Skip to main content
Protein Science : A Publication of the Protein Society logoLink to Protein Science : A Publication of the Protein Society
. 2019 Dec 9;29(4):856–871. doi: 10.1002/pro.3794

Engineering the serpin α1‐antitrypsin: A diversity of goals and techniques

Benjamin M Scott 1,2, William P Sheffield 3,4,
PMCID: PMC7096715  PMID: 31774589

Abstract

α1‐Antitrypsin (α1‐AT) serves as an archetypal example for the serine proteinase inhibitor (serpin) protein family and has been used as a scaffold for protein engineering for >35 years. Techniques used to engineer α1‐AT include targeted mutagenesis, protein fusions, phage display, glycoengineering, and consensus protein design. The goals of engineering have also been diverse, ranging from understanding serpin structure–function relationships, to the design of more potent or more specific proteinase inhibitors with potential therapeutic relevance. Here we summarize the history of these protein engineering efforts, describing the techniques applied to engineer α1‐AT, specific mutants of interest, and providing an appended catalog of the >200 α1‐AT mutants published to date.

Keywords: protein design, protein engineering, proteinase inhibitor, serpin, α1‐antitrypsin, α1‐proteinase inhibitor

1. INTRODUCTION

1.1. The abundant serpin α1‐antitrypsin

α1‐Antitrypsin (α1‐AT), also known as α1‐proteinase inhibitor (α1‐PI), is a member of the serine proteinase inhibitor (“serpin”) superfamily of proteins.1, 2, 3 Serpins are primarily inhibitors of serine proteinases in the PA clan (Proteinases of mixed nucleophile, superfamily A) chymotrypsin‐like family,4 and play vital roles in regulating serine proteinase cascades by limiting the amount of active proteinase present.3, 5 Over 1,500 serpin genes organized into 16 clades have been identified in the genomes of all forms of life, including viruses; there are 36 human serpins.6 A serpin nomenclature identifying α1‐AT as SERPINA1 has been proposed.3 The core structure of serpins includes three β‐sheets and eight to nine α‐helices,3 is highly conserved across phyla, and may have arisen due to the combined effects of gene duplication and pressures to retain functional structures.6 A surface loop of 20–24 residues, called the reactive center loop (RCL), serves as the initial recognition motif for proteinases destined for serpin inhibition and contributes importantly to serpin specificity.

Mature α1‐AT is a 394‐residue 52 kDa glycoprotein synthesized primarily in the liver, and is the most abundant serpin in human plasma, with typical concentrations between 20 and 53 μmol/L that are further elevated during an acute phase response.7, 8 The principal physiological function of α1‐AT is to inhibit the serine proteinase neutrophil elastase, which is crucial for controlling the action of this proteinase in the lungs and liver. In individuals with α1‐AT deficiency, an autosomal co‐dominant disease, circulating levels of α1‐AT are as low as 10% of normal, contributing to a high risk for chronic obstructive pulmonary disorder (COPD)/emphysema and cirrhosis of the liver.9 α1‐AT deficiency has been linked to over 30 pathogenic variants,10, 11, 12 with the E264V “S” and E342K “Z” variants accounting for most cases due to a particularly high frequency among people of European decent.13, 14

Unraveling the molecular mechanism underlying α1‐AT deficiency and the inhibitory mechanism of serpins, in general, have been goals making α1‐AT the subject of extensive study for over 50 years.15 In the early 1980s, α1‐AT became the first serpin for which full‐length cDNA was isolated,16 and the first serpin to have a solved X‐ray crystal structure (albeit in a cleaved form).17 At the same time, advances in DNA manipulation and recombinant protein expression methods made it possible to purify large amounts of α1‐AT, with the initial goal of using it for replacement therapy for individuals with α1‐AT deficiency.18, 19 Recombinant α1‐AT has since been expressed in cultured mammalian cells, bacteria, yeast, insect cells, plants, and in transgenic animals (reviewed in Reference 20). Expression in bacteria and yeast is inexpensive and particularly robust, achieving yields of several mg/L up to 1 g/L.21, 22 Optimizing the expression of recombinant α1‐AT remains a major area of study, as current replacement therapy is dependent on expensive plasma‐derived α1‐AT.23

Beyond replacement therapy, α1‐AT has been investigated as a protein therapeutic for many other diseases including diabetes, graft‐versus‐host disease, cystic fibrosis, and arthritis among others.24, 25 This versatility stems from α1‐AT's ability to inhibit several proteinases tied to inflammation, including neutrophil elastase as well as proteinase 3,26 cathepsin G,27 and several caspases which are cysteine proteinases.28 α1‐AT has also been investigated as an anti‐infective agent, displaying an ability to suppress HIV‐1 infection,29 and overexpression of α1‐AT reduces bacterial load in transgenic mice,30 likely by suppressing proteinases required for pathogen proliferation. The production of pro‐inflammatory cytokines is also lowered by α1‐AT, however, the molecular mechanism contributing to this effect is not yet well‐defined and may be unrelated to proteinase inhibition.31, 32, 33, 34 The globular nature of α1‐AT may also contribute to its anti‐inflammatory properties, as it has been found to bind pro‐inflammatory cytokines and other proteins outside of the serpin mechanism.24, 35, 36

These potential therapeutic applications and relative ease of recombinant expression, combined with a detailed understanding of α1‐AT biochemistry, has made α1‐AT an attractive and versatile scaffold for protein engineering. In 1984, Rosenberg et al. specifically introduced the M358V mutation in an effort to create an oxidation‐resistant α1‐AT variant for replacement therapy, representing one of the earliest examples of protein engineering.37 Since then, over 200 engineered α1‐AT variants have been reported, modified using a wide variety of techniques. In this review, we provide an overview of the various methods and rationales for engineering α1‐AT to have novel and desired properties, highlighting key examples spanning the history of protein engineering as a discipline.

1.2. Structural and biochemical studies of α1 AT and the serpin mechanism

Crystal structures of intact α1‐AT revealed that it is composed of nine alpha helices, three beta sheets, and an exposed RCL loop.38 Numerous structural and biochemical studies have shown that α1‐AT shares both this general structure and a “suicide‐substrate” mechanism of inhibition39 with inhibitory serpins.40, 41 Unlike some serpins, α1‐AT does not require a cofactor (such as heparin or vitronectin) to maximize its rate of inhibition of target proteinases. Synthesis of α1‐AT occurs primarily in hepatocytes, where its 24‐residue signal peptide directs the nascent chain to the endoplasmic reticulum for folding and glycosylation prior to maturation in the Golgi apparatus and secretion.42 The protein contains three sites of N‐linked glycosylation (N46, N83, and N247) that help provide quality control of α1‐AT folding through the glycosylation‐dependent chaperones calreticulin and calnexin,42 and which extend the circulating half‐life of α1‐AT.43

Detailed biochemical characterization of α1‐AT has revealed a diverse range of inhibitory targets: neutrophil elastase is inhibited rapidly (with reported rate constants of inhibition of 1.2 × 107 to 7 × 107 L·mol−1·s−1),44, 45 however α1‐AT also has significant inhibitory activity toward the human serine proteinases chymotrypsin,46 kallikrein 7,47 neutrophil proteinase 3,26 pancreatic elastase,48 kallikrein 14,49 trypsin,45 and cathepsin G27 (listed in descending order of the reported rate of inhibition). Limited inhibitory activity toward other serine proteinases has also been reported, including coagulation factor Xa (FXa),50 coagulation factor XIa (FXIa),51 plasmin,48 thrombin,48 granzyme M,52 and activated protein C (APC).53 Even certain nonserine proteinases such as the metalloproteinase ADAM‐17,35 and the cysteine proteinases calpain‐154 and various caspases28 are reported to be inhibited by α1‐AT.

The mechanism by which α1‐AT inhibits all targets has been elucidated by biochemical and structural studies and is thought to be common to all inhibitory serpins.40, 41 Attacking proteinases first form an encounter or Michaelis complex with serpins, in which the RCL contacts the proteinase active site, and a tetrahedral intermediate is formed. Attack of the active site nucleophile (either serine or cysteine) on the serpin reactive center bond (termed P1‐P1′ in the Schechter and Berger nomenclature,55 M358‐S359 in α1‐AT) releases stored energy and powers a translocation of the proteinase to the opposite pole of the serpin. The proteinase remains tethered to the serpin via an acyl ester bond between the hydroxyl group of the active site serine and the P1 residue's carbonyl group, and the N‐terminal remainder of the RCL is inserted into β‐sheet A as a sixth strand. The rapidity of translocation of the proteinase and RCL loop insertion leaves the proteinase active site distorted, such that it cannot hydrolyze the acyl intermediate in a physiologically meaningful length of time but is instead cleared and degraded via scavenger receptors.56, 57

Studies employing α1‐AT were critical to solving the serpin mechanism (see Figure 1). Crystal structures of cleaved plasma‐derived α1‐AT,17 recombinant α1‐AT M358R non‐covalently bound to S195A‐trypsin,58 recombinant α1‐AT covalently bound to trypsin,59 and recombinant α1‐AT M358R covalently bound to pancreatic elastase60 provided static snapshots of different stages of inhibition. Fluorescence resonance experiments demonstrated a major conformational change in α1‐AT M358R complexed with trypsin but not with proteolytically inactive anhydrotrypsin, and full insertion of the RCL into β‐sheet A.57, 61 These studies helped rationalize earlier results from mutational studies, which identified a critical role for residues in the hinge region of the RCL, defined as stretching from residues P15 to P8. Alterations in this region converted α1‐AT from an inhibitor to a proteinase substrate or shifted the balance of the branched pathway to favor substrate behavior.44, 62, 63 The full loop insertion revealed by the two covalent α1‐AT complex structures and these mutagenic studies supported the conclusion, first made using P14 variants of plasminogen activator inhibitor 1 (PAI‐1, SERPINE1), that the rate of loop insertion into β‐sheet A determines whether a stable complex will form, or whether cleaved serpin and regenerated active proteinase will be liberated.64 The α1‐AT portions of the structures faithfully reproduced the original RCL‐cleaved structure, underlining that the conversion from a stressed to a relaxed state, accompanied by large differences in thermal melting temperatures of 50–60°C,65 is an integral part of the serpin mechanism.

Figure 1.

