicon wafer and ramping

annealed it from room temperature to 350oC to

make it chemical inert. The solidified polymer

layer would have very low adhesion with the

silicon wafer but enough to bond them together

during fabrication so that easy separation of them

could be achieved (Figure 3(b)). We fabricated

the Au microheaters (Figure 3 (c)), the polyimide

dielectric layer, sputtered vanadium oxide

thermistors, Au traces for thermistor (Figure

3(d)), and the polyimide moisture protection

layer sequentially. The 100nm vanadium oxide

layer was deposited by DC sputter and Au trace

was deposited by e-beam evaporation. In order to

enhance the bonding strength of the flexible

sensor and microfluidics chamber, a thin layer of

PDMS diluted with toluene would be spun coated

before peeling off from the silicon substrate.

After oxygen plasma treatment for 30s under

20mTorr, the microfluidic chamber and flexible

sensor would be bonded together. Figure 3 (a)

showed the fabricated device after bonding.

Page 6: Micro-differential scanning calorimeter for liquid ...for protein liquid sample and the other for reference buffer liquid sample. The chamber had a volume of 1µL (height 200µm, radius

Figure 1.Schematics of the micro-DSC, showing the

overall structure

Figure 2. Cross-section view of the micro-DSC design

Figure 3. (a) Fabricated micro-DSC device (b) flexible

substrate peeling off from the silicon wafer (c) Optical

microscopy image of the heater (d) thermistor metal trace

Experimental methods

The electrical circuit connection was illustrated in

Figure 4. The two thermistors for sample and

reference liquid and two outside variable resistors

were connected as a Wheatstone bridge in order

to provide common mode rejection. Such bridge

circuit could pick up the temperature difference

between the sample and reference side. The

voltage output of the bridge circuit represented

the temperature difference between the sample

and reference chamber. We used the lock-in

amplifier (Stanford SR830) to provide the voltage

input (V+) and differentiate the voltage output

from the bridge(Vout). The sourcemeter (Keithley

2636) was the power source for the microheaters

and measures the resistance change during the

thermal response test. To calibrate the resistance

of the thermistors and do temperature scanning,

we placed the microDSC inside a high

temperature probe station and further connect the

wire outside to the equipment. This high

temperature probe station could raise the

temperature with a program and provide the

thermal shielding at the same time. We used

lysozyme samples (molar weight is14307Da) at a

concentration of 100mg/mL to demonstrate the

device’s performance. The buffer used was 10

Page 7: Micro-differential scanning calorimeter for liquid ...for protein liquid sample and the other for reference buffer liquid sample. The chamber had a volume of 1µL (height 200µm, radius

mM HEPES pH 7.4 and 150 mM NaCl. The

whole experimental measurement and control

were achieved with LabVIEW programs.

Figure 4. Schematic view of the micro-DSC circuit

connection. The thermistors and outside variable resistors

form a Wheatstone bridge.

Results and discussions

Before doing the measurement, we first

calibrated the vanadium oxide thermistors. The

resistance and temperature’s relation is plotted in

log scale (Figure 5). The thermistor’s resistance

is normally in the range of 15-40KΩ, meaning the

designed gold trace pattern can effectively reduce

the resistance to an appropriate range. The

temperature coefficient of resistance (TCR) can

be obtained from the slope of LnR versus

temperature as -2.8%/oC, which is one order

higher than a traditional platinum thermistor. We

also observed good uniformity of the thermistor

material; the sample and reference thermistors

could have 1-2% variation in resistance.

Although the thermistors could introduce self-

heating during measurement, the thermistor on

the reference side can counterbalance it, and such

self-heat can be further neglected during

temperature scanning. Unlike the thermopiles

that require large numbers of them to gain high

sensitivity, the high-performance thermistors can

achieve compactness with simplified

fabrication[23]. Moreover, thermistors can detect

the absolute temperature within the small

chamber, which is necessary for DSC

measurement.

Figure 5. Temperature calibration of a thermistor and

vanadium oxide resistivity measurement as a function of

temperature

We further calibrated the microcalorimeter using

electric power applied to the microheater. The

thermal response to a step function power can be

predicted by /( )( ) (1 )tP t

T t eG

. By fitting curve

of data in Figure 6 to exponential increase

function, we could extract the thermal time τ

constant to be 6 s. Due to the well-designed air

gap and the high thermal insulation of PDMS, the

affected addenda thermal mass was minimized to

be much smaller than the liquid samples

especially when small amount of heat was

applied. Therefore, the thermal conductance

(G=𝐶

τ )could be calculated to be around

0.7mW/K. Such low thermal conduction could be

comparable to the open-type calorimeters [12,

24].

