biothermodynamics : the role of thermodynamics in ... parti fundamentals 1...

12
Biothermodynamics With contributions by: John F. Carpenter, Eva Y. Chi, A. Espah Borujeni, M.T. Gude, W.M. van Gulik, Charles Haynes, Sef J. J. Heijnen, Curtis B. Hughesman, Gunnar Karlstrom, Jiirgen U. Keller, E. Jamalzadeh, Marcel L. Jansen, Hans-Olof Johansson, Karel Ch.A. M. Luyben, Thomas Maskow, Willem Norde, John M. Prausnitz, Theodore W. Randolph, E.S.J. Rudolph, Urs von Stockar, Adrie Straathof, H. Taymaz- Nikerel, Folke Tjerneld, Robin F. B. Turner, Patricia Vazquez Villegas, Voja Vojinic, P.J.T. Verheijen, and Luukvan derWielen. EPFL Press A Swiss academic publisher distributed by CRC Press

Upload: vothien

Post on 17-May-2018

219 views

Category:

Documents


1 download

TRANSCRIPT

Biothermodynamics

With contributions by: John F. Carpenter, Eva Y. Chi, A. Espah Borujeni, M.T. Gude, W.M. van Gulik,

Charles Haynes, Sef J. J. Heijnen, Curtis B. Hughesman, Gunnar Karlstrom, Jiirgen U. Keller, E. Jamalzadeh,

Marcel L. Jansen, Hans-Olof Johansson, Karel Ch.A. M. Luyben, Thomas Maskow, Willem Norde,

John M. Prausnitz, Theodore W. Randolph, E.S.J. Rudolph, Urs von Stockar, Adrie Straathof, H. Taymaz-

Nikerel, Folke Tjerneld, Robin F. B. Turner, Patricia Vazquez Villegas, Voja Vojinic, P.J.T. Verheijen, and

Luukvan derWielen.