Figure 1

Branched inhibitory mechanism of α1‐AT. X‐ray crystal structures of α1‐AT with PDB codes are shown. In Step 1, α1‐AT is shown oriented with the RCL (light blue‐dark blue) at the top, forming a noncovalent encounter complex with trypsin (red). The PDB model was modified to convert both proteins to their respective wild type sequences, with the trypsin catalytic triad (H57, D102, and S195) and α1‐AT P1 and P1′ residues (M358, S359) shown as sticks. The encounter complex can then proceed to form the covalent serpin‐proteinase complex (Step 2a), via reactive center cleavage, RCL insertion into β‐sheet A, and proteinase translocation. In this complex, the proteinase is trapped in the acyl‐enzyme state (trypsin S195, α1‐AT M358 shown as sticks). Or, the complex decays into released proteinase (not shown) and cleaved serpin (Step 2b)

If the rate of loop insertion determines whether the covalent serpin‐proteinase complex forms, RCL length should be critical. In support of this idea, most serpins have an RCL of the same length, with only a few (e.g., antiplasmin [SERPINF2] and C1 esterase inhibitor, C1INH [SERPING1]) having an RCL one residue shorter. Zhou et al. tested this deduction by inserting one (Add‐1) or two Ala (Add‐2) residues into α1‐AT M358R between P4 and P3, and by stepwise deletions of P4 (Del‐1) up to P3‐P6 (Del‐4).66 Add‐1 and Add‐2 mutations reduced the rate of FXa inhibition by only twofold, but increased the dissociation rate of α1‐AT‐FXa complexes by factors of 105 to 106 With thrombin, Add‐1 and Del‐1 were minimally affected, while Add‐2 exhibited complex instability and elevated SI. Del‐2, ‐3, and ‐4 were substrates of either proteinase. Im et al. introduced larger insertions into the RCL, from 3 up to 30 residues, to investigate the underlying strain in the metastable native state.67 Each successive insertion increased the stability of α1‐AT, by releasing the energy stored by the strained native conformation, which the authors proposed generates the inactive “latent” form which features the uncleaved RCL inserted into β‐sheet A. These insertions also eliminated anti‐proteinase activity. These results provided independent support for the mechanism inferred from the structural studies and imposed a length restriction on RCL engineering.

The α1‐AT‐trypsin59 and α1‐AT‐elastase60 complexes remain the only examples of solved crystal structures of covalently linked serpin‐proteinase complexes. In both cases, there is full loop insertion, a tight interface between serpin and proteinase, and enough distortion of the translocated proteinase active site to explain its failure to leave the complex. Where the two structures differ is in the degree of proteinase disordering and proteolytic susceptibility. In the α1‐AT‐trypsin complex, 37% of the trypsin polypeptide was disordered, while in the α1‐AT‐elastase complex, less than 10% of the elastase moiety was crystallographically unresolved. The extent to which cognate proteinases are denatured by crushing up against the body of the serpin therefore likely varies in different serpin‐proteinase combinations.40

Mechanistic understanding of how α1‐AT executes the serpin mechanism of proteinase inhibition is indirectly important for protein engineering efforts and has productively informed multiple efforts to re‐direct the inhibitor's specificity. Gettins and Olson have noted that the complexities of the serpin mechanism place great demands on serpins in general,68 and these demands are increased for engineered serpins that have not benefited from evolutionary selection. Such demands include: metastability; optimal RCL for cognate proteinase recognition; correct length of the RCL for rapid loop insertion; compatibility of RCL residues for integration into β‐sheet A, with the burial of alternate side chains into the hydrophobic interior; and stability of the ensuing complex. There are numerous ways in which a serpin can be inactivated by mutation, either by nature (reviewed in References 11, 12) or in the course of nonproductive protein engineering efforts. Such engineering efforts can be refined by taking advantage of the large literature of loss‐of‐function mutations generated during structure and function studies, such as those involving the proximal44, 62, 63 and distal69 regions of the RCL, changes near the “shutter” region where two strands of β‐sheet A must open to allow RCL insertion,70 RCL length,66 and extensive loop exchange variants in which all or much of the RCL of one serpin has been substituted for the native RCL of another.71, 72, 73, 74 Future efforts may also be aided by advances in molecular modeling, such as that exemplified by a molecular dynamics simulation that predicted and confirmed enhanced inhibition of elastase by α1‐AT E346K.75

1.3. Measuring the kinetics of inhibition by α1‐AT

Any effort to enhance α1‐AT inhibition or re‐direct its specificity requires quantitative tools to characterize variant protein properties. Proteinase inhibition by all inhibitory serpins, including α1 AT, is a second‐order reaction in which reactants A and B combine to yield product AB, and is generally considered irreversible. The velocity of the reaction is the product of the second‐order rate constant (k2) and the molar concentrations of the serpin (A) and proteinase (B) reactants. This constant can be determined either continuously under second‐order conditions, using iterative curve‐fitting methods,58 or discontinuously under pseudo‐first‐order conditions.72, 76, 77, 78 In the latter simplification, a large molar excess of serpin to proteinase (typically at least 10‐fold) is employed, and serpin consumption is therefore considered to be negligible. The pseudo‐first‐order reaction velocity becomes the product of the observed pseudo‐first‐order rate constant kobs and the proteinase concentration, which can be readily determined using amidolysis of chromogenic or fluorogenic substrates to generate product P concentrations at time zero or time t. This constant is readily determined from the slope of a ln(P0/Pt) versus time in seconds plot; division by the initial serpin concentration then yields k2. The units of k2 are L·mol−1·s−1 (or M−1·s−1). Note that some investigators have used different terms from k2 to describe the rate constant, including kass (association) or kapp (apparent) while using the same methodology and units to determine the value of the constant (e.g., 66, 69).

Another parameter of critical importance in characterizing the kinetics of α1 AT‐proteinase interaction is the stoichiometry of inhibition (SI), defined as the number of moles of serpin required to inhibit a mole of proteinase.76 For plasma‐derived α1 AT and its arguably most relevant cognate proteinase, neutrophil elastase, this value at physiological ionic strength and pH, should be 1, indicating ideal inhibitor behavior with no substrate activity. Altering the reaction conditions or the serpin's primary structure via mutation can result in an increased stoichiometry as noted above for α1 AT hinge variants. The SI is determined by incubating varying ratios of α1 AT with proteinase until the reaction reaches equilibrium (i.e., for several hours) and then determining the residual proteolytic activity versus the serpin/proteinase ratio, extrapolated to zero proteolytic activity.72, 76, 77, 78 SI values are dimensionless.

Although some investigators have employed more complex kinetic models to generate additional rate constants specific for reversible or irreversible steps in the overall serpin pathway,73 k2 and SI seem to be the most commonly reported parameters. Some investigators have also argued that a true measure of the reaction constant for the branched pathway of serpin inhibition is an apparent association rate constant that is the product of k2 and SI. While this might be a better description of the combined inhibitory and substrate reactivity of a given serpin and proteinase, readers of this literature must take care to note what kind of constant is being used. For protein engineering purposes, investigators typically attempt to maximize k2 without elevating SI, to select mutant proteins with the most inhibitory predicted in vivo properties.

2. RATIONALE FOR Α1‐AT MUTAGENESIS AND ENGINEERING OF NOVEL PROPERTIES

Efforts to engineer α1 AT have spanned a spectrum of different goals, loosely related to general biotechnological aims such as enhancing protein stability, increasing expression yield, and prolonging circulatory half‐life, and more specific objectives such as re‐orienting α1 AT specificity to inhibit novel proteinase targets. Different regions of α1 AT have been mutated to advance these different goals, as highlighted in Figure 2. A nonexhaustive list of α1 AT mutants and their properties reported to date are provided in Table S1.

Figure 2.

Figure 2

Human α1‐AT labeled with regions of interest, and examples of protein engineering targeted to these regions. Colors of the text boxes correspond with colored residues and regions in the α1‐AT protein structure. P1 (M358) and P1′ (S359) residues are shown as sticks, to illustrate where the RCL is cleaved. PDB 3NE4 was used to generate the figure

2.1. Altering stability

Increasing the stability of recombinant proteins is a standard biotechnological goal that can be necessary to produce a robust product for pharmaceutical purposes. Serpins depart from this generality because their inhibitory mechanism demands a release of stored energy on reactive center cleavage, to transpose a target proteinase from one pole to the other. The difference in free energy between native α1 AT and RCL‐inserted α1 AT is 32.2 kcal mol−1, suggesting significant strain in the protein.65, 67 This obligatory metastability can leave α1 AT prone to misfolding into nonfunctional conformations, such as the latent conformation or polymerized conformations adopted by the pathogenic Z and S variants.13, 14

It was initially unknown whether stabilizing α1 AT against thermal or chemical denaturation would impair its function. Kwon et al. screened randomly mutated α1 AT variants for retention of function after heating of crude Escherichia coli lysates, finding a 13‐fold slower rate of inactivation for F51C but no effect on inhibitory function.79 Similarly, Kim et al. noted that F51L reduced misfolding of the Z variant,80 helping to link thermostability and the propensity for polymerization, a trend that has since been supported by extensive mutagenesis.81 Lee et al. added six additional mutations to F51L to produce “Multi‐7” α1 AT, which gained exceptional stability to chemical denaturation or conversion to latent form without losing functionality.82, 83, 84 Multi‐7 α1 AT was used to show proof‐of‐concept for the use of M13 phage display to screen for hyperstable α1 AT mutants, however, novel mutations were not reported in this study.85 Seo et al. also used random mutagenesis to identify 50 single amino acid substitutions in α1 AT that increased its thermal stability; only those stabilizing mutations lying near the RCL or β‐sheet A impaired anti‐proteinase function.84 Taken together these observations suggest that maintaining localized strain is necessary for α1 AT function, but that global strain can be reduced without impairment.

A simpler form of stabilization has also been applied, by replacing the lone cysteine residue in α1 AT, C232. This residue is highly reactive due to a positively charged local environment, which biases its thiol group toward thiolate formation, which could increase crosslinking with other cysteine residues.86 The C232S mutation ensures α1 AT cannot form disulfide‐bonded homodimers or heterodimers, but does not affect the rate of proteinase inhibition.87 This mutation is often included when overexpressing α1 AT, which improves soluble yields,88 and is treated as wild type protein in assays. While few investigators focusing on protein engineering have incorporated stabilizing mutations other than C232S into their strategies, these studies have been indirectly important because they have facilitated α1 AT crystallization. Multi‐7 mutations combined with M358R and C232S (i.e., “Multi‐9” α1 AT) were exploited to obtain crystals of α1 AT complexes with S195A‐trypsin58 and porcine pancreatic elastase.60

Engineering α1 AT stability is limited by the serpin inhibitory mechanism itself, and the inherent structure of the starting scaffold which has evolved to retain a certain level of strain. To explore these constraints, Porebski employed consensus protein design; using a multiple sequence alignment of serpins to arrive at a synthetic amino acid sequence that contains the most common residue at each position.89 This consensus serpin, conserpin, contained 137 residue differences versus α1 AT throughout the protein. Impressively, conserpin folded reversibly in response to chemical denaturation, which α1 AT cannot, by avoiding intermediate states as it folds. Conserpin also remained folded at temperatures up to 110°C, and resisted polymerization even with the addition of the destabilizing Z mutation. Counterintuitively, the crystal structure of conserpin revealed a lower number of intramolecular noncovalent interactions versus other serpins, but had increased electrostatic and hydrogen bonding at regions key for folding.

Although conserpin was able to inhibit proteinases rapidly, the covalent serpin‐proteinase complexes were not stable.89, 90 Replacing conserpin RCL residues P7–P1′ with P7‐P2′ from α1 AT, so that the RCL length and identity were closer to a natural serpin, improved SI values although complexes continued to dissociate faster than wild type α1 AT.90, 91 Interestingly, the protein remained unable to efficiently inhibit the primary cognate proteinase neutrophil elastase, which may be due to changes in RCL structural dynamics and the modified electrostatic potential of the serpin body.91 These studies further stress the important balance between the inhibitory properties of α1 AT and its stability, which remain challenging engineering independent of one another.

