You are on page 1of 26

Preparative Biochemistry & Biotechnology

ISSN: (Print) (Online) Journal homepage: www.tandfonline.com/journals/lpbb20

Bioprocessing of recombinant proteins from


Escherichia coli inclusion bodies: insights from
structure-function relationship for novel
applications

Kajal Kachhawaha, Santanu Singh, Khyati Joshi, Priyanka Nain & Sumit K.
Singh

To cite this article: Kajal Kachhawaha, Santanu Singh, Khyati Joshi, Priyanka Nain & Sumit K.
Singh (2023) Bioprocessing of recombinant proteins from Escherichia coli inclusion bodies:
insights from structure-function relationship for novel applications, Preparative Biochemistry &
Biotechnology, 53:7, 728-752, DOI: 10.1080/10826068.2022.2155835

To link to this article: https://doi.org/10.1080/10826068.2022.2155835

Published online: 19 Dec 2022.

Submit your article to this journal

Article views: 721

View related articles

View Crossmark data

Citing articles: 1 View citing articles

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=lpbb20
PREPARATIVE BIOCHEMISTRY & BIOTECHNOLOGY
2023, VOL. 53, NO. 7, 728–752
https://doi.org/10.1080/10826068.2022.2155835

Bioprocessing of recombinant proteins from Escherichia coli inclusion bodies:


insights from structure-function relationship for novel applications
Kajal Kachhawahaa, Santanu Singha, Khyati Joshia, Priyanka Nainb, and Sumit K. Singha
a
School of Biochemical Engineering, Indian Institute of Technology (Banaras Hindu University), Varanasi, India; bDepartment of Chemical and
Bimolecular Engineering, University of Delaware, Newark, DE, USA

ABSTRACT KEYWORDS
The formation of inclusion bodies (IBs) during expression of recombinant therapeutic proteins Inclusion bodies; refolding;
using E. coli is a significant hurdle in producing high-quality, safe, and efficacious medicines. The E. coli; downstream
improved understanding of the structure-function relationship of the IBs has resulted in the devel- purification; thera-
peutic proteins
opment of novel biotechnologies that have streamlined the isolation, solubilization, refolding, and
purification of the active functional proteins from the bacterial IBs. Together, this overall effort
promises to radically improve the scope of experimental biology of therapeutic protein production
and expand new prospects in IBs usage. Notably, the IBs are increasingly used for applications in
more pristine areas such as drug delivery and material sciences. In this review, we intend to pro-
vide a comprehensive picture of the bio-processing of bacterial IBs, including assessing critical
gaps that still need to be addressed and potential solutions to overcome them. We expect this
review to be a useful resource for those working in the area of protein refolding and therapeutic
protein production.

Introduction efficacy at a large scale. Despite these developments, protein


refolding, for a large part, is still carried out using conven-
Protein-based therapeutics have become the cornerstone of
modern-day healthcare. Both mammalian[1] and microbial tional time-based dilution methods.[7]
Some recent encouraging developments include the
expression systems[2] are mainly utilized to express recom-
binant proteins. However, a comprehensive understanding broader acceptance among the stakeholders that the IBs are
of the complete physiology of E. coli and the availability of a inevitable during recombinant protein production. With this
plethora of strains that are easier to grow makes microbial reality, the community is striving to innovate and develop
hosts the preferred platform for expressing more than 25% bioprocesses that are envisioned as a universal method for
of the recombinant proteins.[3] recovering active functional protein from bacterial IBs.
A recombinant protein must be expressed in its native Further, several ways to isolate IBs have been developed that
3D form to be an effective therapeutic. However, the higher are harsh enough to break open the bacterial cells without
productivity achievable with E. coli as host results in the disturbing the structural integrity of the proteins. The use of
over-expression of recombinant proteins.[4] The overexpres- mild solubilization agents has resulted in a multi-fold
sion of proteins invariably results in an inactive, misfolded improvement in the efficiency of bioprocessing of the IBs.[8]
form of the protein, commonly referred to as inclusion Additionally, the advent and wide use of novel chromatog-
bodies (IBs).[5] This necessitates refolding the IBs under ena- raphy formats such as multi-modal chromatography[9] have
bling conditions to generate an active, functional form of
opened avenues for separating molecular variants of the tar-
the protein.
get product yielding highly homogenous and pure thera-
While IBs are inherently very stable and are easy to tackle
during downstream purification, yet protein refolding under peutic proteins.
in-vitro conditions is a non-trivial process as it involves sev- The availability of sophisticated and cutting-edge analyt-
eral structural transitions from the assembly of the amino ical tools has enabled the study of several proteins’ struc-
acid chain to the formation of the higher order structure.[6] ture-function relationships with IB.[10] This information has
With the ever-growing demand for biotherapeutics for cater- not only facilitated to design of efficient bioprocessing
ing to several unmet medical needs, considerable attention schemes for IBs, but also opened newer frontiers for the
has been given to innovation of process development for application of the IBs in areas such as drug delivery and
producing protein therapeutics of desired quality, safety, and material sciences.[11]

CONTACT Sumit K. Singh sumit.bce@iitbhu.ac.in School of Biochemical Engineering, Indian Institute of Technology (Banaras Hindu University), Varanasi,
221005, India.
Equal contribution
ß 2022 Taylor & Francis Group, LLC
PREPARATIVE BIOCHEMISTRY & BIOTECHNOLOGY 729