EPFL Press

A Swiss academic publisher distributed by CRC Press

CONTENTS

PARTI

FUNDAMENTALS

1 THE ROLE OF THERMODYNAMICS IN BIOCHEMICAL

ENGINEERING 3

1.0 Basic remarks on thermodynamics in biochemical engineering 3

1.1 Fundamental concepts in equilibrium thermodynamics 5

1.2 Charged species, gels and other soft systems 5

1.3 Stability and activity of biomacromolecules 8

1.4 Thermodynamics of live cells 9

1.5 Thermodynamic analysis of metabolism 10

1.6 Conclusions 11

1.7 References 12

2 PHASE EQUILIBRIUM IN NON-ELECTROLYTE SYSTEMS 13

2.1 Introduction 13

2.2 Essential formal relations 14

2.2.1 Criteria for equilibrium 14

2.3 Liquid-liquid equilibria 19

2.4 Solid-liquid equilibria 21

2.5 References 23

3 VIRIAL EXPANSION FOR CHEMICAL POTENTIALS IN A

DILUTE SOLUTION FOR CALCULATION OF LIQUID-LIQUID

EQUILIBRIA 25

3.1 Introduction 25

3.2 Example of protein separation 25

3.3 References 31

4 THE THERMODYNAMICS OF ELECTRICALLY CHARGED

MOLECULES IN SOLUTION 33

4.1 Why do electrically charged molecules call for a particular

thermodynamic treatment? 33

xii B iothermodynamics

4.2 The thermodynamics of electrolytes 35

4.2.1 The electrochemical potential 35

4.2.2 Equilibrium conditions 36

4.2.3 Appropriate concentration measures and non-ideality 37

4.3 Electrostatics 38

4.3.1 Coulombs law, force of interactions 38

4.3.2 Short and long range interactions 38

4.3.3 Simplifications within the Debye-Hiickel theory 40

4.3.4 Derivation of the simple Debye-Huckel (DH) model 42

4.4 Empirical and advanced ion activity coefficient models 48

4.4.1 Empirical extension of Debye-Huckel theory 48

4.4.2 The Bjerrum theory of ion association 49

4.4.3 Modern developments in electrolyte theory 51

4.4.4 Pitzer's Model 52

4.4.5 Guggenheims numerical integration 53

4.4.6 Integral theory of electrolyte solutions 54

4.4 Equations of state for strong electrolyte thermodynamics 56

4.5 References 60

5 WATER 63

5.1 Introduction 63

5.2 Phenomenological aspects of water 64

5.3 Molecular properties of water 67

5.4 Water as a solvent 70

5.4.1 Electrolytes 71

5.4.2 Uncharged components 72

5.5 Further reading 75

PART II

CHARGED SPECIES, GELS, AND OTHER SOFT SYSTEMS

6 POLYMERS, POLYELECTROLYTES AND GELS 79

6.1 Flory's Theory of polymer solutions 79

6.2 Electric Charge on a weak polyelectrolyte 86

6.3 Hydrogels: Elementary Equations for Idealized Networks and

Their Swelling Behavior 96

6.3.1 Affine Network Model'

97

6.3.2 Phantom Network Model 98

6.3.3 Swelling theory for hydrogels 98

6.3.4 Illustration for a perfect tetrafunctional network 100

6.3.5 Effect of chain entanglement on swelling 104

6.3.6 Polyelectrolyte hydrogels 107

6.3.7 Hydrogel collapse: phase transition 112

Contents xiii

6.4 Appendix: Entropy of mixing for polymer solutions 115

6.5 References 121

7 SELF-ASSEMBLY OF AMPHIPHILIC MOLECULES 123

7.1 Introduction 123

7.2 Self-assembly as phase separation 124

7.3 Different types of self-assembled structures 128

7.4 Aggregation as a "start-stop" process: size and shape of

self-assembled structures 130

7.5 Mass action model for micellization 132

7.6 Factors that influence the critical micelle concentration 135

7.7 Bilayer structures 138

7.8 Reverse micelles 141

7.9 Microemulsions 142

7.10 Self-assembled structures in applications 146

7.11 References 147

8 MOLECULAR THERMODYNAMICS OF PARTITIONING IN

AQUEOUS TWO-PHASE SYSTEMS 149 •

8.1 Introduction 149

8.2 Flory-Huggins theory applied to aqueous two-phase

partition systems 152

8.3 Dependence of partitioning on system variables 154

8.3.1 Entropic contribution to the partition coefficient 156

8.3.2 Enthalpic contribution to the partition coefficient 158

8.3.3 Relative magnitudes of enthalpic and entropic

contributions to partitioning 160

8.3.4 Effect of polymer molecular mass on partitioning 162

8.3.5 Effect of tie-line length on partitioning 163

8.4 Simple interpretation of the effects of added electrolyte 165

8.4.1 Ion partitioning in systems containing a single salt 166

8.4.2 Ion partitioning in systems containing a single salt of a

polyelectrolyte with monovalent counter ions 168