2.2. Modifications to increase recombinant protein yield

With the many systems used to express recombinant human α1‐AT, there have been several strategies used to increase the functional yield. This section will outline specific modifications made to the α1‐AT DNA sequence to increase yield, however, there are other variables such as the organism and strain selected, plasmid copy number, codon usage, culturing conditions, and purification methods (as reviewed by others20, 21, 92).

Initial attempts to express α1‐AT in E. coli, using a DNA sequence lacking the endogenous secretion signal peptide, resulted in functional protein but with relatively low yields.18, 93 Tessier et al. noted that the sequence immediately following this new start codon likely forms a stem‐loop secondary structure, obstructing ribosome initiation in E. coli.94 Modifications to this region by either adding a N‐terminal peptide, introducing silent mutations, or by deleting up to the initial 15 amino acids, resulted in a 10‐ to 200‐fold increase in expression.94, 95, 96 Deleting N‐terminal residues was subsequently found to be crucial for the expression of α1‐AT RCL hinge mutants in E. coli, which otherwise had negligible expression.62

α1‐AT sequences containing a truncated N‐terminus, the addition of an N‐terminal affinity tag, or containing various silent mutations are the most commonly used sequences for expression in E. coli, which escape the issue of an obstructed or improper start codon.21 However, the overexpression of recombinant proteins in E. coli often results in the formation of inclusion bodies, insoluble aggregates of protein that accumulate in the cytoplasm, which significantly reduces the recoverable yield of α1‐AT. Schulze et al. investigated the importance of RCL mutations on α1‐AT solubility in E. coli,97 noting that that the need for the RCL to insert into β‐sheet A may lead to dimers forming between unstable intermediate molecules of α1‐AT as they fold, akin to a molecular mechanism proposed for the pathogenicity of the Z E342K mutation.14, 98 The M351E mutation at position P8 in the RCL, significantly reduced aggregation in E. coli, increasing soluble protein yield by over twofold.97 This mutation likely prevents the formation of stabilizing hydrophobic interactions with β‐sheet A, but importantly the M351E mutation did not disrupt inhibitory function.99

The M351E mutation has also been used to increase expression in insect cells, with the additional modification of an N‐terminal fusion protein to facilitate α1‐AT secretion.100, 101 The addition of a signal peptide or fusion protein is a common technique used to increase recombinant protein expression, and has been used to improve α1‐AT yields from E. coli,102 yeast,103 and plants.104, 105 In the first study to overexpress α1‐AT using cultured mammalian cells, modification to the 5′ untranslated region and endogenous signal peptide improved expression up to fivefold,106 however, more recent studies using the wild type endogenous signal peptide achieved superior yields.107, 108 There are therefore a variety of protein and DNA modifications that can be made to α1‐AT to improve the yield, which is largely dependent on the recombinant expression system used.

2.3. Oxidation resistance

Individuals with α1‐AT deficiency are predisposed to COPD and emphysema, which is worsened by cigarette smoking, due to the smoke causing oxidation of the remaining α1‐AT.9, 109 This effect highlights α1‐AT's susceptibility to oxidation, which may underlay the pathogenesis of emphysema even in individuals without α1‐AT deficiency.110 α1‐AT oxidation may additionally be exacerbated by neutrophil‐mediated generation of free radicals during lung inflammation, leading to a further imbalance in the elastase/α1‐AT stoichiometry in the lungs.8, 111 α1‐AT's susceptibility to oxidation results from the protein containing nine methionine residues, two of which occur in the RCL: M358 (being the crucial P1 residue recognized by target proteinases); and M351 close to the hinge region of the RCL. That both methionines remain intact, without being oxidized to methionine sulfoxide, is a requirement for efficient inhibition of elastase.111

As mentioned previously, M358V was the first mutation deliberately introduced into α1‐AT to engineer its properties.37 M358V has since been characterized by many others, revealing that although this mutation modestly reduces the rate of neutrophil elastase inhibition (1.2‐fold to 2.7‐fold lower), it retains inhibitory function after being exposed to chemical oxidation while its wild type counterpart does not.112, 113 M358V also improves the rate of pancreatic elastase inhibition by ≈10‐fold, likely due to this proteinase's preference for valine at P1.4, 112 The double mutant M351V M358V significantly improves oxidation resistance, and maintains neutrophil elastase inhibition.111

Substituting M358 with other hydrophobic residues has also been explored as a way of improving oxidation resistance. Cysteine at this position is not resistant to oxidation, due to the exposed terminal sulfur, while other hydrophobic residues (A/F/I/L) are resistant but with modestly reduced rates of elastase inhibition compared to M358V.113 This finding is somewhat surprising, given neutrophil elastase's preference for A/I/T/V at P1,4 which may suggest a fundamental requirement for methionine for efficient inhibition via the serpin mechanism.

Silberstein et al. recently reported the high‐yield expression of α1‐AT M358V in Nicotiana benthamiana, and argued that this mutant could be used as an improved biologic drug, a “biobetter,” to replace therapeutic preparations of wild type α1‐AT currently obtained from plasma.114 The plant‐derived M358V inhibited neutrophil elastase in vitro as well as a current therapeutic preparation of α1‐AT, tradename Prolastin‐C®, with the additional property of being oxidation resistant.114 Similarly, Zhu et al. reported combining efforts to create a more thermostable α1‐AT with oxidation resistance, also to be used as a bio‐better.115 The expression of F51L/M351V/M358V in E. coli was robust, with the F51L stabilizing mutation increasing the denaturation temperature by 6°C, in addition to being oxidation resistant.115 These developments represent steps toward engineered α1‐AT being used for replacement therapy, benefiting from the extensive work understanding α1‐AT inactivation by oxidation, and efforts to improve the overall stability of the protein.

2.4. Thrombin inhibition

Over 40 years ago, the rare α1‐AT “Pittsburgh” variant was found to cause a severe and ultimately fatal bleeding disorder by converting the protein into a potent thrombin inhibitor.116 The discovery that a single α1‐AT mutation was responsible, M358R, confirmed the suspected location of the reactive site and indicated that mutations in this region could significantly change α1‐AT's inhibitory properties.15, 117 It was subsequently noted that α1‐AT M358R also inhibited the contact factor enzymes FXIa, FXIIa, and kallikrein.51, 118 This profile encouraged investigators to express α1‐AT M358R in yeast and to test in a piglet model of Pseudomonas aeruginosa septicemia, where it reduced mortality and prevented consumption of antithrombin (SERPINC1) and FXI.119 However, yeast‐derived α1‐AT M358R failed to prevent mortality in an E. coli septicemia model in baboons and may have exacerbated coagulopathy.120 The negative outcome may have arisen due to the use of recombinant α1‐AT M358R preparations containing cleaved serpin and yeast‐specific glycans, and/or due to additional properties of α1‐AT M358R such as the ability to inhibit APC121 and plasmin.

The difficulties encountered in these initial attempts at preclinical use of α1‐AT M358R prompted several investigators to hone α1‐AT M358R to target thrombin more specifically, in hopes of developing a heparin‐independent thrombin inhibitor and antithrombotic agent. Single and double substitutions of the P2 and P1' residues of α1‐AT for the corresponding residues of antithrombin or C1INH, both very poor inhibitors of APC, yielded no increase in selectivity (defined quantitatively as the quotient of the k2 for thrombin and the k2 for APC).122 In contrast, Hopkins et al. found that substituting the P7‐P2 residues of antithrombin for those of α1‐AT improved selectivity; the most selective variant found in this study, α1‐AT LS, replaced α1‐AT M358R's P7‐P2 and P2′‐P3′ residues with those of antithrombin (FLEAIPRSIP to AVVIAGRSLN).122 The LS variant exhibited a 10,000‐fold reduction in its rate of APC inhibition, at the cost of a twofold reduction in the rate of thrombin inhibition, for a selectivity factor of ≈8,000. A follow‐up study demonstrated faster inhibition of thrombin for a less extensive variant (lacking the I361N mutation) but greater residual anti‐APC activity, for a selectivity factor of ≈3,200.123

Our group initially attempted to transfer the thrombin specificity of heparin cofactor II (HCII; SERPIND1) by substituting its RCL for that of α1‐AT from P16 to P3′, excepting the P1 Arg. This mutagenic effort reduced the rate of thrombin inhibition by 3.3‐fold, but that of APC by 68‐fold, with a selectivity factor of ≈160; the SI was also elevated threefold by this manipulation.72 Appending residues 1–75 of HCII, a negatively charged extension that binds thrombin exosite 1, to the N‐terminus of α1‐AT M358R enhanced the rate of thrombin inhibition by α1‐AT M358R by 21‐fold.77 The fusion protein was termed HAPI M358R for HCII(1–75)‐ α1PI M358R. Our group subsequently combined the HCII fusion approach with additional RCL mutations: HAPI RCL4 (incorporating P16‐P3′ HCII residues and M358R); and HAPI RCL5 (incorporating the LS variant RCL from122).124 HAPI RCL5 inhibited thrombin 13‐fold more rapidly than α1‐AT M358R, and APC 108‐fold less rapidly, for an overall selectivity factor of ≈10,000. The chimera inhibited FXa and FXIa 2‐ to threefold less rapidly than thrombin, and FXIIa 14‐fold less rapidly than α1‐AT M358R.125 At a dose of 7.2 mg/kg body weight, E. coli‐derived HAPI RCL5 was superior to α1‐AT M358R in reducing thrombus weight in the vena cava of mice treated with ferric chloride and in minimizing fibrin deposition in murine endotoxemia.125 Grafting another thrombin exosite 1‐binding domain derived from the leech inhibitor hirudin onto α1‐AT M358R also enhanced its rate of thrombin inhibition, but to a lesser extent than HCII 1–75.126

Our group also used a T7 phage display system to fully randomize the P7‐P3 residues of α1‐AT M358R, and to select two novel variants that enhanced the rate of thrombin inhibition by twofold via directed evolution: DITMA and AAFVS (compared to wild type FLEAI residues).78 This system can be exploited to vary different portions of the RCL and/or to probe with different proteinases via positive or negative selection.127

2.5. Inhibition of factors IXa and Xa

While most α1‐AT protein engineering efforts have focused on the RCL, alterations to strand 3 of β‐sheet C have been shown to create an exosite facilitating its interactions with FXa and FIXa.128 Building on their previous demonstration of such an exosite in antithrombin,129 Izaguirre et al. altered two residues in α1‐AT M358R (K222Y and L224E) lying under the RCL in the body of the serpin, and recreated the exosite. The rate of inhibition of FXa was increased 14‐fold versus α1‐AT M358R in the variant, and 11‐fold with respect to FIXa, without altering the reaction stoichiometry. Substituting a FXa recognition site IEG for the P4‐P2 sites of α1‐AT M358R increased the k2 for FXa 10‐fold, and combining this motif with K222Y and L224E effected an overall 20‐fold and 26‐fold increased rate of Xa and IXa inhibition versus α1‐AT M358R, respectively.128 Much of the increased rate of inhibition could be ascribed to the increased affinity of the reactants and the resulting increased ease of forming a Michaelis complex.