This article discusses different types of bacterial IBs formed reversible[26] and achieved to its native state by downstream
during the recombinant expression of proteins and their mor- processing (Figure 1).
phological, biochemical, and biophysical characteristics. We
also discuss various aspects of the bioprocessing of IBs,
including isolation, solubilization, refolding, purification, and Composition and localized cellular formation of IBs
characterization. A great emphasis is given to innovations and The bacterial IBs are heterogeneous, comprising an ensem-
developments made in the last 5 years. Further, we analyzed ble of target protein conformations and host cell protein
the current challenges in the bioprocessing of IBs and sug- contaminants.[27] Various protein conformations include
gested approaches to overcome these hurdles. The use of IBs amyloid-like fibrils, protofibrils, misfolded proteins, partially
to design novel biomaterials and as a self-releasing drug deliv- folded proteins, and a fraction of the native target pro-
ery vehicle is also discussed. We expect that the information tein.[28] On the other hand, host cell contaminants include
consolidated in this paper will generate a deeper understand- membrane proteins, cytoplasmic proteins, chaperones, and
ing of the current developments, challenges, and opportunities genetic material wrapped in IBs via nonspecific intermolecu-
in the bioprocessing of bacterial IBs and allow them to suc- lar interactions[29] (Figure 2). It is to be noted that conform-
cessfully capitalize on these aspects to develop therapeutic ational impurities overwhelm cellular impurities as bacterial
proteins with consistent quality, safety, and efficacy during IBs contain very low levels of host cell proteins, ribosomal
commercial manufacturing. components, and DNA/RNA fragments.[30]
IBs are deposited at bacterial cells’ center and polar
end.[31] There are two hypotheses about IB formation in
General morphological characteristics of bacterial
bacterial cells. According to one view, the IB formation
inclusion bodies
occurs passively (without any energy expenditure) by the
Bacterial IBs are expressed in cytoplasmic or periplasmic bacterial nucleoid’s volumetric exclusion effect, leading to
space inside the bacterial cells.[12,13] The diameter of IBs the accumulation of IBs in the polar ends of the host bacter-
spans in the range of 0.2–1.2 mm[14] encompassing round, ial cell.[32] The localization of IBs within the bacterial cells
oval, spherical, cylindrical, and ellipsoidal shapes. IBs exist depends on nucleoid size, IBs aggregate size, and cytoplas-
as dense electron refractile particles of aggregated protein mic mobility of the bacterial cell.[33] Cytoplasmic mobility
during high-level expression of recombinant proteins. reduces the anisotropy of IB aggregates at the midpoint of
Depending on the localization, the IBs exhibit amorphous or the cell (near the nucleoid border), eventually leading to
para-crystalline nature.[15] their accumulation along the polar ends of the cell.[34]
Inclusion bodies have a higher density (1.3 mg/mL)[16] The other hypothesis postulates an energy-dependent
than many of the cellular components and thus can be easily process[35] where IBs formation is believed to be a multi-
separated by high-speed centrifugation after cell disruption. step process. The series of steps includes the formation of
Despite being dense particles that are highly hydrated, IBs several small aggregates that eventually agglomerates into
have a porous architecture.[17] Another characteristic of IB the large aggregate size and localization in the polar ends of
shape is the presence of either rough or smooth surfaces, the bacterial cells. There is a substantial expenditure of
which affects the solubilization process.[18] The degree of solu- energy in the ensuing process that is modulated by chaper-
bilization depends on the protein-protein interaction (hydro- ones (DnaK and DnaJ), cytoskeleton (MreB), and proton
phobic interaction, electrostatic forces, hydrogen bonding, and motive force (pmf).[36,37] Both hypotheses (energy-depend-
van-der Waal forces) between IB molecules. Cytoplasmic IBs ent/energy-independent) are accepted and hold for various
have stronger protein-protein interaction than periplasmic bioprocess conditions during large-scale heterologous pro-
IBs. Thus, periplasmic IBs are solubilized with low denaturant tein expression in E. coli.[32,35]
concentration compared to cytoplasmic IBs.[19]
IBs lack well-defined 3D structures like functional pro-
Classification of inclusion bodies
teins. The origins of the IB formation during protein expres-
sion can be traced to the lack of post-translational The bacterial IBs formed during heterologous expression[38]
modifications such as glycosylation in E. coli.[20] The role of of recombinant proteins, often called classical IBs, are dense
glycosylation in protein folding through various cellular particle-like entities that completely lack functional bioactive
compartments of a eukaryotic cell is well established.[21] The proteins. They also uniquely resist any protease-mediated
lack of cellular organelles, such as the endoplasmic reticu- degradation under a dense cellular environment.[39] These
lum and golgi apparatus in E. coli, impairs glycosylation IBs usually require very high concentrations of denaturants
yielding misfolded proteins.[22] Recent studies on engineered for solubilization and subsequent dilution of solubilized IBs
E. coli cells for the presence of glycosylation machinery also for refolding into the 3D active form of the protein.[40] The
indicate the formation of recombinant glycosylated pro- purified form of the target protein is obtained by subjecting
teins.[23] In general, pglB N-glycosylation machinery the refolded output of the protein to a series of purification
(obtained from C. jejuni)[24] and lsg locus machinery steps involving orthogonal chromatography-based separation
(obtained from H. influenzae)[25] are commonly used from the product (molecular and conformational variants)
machinery for transferring glycans to the acceptor sites of and process-related (host-cell proteins and host-cell DNA)
proteins in E. coli. IBs deposition in bacterial cells is impurities. Overall obtaining a purified protein from
730 K. KACHHAWAHA ET AL.

Figure 1. Bioprocessing steps of recombinant proteins expressed in E. coli. (1) Induction of recombinant proteins. (2) Isolation of expressed recombinant proteins.
(3) Washing of isolated proteins which contains the IBs and cellular contaminants, washed IB pellets was removed out from cellular contaminants. (4) Solubilization
of washed IBs which will denature the IBs aggregates. (5) Refolding of solubilized IBs. (6) Purification of refolded proteins, the native proteins are separated from
the mixture of misfolded, partially folded and aggregated proteins.

Figure 2. Composition of Inclusion bodies (IBs). IBs contain cellular protein contaminants and recombinant protein conformations. (a) Cellular protein contaminants
are composed of bacterial genetic material, cytoplasmic proteins, membrane proteins, and chaperones. (b) Recombinant protein conformations are composed of
native, misfolded, amyloid like fibrils and partially folded proteins.

classical IBs is a time-consuming, cost-intensive, and tech- of IBs referred to as non-classical IBs. In contrast to the
nically complex process.[41] classical IBs, the non-classical IBs are relatively less dense
To simplify the above mentioned issues, researchers have and often contain a small fraction of bioactive proteins.[42]
attempted to express the target proteins as a particular class The non-classical IBs require relatively very mild
PREPARATIVE BIOCHEMISTRY & BIOTECHNOLOGY 731

solubilization conditions. Active 3D correctly folded proteins Upstream processing aspects