Page 8: Micro-differential scanning calorimeter for liquid ...for protein liquid sample and the other for reference buffer liquid sample. The chamber had a volume of 1µL (height 200µm, radius

Figure 6. Thermal response of the device after applying a

constant power from the microheater

We could also obtain the sensitivity of the device

by varying the power dissipated in the heater. The

data showed good linearity and we measured the

slope of the plotted data (Figure 7) which

indicated the sensitivity to be 6V/W. The result

proved the device was highly sensitive to

measure a small amount of heat and it was further

used to determine the differential thermal power

between the calorimeter chambers for protein

tests.

Figure 7. Micro-DSC’s voltage output responding to a

constant differential power input from the heater. sensitivity

of the microcalorimeter is obtained from the slope

The performance about noise level of the device

was also evaluated. We filled the two chambers

with deionized water and tested it at a constant

temperature in a well-defined environment. Due

to the inevitable asymmetry of the two sides, the

measured voltage signals might drift. Figure

Figure 8 was the result after subtracting the raw

data from three order fit of the data. The rms noise

at 1 Hz bandwidth is 250nV, corresponding to

40nW of power resolution and 60µK of

temperature resolution (the results are calculated

based on the sensitivity S and thermal conduction

value G). This noisy behavior could be related to

the environment temperature fluctuation control,

the extent of the common mode rejection and the

thermistors’ Johnson noise.

Figure 8 noise measurement of the micro-DSC at a constant

temperature (raw data subtracted from three order fit)

We demonstrated the micro-DSC’s performance

with the lysozyme protein (100mg/mL) for

consecutively two times. We raised the

temperature of the chamber from 25oC to 80oC at

the rate of 7.5 oC /min. After baseline subtraction,

the measured voltage outputs from the

Wheatstone bridge showed the typical DSC curve

of protein denaturation. We could further obtain

the partial specific heat capacity of lysozyme as a

function of temperature (Figure 9) based on Eq3.

The melting temperature were at around

61oC.These results were further integrated to

obtain the enthalpy change (∆H) of the lysozyme

samples (Figure 10). The values obtained in this

Page 9: Micro-differential scanning calorimeter for liquid ...for protein liquid sample and the other for reference buffer liquid sample. The chamber had a volume of 1µL (height 200µm, radius

study were around 415KJ/mol, which

corresponded well with the published data 377-

439 KJ/mol [25]. Apart from these, we could see

the second test could almost repeat the first time

test. Such variation of the results might be related

to the difference in the protein samples.

Figure 9. micro-DSC’s two tests with lysozyme sample

(molar weight:14307 Da). Specific heat capacity of

lysozyme vs. Temperature. (baseline subtracted)

Figure 10 enthalpy change of two lysozyme samples vs.

Temperature during unfolding. The enthalpy is obtained by

integrating the data in figure 9.

Conclusion

This paper reported a micro-DSC applied for

liquid biological sample characterization. This

sensor implemented temperature sensitive

vanadium oxide as the thermistor material with

TCR value of -2.8%/oC. With the help of the high

thermal insulation provided by the thin polyimide

membrane and the well-designed microfluidics

chamber, a sensitivity of 6V/W and power

resolution of 40nW were achieved. This high-

performance micro-DSC successfully tested the

lysozyme unfolding with the minimum sample

consumption (1µL) and this technology could be

further applied to expand the devices into arrays

so that high throughput measurement in the drug

discovery industry might be attained.

Acknowledge

The authors thank the funding support from NSF

(IDBR #1530508) and AbbVie Inc. The

fabrication was carried out at the Center for

Functional Nanomaterials (CFN) at Brookhaven

National Laboratory, which is supported by the

U.S. Department of Energy, Office of Basic

Energy Sciences, under Contract No. DE-AC02-

98CH10886. We also thank Dr. Fernando

Camino and Kuan Hu for the help and useful

discussion.

Reference

[1] Park BK, Yi N, Park J, Kim D. Monitoring protein denaturation using thermal conductivity probe. Int J Biol Macromol. 2013;52:353-7. [2] Olson EA, Efremov MY, Kwan AT, Lai S, Petrova V, Schiettekatte F, et al. Scanning calorimeter for nanoliter-scale liquid samples. Applied Physics Letters. 2000;77:2671. [3] Zhang Y, Tadigadapa S. Calorimetric biosensors with integrated microfluidic channels. Biosens Bioelectron. 2004;19:1733-43. [4] Lee W, Fon W, Axelrod BW, Roukes ML. High-sensitivity microfluidic calorimeters for biological and chemical applications. Proc Natl Acad Sci U S A. 2009;106:15225-30. [5] Wang B, Lin Q. Temperature-modulated differential scanning calorimetry in a MEMS device. Sensors and Actuators B: Chemical. 2013;180:60-5. [6] Mariana F, Buchholz F, Lerchner J, Neu TR, Harms H, Maskow T. Chip-calorimetric monitoring of biofilm eradication with