In another example of extra‐RCL engineering, Yang et al. substituted the heparin‐binding D helix of antithrombin for the native residues in α1‐AT. The α1‐AT/D helix chimera acquired a 100‐fold acceleration of thrombin inhibition in the presence of heparin, as well as in vivo anti‐inflammatory properties thought to arise from its new ability to bind to heparan sulfate proteoglycans of the endothelium and mediate vascular protective effects.130

2.6. Inhibition of activated protein C

Polderdijk et al. sought to enhance the inhibition of APC by α1‐AT, in hopes of developing a new treatment agent for hemophilia.131 Extrapolating from clinical observations that the severity of hemophilia A or B was lessened in patients whose factor V (Leiden) was resistant to inactivation by APC, these investigators reasoned that effectively inhibiting APC could preserve enough prothrombinase activity to remedy either bleeding diathesis. Using structural comparisons of the active sites of thrombin and APC, they designed an α1‐AT M358R variant with Lys substitutions at P2 and P1′ (α1‐AT KRK). This variant exhibited no detectable inhibition of thrombin, a sevenfold slower inhibition of APC than α1‐AT M358R, a 400‐fold reduced rate of FXa inhibition, and a 1,000‐fold reduction in the rate of FXIa inhibition. In hemophilia B fIX knockout mice, α1‐AT KRK administered subcutaneously at 15 mg/kg body weight eliminated the bleeding diathesis and reduced bleeding to levels seen in wild type mice, using a tail clip model. If these promising results hold in further preclinical development, α1‐AT KRK could become a drug candidate for hemophilia A and B treatment, one of a new class of “inhibitors of coagulation inhibitors.”132 The same group, in a follow‐up study, randomized the P2 and P1' residues and screened a library of α1‐AT M358R variants, but did not find one superior to α1‐AT KRK for APC inhibition.133

2.7. Inhibition of contact pathway proteinases

Researchers recently engineered α1‐AT M358R to inhibit plasma kallikrein (PKa).134 de Maat et al. sought a novel therapeutic agent to control disorders of excessive bradykinin generation, which arises from PKa digestion of high molecular weight kininogen. One such disorder is hereditary angioedema, which results from C1 esterase inhibitor (C1INH, SERPING1) deficiency, which releases FXIIa, FXIa, and PKa from C1INH control. Double mutant α1‐AT P357A/M358R exhibited a fivefold increase in the rate of PKa inhibition, and a similar decrease in thrombin inhibition.135 Similarly, synthetic peptide cleavage studies were leveraged to alter α1‐AT to PFR at P3‐P1, which retained much of α1‐AT M358R's activity against PKa, but not complement C1s, FXIIa, or APC.136 Recently, investigators selected P4‐P1′ SMTRS and SLLRS, from the activation peptide of FXII and peptide substrate libraries, respectively, to re‐orient α1‐AT specificity. These variants inhibited PKa, FXIIa, and plasmin, but also retained residual anti‐thrombin and anti‐APC activities. The latter were eliminated in α1‐AT‐SLLRV, which exhibited k2 values of 1.88 × 105 M−1 s−1 for PKa and 1.34 × 104 M−1 s−1 for FXIIa. This variant, when given to mice at 8 mg/kg body weight, reduced ferric chloride‐induced arterial thrombosis, prevented carrageenan‐induced paw swelling, and reduced epithelial leakage of fluorescent dextran in the dextran sulfate colitis model.134

2.8. Inhibition of other proteinases

The well‐characterized structure and function of α1‐AT, the lack of a requirement for a GAG cofactor for optimal activity, and its promiscuous activity, has also made this serpin a useful scaffold for engineering inhibition of proteinases beyond the coagulation cascade. The human serine proteinase furin, a proprotein convertase, was an early target for engineered inhibition, as furin‐mediated cleavage of viral glycoproteins (gp) facilitates HIV‐1, measles, and influenza infection.137 By introducing a furin consensus sequence into the α1‐AT RCL via two mutations (A355R M358R), Anderson et al. demonstrated that α1‐AT could be converted to a furin inhibitor, which blocked the processing of HIV‐1 gp160 required for cellular invasion.138 This engineered “α1‐PDX” protein has since been shown reduce the infectivity of HIV‐1 and measles in vitro,139, 140 as well as reduce tumor growth and atherosclerosis in mice.141, 142 The latter findings likely stem from furin's role in processing the pro‐peptide of human metalloproteinases involved in tumor cell migration and endothelial remodeling.141

Efforts to improve the inhibition of furin and other proprotein convertases, by adding additional positively charged residues to the RCL, have had mixed results.143, 144 However, transplanting the P6‐P1 RCL residues from the natural furin inhibitor Serpin B8 into α1‐AT improved the rate of furin inhibition twofold versus α1‐PDX, and surprisingly 10‐fold versus the original Serpin B8.145 Selectivity toward other proprotein convertases could be tuned via additional mutations, by introducing P′ RCL residues from Serpin B8 or Serpin B8 exosite residues.146 α1‐AT, therefore, appears to be an attractive and tunable scaffold for engineering inhibitors of a variety of proprotein convertases.

Serpins are also capable of inhibiting cysteine proteinases, which are not primary targets of α1‐AT. To understand the basis for cysteine proteinase inhibition, Irving et al. introduced RCL residues from the serpin SCCA‐1, which inhibits papain‐like cysteine proteinases, into α1‐AT.147 The resulting chimeric serpins inhibited cathepsins L, V, and K rapidly (k2 up to 1.32 × 106 M−1 s−1) with low SI, provided that the RCL length remained similar to α1‐AT.

To engineer α1‐AT to inhibit kallikrein‐14, rather than borrowing RCL residues from known serpins, Felber et al. used an alternative approach.49 Peptides first identified as optimal kallikrein‐14 substrates by phage display were inserted into the α1‐AT RCL, yielding novel serpins with inhibitory activity toward several human kallikreins. However, the serpin anti‐chymotrypsin was ultimately found to be a better scaffold than α1‐AT for engineering kallikrein‐14 inhibition, illustrating the importance of the original serpin sequence.

2.9. Increasing circulation half‐life

Pharmacokinetic studies of plasma‐derived α1‐AT concentrates administered to α1‐AT‐deficient patients have established a long circulatory half‐life (t1/2) of 7.7 to 8.9 days.148 While nonglycosylated recombinant α1‐AT produced in E. coli demonstrates highly similar kinetics of enzyme inhibition in vitro,20 its t1/2 is much shorter.149 This accelerated clearance can be ameliorated by chemical modification. Cantin et al. exploited the lone free thiol (C232) and conjugated polyethylene glycol (PEG) chains of varying lengths to this residue.149 Although these investigators did not calculate pharmacokinetic parameters, they found that the residual amount of recombinant α1‐AT conjugated to 20 or 40 kDa PEG, but not 5 kDa PEG, did not differ from that of plasma‐derived α1‐AT in mice up to 24 hr post‐injection.

Chinese Hamster Ovary (CHO) cells and more recently human embryonic kidney (HEK293) cells have been used to express recombinant human proteins that have been approved by regulators for clinical use. Functional expression of wild type recombinant α1‐AT in CHO and HEK293 cells has been reported, although without pharmacokinetic information.90, 91 Amann et al. engineered CHO cells, combining CRISPR/Cas9‐mediated disruption of 10 rodent glycosylation‐related genes and overexpression of human St6 β galactoside α‐2,6‐sialyltransferase 1 (ST6GAL1), to produce recombinant α1‐AT with N‐linked glycan structures more closely resembling those in plasma‐derived α1‐AT than in standard CHO‐derived α1‐AT.108

Several groups have attempted to prolong α1‐AT t1/2 via glycoengineering, which is the addition or modification of glycans on the protein. By modifying α1‐AT's existing sugars with sialylation enzymes, Lindhout et al. increased the t1/2 from 5 to 27 hr in mice.150 Lusch et al. introduced additional glycosylation consensus motifs N‐x‐S/T at residues N90, N123, and N201, finding that that additional glycosylation at only N123 was superior, which increased the serum t1/2 of HEK293‐derived α1‐AT by 63%.151 Similarly, Chung et al. tested the introduction of seven different N‐x‐S/T motifs, finding that the fourth glycan at Q9N or D12N/S14T increased the t1/2 of CHO‐derived α1‐AT by 2.2‐fold versus the wild type in rats.152

The constant portion of human IgG1 (Fc) has been fused to proteins of interest to prolong their t1/2 by imparting the property of recycling via the neonatal Fc receptor.153 An α1‐AT‐Fc fusion protein has been reported to lack anti‐elastase activity154, 155; presumably, this arises either from steric hindrance due to the dimerization of the α1‐AT‐Fc protein engendered by two disulfide bonds in the Fc hinge region near the fusion point, or an indirect conformational distortion of the α1‐AT moiety. The strategy would, therefore, be unlikely to be effective for recombinant α1‐AT proteins being developed as novel inhibitors of any proteinase. The fusion protein is instead being developed by those interested in anti‐inflammatory effects of α1‐AT that are independent of anti‐proteinase function, such as regulation of tissue necrosis factor and neutrophil activation.34, 154, 155 Pharmacokinetic studies have not been reported, although the α1‐AT‐Fc fusion exhibited prolonged anti‐inflammatory effects in mouse models of gouty arthritis and ischemia–reperfusion.156, 157

3. CONCLUDING REMARKS

Considerable research effort has gone into engineering the function and specific properties of the serpin α1‐AT. While these efforts have reaped many dividends, including an expanded understanding of serpin structure and function, the field is not yet at the point where α1‐AT variants can be designed a priori and then confirmed to function as desired. Attaining such an ideal state will require, at a minimum, new advances in protein structural modeling and molecular dynamics simulations in the long term. Even in the case of the synthetic thermostable conserpin, it remains difficult to alter specificity productively due to the many demands of the serpin mechanism.91 In the medium term, it is likely that combining different approaches, such as phage display or serpin‐proteinase docking data to select α1‐AT variants and then reconfiguring them into long‐lasting injectable protein drugs, will be undertaken. In the shorter term, it is likely that protein engineering modifications improving the pharmacokinetics of wild type α1‐AT will make feasible clinical trials for new indications. With respect to engineered serpins, Serpin PC (also called α1‐AT KRK),131, 158 and the α1‐AT‐Fc fusion protein are poised at this writing to enter early clinical trials.159 In so doing they will join only one other engineered serpin that has taken these steps to the clinic, the viral serpin Serp1, which showed promise in a small phase II randomized clinical trial in patients with acute coronary syndromes.160

In spite of the intensity of mutagenesis undertaken to date (partially summarized in Table S1), there remain areas of the protein in which more investigation should be focused: the salt bridge between P5 and the body of the protein, which is key to RCL flexibility161; the “south pole” of α1‐AT, an interface that could be modified to facilitate proteinase migration and retention; and the β‐sheet A strands into which the RCL must insert to form complexes. Few investigators have considered the possibility of immunogenicity of engineered serpins, and this possibility should be addressed in silico and in animal studies to see if this is a likely scenario, and one for which immune‐camouflage approaches like PEGylation might be appropriate.