can be obtained directly from diluting solubilized proteins
E. coli is the most preferred host for recombinant protein
without requiring additional refolding and purification
production.[50] This is attributed to its ability to express
steps.[43] For example, Trinh et al. reported a feasible E.
high titers of the target proteins (50% of bacterial proteins
coli-based expression platform for granulocyte colony-
and 15% of non-bacterial proteins[10] in the soluble form
stimulating factor (GCSF) production at low temperatures in under relatively simple growth conditions.[51] Over 120
the form of non-classical IBs. With the said approach, the approved therapeutic proteins have been expressed using E.
GCSF was expressed, IBs were effectively isolated, solubilized coli as a host, which bears testimony to their prominent
under very mild conditions, and refolded into native form roles as a potential expression system.[52]
by dilution.[44] The genome of E. coli is very simple, comprising 4400
The non-classical IBs, by their unique biophysical prop- genes.[53] This is significantly smaller than other commonly
erties, have been utilized for a wide range of applications, used expression systems, such as Pichia pastoris and Chinese
including drug delivery and the development of novel bio- Hamster Ovary (CHO) cells that are larger and more com-
materials for tissue engineering applications.[45] For plex in size.[15] The smaller genomic size of E. coli makes it
instance, nano-pills were synthesized using the non- amenable to uptake diverse genes of interest without com-
classical IBs used for the sustained release of protein promising productivity.[14] The doubling time of just
drugs.[46] Similarly, pegylation of GCSF was carried out 20 minutes in E. coli also facilitates faster culture campaigns
starting from the non-classical IBs for improved pharmaco- versus other expression systems, where a typical batch turn-
kinetic performance.[44] around time is 10–14 days.[51] With a deep understanding of
the E. coli genome, it is now possible to construct an opti-
mal expression platform for recombinant protein synthesis
Bioprocessing aspects of IB for therapeutic using E. coli as a host cell. It can be designed in such a way
protein production that it encompasses efficient transcription and translation
machinery.[54] In addition, it can also form proteins of inter-
E. coli is a primary workhorse for therapeutic protein pro- est in higher amounts while maintaining the transcript’s
duction. Because of the ensuing reducing environment, most stability.[54]
of the proteins expressed in E. coli are located in the cyto- Examples of therapeutic proteins expressed in E. coli
plasmic space. Under such reducing conditions, proteins include insulin, growth factors, hormones, cytokines, anti-
generally exist in the form of IBs due to non-native interac- body fragments, and nanobodies.[11,55] While E. coli systems
tions between amino acid residues leading to misfolded or exhibit excellent versatility in the heterologous expression of
aggregated proteins.[47] According to an estimate, >80% of proteins, several issues are discussed below that render the
recombinant proteins expressed in E. coli exist in the form expression of specific proteins a non-trivial task.
of IBs, indicating an inevitable feature of IB formation asso-
ciated with E. coli expression systems.[12]
The existing body of literature indicates that there have Parameters affecting therapeutic protein productivity in
been several attempts to minimize IB formation during pro- E. coli
Clone design. Plasmids are the most common vectors uti-
tein expression in E. coli. However, it has been realized that
lized in recombinant DNA technology. Properties such as its
embracing the IB formation during protein expression could
size, copy number and promotor strength significantly con-
offer several advantages for large-scale protein expression.
tribute to the recombinant protein production in the host
The primary benefits associated with the formation of inclu-
cell. The recombinant gene dosage is defined by the copy
sion bodies are (i) expression of a very high level of protein,
number of the plasmid. High gene dosage can either be
more than 30% of the cellular protein,[13] (ii) easy isolation
favorable or unfavorable for the expression of the recombin-
of the inclusion bodies from cells due to differences in their ant protein. Thus, a suitable plasmid should be selected,
size and density as compared with cellular contaminants,[48] keeping it mind the characteristics it possesses.[56,57] To
(iii) lower degradation of the expressed protein, (iv) resist- achieve a higher recombinant protein expression, the inter-
ance to proteolytic attack by cellular proteases on classical est gene should be cloned immediately downstream to a
IBs[49] and (v) homogeneity of the protein of interest in strong promoter.[58] Strong transcriptional promoters con-
inclusion bodies (lesser contaminants) which helps in reduc- trol the foreign gene in various plasmids and bacteriophage
ing the number of purification steps to recover pure pro- vectors.[58] These promoters do not show constitutive
tein.[13] Being an advantageous feature during the expression expression and are governed by adding specific metabolites
and bioprocessing of therapeutic proteins, IB formation, at or changing culture conditions.[59] These are highly regu-
least in a theoretical sense, is no longer considered a limit- lated in nature and also control the expression of the foreign
ing bottleneck in obtaining higher titers of therapeutic pro- gene in such a way that they do not interfere with the nor-
teins. This review provides a comprehensive overview of the mal functioning of cellular genes and are not toxic to the
key developments, challenges, and opportunities, including host cell.[60] If these promoters are not regulated, they may
upstream, downstream, and analytical characterization result in loss of plasmid carrying the strong promoter or
aspects of IB bioprocessing. may be expressed constitutively, which shows detrimental
732 K. KACHHAWAHA ET AL.