Page 10: Micro-differential scanning calorimeter for liquid ...for protein liquid sample and the other for reference buffer liquid sample. The chamber had a volume of 1µL (height 200µm, radius

antibiotics provides mechanistic information. Int J Med Microbiol. 2013;303:158-65. [7] Iervolino E, van Herwaarden AW, Sarro PM. Calorimeter chip calibration for thermal characterization of liquid samples. Thermochimica Acta. 2009;492:95-100. [8] Lopeandia AF, Andre E, Garden JL, Givord D, Bourgeois O. Highly sensitive parylene membrane-based ac-calorimeter for small mass magnetic samples. Rev Sci Instrum. 2010;81:053901. [9] Yi F, LaVan DA. Electrospray-assisted nanocalorimetry measurements. Thermochimica Acta. 2013;569:1-7. [10] Gotzen N-A, Huth H, Schick C, Van Assche G, Neus C, Van Mele B. Phase separation in polymer blend thin films studied by differential AC chip calorimetry. Polymer. 2010;51:647-54. [11] Kyoo Park B, Yi N, Park J, Kim D. Thermal conductivity of single biological cells and relation with cell viability. Applied Physics Letters. 2013;102:203702. [12] Torres FE, Kuhn P, De Bruyker D, Bell AG, Wolkin MV, Peeters E, et al. Enthalpy arrays. Proc Natl Acad Sci U S A. 2004;101:9517-22. [13] Lee W, Lee, Koh. Development and applications of chip calorimeters as novel biosensors. Nanobiosensors in Disease Diagnosis. 2012:17. [14] Lubbers B, Baudenbacher F. Isothermal titration calorimetry in nanoliter droplets with subsecond time constants. Anal Chem. 2011;83:7955-61. [15] Lerchner J, Wolf A, Schneider HJ, Mertens F, Kessler E, Baier V, et al. Nano-calorimetry of small-sized biological samples. Thermochimica Acta. 2008;477:48-53. [16] Sami Youssef JP, Roy Al Asmar, Brice Sorli, Oliver Cyril, and Alain Foucaran. MEMS Scanning Calorimeter With Serpentine-Shaped Platinum Resistors for Characterizations of Microsamples. JOURNAL OF MICROELECTROMECHANICAL SYSTEMS. 2009;18. [17] Yao H, Ema K, Fukada H, Takahashi K, Hatta I. ac nanocalorimeter for measuring heat capacity of biological macromolecules in solution. Review of Scientific Instruments. 2003;74:4164.

[18] Garden JL, Chateau E, Chaussy J. Highly sensitive ac nanocalorimeter for microliter-scale liquids or biological samples. Applied Physics Letters. 2004;84:3597. [19] C.H.Lee GW, A. O’Neill, and D. K. Manikkam. MICRO-CALORIMETER WITH ENCLOSED PARYLENE CHAMBERS FOR BIO/CHEMICAL APPLICATIONS. Twelfth International Conference on Miniaturized Systems for Chemistry and Life Sciences. San Diego, California, USAOctober 12 - 16, 2008. [20] Wang L, Wang B, Lin Q. Demonstration of MEMS-based differential scanning calorimetry for determining thermodynamic properties of biomolecules. Sensors and Actuators B: Chemical. 2008;134:953-8. [21] Jia Y, Wang B, Zhang Z, Lin Q. A polymer-based MEMS differential scanning calorimeter. Sensors and Actuators A: Physical. 2015;231:1-7. [22] G. W. H. Höhne WFH, H.-J. Flammersheim,. Differential Scanning

Calorimetry. Berlin Heidelberg: Springer-Verlag; 2010. [23] Torres FE, Recht MI, Coyle JE, Bruce RH, Williams G. Higher throughput calorimetry: opportunities, approaches and challenges. Curr Opin Struct Biol. 2010;20:598-605. [24] Recht MI, De Bruyker D, Bell AG, Wolkin MV, Peeters E, Anderson GB, et al. Enthalpy array analysis of enzymatic and binding reactions. Anal Biochem. 2008;377:33-9. [25] Hinz H-J, Schwarz FP. Measurement and analysis of results obtained on biological substances with d.s.c. The Journal of Chemical Thermodynamics. 2001;33:1511-25.