8.4.3 Protein (biologic solute) partitioning in systems

containing a dominant salt 169

8.5 Calculation of phase diagrams and partitioning 172

8.6 Conclusions 175

8.7 References 178

9 GENERALIZATION OF THERMODYNAMIC PROPERTIES FOR

SELECTION OF BIOSEPARATION PROCESSES: 181

9.1 Phase behavior in Bioseparation Processes 181

9.1.1 Phase behavior of 'Bio '-molecules 181

9.1.2 Intermolecular interactions and molecular structure 184

xiv Biothermodynamics

9.1.3 Physical interactions 184

9.1.4 Chemical interactions 187

9.1.5 Relative strength of molecular interactions 188

9.1.6 Effect of molecular structure 188

9.1.7 Pure 'bio'-molecules: crystalline and amorphoussolids 193

9.2 Generalized correlation 196

9.2.1 Basic model development 196

9.2.2 Solubilities in mixed solvents 198

9.2.3 Partitioning in mixed solvents and aqueous two

phase systems 202

9.2.4 Sorption in mixed solvents 210

9.2.5 Selectivity of ion exchange resins 212

9.3 Generalized polarity scales 213

9.4 Conclusions 215

9.A APPENDIX 215

9.A.1 An estimation of log P 215

9.A.2 Pure components and mixtures 221

9.5 References 228

10 PROTEIN PRECIPITATION WITH SALTS AND/OR POLYMERS....

231

10.1 Introduction 231

10.2 Equation of state 234

10.3 The potential of mean force 236

10.4 Precipitation calculations 238

10.5 Generalization to a multicomponent solution 239

10.6 Crystallization 241

10.7 References 243

11 MULTICOMPONENT ION EXCHANGE EQUILIBRIA OFWEAK

ELECTROLYTE BIOMOLECULES 245

11.1 Introduction 245

11.2 Multi-component ion exchange of weak electrolytes 247

11.2.1 Thermodynamic framework 247

11.2.2 The DIX-model for monovalent ions 249

11.3 Experimental case studies 250

11.3.1 Ion exchange of carboxylic and acetyl amino acids....

251

11.3.2 Anion exchange of B-lactam antibiotics 253

11.4 Conclusions 256

11.5 References 257

Contents xv

PART III

STABILITY AND ACTIVITY OF BIOMACROMOLECULES

12 PROTEINS 261

12.1 Introduction 261

12.2 The amino acids in proteins 262

12.3 The three-dimensional structure of protein molecules in

aqueous solution 266

12.4 Non-covalent interactions that determine the structure of a

protein molecule in water 271

12.4.1 Hydrophobic interaction 272

12.4.2 Electrostatic interactions 273

12.4.3 Dipolar interactions 276

12.4.4 Dispersion interactions 277

12.4.5 Hydrogen bonding 277

12.4.6 Bond lengths and angles 279

12.5 Stability of protein structure in aqueous solution 279

12.6 Thermodynamic analysis of protein structure stability 281

12.7 Reversibility of protein denaturation aggregation of

unfolded protein molecules 287

12.8 References 288

13 THERMODYNAMICS IN MULTIPHASE BIOCATALYSIS 289

13.1 Why multiphase biocatalysis? 289

13.2 Thermodynamics of enzymatic reactions in aqueous

systems 290

13.3 Non-aqueous media for biocatalysis 296

13.3.1 Fluid phase systems 297

13.3.2 Solid-fluid systems 298

13.4 Using enyzmes in organic solvents 299

13.4.1 Enzyme inactivation in organic solvents 299

13.4.2 Predicting solvent effects on enzyme stability 300

13.4.3 Water activity control 30

13.5 Phase equilibria in multiphase enyzmatic reactions 303

13.5.1 Partition coefficients 303

13.5.2 Aqueous solubilities 304

13.5.3 Calculation of reaction yields at equilibrium 305

13.5.4 Suspension-to-suspension reactions 308

13.6 Whole cells in organic solvents 31

13.7 List of symbols 31

13.8 References 31

xvi Biothermodynamics

14 THERMODYNAMICS OF THE PHYSICAL STABILITY OF

PROTEIN SOLUTIONS 315

14.1 Introduction 315

14.2 Factors influencing protein stability 316

14.2.1 Temperature 316

14.2.2 pH effects on protein stability 317

14.2.3 Ligands and co-solutes 318

14.2.4 Salt type and concentration 327

14.2.5 Antimicrobial agents 328

14.2.6 Surfactants 328

14.3 Mechanism of protein aggregation 329

14.3.1 Structural transitions accompanying aggregation 329

14.3.2 Characterization of the aggregation competent species ...

329

14.3.3 Aggregation models, energetics, and rates 330

14.3.4 Role of conformational stability 332

14.3.5 Role of colloidal stability 338

14.4 Summary and conclusions 344

14.5 References 345

15 MEASURING, INTERPRETING AND MODELING THE STABILITIES