effects on the cell.[59] The tae promotor is the most com- glucose as the primary carbon source is preferred.[10]
monly used strong promotor.[58] Similarly, for T7-based systems, using pLysS/pLysE bearing
The selection of terminator sequence also plays an E. coli BL21(DE3) cells help in reducing protein toxicity by
important role in regulating recombinant protein synthesis. controlling the expression levels of T7 RNA polymerase.[13]
In most organisms, TAA, TAG, and TGA are utilized as On the contrary, for the cases of reduced expression due to
stop codons, but in E. coli, TAA is more preferentially used protein toxicity after induction, using tunable promoters
than the other two codons.[61] Efficient termination of tran- and reducing induction time are some of the reported
scription may lead to lower cellular energy expenditure approaches to enhance protein expression.[58] In addition,
reducing the host cell’s metabolic load.[41] There is one directing the protein expression to the E. coli periplasm and
more additional advantage of the transcription terminator as using a lower plasmid copy number during recombinant
it forms a secondary structure at the 30 end of mRNA, protein expression allows for utilizing the oxidative environ-
which helps improve mRNA stability and thereby enhances ment of the former and reducing the metabolic burden on
recombinant protein production.[62] Occasionally, the trans- the cells owing to the latter.[20] Further, increasing biomass
location of the protein to the periplasm is required for by optimizing and using novel media additives and provid-
proper protein folding and formation of disulfide bonds. ing appropriate culture conditions (aeration and anti-foam-
Signal peptides are added before target gene to direct the ing) also boosts the recombinant protein expression.[64]
movement of the protein toward the periplasm. Some plas- Inducers such as isopropyl b- d-1-thiogalactopyranoside
mids such as p5 series of pMAL vector and pET-22 vector (IPTG) also play an essential role in determining recombin-
contain the signal peptide at the 50 region of the MCS. ant protein yield. Inducer concentration and duration sig-
Addition of a signal peptide upstream of the target gene can nificantly impact overall production.[67] Using a lower
improve the recombinant protein expression.[63] concentration of inducer will lead to lower protein yield
resulting in inefficient induction, whereas using a higher
Codon optimization. Despite the excellent ability to accom- concentration of inducer will result in toxic effects on cells
modate a plethora of foreign genes, a major impediment and ultimately interfere with product yield.[50] It is also eco-
often observed during the expression of target proteins in E. nomically not feasible to use higher amounts of inducer.[50]
coli is codon bias. This bottleneck arises when significant The most suitable time for induction is the early mid-log
differences exist between the synonymous codon of the for- phase when cell density is at the maximum concentration
eign genes and the host.[17] An example of such a difference because cell density plays a crucial role in protein synthe-
is the occurrence of rare codons such as GGA (glycine), sis.[68] Some studies have also suggested that induction can
CUA (leucine), and AAG (lysine), which are comparatively occur in mid-log or stationary phases.[69] Reducing the
found in lesser frequency in E. coli than in eukaryotic induction temperature is also a frequently used method for
organisms.[64] obtaining soluble expressed proteins. At reduced tempera-
The low abundant rare codons in E. coli pose issues tures, bacteria exhibit a decreased rate of translation and
regarding misincorporation/truncation of amino acids in the correspondingly, less likelihood of misfolding and
newly synthesized polypeptide chain, thereby affecting over- aggregation.[70]
all yields.[17] Several strategies have been employed to solve
the issue of codon bias during heterologous protein expres- Growth conditions & IB formation. Although a higher rate
sion in E. coli. The most common strategy used is codon of protein synthesis is desirable in therapeutic production, it
optimization in the cDNA- a method that maximizes the can often lead to protein aggregate formation in the form of
codon usage of the host.[20] Codon optimization is usually inclusion bodies. When foreign DNA is introduced into E.
performed by site-directed mutagenesis of the rare codons coli, spatio-temporal regulation of its expression is lost. The
with the more abundant codons of E. coli. However, site- recombinant polypeptide is expressed in the E. coli micro-
directed mutagenesis is very costly and time-consuming.[65] environment, which may not be exactly the same as the
Therefore, a more attractive and economical approach source in terms of pH, temperature, redox potential, cofac-
recently gaining attention is the co-expression of a plasmid tors, and folding mechanisms. Additionally, in high level
containing the tRNA for rare codons, thereby minimizing expression, hydrophobic sections in the polypeptide are pre-
the ensuing loss in yields owing to codon biases.[66] Further, sent in large amounts and ready for interaction with related
several codon bias-adjusted strains of E. coli, such as BL21 areas during high level expression. All these parameters col-
(DE3), Codon Plus, and Rosetta (DE3), are commercially lectively cause the protein to aggregate.[71]
available, which effectively minimizes the issues of codon Such insoluble aggregate formation can be prevented by
bias during protein expression.[50] slowing down the protein synthesis rate by using a weak
promoter, using a low copy number plasmid, or production
Induction. In addition to the codon bias, low expression of at lower temperatures and extreme pH ranges.[54] Culture
proteins could be caused by protein toxicity either before or conditions can be manipulated to slow down or enhance the
after induction.[15] For the former, controlling basal induc- expression of recombinant protein in E. coli. Reducing the
tion by tightly regulated promoters enhances the protein growth temperature may help decrease the formation of
expression multi-fold. Also, for expression vectors contain- inclusion bodies because of the slower protein synthesis
ing lac-based promoters, the use of defined media with rate.[72] Moreover, the decrease in temperature prevents
PREPARATIVE BIOCHEMISTRY & BIOTECHNOLOGY 733

Figure 3. An illustration showing different cell compartments for protein synthesis and trafficking where cytoplasm being the most reducing environment and
both periplasmic and extracellular spaces are oxidizing in nature. The inner membrane incorporates transport proteins namely Tat and Sec complexes. Tat is capable
of transporting folded protein via TAT pathway whereas Sec is capable of transporting both unfolded/partially folded proteins via Sec-B pathway and co-transla-
tional transport via SRP pathway. N-terminal signal sequences are responsible for targeting their location and are cleaved once protein gets transported.

aggregation by reducing the hydrophobic interactions.[73] another study, the presence of metallic cofactor Ca2þ hemin
Reduced temperature can also be responsible for the pro- was found to positively influence the refolding of horserad-
longed induction time, increasing the expressed protein’s ish peroxidase.[79]
solubility.[58] This is due to the inactivity of most of the pro-
teases at lower temperatures, so they cannot cleave or
Strategies to minimize IB formation
degrade newly synthesized proteins.[72] Several studies report
Inclusion body (IB) formation is a major feature of protein
the improvement in expression of recombinant protein in E.
expression using E. coli that impacts productivity. IBs are
coli with the reduction in incubation temperature. Huang
formed for various reasons, including incorrect or non-clas-
et al., reported improvement in protein folding when the
sical disulfide bond formation, inability to attain the unique
incubation temperature was decreased from 37  C to 25  C. native conformation of the protein, low solubility, and lack
This led to an increase in the yield of nonstructural antigen of glycosylation in E. coli.[51]
protein 3 of the hepatitis C virus in E. coli from 4.15 to The current efforts to capitalize bacterial IBs without
11.1 mg/L.[74] In another study, higher fraction of soluble compromising product yield principally encompasses strat-
anti-HER2 scFv was attained when the E. coli culture was egies to translocate the newly formed IBs from the reducing
carried out at 25  C.[75] pH is also known to play an import- cytoplasmic environment to relatively more oxidative peri-
ant role in the correct folding of recombinant protein. pH plasmic or extracellular space.[80] The underlying reason is
manipulation influences IB formation and their physical as several advantages the periplasmic and extracellular space
well as chemical characteristics. Thus, pH should be opti- offers during the expression of functional therapeutic pro-
mized in such a way that IB formation can be minimized. Li teins. For instance, periplasmic space confers resistance to
and Cheng reported an increase in the aggregation of blue the protease-mediated degradation of the target proteins.[47]
fluorescent protein with decrease in pH.[76] In contrast, an Further, it streamlines downstream processing because very
increase in the culture pH was shown to increase the forma- few naturally occurring cellular proteins are present in E.
tion of inclusion bodies during salmon growth hormone coli periplasm[81] (Figure 3).
(SGH) production using E. coli.[77] The addition of cofactors However, since the volume of periplasm is only about
in the growth medium may also help in the proper folding 20% of the overall cell volume,[81] there is a limit to the
of some proteins, and a change in pH can affect the proteo- amount of expressed protein to be transported and accom-
lytic activity and secretion of specific proteins. It has been modated into this space. The issue assumes greater signifi-
reported that addition of FMN in the unfolded form of fla- cance under conditions of high protein expression when the
vodoxin speeds up its folding process by several times.[78] In target protein may start to leach out into the extracellular
734 K. KACHHAWAHA ET AL.