The abundance of α1‐AT, its ease of purification, and its early association with human disease made it an archetype for serpins. Research to date has identified its value as an adaptable scaffold for serpin investigation, and in the future, this value will likely be extended into clinical use along the lines outlined in this review.

CONFLICT OF INTEREST

The authors have no conflicts of interest to declare.

Supporting information

Supplemental Table 1 An Excel file containing a descriptive list of over 200 published engineered α1‐AT variants (Scott_Sheffield_SupTable1_191112.xlsx). The list is non‐exhaustive, meant to provide an overview of the diversity of α1‐AT mutants published. The most recent version is available via Figshare https://doi.org/10.6084/m9.figshare.10294889

ACKNOWLEDGMENTS

B.M.S. is an International Associate of the National Institute of Standards and Technology (NIST). NIST notes that certain commercial materials are identified in this article to specify an experimental procedure as completely as possible. In no case does the identification of particular materials imply a recommendation or endorsement by NIST, nor does it imply that the particular materials are necessarily the best available for the purpose. The opinions expressed in this article are the authors' own and do not necessarily represent the views of NIST. W.P.S. is Associate Director, Research, Centre for Innovation, Canadian Blood Services. Since the Centre for Innovation receives funds from Health Canada, a department of the federal government of Canada, this article must contain the statement, “The views expressed herein do not necessarily represent the views of the federal government.”

Scott BM, Sheffield WP. Engineering the serpin α1‐antitrypsin: A diversity of goals and techniques. Protein Science. 2020;29:856–871. 10.1002/pro.3794

Funding information Canadian Blood Services, Grant/Award Number: WS‐IG2019; Heart and Stroke Foundation of Canada, Grant/Award Number: G‐19‐0026318