AND MELTING TEMPERATURES OF B-FORM DNAS THAT

EXHIBIT A TWO-STATE HELIX-TO-COIL TRANSITION 355

15.1 Introduction 355

15.2 Methods for measuring duplex DNA melting thermodynamics... .

358

15.2.1 UV absorption spectroscopy 359

15.2.2 Calorimetry 365

15.3 Modeling dsDNA stability and the melting transition 370

15.3.1 Statistical mechanical models of the melting transition...

370

15.3.2 Linear nearest-neighbor thermodynamic models of

B-form DNA stability and melting 372

15.3.3 Non-linear NNT models of B-form DNA stability

and melting 375

15.4 Comparing and further improving the performance of

NNT models 377

15.4.1 Duplexes terminating in a 5'-TA group have statisticallysignificant ATm errors 381

15.4.2 Correcting Tm predictions for duplexes containing5'-TA type termini 382

15.4.3 The dependence of B-DNA melting temperatures on

ionic strength 384

15.4.4 Correcting Tm predictions for common features and

modifications of probes and primers 385

15.5 Final thoughts 387

15.6 References 389

Contents xvii

PART IV

THERMODYNAMICS IN LIVING SYSTEMS

16 LIVE CELLS AS OPEN NON-EQUILIBRIUM SYSTEMS 399

16.1 Introduction 399

16.2 Balances for open systems 400

16.2.1 General remarks 400

16.2.2 Molar balances 401

16.2.3 Energy balances 402

16.2.4 Entropy balance 404

16.2.5 Gibbs energy balance 404

16.3 Entropy production, forces and fluxes 405

16.3.1 Entropy production in closed systems 405

16.3.2 Entropy production in non-reactive and reactive

flow-systems 406

16.3.3 Entropy production in steady-state heat conduction....

407

16.3.4 Total entropy production 407

16.4 Flux-force relationships and coupled processes 408

16.5 The linear energy converter as a model for living systems 409

16.5.1 Reactions driven against their driving force

through coupling 409

16.5.2 Efficiency of energy converters 412

16.5.3 Driving output reactions up-hill and the principle of

minimum entropy production 415

16.5.4 Predicting growth kinetics from irreversible

thermodynamics 416

16.5.5 Maintenance as static head situation 417

16.6 Conclusions 419

16.7 References 421

17 MINIATURIZATION OF CALORIMETRY: STRENGTHS

AND WEAKNESSES FOR BIOPROCESS MONITORING

AND CONTROL 423

17.1 Why miniaturization of calorimeters? 423

17.2 Historical roots 425

17.3 Measurement principle 426

17.3.1 Assembly of chip-calorimeter 426

17.3.2 Miniaturization limits 427

17.3.3 Signal evaluation 430

17.4 Calorimetry versus off-gas analysis 432

17.5 Applications of chip-calorimetry 434

17.5.1 Monitoring of discontinuous bioprocesses 434

17.5.2 Monitoring and control of continuous bioprocesses 435

17.5.3 Application for biofilm analysis 436

xviii B iothermodynamics

17.6 Outlook 438

17.7 References 438

18 A THERMODYNAMIC APPROACH TO PREDICT BLACK BOX

MODEL PARAMETERS FOR MICROBIAL GROWTH 443

18.1 Introduction 443

18.2 Catabolic energy production 445

18.2.1 Catabolic Gibbs energy under standard conditions 445

18.2.2 Catabolic Gibbs energy under non-standard

conditions 447

18.2.3 Threshold inhibition concentrations of catabolic

reactants 452

18.3 Thermodynamic prediction of the parameters in the

Herbert-Pirt substrate distribution relation 453

18.3.1 The substrate consumption rate for organismmaintenance, ms 453

18.3.2 The biomass reaction substrate parameter, a 454

18.3.3 The anabolic product reaction substrate parameter b.... 458

18.3.4 Stoichiometry of the biomass, product and catabolic

reactions 460

18.4 Prediction of the qp{\i) relationship 461

18.5 Prediction of the process reaction 462

18.5.1 Catabolic products 462

18.5.2 Anabolic products 464

18.5.3 Conclusion 465

18.6 Prediction of the hyperbolic substrate uptake kinetic parameters ...465

18.6.1 The parameters «7smax (or |amax) 465

18.6.2 Affinity, Ks 467

18.6.3 Other mechanisms putting thermodynamically based

upper limits on qs and qp 467

18.7 Influence of temperature and pH on Black Box model parameters.. 468

18.7.1 Effect of temperature 468

18.7.2 Effect of pH 469

18.7.3 Conclusion on temperature and pH- related kinetic

effects 471

18.8 Heat production in biological systems 471

18.9 Conclusion 472

18.10 References 472