Figure 4. A simplified representation of cell-free protein expression system by using E. coli crude extract. Host cell DNA and cell debris are removed followed by
supplementing the reaction mixture with an energy source, cofactors, DNA template, and other components required for protein synthesis. The protein of interest
is expressed and further purified for bio- therapeutic development.

medium due to enhanced periplasmic permeability.[82] High cell density cultures


Under such conditions, extracellular secretion of the In the case of recombinant protein synthesis, cell density is
expressed proteins is the preferred alternative. The extracel- a major determinant of the overall yield of the product. Cell
lular secretion of the target proteins can be achieved by the density reflects the optimum cell growth by regulating cell-
fusion of the target protein with an N-terminal signal to-cell signaling and communication.[84] Cell growth will be
sequence, using chemical additives, modifying the induction sluggish without such interaction, leading to cell death.
procedure, or modulating the promoter of the expres- Further, such cellular interactions are also responsible for
sion vector.[82] transporting small molecules required for the proliferation
Another approach to reducing IB formation is by co- of cells in a growth medium.[85] Therefore, maintaining high
expression of molecular chaperones.[47] However, this comes cell density is a primary requisite for the growth of micro-
at the cost of enhanced process complexity and issues in bial cells and the production of recombinant proteins in
downstream purification of the target protein. The fusion of high titers.[86] Cells are usually grown at high cell density by
solubility enhancer molecules addresses the problem of the using certain inducer molecules such as IPTG, anhydrotetra-
low solubility of the expressed protein (e.g., MBP, NusA, cycline (Atc), and appropriately optimizing the cellular
SUMO, Halo, and GST).[59] Further, there have been signifi- growth conditions.[87] Nevertheless, high cell density cul-
cant improvements in the solubilization efficiency using tures also lead to the accumulation of acetate and lactate,
detergents, urea, guanidium hydrochloride, and SDS during nutrients, and metabolic load, which is detrimental to the
in-vitro refolding in recent years.[30] The lack of post-trans- overall process efficiency.[88] Further, the expressed proteins
lational modifications such as glycosylation is addressed by are found to be highly susceptible to proteolysis by the host
co-expression of the glycosylation machinery of cell proteases.[30] In addition, aggregation and IB formation
Campylobacter jejuni in E. coli.[28] However, this strategy also pose a major issue in successfully adopting high cell
density cultures for enhanced target protein productivity.
has not been as successful as anticipated owing to complex
process machinery, the requirement of multiple vectors, and
inappropriate co-expression of proteins.[83] Cell-free protein synthesis
A major source of the enhanced propensity of IB forma- Cell-free synthesis of target proteins offers an attractive solu-
tion during protein expression is incorrect/scrambled disul- tion to the aforementioned issues associated with high cell
fide bonding in the expressed proteins.[10] Such proteins are density fermentation. Under this paradigm, the cytoplasmic
usually refolded in-vitro using conditions that favor correct constituents of the cells are used to express the target pro-
disulfide bond formation (by optimizing the cell’s redox tein.[80] Unlike the cell-based protein expression, this
environment), thereby facilitating the folding pathway to method does not utilize a live cell and thereby re-routing all
attain the final native folded state.[58] growth factors toward target protein synthesis (Figure 4).
PREPARATIVE BIOCHEMISTRY & BIOTECHNOLOGY 735

The downstream purification of cell-free expressed proteins how these techniques, when adopted for a variety of situa-
is significantly simpler as compared to the cell-based protein tions, impact the quality of the target product.
expression.[89] This is primarily attributed to the lack of the As a general rule of thumb, the efficient isolation of IBs
cell membrane and relatively simpler host cell impurity pro- defines the overall process performance as more proteins
files in the former. The lack of cell membrane facilitates dir- would be available for refolding, which would add to the
ect extraction of target protein without the need for cell overall yield of the process. Further, any IB isolation method
lysis.[90] Similarly, the fact that only proteins required for used should be able to preserve the quality and structure of
the target protein expression are included in the input cellu- the IBs, show scalability and process robustness characteris-
lar soup overcomes the issues of the auto-proteolysis of the tics and minimize additional impurities in the target protein
target proteins.[90] Further, the downstream recovery of the product.[96] The choice of the IB isolation method signifi-
protein post-harvest is also enhanced by minimizing product cantly impacts the quality of the target product that is being
expressed.[97] For instance, the use of lysozyme for bacterial
loss at this stage of operation.[89] Several types of proteins,
cell lysis inadvertently gets associated with the IBs, necessi-
such as misfolded, cytotoxic, and insoluble proteins that are
tating frequent washing steps to remove additional impur-
difficult to express using conventional cell-based systems,
ities during process development. Added to this, several
have been successfully synthesized using cell-free systems.[89]
bacterial cells, especially gram-negative bacteria are less sus-
The ability to use small molecular additives such as denatur-
ceptible to lysis by lysozymes, thereby significantly affecting
ants (sulfobetaine, substituted pyridines, and pyrroles), pro- the overall efficacy of cell lysis.[98] Similarly, sonication often
tein stabilizers (polyethylene glycol), and aggregation disturbs the active site of the protein present in the bacterial
inhibitors (L-arginine, proline) in a relatively less crowded IBs, reducing the overall reported biological activity.[99] In
cellular environment in the cell-free systems facilitates the contrast to these techniques, high-pressure homogenization
prevention of protein aggregation and misfolding by the cor- does not show any of the issues associated with the enzym-
rect formation of disulfide bonds and 3D packing of the atic and sonication-based methods, making it a method of
proteins to its native conformation.[30] Although the cell- choice for IB isolation from bacterial cells.[100]
free expression system provides a high-quality platform for One of the major impediments during IB isolation is the
protein synthesis, it also offers some disadvantages as it is a possibility of contamination of IBs with viable bacterial
very expensive process because it requires purified cell com- cells.[101] Such issues have been solved using the combina-
ponents for protein expression.[91] Therefore, more research torial approach of simultaneously using two orthogonal
is required to lower the cost of the cell-free system, and methods for IB isolation.[102] For example, using both sonic-
other strategies are to be employed for recombinant pro- ation and lysozyme treatment resulted in negligible (10–1
tein synthesis. CFU/mL) viable cell contamination. In this approach, lyso-
zyme treatment followed by frequent washing resulted in the
removal of cellular debris, while sonication caused the
Downstream processing aspects destruction of the remaining viable cell contaminants that
The recovery of biotherapeutics from IBs is a very crucial were originally lysozyme resistant.[103,104]
step and is accomplished through a train of unit operations,
including isolation, solubilization, refolding, and purification Solubilization of IBs
of IBs.[49] In the following sections, we discuss all aspects of Subsequent to the isolation of IBs, the next step in the train
IB downstream processing, from Isolation to obtaining a of bioprocessing is to solubilize them so that, under appro-
purified functional protein. priate conditions, they can be refolded to their native struc-
ture. The product yield is intrinsically linked to the
solubilization efficiency. The solubilization process is aided
Isolation of IBs
by adding solubilizing agents such as detergents, alkali
The recombinant protein expression using bacterial systems
medium, chaotropes, organic solvents, and high pressure
inadvertently leads to the formation of IBs in the E. coli
that break the non-native H-bonds in the IBs.[105] The
cytoplasm or periplasm.[15] In order to recover the active
choice of a particular method for solubilization of IBs is
protein from the IBs, the first step is to isolate the IBs and
generally guided by the prior knowledge of the structure-
subsequently subject them to a train of bioprocessing steps. function relationship of the proteins trapped in the IBs, if
Isolation of IBs from the bacterial cytoplasmic soup is car- available. However, in the lack of such information, a com-
ried out using a plethora of techniques broadly categorized bination of methods that maximize product recovery and
as physical, chemical, and enzymatic methods.[92,93] In sev- fully functional 3D folded protein is employed. The solubil-
eral instances of problematic IBs, a combination of any of ization process is described aptly by the homogenous layer
these methods is also employed to isolate IBs.[94] The choice model.[106] According to this model, the denaturants pene-
of the method is defined based on several parameters such trate the core of the IB, and the free monomeric and multi-
as scalability, efficiency, the time required for bioprocessing, meric peptides diffuse back into the solution.
and cost-effectiveness.[95] While a detailed review of the Typically, the turnaround time (a few to several hours)
underlying basics of these methods is discussed elsewhere for the solubilization process depends on the nature of
and summarized in Table 1, we limit our discussion here to the IBs (classical or non-classical) and the choice of the
736 K. KACHHAWAHA ET AL.