REFERENCES

  • 1. Hunt LT, Dayhoff MO. A surprising new protein superfamily containing ovalbumin, antithrombin‐III, and alpha 1‐proteinase inhibitor. Biochem Biophys Res Commun. 1980;95:864–871. [DOI] [PubMed] [Google Scholar]
  • 2. Huber R, Carrell RW. Implications of the three‐dimensional structure of alpha 1‐antitrypsin for structure and function of serpins. Biochemistry. 1989;28:8951–8966. [DOI] [PubMed] [Google Scholar]
  • 3. Law RH, Zhang Q, McGowan S, et al. An overview of the serpin superfamily. Genome Biol. 2006;7:216. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Rawlings ND, Barrett AJ, Thomas PD, Huang X, Bateman A, Finn RD. The MEROPS database of proteolytic enzymes, their substrates and inhibitors in 2017 and a comparison with peptidases in the PANTHER database. Nucleic Acids Res. 2018;46:D624–D632. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Silverman GA, Whisstock JC, Bottomley SP, et al. Serpins flex their muscle: I. putting the clamps on proteolysis in diverse biological systems. J Biol Chem. 2010;285:24299–24305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Heit C, Jackson BC, McAndrews M, et al. Update of the human and mouse SERPIN gene superfamily. Hum Genomics. 2013;7:22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Baumann H, Gauldie J. The acute phase response. Immunol Today. 1994;15:74–80. [DOI] [PubMed] [Google Scholar]
  • 8. Crystal RG. The alpha 1‐antitrypsin gene and its deficiency states. Trends Genet. 1989;5:411–417. [DOI] [PubMed] [Google Scholar]
  • 9. Perlmutter DH. Alpha‐1‐antitrypsin deficiency: Importance of proteasomal and autophagic degradative pathways in disposal of liver disease‐associated protein aggregates. Annu Rev Med. 2011;62:333–345. [DOI] [PubMed] [Google Scholar]
  • 10. Landrum MJ, Lee JM, Benson M, et al. ClinVar: Improving access to variant interpretations and supporting evidence. Nucleic Acids Res. 2018;46:D1062–D1067. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Salahuddin P. Genetic variants of alpha1‐antitrypsin. Curr Protein Pept Sci. 2010;11:101–117. [DOI] [PubMed] [Google Scholar]
  • 12. Hazari YM, Bashir A, Habib M, et al. Alpha‐1‐antitrypsin deficiency: Genetic variations, clinical manifestations and therapeutic interventions. Mutat Res. 2017;773:14–25. [DOI] [PubMed] [Google Scholar]
  • 13. de Serres FJ, Blanco I, Fernandez‐Bustillo E. PI S and PI Z alpha‐1 antitrypsin deficiency worldwide. A review of existing genetic epidemiological data. Monaldi Arch Chest Dis. 2007;67:184–208. [DOI] [PubMed] [Google Scholar]
  • 14. Gooptu B, Dickens JA, Lomas DA. The molecular and cellular pathology of alpha(1)‐antitrypsin deficiency. Trends Mol Med. 2014;20:116–127. [DOI] [PubMed] [Google Scholar]
  • 15. Carrell RW. What we owe to alpha(1)‐antitrypsin and to Carl‐Bertil Laurell. COPD. 2004;1:71–84. [DOI] [PubMed] [Google Scholar]
  • 16. Long GL, Chandra T, Woo SL, Davie EW, Kurachi K. Complete sequence of the cDNA for human alpha 1‐antitrypsin and the gene for the S variant. Biochemistry. 1984;23:4828–4837. [DOI] [PubMed] [Google Scholar]
  • 17. Loebermann H, Tokuoka R, Deisenhofer J, Huber R. Human alpha 1‐proteinase inhibitor. Crystal structure analysis of two crystal modifications, molecular model and preliminary analysis of the implications for function. J Mol Biol. 1984;177:531–557. [PubMed] [Google Scholar]
  • 18. Bollen A, Herzog A, Cravador A, et al. Cloning and expression in Escherichia coli of full‐length complementary DNA coding for human alpha 1‐antitrypsin. DNA. 1983;2:255–264. [DOI] [PubMed] [Google Scholar]
  • 19. Cabezon T, De Wilde M, Herion P, Loriau R, Bollen A. Expression of human alpha 1‐antitrypsin cDNA in the yeast Saccharomyces cerevisiae . Proc Natl Acad Sci U S A. 1984;81:6594–6598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Karnaukhova E, Ophir Y, Golding B. Recombinant human alpha‐1 proteinase inhibitor: Towards therapeutic use. Amino Acids. 2006;30:317–332. [DOI] [PubMed] [Google Scholar]
  • 21. Krishnan B, Hedstrom L, Hebert DN, Gierasch LM, Gershenson A. Expression and purification of active recombinant human Alpha‐1 antitrypsin (AAT) from Escherichia coli . Methods Mol Biol. 2017;1639:195–209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Tamer IM, Chisti Y. Production and recovery of recombinant protease inhibitor α1‐antitrypsin. Enzyme Microb Technol. 2001;29:611–620. [Google Scholar]
  • 23. Karnaukhova E. Recent advances in the research and development of alpha‐1 proteinase inhibitor for therapeutic use In: Irusen M, editor. Lung diseases ‐ selected state of the art reviews. London, United Kingdom: InTech, 2012; p. 83–104. [Google Scholar]
  • 24. Janciauskiene S, Welte T. Well‐known and less well‐known functions of alpha‐1 antitrypsin. Its role in chronic obstructive pulmonary disease and other disease developments. Ann Am Thorac Soc. 2016;13(Suppl 4):S280–S288. [DOI] [PubMed] [Google Scholar]
  • 25. Wanner A. Alpha‐1 antitrypsin as a therapeutic agent for conditions not associated with alpha‐1 antitrypsin deficiency In: Wanner A, Sandhaus RA, editors. Alpha‐1 antitrypsin: Role in health and disease. Cham: Springer International Publishing, 2016; p. 141–155. [Google Scholar]
  • 26. Duranton J, Bieth JG. Inhibition of proteinase 3 by [alpha]1‐antitrypsin in vitro predicts very fast inhibition in vivo. Am J Respir Cell Mol Biol. 2003;29:57–61. [DOI] [PubMed] [Google Scholar]
  • 27. Duranton J, Adam C, Bieth JG. Kinetic mechanism of the inhibition of cathepsin G by alpha 1‐antichymotrypsin and alpha 1‐proteinase inhibitor. Biochemistry. 1998;37:11239–11245. [DOI] [PubMed] [Google Scholar]
  • 28. Lockett AD, Van Demark M, Gu Y, et al. Effect of cigarette smoke exposure and structural modifications on the alpha‐1 antitrypsin interaction with caspases. Mol Med. 2012;18:445–454. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Shapiro L, Pott GB, Ralston AH. Alpha‐1‐antitrypsin inhibits human immunodeficiency virus type 1. FASEB J. 2001;15:115–122. [DOI] [PubMed] [Google Scholar]
  • 30. Kaner Z, Ochayon DE, Shahaf G, et al. Acute phase protein alpha1‐antitrypsin reduces the bacterial burden in mice by selective modulation of innate cell responses. J Infect Dis. 2015;211:1489–1498. [DOI] [PubMed] [Google Scholar]
  • 31. Ehlers MR. Immune‐modulating effects of alpha‐1 antitrypsin. Biol Chem. 2014;395:1187–1193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Pott GB, Chan ED, Dinarello CA, Shapiro L. Alpha‐1‐antitrypsin is an endogenous inhibitor of proinflammatory cytokine production in whole blood. J Leukoc Biol. 2009;85:886–895. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Janciauskiene SM, Nita IM, Stevens T. Alpha1‐antitrypsin, old dog, new tricks. Alpha1‐antitrypsin exerts in vitro anti‐inflammatory activity in human monocytes by elevating cAMP. J Biol Chem. 2007;282:8573–8582. [DOI] [PubMed] [Google Scholar]
  • 34. Bergin DA, Reeves EP, Hurley K, et al. The circulating proteinase inhibitor alpha‐1 antitrypsin regulates neutrophil degranulation and autoimmunity. Sci Transl Med. 2014;6:217ra211. [DOI] [PubMed] [Google Scholar]
  • 35. Bergin DA, Reeves EP, Meleady P, et al. Alpha‐1 antitrypsin regulates human neutrophil chemotaxis induced by soluble immune complexes and IL‐8. J Clin Invest. 2010;120:4236–4250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Gordon SM, McKenzie B, Kemeh G, et al. Rosuvastatin alters the proteome of high density lipoproteins: Generation of alpha‐1‐antitrypsin enriched particles with anti‐inflammatory properties. Mol Cell Proteomics. 2015;14:3247–3257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Rosenberg S, Barr PJ, Najarian RC, Hallewell RA. Synthesis in yeast of a functional oxidation‐resistant mutant of human alpha‐antitrypsin. Nature. 1984;312:77–80. [DOI] [PubMed] [Google Scholar]
  • 38. Elliott PR, Abrahams JP, Lomas DA. Wild‐type alpha 1‐antitrypsin is in the canonical inhibitory conformation. J Mol Biol. 1998;275:419–425. [DOI] [PubMed] [Google Scholar]
  • 39. Patston PA, Gettins PG, Schapira M. Serpins are suicide substrates: Implications for the regulation of proteolytic pathways. Semin Thromb Hemost. 1994;20:410–416. [DOI] [PubMed] [Google Scholar]
  • 40. Huntington JA. Serpin structure, function and dysfunction. J Thromb Haemost. 2011;9(Suppl 1):26–34. [DOI] [PubMed] [Google Scholar]
  • 41. Huntington JA. Thrombin inhibition by the serpins. J Thromb Haemost. 2013;11(Suppl 1):254–264. [DOI] [PubMed] [Google Scholar]
  • 42. Sifers RN. Intracellular processing of alpha1‐antitrypsin. Proc Am Thorac Soc. 2010;7:376–380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Casolaro MA, Fells G, Wewers M, et al. Augmentation of lung antineutrophil elastase capacity with recombinant human alpha‐1‐antitrypsin. J Appl Physiol. 1987;63:2015–2023. [DOI] [PubMed] [Google Scholar]
  • 44. Hopkins PC, Carrell RW, Stone SR. Effects of mutations in the hinge region of serpins. Biochemistry. 1993;32:7650–7657. [DOI] [PubMed] [Google Scholar]
  • 45. Avron A, Reeve FH, Lickorish JM, Carrell RW. Effect of alanine insertion (P'5) on the reactive centre of alpha 1‐antitrypsin. FEBS Lett. 1991;280:41–43. [DOI] [PubMed] [Google Scholar]
  • 46. Djie MZ, Stone SR, Le Bonniec BF. Intrinsic specificity of the reactive site loop of alpha1‐antitrypsin, alpha1‐antichymotrypsin, antithrombin III, and protease nexin I. J Biol Chem. 1997;272:16268–16273. [DOI] [PubMed] [Google Scholar]
  • 47. Luo LY, Jiang W. Inhibition profiles of human tissue kallikreins by serine protease inhibitors. Biol Chem. 2006;387:813–816. [DOI] [PubMed] [Google Scholar]
  • 48. Beatty K, Bieth J, Travis J. Kinetics of association of serine proteinases with native and oxidized alpha‐1‐proteinase inhibitor and alpha‐1‐antichymotrypsin. J Biol Chem. 1980;255:3931–3934. [PubMed] [Google Scholar]
  • 49. Felber LM, Kundig C, Borgono CA, et al. Mutant recombinant serpins as highly specific inhibitors of human kallikrein 14. FEBS J. 2006;273:2505–2514. [DOI] [PubMed] [Google Scholar]
  • 50. Ellis V, Scully M, MacGregor I, Kakkar V. Inhibition of human factor Xa by various plasma protease inhibitors. Biochim Biophys Acta. 1982;701:24–31. [PubMed] [Google Scholar]
  • 51. Scott CF, Schapira M, James HL, Cohen AB, Colman RW. Inactivation of factor XIa by plasma protease inhibitors: Predominant role of alpha 1‐protease inhibitor and protective effect of high molecular weight kininogen. J Clin Invest. 1982;69:844–852. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Mahrus S, Kisiel W, Craik CS. Granzyme M is a regulatory protease that inactivates proteinase inhibitor 9, an endogenous inhibitor of granzyme B. J Biol Chem. 2004;279:54275–54282. [DOI] [PubMed] [Google Scholar]
  • 53. van der Meer FJ, van Tilburg NH, van Wijngaarden A, van der Linden IK, Briet E, Bertina RM. A second plasma inhibitor of activated protein C: Alpha 1‐antitrypsin. Thromb Haemost. 1989;62:756–762. [PubMed] [Google Scholar]
  • 54. Al‐Omari M, Korenbaum E, Ballmaier M, et al. Acute‐phase protein alpha1‐antitrypsin inhibits neutrophil calpain I and induces random migration. Mol Med. 2011;17:865–874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Schechter I, Berger A. On the size of the active site in proteases. I. Papain. Biochem Biophys Res Commun. 1967;27:157–162. [DOI] [PubMed] [Google Scholar]
  • 56. Kounnas MZ, Church FC, Argraves WS, Strickland DK. Cellular internalization and degradation of antithrombin III‐thrombin, heparin cofactor II‐thrombin, and alpha 1‐antitrypsin‐trypsin complexes is mediated by the low density lipoprotein receptor‐related protein. J Biol Chem. 1996;271:6523–6529. [DOI] [PubMed] [Google Scholar]
  • 57. Strickland DK, Ranganathan S. Diverse role of LDL receptor‐related protein in the clearance of proteases and in signaling. J Thromb Haemost. 2003;1:1663–1670. [DOI] [PubMed] [Google Scholar]
  • 58. Dementiev A, Simonovic M, Volz K, Gettins PG. Canonical inhibitor‐like interactions explain reactivity of alpha1‐proteinase inhibitor Pittsburgh and antithrombin with proteinases. J Biol Chem. 2003;278:37881–37887. [DOI] [PubMed] [Google Scholar]
  • 59. Huntington JA, Read JR, Carrell RW. Structure of a serpin‐protease complex shows inhibition by deformation. Nature. 2000;407:923–926. [DOI] [PubMed] [Google Scholar]
  • 60. Dementiev A, Dobo J, Gettins PG. Active site distortion is sufficient for proteinase inhibition by serpins: Structure of the covalent complex of alpha1‐proteinase inhibitor with porcine pancreatic elastase. J Biol Chem. 2006;281:3452–3457. [DOI] [PubMed] [Google Scholar]