18.11 Further reading 472

19 BIOTHERMODYNAMICS OF LIVE CELLS: ENERGY DISSIPATION

AND HEAT GENERATION IN CELLULAR CULTURES 475

19.1 Why study heat generation and energy dissipation in

biotechnology? 475

Contents xix

19.2 The first law: measuring, interpreting and exploiting heat

generation in live cultures 477

19.2.1 Applying heat balances to bioreactors and calorimeters... 477

19.2.2 Calorimeters 479

19.2.3 Typical heat generation rates during microbial growth

and their interpretation 481

19.2.4 On-line monitoring and control of bioprocesses by

heat dissipation measurements 484

19.3 The second law: energy dissipation, driving force and growth ....486

19.3.1 Energy dissipation and the driving force for growth in

chemotrophes 486

19.3.2 The relationship between the driving force for growthand the biomass yield in chemotrophes 489

19.3.3 Summary of the thermodynamics of chemotrophic

growth 491

19.4 Predicting energy and heat dissipation by calculation 492

19.4.1 Problem statement 492

19.4.2 Standard states 493

19.4.3 Reference states 493

19.4.4 Stoichiometry of the growth reaction and split into

catabolic and biosynthetic reactions 498

19.5 Results: heat generation and Gibbs energy dissipation as a

function of biomass yield 501

19.5.1 Aerobic growth 501

19.5.2 Ethanol fermentation 502

19.5.3 Lactic acid fermentation 504

19.5.4 Acetotrophi£ mefhanogenesis 505

19.5.5 Autotrophic methanogenesis 506

19.5.6 The relationship between heat generation and free

energy dissipation for chemotrophic growth 507

19.5.6 Mixotrophic and phototrophic growth 508

19.6 Application: prediction of yield coefficients 512

19.6.1 Growth efficiency and irreversible thermodynamics.... 513

19.6.2 Gibbs energy correlations 514

19.6.3 Product and energy yields for biofuels and

biorefineries 517

19.7 Discussion and conclusions 519

19.A Appendix: Example calculation for prediction of growth

stoichiometry 523

19.A.1 Statement of the problem 523

19.A.2 Thermodynamic data 524

19.A.3 Solution 525

19.A.4 Discussion 528

19.8 References 531

XX Biothermodynamics

20 THERMODYNAMIC ANALYSIS OF PHOTOSYNTHESIS 535

20.1 Introduction 535

20.2 References 543

PART V

THERMODYNAMICS OF METABOLISM

21 A THERMODYNAMIC ANALYSIS OF DICARBOXYLIC ACID

PRODUCTION IN MICROORGANISMS 547

21.1 Introduction 547

21.2 Outline of the approach 548

21.2.1 Black Box thermodynamic analysis of the theoretical

dicarboxylic acid product reaction 548

21.2.2 Maximal theoretical product yield 549

21.2.3 Stoichiometry of the theoretical product reaction 549

21.2.4 Alkali consumption, osmotic stress and ionic

strength 552

21.2.5 Thermodynamics of product formation 555

21.3 Thermodynamics of dicarboxylic acid transport 556

21.3.1 Thermodynamically feasible transport mechanisms ....556

21.3.2 Metabolic energy required for dicarboxylicacid export 560

21.3.3 Converting Gibbs energy of the theoretical product

reaction into ATP for growth 561

21.3.4 Fumaric acid 561

21.3.5 Succinic acid 565

21.3.6 Acid back diffusion 566

21.4 Genetic engineering of target systems based upon thermodynamic

analysis results 569

21.5 Conclusion 569

21.A Appendices 571

21 .A. 1 Acid/alkali cost 571

21.A.2 Standard AfG values 571

21 .A.3 In vivo energy aspects of ATP, proton motive force,

and fumarate reductase ., 572

21 .A.4 Effect of acid back-diffusion on the product yield of

dicarboxylic acid 574

21.6 References 577

22 THERMODYNAMIC ANALYSIS OF METABOLIC PATHWAYS 581

22.1 Introduction 581

22.2 Thermodynamic feasibility analysis of individual

metabolic pathways 582

Contents xxi

22.3 Estimation of observable standard Gibbs energies of reaction 585

22.3.1 K vs. K' - accounting for the non-ideality of the

reaction medium 587

22.3.2 K vs. K" - accounting for the pH and pMg of the

reaction medium 588

22.4 Materials and methods [22] 594

22.4.1 Data for standard Gibbs energies of reaction 594

22.4.2 Data for physiological conditions in the cytosol 595

22.4.3 Computational software 596

22.5 Results and discussion 596

23.5.1 Influence of the metabolite concentration range 596

22.6 Conclusions 600

22.7 References 603

INDEX 605