Table 1. Techniques of IBs isolation from bacterial cells.


Method Technique Advantages Challenges Ref.
Physical method High pressure - No loss in biological activity of IB protein - Generates heat (approx. 1.5  C/1000 psi) [200]

(Mechanical) homogenization during cell lysis - Not suitable for heat sensitive proteins
- Applicable for both small- and large-scale
cell lysis
[93,201]
Bead mill - Efficient for microscale cell lysis - Not efficient cell lysis for large scale
- Smaller beads (0.10  0.15 mm) are optimum - Heating and power consumption increases
for bacterial cell lysis with increase in number of beads
[202]
Physical method Sonication - Effective for small scale cell lysis (below - High frequency sonication (20–40 kHz)
(Non-Mechanical) 100 mL) damages the native proteins in IBs
- Time effective (Generally, it takes 2–5 min for - Increases the temperature (approx. 50–90 K)
cell lysis) of sample which damages the proteins
[203]
Thermolysis / Heating - Efficient for isolation of heat stable proteins - Not suitable for thermolabile proteins
- Able to release cytoplasmic proteins in less - Proteins solubility changes due to
than 10 min temperature fluctuation
[204,205]
Freeze-thaw - Suitable for isolation of thermolabile proteins - Cold denaturation may occur due to
- Did not require complex instrumentation, weakening of H-bonds
thus simple and cost-effective method for - Time consuming (usually requires 3–5 cycles)
cell lysis for cell lysis
[206,207]
Chemical method Solvents (Alcohol, DMSO - Effective method for wide range of microbial - Flammable & toxic (requires special
& Toluene) cell lysis consideration)
- Suitable for classical IBs - Denatures the IB protein
- Not suitable for non-classical IBs
[207,208]
Detergents (SLS, SDS & - Effective for cytoplasmic protein release - Detergents will denature the IB protein
Triton-X-100) (more than 75% in 1 h) - Not suitable for non-classical IBs
- Variability of detergent offers the appropriate - Additional impurity of detergents added
selection of detergent for different proteins. in proteins
[209]
Alkaline lysis - Alkaline buffers (10.5–12.5 pH) efficiently lyse - High chemical cost for neutralization of alkali
the bacterial cell by hydrolyzing the
cell wall.
[206,210]
Osmotic shock - No loss in biological activity of IB protein - Product may be diluted (which can increase
during cell lysis the down streaming cost)
 1 M salt concentration is sufficient for small - Less efficient & requires pre-enzymatic
scale cell lysis (below 100 mL) treatment for cell lysis
[211]
Enzymatic method Lysozyme - No loss in biological activity of IB protein - Not very efficient for gram negative bacteria
during cell lysis - Enzymatic impurity complexes the
- Suitable for gram positive bacteria downstream process.
[212]
Lysis protein E - Upstream In-process disruption of bacterial - Quality loss (10%) in IB purity
(Bacteriophage cell
UX174-derived) - Also, bacterial ghost can be used as
desirable product which contains active
IB proteins.
[103]
Combinational Sonication & Lysozyme - No impurity of viable bacterial cell in IB - Requires frequent washing steps to remove
method protein enzymatic impurity
- Effective cell disruption technique due to
synergistic effect of combined methods
[94]
High pressure - Efficient cell lysis - Sonication with High pressure
homogenization - No viable bacteria will remain in medium homogenization is simply not possible
& sonication because cavitation is substantially inhibited
at above 100 kPa pressures.
[213]
Novel approaches High pressure jet device - High throughput and cost-effective - Complex instrumentation
(Frictional - Efficient cell lysis
shear þ cavitation
þ collision)

solubilization agent.[107] This constitutes most of the proc- successfully refolded into their native conformations under
essing time required for the production of protein therapeu- appropriate conditions.[109]
tics. Recent efforts to reduce the time required for One of the major challenges during the solubilization of
solubilization of IBs indicate the excellent capability of the IBs is an aggregation of recombinant proteins. Aggregation
microwave-assisted method to carry out the process in less during the solubilization mainly occurs due to hydrophobic
than 5 minutes.[108] This method utilizes brief exposure of interactions and non-classical disulfide bond formation.[49]
the IBs treated with detergents to microwave radiation (with As aggregation follows higher order reaction kinetics, a high
a wavelength range of 300 mm–1 mm) at 25–30  C. The concentration of target protein or other product-related
exposure to microwave radiation results in the re-alignment impurities also contributes significantly to the aggregation
of the polar molecules (protein IBs) in solution, leading to during solubilization and refolding.[39]
efficient heating that enhances the solubility of the proteins. A variety of agents have been used that assist in the solu-
The method works for various IBs, as evidenced by the bilization of the isolated IBs. These agents differ in terms of
work of Dutta et al.,[108] who showed that seven different the strength with which they mediate solubilization. For
types of IBs were successfully solubilized in less than 2 min. example, while strong denaturing agents like urea and guan-
The solubilized IBs were completely unfolded and idine hydrochloride are more suitable for solubilizing
PREPARATIVE BIOCHEMISTRY & BIOTECHNOLOGY 737