  • 61. Stratikos E, Gettins PG. Formation of the covalent serpin‐proteinase complex involves translocation of the proteinase by more than 70 A and full insertion of the reactive center loop into beta‐sheet A. Proc Natl Acad Sci U S A. 1999;96:4808–4813. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Hopkins PC, Stone SR. The contribution of the conserved hinge region residues of alpha1‐antitrypsin to its reaction with elastase. Biochemistry. 1995;34:15872–15879. [DOI] [PubMed] [Google Scholar]
  • 63. Hood DB, Huntington JA, Gettins PG. Alpha 1‐proteinase inhibitor variant T345R. Influence of P14 residue on substrate and inhibitory pathways. Biochemistry. 1994;33:8538–8547. [DOI] [PubMed] [Google Scholar]
  • 64. Lawrence DA, Olson ST, Muhammad S, et al. Partitioning of serpin‐proteinase reactions between stable inhibition and substrate cleavage is regulated by the rate of serpin reactive center loop insertion into beta‐sheet A. J Biol Chem. 2000;275:5839–5844. [DOI] [PubMed] [Google Scholar]
  • 65. Kaslik G, Kardos J, Szabo E, et al. Effects of serpin binding on the target proteinase: Global stabilization, localized increased structural flexibility, and conserved hydrogen bonding at the active site. Biochemistry. 1997;36:5455–5464. [DOI] [PubMed] [Google Scholar]
  • 66. Zhou A, Carrell RW, Huntington JA. The serpin inhibitory mechanism is critically dependent on the length of the reactive center loop. J Biol Chem. 2001;276:27541–27547. [DOI] [PubMed] [Google Scholar]
  • 67. Im H, Ahn HY, Yu MH. Bypassing the kinetic trap of serpin protein folding by loop extension. Protein Sci. 2000;9:1497–1502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Gettins PG, Olson ST. Inhibitory serpins. New insights into their folding, polymerization, regulation and clearance. Biochem J. 2016;473:2273–2293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Bottomley SP, Lawrenson ID, Tew D, Dai W, Whisstock JC, Pike RN. The role of strand 1 of the C beta‐sheet in the structure and function of alpha(1)‐antitrypsin. Protein Sci. 2001;10:2518–2524. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Quinsey NS, Fitton HL, Coughlin P, et al. Introduction of a mutation in the shutter region of antithrombin (Phe77 → Leu) increases affinity for heparin and decreases thermal stability. Biochemistry. 2003;42:10169–10173. [DOI] [PubMed] [Google Scholar]
  • 71. Bottomley SP, Stone SR. Protein engineering of chimeric serpins: An investigation into effects of the serpin scaffold and reactive centre loop length. Protein Eng. 1998;11:1243–1247. [DOI] [PubMed] [Google Scholar]
  • 72. Filion ML, Bhakta V, Nguyen LH, Liaw PS, Sheffield WP. Full or partial substitution of the reactive center loop of alpha‐1‐proteinase inhibitor by that of heparin cofactor II: P1 Arg is required for maximal thrombin inhibition. Biochemistry. 2004;43:14864–14872. [DOI] [PubMed] [Google Scholar]
  • 73. Chaillan‐Huntington CE, Gettins PG, Huntington JA, Patston PA. The P6‐P2 region of serpins is critical for proteinase inhibition and complex stability. Biochemistry. 1997;36:9562–9570. [DOI] [PubMed] [Google Scholar]
  • 74. Plotnick MI, Schechter NM, Wang ZM, Liu X, Rubin H. Role of the P6‐P3' region of the serpin reactive loop in the formation and breakdown of the inhibitory complex. Biochemistry. 1997;36:14601–14608. [DOI] [PubMed] [Google Scholar]
  • 75. Andersen OJ, Risor MW, Poulsen EC, et al. Reactive center loop insertion in alpha‐1‐antitrypsin captured by accelerated molecular dynamics simulation. Biochemistry. 2017;56:634–646. [DOI] [PubMed] [Google Scholar]
  • 76. Olson ST, Bjork I, Shore JD. Kinetic characterization of heparin‐catalyzed and uncatalyzed inhibition of blood coagulation proteinases by antithrombin. Methods Enzymol. 1993;222:525–559. [DOI] [PubMed] [Google Scholar]
  • 77. Sutherland JS, Bhakta V, Filion ML, Sheffield WP. The transferable tail: Fusion of the N‐terminal acidic extension of heparin cofactor II to alpha1‐proteinase inhibitor M358R specifically increases the rate of thrombin inhibition. Biochemistry. 2006;45:11444–11452. [DOI] [PubMed] [Google Scholar]
  • 78. Scott BM, Matochko WL, Gierczak RF, Bhakta V, Derda R, Sheffield WP. Phage display of the serpin alpha‐1 proteinase inhibitor randomized at consecutive residues in the reactive Centre loop and biopanned with or without thrombin. PLoS One. 2014;9:e84491. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Kwon KS, Kim J, Shin HS, Yu MH. Single amino acid substitutions of alpha 1‐antitrypsin that confer enhancement in thermal stability. J Biol Chem. 1994;269:9627–9631. [PubMed] [Google Scholar]
  • 80. Kim J, Lee KN, Yi GS, Yu MH. A thermostable mutation located at the hydrophobic core of alpha 1‐antitrypsin suppresses the folding defect of the Z‐type variant. J Biol Chem. 1995;270:8597–8601. [DOI] [PubMed] [Google Scholar]
  • 81. Irving JA, Haq I, Dickens JA, Faull SV, Lomas DA. Altered native stability is the dominant basis for susceptibility of alpha1‐antitrypsin mutants to polymerization. Biochem J. 2014;460:103–115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Lee KN, Im H, Kang SW, Yu MH. Characterization of a human alpha1‐antitrypsin variant that is as stable as ovalbumin. J Biol Chem. 1998;273:2509–2516. [DOI] [PubMed] [Google Scholar]
  • 83. Lee KN, Park SD, Yu MH. Probing the native strain in alpha1‐antitrypsin. Nat Struct Biol. 1996;3:497–500. [DOI] [PubMed] [Google Scholar]
  • 84. Seo EJ, Im H, Maeng JS, Kim KE, Yu MH. Distribution of the native strain in human alpha 1‐antitrypsin and its association with protease inhibitor function. J Biol Chem. 2000;275:16904–16909. [DOI] [PubMed] [Google Scholar]
  • 85. Shin JS, Ryu SH, Lee C, Yu MH. Misfolding‐assisted selection of stable protein variants using phage displays. J Biochem Mol Biol. 2006;39:55–60. [DOI] [PubMed] [Google Scholar]
  • 86. Griffiths SW, King J, Cooney CL. The reactivity and oxidation pathway of cysteine 232 in recombinant human alpha 1‐antitrypsin. J Biol Chem. 2002;277:25486–25492. [DOI] [PubMed] [Google Scholar]
  • 87. Bottomley SP, Hopkins PC, Whisstock JC. Alpha 1‐antitrypsin polymerisation can occur by both loop A and C sheet mechanisms. Biochem Biophys Res Commun. 1998;251:1–5. [DOI] [PubMed] [Google Scholar]
  • 88. Peterson FC, Gordon NC, Gettins PG. Formation of a noncovalent serpin‐proteinase complex involves no conformational change in the serpin. Use of 1H‐15N HSQC NMR as a sensitive nonperturbing monitor of conformation. Biochemistry. 2000;39:11884–11892. [DOI] [PubMed] [Google Scholar]
  • 89. Porebski BT, Keleher S, Hollins JJ, et al. Smoothing a rugged protein folding landscape by sequence‐based redesign. Sci Rep. 2016;6:33958. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Yang L, Irving JA, Dai W, Aguilar MI, Bottomley SP. Probing the folding pathway of a consensus serpin using single tryptophan mutants. Sci Rep. 2018;8:2121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Marijanovic EM, Fodor J, Riley BT, et al. Reactive centre loop dynamics and serpin specificity. Sci Rep. 2019;9:3870. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Pearce MC, Cabrita LD. Production of recombinant serpins in Escherichia coli . Methods Enzymol. 2011;501:13–28. [DOI] [PubMed] [Google Scholar]
  • 93. Courtney M, Buchwalder A, Tessier LH, et al. High‐level production of biologically active human alpha 1‐antitrypsin in Escherichia coli . Proc Natl Acad Sci U S A. 1984;81:669–673. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Tessier LH, Jallat S, Sauvageot M, Crystal RG, Courtney M. RNA structural elements for expression in Escherichia coli. Alpha 1‐antitrypsin synthesis using translation control elements based on the cII ribosome‐binding site of phage lambda. FEBS Lett. 1986;208:183–188. [DOI] [PubMed] [Google Scholar]
  • 95. Johansen H, Sutiphong J, Sathe G, et al. High‐level production of fully active human alpha 1‐antitrypsin in Escherichia coli . Mol Biol Med. 1987;4:291–305. [PubMed] [Google Scholar]
  • 96. Sutiphong J, Johansen H, Sathe G, Rosenberg GS, Shatzman A. Selection of mutations that increase alpha 1‐antitrypsin gene expression in Escherichia coli . Mol Biol Med. 1987;4:307–322. [PubMed] [Google Scholar]
  • 97. Schulze AJ, Degryse E, Speck D, Huber R, Bischoff R. Expression of alpha 1‐proteinase inhibitor in Escherichia coli: Effects of single amino acid substitutions in the active site loop on aggregate formation. J Biotechnol. 1994;32:231–238. [DOI] [PubMed] [Google Scholar]
  • 98. Haq I, Irving JA, Faull SV, et al. Reactive Centre loop mutants of alpha‐1‐antitrypsin reveal position‐specific effects on intermediate formation along the polymerization pathway. Biosci Rep. 2013;33:e00046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Schulze AJ, Huber R, Degryse E, Speck D, Bischoff R. Inhibitory activity and conformational transition of alpha 1‐proteinase inhibitor variants. Eur J Biochem. 1991;202:1147–1155. [DOI] [PubMed] [Google Scholar]
  • 100. Curtis H, Sandoval C, Oblin C, Difalco MR, Congote LF. Insect cell production of a secreted form of human alpha(1)‐proteinase inhibitor as a bifunctional protein which inhibits neutrophil elastase and has growth factor‐like activities. J Biotechnol. 2002;93:35–44. [DOI] [PubMed] [Google Scholar]
  • 101. Sandoval C, Curtis H, Congote LF. Enhanced proliferative effects of a baculovirus‐produced fusion protein of insulin‐like growth factor and alpha(1)‐proteinase inhibitor and improved anti‐elastase activity of the inhibitor with glutamate at position 351. Protein Eng. 2002;15:413–418. [DOI] [PubMed] [Google Scholar]
  • 102. Agarwal S, Jha S, Sanyal I, Amla DV. Expression and purification of recombinant human alpha1‐proteinase inhibitor and its single amino acid substituted variants in Escherichia coli for enhanced stability and biological activity. J Biotechnol. 2010;147:64–72. [DOI] [PubMed] [Google Scholar]
  • 103. Kang HA, Nam SW, Kwon KS, Chung BH, Yu MH. High‐level secretion of human alpha 1‐antitrypsin from Saccharomyces cerevisiae using inulinase signal sequence. J Biotechnol. 1996;48:15–24. [DOI] [PubMed] [Google Scholar]
  • 104. Agarwal S, Singh R, Sanyal I, Amla DV. Expression of modified gene encoding functional human alpha‐1‐antitrypsin protein in transgenic tomato plants. Transgenic Res. 2008;17:881–896. [DOI] [PubMed] [Google Scholar]
  • 105. Zhang N, Wright T, Caraway P, Xu J. Enhanced secretion of human alpha1‐antitrypsin expressed with a novel glycosylation module in tobacco BY‐2 cell culture. Bioengineered. 2019;10:87–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Paterson T, Innes J, Moore S. Approaches to maximizing stable expression of alpha 1‐antitrypsin in transformed CHO cells. Appl Microbiol Biotechnol. 1994;40:691–698. [DOI] [PubMed] [Google Scholar]
  • 107. Blanchard V, Liu X, Eigel S, et al. N‐glycosylation and biological activity of recombinant human alpha1‐antitrypsin expressed in a novel human neuronal cell line. Biotechnol Bioeng. 2011;108:2118–2128. [DOI] [PubMed] [Google Scholar]
  • 108. Amann T, Hansen AH, Kol S, et al. Glyco‐engineered CHO cell lines producing alpha‐1‐antitrypsin and C1 esterase inhibitor with fully humanized N‐glycosylation profiles. Metab Eng. 2019;52:143–152. [DOI] [PubMed] [Google Scholar]
  • 109. Janus ED, Phillips NT, Carrell RW. Smoking, lung function, and alpha 1‐antitrypsin deficiency. Lancet. 1985;1:152–154. [DOI] [PubMed] [Google Scholar]
  • 110. Li Z, Alam S, Wang J, Sandstrom CS, Janciauskiene S, Mahadeva R. Oxidized {alpha}1‐antitrypsin stimulates the release of monocyte chemotactic protein‐1 from lung epithelial cells: Potential role in emphysema. Am J Physiol Lung Cell Mol Physiol. 2009;297:L388–L400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Taggart C, Cervantes‐Laurean D, Kim G, et al. Oxidation of either methionine 351 or methionine 358 in alpha 1‐antitrypsin causes loss of anti‐neutrophil elastase activity. J Biol Chem. 2000;275:27258–27265. [DOI] [PubMed] [Google Scholar]
  • 112. Travis J, Owen M, George P, et al. Isolation and properties of recombinant DNA produced variants of human alpha 1‐proteinase inhibitor. J Biol Chem. 1985;260:4384–4389. [PubMed] [Google Scholar]
  • 113. Jallat S, Carvallo D, Tessier LH, et al. Altered specificities of genetically engineered alpha 1 antitrypsin variants. Protein Eng. 1986;1:29–35. [DOI] [PubMed] [Google Scholar]