Table 2. List of fusion tags to improve solubility and expression of recombinant proteins in E. coli.
Amino acid
Fusion tags Protein Organism Type Isoelectric point chain length Size (kDa) Ref.
[214]
MBP Maltose-binding protein E. coli chaperone based tag 4.97 396 40
[215]
GST Glutathione-S-transferase S. japonicum chaperone based tag 6.51 211 26
[216]
Trx Thioredoxin E. coli chaperone based tag 4.46 109 12
[217]
Spy Spheroplast protein Y E. coli chaperone based tag 9.45 161 18
SUMO Small ubiquitin modified Homo sapiens chaperone based tag 4.96 100 11 [218]

Fh8 F. hepatica antigen (8 kDa) F. hepatica Peptide tag – 69 8 [219]


[220]
Halo (DhA) Mutated dehalogenase Rhodococcus Peptide/affinity tag 4.83 293 33
[221]
NusA N-utilization substance A E. coli Fusion protein 4.44 495 54
[222]
Ecotin E. coli trypsin inhibitor E. coli Fusion protein 6.53 162 18
[223]
GB1 IgG domain B1 of protein G streptococcus Fusion protein 4.68 593 63
[224]
Skp Seventeen kilodalton protein E. coli Fusion protein 8.86 161 17

classical IBs,[12] their use is limited owing to the damaging electrostatic repulsion. Similarly, the IBs of hEGF protein
effect they exert on the secondary and high-order structures were expressed as CBD-Ssp DnaB-hEGF protein in E. coli. It
of the IBs. Further, strong denaturants also contribute to the was also solubilized via a mild denaturation method by the
formation of soluble and insoluble aggregates. The non- addition of urea followed by freeze-thaw. Urea breaks the
denaturants like DMSO and n-propanol, on the other hand, hydrophobic interactions of IBs peptides. Freeze-thaw allows
solubilize the non-classical IBs. As these agents have mild the IBs peptides to separate due to the formation of ice crys-
effects, the IBs retain their bioactivity and generally do not tals.[115] Another effort to get solubilized IBs is adding
require a downstream refolding step.[110] This, in turn, saves fusion tags to the expression vectors.[116] These Fusion tags
both time and cost involved in the bioprocessing of the IBs. (Table 2) will increase the solubility and inhibits the aggre-
Despite their advantages, non-denaturing agents often fail to gation of recombinant proteins by electrostatic repulsion
solubilize tough/classical IBs. The disadvantages associated between fusion tags, the interaction of hydrophobic portion
with the denaturing and non-denaturing agents necessitated of fusion tags with recombinant proteins, and attraction of
using mild solubilization agents that could solubilize both chaperones (trigger factor, GroEL, DnaK and ClpB) by
classical and non-classical IBs and preserve their residual fusion tags to mediate chaperones-assisted solubilization and
3 D structure, if any. Some mild solubilization methods refolding.[117] Recently a new retro-protein XXA has been
include subjecting the IBs to heat, alkaline pH, acid, organic developed as a fusion tag for the expression of soluble IBs.
solvents, freeze-thaw, and high hydrostatic pressure.[18] Retro-protein XXA is derived from antifreeze protein AXX
Nevertheless, these methods introduce artifacts by modifying and contains 192 amino acids. This retro-protein XXA
amino acid side chains, acid-mediated cleavage, and the fusion tag is more efficient than other fusion tags because it
introduction of impurities emanating from the use of can express Chrono, Notch2NL, and nCLu soluble proteins,
organic solvents and detergents. Further, it is difficult to which are difficult for other fusion tags.[118] Fusion tags are
make an appropriate decision as to which mild solubilization removed by enzymatic cleavage (enterokinase, aminopepti-
method would be useful in solubilizing a given IB.[111] dase) or chemical cleavage (formic acid).[119]
Efforts to develop correlations between various attributes of
solubilizing agents and bacterial IBs are currently in pro- Refolding of IBs
gress to overcome the uncertainty in choosing appropriate The thermodynamic hypothesis of protein folding states all
mild solubilization agents during the bioprocessing of bac- that a protein needs to fold into its native 3 D structure is
terial IBs.[112] In addition, a combination of mild solubiliza- its primary amino acid sequence.[6] However, this is true
tion methods, such as simultaneously subjecting IBs to only under dilute conditions. Within the cells (in-vivo), the
extreme pH and pressure/temperature conditions, can also protein translation occurs under very crowded conditions
generate conditions wherein the IBs get solubilized without and is generally assisted by molecular chaperones. However,
altering the existing native-3D structure of the proteins.[113] the in-vivo protein folding is non-trivial, and hence methods
While the use of mild solubilization methods is yet to of refolding proteins outside the cell (in-vitro) are
become a norm in the industry, their successful adoption preferred.[49]
would eventually pave the way for a reduction in the time In-vitro protein folding is done by decreasing the
and cost involved in the production of protein therapeutics. denaturant concentration of the solubilized IB, thereby driv-
However, efforts have been made for effective solubiliza- ing the equilibrium toward the native folded state of the
tion of the IBs, i.e., getting more soluble IBs with less or no protein. However, the transition from the solubilized,
formation of aggregates. Recently, the nucleocapsid fragment unfolded, to the natively folded form of the protein is asso-
IB of SARS-CoV-2 expressed in E. coli was efficiently solubi- ciated with instances of protein misfolding and aggrega-
lized under mild denaturing conditions, i.e., high pressure at tion.[120] As such, aggregation and protein misfolding
2.4 kbar and alkaline pH 9.[114] High-pressure solubilizes constitute the biggest challenge during protein refolding as
protein aggregates by weakening hydrophobic and ionic it impacts the final product’s overall process economics and
bonds, and alkaline pH promotes solubilization by quality. Protein refolding is a strong function of the initial
738 K. KACHHAWAHA ET AL.