  • 114. Silberstein DZ, Karuppanan K, Aung HH, Chen CH, Cross CE, McDonald KA. An oxidation‐resistant, recombinant alpha‐1 antitrypsin produced in Nicotiana benthamiana . Free Radic Biol Med. 2018;120:303–310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Zhu W, Li L, Deng M, et al. Oxidation‐resistant and thermostable forms of alpha‐1 antitrypsin from Escherichia coli inclusion bodies. FEBS Open Bio. 2018;8:1711–1721. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Lewis JH, Iammarino RM, Spero JA, Hasiba U. Antithrombin Pittsburgh: An alpha1‐antitrypsin variant causing hemorrhagic disease. Blood. 1978;51:129–137. [PubMed] [Google Scholar]
  • 117. Owen M, Brennan S, Lewis J, Carrell R. Mutation of antitrypsin to antithrombin. Alpha 1‐antitrypsin Pittsburgh (358 Met leads to Arg), a fatal bleeding disorder. N Engl J Med. 1983;309:649–698. [DOI] [PubMed] [Google Scholar]
  • 118. Scott CF, Carrell RW, Glaser CB, Kueppers F, Lewis JH, Colman RW. Alpha‐1‐antitrypsin‐Pittsburgh. A potent inhibitor of human plasma factor XIa, kallikrein, and factor XIIf. J Clin Invest. 1986;77:631–634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Colman RW, Flores DN, De La Cadena RA, et al. Recombinant alpha 1‐antitrypsin Pittsburgh attenuates experimental gram‐negative septicemia. Am J Pathol. 1988;130:418–426. [PMC free article] [PubMed] [Google Scholar]
  • 120. Harper PL, Taylor FB, DeLa Cadena RA, Courtney M, Colman RW, Carrell RW. Recombinant antitrypsin Pittsburgh undergoes proteolytic cleavage during E. coli sepsis and fails to prevent the associated coagulopathy in a primate model. Thromb Haemost. 1998;80:816–821. [PubMed] [Google Scholar]
  • 121. Heeb MJ, Bischoff R, Courtney M, Griffin JH. Inhibition of activated protein C by recombinant alpha 1‐antitrypsin variants with substitution of arginine or leucine for methionine358. J Biol Chem. 1990;265:2365–2369. [PubMed] [Google Scholar]
  • 122. Hopkins PC, Crowther DC, Carrell RW, Stone SR. Development of a novel recombinant serpin with potential antithrombotic properties. J Biol Chem. 1995;270:11866–11871. [DOI] [PubMed] [Google Scholar]
  • 123. Hopkins PC, Pike RN, Stone SR. Evolution of serpin specificity: Cooperative interactions in the reactive‐site loop sequence of antithrombin specifically restrict the inhibition of activated protein C. J Mol Evol. 2000;51:507–515. [DOI] [PubMed] [Google Scholar]
  • 124. Sutherland JS, Bhakta V, Sheffield WP. The appended tail region of heparin cofactor II and additional reactive Centre loop mutations combine to increase the reactivity and specificity of alpha1‐proteinase inhibitor M358R for thrombin. Thromb Haemost. 2007;98:1014–1023. [PubMed] [Google Scholar]
  • 125. Sheffield WP, Eltringham‐Smith LJ, Bhakta V, Gataiance S. Reduction of thrombus size in murine models of thrombosis following administration of recombinant alpha1‐proteinase inhibitor mutant proteins. Thromb Haemost. 2012;107:972–984. [DOI] [PubMed] [Google Scholar]
  • 126. Roddick LA, Bhakta V, Sheffield WP. Fusion of the C‐terminal triskaidecapeptide of hirudin variant 3 to alpha1‐proteinase inhibitor M358R increases the serpin‐mediated rate of thrombin inhibition. BMC Biochem. 2013;14:31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. de Souza LR, Scott BM, Bhakta V, Donkor DA, Perruzza DL, Sheffield WP. Serpin phage display: The use of a T7 system to probe reactive center loop libraries with different serine proteinases. Methods Mol Biol. 2018;1826:41–64. [DOI] [PubMed] [Google Scholar]
  • 128. Izaguirre G, Rezaie AR, Olson ST. Engineering functional antithrombin exosites in alpha1‐proteinase inhibitor that specifically promote the inhibition of factor Xa and factor IXa. J Biol Chem. 2009;284:1550–1558. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129. Izaguirre G, Olson ST. Residues Tyr253 and Glu255 in strand 3 of beta‐sheet C of antithrombin are key determinants of an exosite made accessible by heparin activation to promote rapid inhibition of factors Xa and IXa. J Biol Chem. 2006;281:13424–13432. [DOI] [PubMed] [Google Scholar]
  • 130. Yang L, Dinarvand P, Qureshi SH, Rezaie AR. Engineering D‐helix of antithrombin in alpha‐1‐proteinase inhibitor confers antiinflammatory properties on the chimeric serpin. Thromb Haemost. 2014;112:164–175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131. Polderdijk SG, Adams TE, Ivanciu L, Camire RM, Baglin TP, Huntington JA. Design and characterization of an APC‐specific serpin for the treatment of hemophilia. Blood. 2017;129:105–113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132. Shapiro AD, Mitchell IS, Nasr S. The future of bypassing agents for hemophilia with inhibitors in the era of novel agents. J Thromb Haemost. 2018;16:2362–2374. [DOI] [PubMed] [Google Scholar]
  • 133. Polderdijk SGI, Huntington JA. Identification of serpins specific for activated protein C using a lysate‐based screening assay. Sci Rep. 2018;8:8793. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134. de Maat S, Sanrattana W, Mailer RK, et al. Design and characterization of alpha1‐antitrypsin variants for treatment of contact system‐driven thromboinflammation. Blood. 2019;134(19):1658–1669. 10.1182/blood.2019000481. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135. Schapira M, Ramus MA, Waeber B, et al. Protection by recombinant alpha 1‐antitrypsin Ala357 Arg358 against arterial hypotension induced by factor XII fragment. J Clin Invest. 1987;80:582–585. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136. Sulikowski T, Bauer BA, Patston PA. Alpha(1)‐proteinase inhibitor mutants with specificity for plasma kallikrein and C1s but not C1. Protein Sci. 2002;11:2230–2236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137. Thomas G. Furin at the cutting edge: From protein traffic to embryogenesis and disease. Nat Rev Mol Cell Biol. 2002;3:753–766. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138. Anderson ED, Thomas L, Hayflick JS, Thomas G. Inhibition of HIV‐1 gp160‐dependent membrane fusion by a furin‐directed alpha 1‐antitrypsin variant. J Biol Chem. 1993;268:24887–24891. [PubMed] [Google Scholar]
  • 139. Bahbouhi B, Bendjennat M, Guetard D, Seidah NG, Bahraoui E. Effect of alpha‐1 antitrypsin Portland variant (alpha 1‐PDX) on HIV‐1 replication. Biochem J. 2000;352:91–98. [PMC free article] [PubMed] [Google Scholar]
  • 140. Watanabe M, Hirano A, Stenglein S, Nelson J, Thomas G, Wong TC. Engineered serine protease inhibitor prevents furin‐catalyzed activation of the fusion glycoprotein and production of infectious measles virus. J Virol. 1995;69:3206–3210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141. Bassi DE, Lopez De Cicco R, Mahloogi H, Zucker S, Thomas G, Klein‐Szanto AJ. Furin inhibition results in absent or decreased invasiveness and tumorigenicity of human cancer cells. Proc Natl Acad Sci U S A. 2001;98:10326–10331. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142. Yakala GK, Cabrera‐Fuentes HA, Crespo‐Avilan GE, et al. FURIN inhibition reduces vascular remodeling and atherosclerotic lesion progression in mice. Arterioscler Thromb Vasc Biol. 2019;39:387–401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143. Dufour EK, Desilets A, Longpre JM, Leduc R. Stability of mutant serpin/furin complexes: Dependence on pH and regulation at the deacylation step. Protein Sci. 2005;14:303–315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144. Tsuji A, Kanie H, Makise H, Yuasa K, Nagahama M, Matsuda Y. Engineering of alpha1‐antitrypsin variants selective for subtilisin‐like proprotein convertases PACE4 and PC6: Importance of the P2' residue in stable complex formation of the serpin with proprotein convertase. Protein Eng Des Sel. 2007;20:163–170. [DOI] [PubMed] [Google Scholar]
  • 145. Izaguirre G, Qi L, Lima M, Olson ST. Identification of serpin determinants of specificity and selectivity for furin inhibition through studies of alpha1PDX (alpha1‐protease inhibitor Portland)‐serpin B8 and furin active‐site loop chimeras. J Biol Chem. 2013;288:21802–21814. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146. Izaguirre G, Arciniega M, Quezada AG. Specific and selective inhibitors of proprotein convertases engineered by transferring serpin B8 reactive‐site and exosite determinants of reactivity to the serpin alpha1PDX. Biochemistry. 2019;58:1679–1688. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147. Irving JA, Pike RN, Dai W, et al. Evidence that serpin architecture intrinsically supports papain‐like cysteine protease inhibition: Engineering alpha(1)‐antitrypsin to inhibit cathepsin proteases. Biochemistry. 2002;41:4998–5004. [DOI] [PubMed] [Google Scholar]
  • 148. Campos MA, Kueppers F, Stocks JM, et al. Safety and pharmacokinetics of 120 mg/kg versus 60 mg/kg weekly intravenous infusions of alpha‐1 proteinase inhibitor in alpha‐1 antitrypsin deficiency: A multicenter, randomized, double‐blind, crossover study (SPARK). COPD. 2013;10:687–695. [DOI] [PubMed] [Google Scholar]
  • 149. Cantin AM, Woods DE, Cloutier D, Dufour EK, Leduc R. Polyethylene glycol conjugation at Cys232 prolongs the half‐life of alpha1 proteinase inhibitor. Am J Respir Cell Mol Biol. 2002;27:659–665. [DOI] [PubMed] [Google Scholar]
  • 150. Lindhout T, Iqbal U, Willis LM, et al. Site‐specific enzymatic polysialylation of therapeutic proteins using bacterial enzymes. Proc Natl Acad Sci U S A. 2011;108:7397–7402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151. Lusch A, Kaup M, Marx U, Tauber R, Blanchard V, Berger M. Development and analysis of alpha 1‐antitrypsin neoglycoproteins: The impact of additional N‐glycosylation sites on serum half‐life. Mol Pharm. 2013;10:2616–2629. [DOI] [PubMed] [Google Scholar]
  • 152. Chung HS, Kim JS, Lee SM, Park SJ. Additional N‐glycosylation in the N‐terminal region of recombinant human alpha‐1 antitrypsin enhances the circulatory half‐life in Sprague‐Dawley rats. Glycoconj J. 2016;33:201–208. [DOI] [PubMed] [Google Scholar]
  • 153. Rath T, Baker K, Dumont JA, et al. Fc‐fusion proteins and FcRn: Structural insights for longer‐lasting and more effective therapeutics. Crit Rev Biotechnol. 2015;35:235–254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154. Lee S, Lee Y, Hong K, et al. Effect of recombinant alpha1‐antitrypsin Fc‐fused (AAT‐Fc)protein on the inhibition of inflammatory cytokine production and streptozotocin‐induced diabetes. Mol Med. 2013;19:65–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155. Jonigk D, Al‐Omari M, Maegel L, et al. Anti‐inflammatory and immunomodulatory properties of alpha1‐antitrypsin without inhibition of elastase. Proc Natl Acad Sci U S A. 2013;110:15007–15012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156. Toldo S, Mauro AG, Marchetti C, et al. Recombinant human alpha‐1 antitrypsin‐fc fusion protein reduces mouse myocardial inflammatory injury after ischemia‐reperfusion independent of elastase inhibition. J Cardiovasc Pharmacol. 2016;68:27–32. [DOI] [PubMed] [Google Scholar]
  • 157. Joosten LA, Crisan TO , Azam T, et al. Alpha‐1‐anti‐trypsin‐fc fusion protein ameliorates gouty arthritis by reducing release and extracellular processing of IL‐1beta and by the induction of endogenous IL‐1Ra. Ann Rheum Dis. 2016;75:1219–1227. [DOI] [PubMed] [Google Scholar]
  • 158. The Safety and Tolerability of SerpinPC in Healthy Men and in Men With Severe Blood Disorders (Haemophilia A and B) . [accessed October 22, 2019]. https://clinicaltrials.gov/ct2/show/NCT04073498.
  • 159. Phase 1 Study to Assess the Safety , PK and PD of INBRX‐101 in Adults With Alpha‐1 Antitrypsin Deficiency. [accessed October 21, 2019]. https://clinicaltrials.gov/show/NCT03815396.
  • 160. Tardif JC, L'Allier PL, Gregoire J, et al. A randomized controlled, phase 2 trial of the viral serpin Serp‐1 in patients with acute coronary syndromes undergoing percutaneous coronary intervention. Circ Cardiovasc Interv. 2010;3:543–548. [DOI] [PubMed] [Google Scholar]
  • 161. Chaillan‐Huntington CE, Patston PA. Influence of the P5 residue on alpha1‐proteinase inhibitor mechanism. J Biol Chem. 1998;273:4569–4573. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplemental Table 1 An Excel file containing a descriptive list of over 200 published engineered α1‐AT variants (Scott_Sheffield_SupTable1_191112.xlsx). The list is non‐exhaustive, meant to provide an overview of the diversity of α1‐AT mutants published. The most recent version is available via Figshare https://doi.org/10.6084/m9.figshare.10294889


Articles from Protein Science : A Publication of the Protein Society are provided here courtesy of The Protein Society

RESOURCES