concentration of the unfolded protein and follows first-order Further, our understanding of the protein folding mech-
kinetics. On the contrary, protein aggregation is a higher- anism, conditions of refolding, and the characterization
order process and is accelerated at the higher initial concen- methods involving biophysical and biochemical approaches
tration of the proteins.[121] Thus, protein refolding is are still incomplete. Consequently, there is still a need for
strongly favored under dilute conditions (approx. the development of a universal refolding buffer that could
0.01–0.1 mg/mL), wherein the molecular collisions between assist in refolding IBs of any characteristics back into their
the unfolded and folding intermediates are minimized. The native folded state. However, practices have been made to
stability of the protein folding intermediates is enhanced by develop the correlation between the refolding parameters
the addition of several molecular excipients (folding (such as temperature and pH) and methods of refolding.
enhancers and aggregation suppressors) such as sucrose, L- These databases can assist in the better prediction of protein
arginine, ethyl ammonium nitrate (EAN), trifluoroethanol, refolding conditions.[125]
cyclodextrins, and alginates.[122]
Several physicochemical factors, such as pressure, tem-
perature, pH, and ionic strength of the refolding buffer, also Purification of refolded proteins
determine the efficiency of protein refolding.[123] For The refolded output generally would be a cocktail of cor-
example, successful refolding of b-lactamase from IBs was rectly folded, misfolded, truncated, and aggregated forms of
done at 2  103 bar for 48 hours.[124] Under this condition, target protein.[28] It is of utmost necessity to separate the
the protein aggregate dissociates and assumes the native correctly folded form of the protein from the other con-
folded conformation of the protein. Similarly, existing data formational impurities for the development of a safe and
from a refolding database[125,126] indicates the existence of efficacious biotherapeutic. While the truncated and aggre-
two temperature conditions for refolding under low temper- gated form of the protein could easily be removed using size
atures (0–5  C) and under ambient conditions (25–30  C). exclusion chromatography (SEC),[131] it is not an attractive
Likewise, the optimum pH for a variety of IB refolding lies option at the industrial scale. The SEC is characterized by
in the broad range of 5–9, with the most common pH of small loading (usually <5% of the column volume) and
8–8.5. It is important to note that refolding should not be operation under slow flow rates (50 cm/h), which exorbi-
carried out under pI conditions to avoid precipitation. tantly adds to the overall cost and time of bioprocessing.[132]
Table 3 lists the commonly used methods of protein The biggest challenge, however, is the separation of the mis-
refolding and their associated advantages/disadvantages. As folded forms from the correctly folded proteins due to
evident from the table, each refolding condition must be nearly the same Physio-chemical properties. The currently
optimized on a case-to-case basis depending on the propen- used approaches to separate protein isoforms include multi-
sity of the IB to aggregate and the scale of the operation. modal chromatography, which uses two orthogonal dimen-
For example, while dilution of denaturants or chromatog- sions of separation based on the intrinsic properties of the
raphy-based processes is an attractive option for protein proteins.[133] The excellent separation efficiency offered by
refolding at a laboratory scale, the large volume of buffer multi-modal chromatography is often marred by difficulties
used necessitates an additional concentration step, which in optimizing the separation conditions and issues with the
adds to the cost and time of bioprocessing. Likewise, refold- method’s reproducibility.[134]
ing proteins at an industrial scale should also yield a high Recently, affinity-based purification using Ni-NTA chem-
concentration of refolded protein output. A dialysis-based istry has been used to separate protein isomers during
method is suitable for obtaining a high concentration of the refolding.[135] The advantage of this method is that the same
refolded protein output.[127] However, the rate-limiting step buffer used for purification of IBs can be used for refolding
of removing denaturants from the membrane makes dialy- after diluting into refolding buffer, which allows the protein
sis-based refolding exceedingly time-consuming.[7] This step to specifically bind to the resin in a denatured state. Upon
could be hastened by increasing the membrane surface area elution under refolding conditions, active function proteins
(i.e., membrane area per volume of denatured protein solu- in milligram amounts could be received per liter of the cell
tion). Kato et al. achieved this objective by employing a culture. Other novel chromatographic modes include the use
chemical engineering approach wherein they incorporated a of monolithic[136] and membrane-based chromatography[137]
dialysis membrane into a microchannel. They successfully which also provides avenues for high-throughput separation
refolded a model protein (known to be difficult to refold) of protein isoforms. However, these methods have low-bind-
with an eight-fold concentration instead of the dilution- ing capacity during separation and therefore are less sought
based method in under 20 min using this custom-designed after at industrial scales.[138] The amalgamation of nano-
dialysis setup.[128,129] However, the size of such refolding technology with separation science has resulted in the devel-
apparatus limits the volume of solubilized IB that could be opment of nano-fiber-based chromatography systems that
handled. Also, rapid mixing of the solubilized IB and refold- have much higher binding capacities than monoliths/mem-
ing buffer in the microchannel may result in protein aggre- brane-based systems.[139,140] However, such systems still
gation. The use of aggregation suppressors such as arginine, need to be validated for robustness, efficiency, and ease of
allantoin, and hydantoin could prevent aggregate formation operation before they can be rolled out for large-scale usage
in such instances.[130] in the purification of correctly folded proteins.
PREPARATIVE BIOCHEMISTRY & BIOTECHNOLOGY 739

Table 3. Techniques of refolding of solubilized IBs.


Methods of refolding Technique Sub-type Advantages Disadvantages Ref.
[7,225]
Conventional Dilution Single step dilution - Simple procedure and ease of - Suitable for low concentration of
Methods operation protein (approx. 0.1 mg/mL)
- Applicable for small scale refolding - Large volume of buffers and
- Time effective reactors increases the refolding
cost
- Non-uniform mixing leads to
aggregate formation
Batch dilution - Less formation of protein - Time-consuming in contrast to
aggregates single step dilution
- Applicable for large scale refolding - Additional step of protein
concentration is required
[7,226]
Dialysis One-step Dialysis - Simple method - Rapid decrease in denaturant
concentration increases the
aggregate formation
- Interaction of denatured protein
with dialysis membrane
Multi-step Dialysis - High refolding efficiency as - Time consuming
compared to one-step dialysis - Small membrane surface area slows
down the denaturant removal
from dialysis bag
[227]
Chemical additives Stabilizers, Aggregation - Stabilizers prevents the reverse - Chemical additives increases the
inhibitors, transition of native proteins overall production cost
Chaotropes - Decreases the rate of aggregation - Imparts the chemical impurity
- Chaotropes promotes the - Additional purification step needs
denaturation and solubilization to be performed
[228]
Non-conventional Microfluidic Chip - Efficient protein refolding due - Not suitable for the large-scale
Methods controlled and regulated diffusion protein refolding
in microchannels - Sometimes protein aggregates will
- Rapid process disrupt the laminar flow in
microchannels
[132]
On-column Size exclusion -Simple and automated process -Large volume of buffer required
chromatography -Combined refolding and purification -Large dilution factor reduces the
steps are performed protein concentration
-High time consumption due to
column retention time
[229]
Periodic Counter-current - Economical process - Continuous columns efficiency
Chromatographic - Continuous process utilizing requires frequent usage
system multiple subsequent columns, so - Sophisticated columns, chances of
chromatography resins is mechanical breakdown and
efficiently used microbial contamination
- Specific buffer consumption is less
as compared to other similar
techniques
- Higher protein refolding efficiency
(11mg/hour)
[230]
Hydrostatic pressure - Able to refold Hydrophobic proteins - Requires preliminary treatment of
- High hydrostatic pressure proteins with detergents and
(100–300 MPa) efficiently denaturants
solubilizes the protein by
disrupting inter and intra
hydrophobic interaction between
protein molecules
[231]
Differential scanning Fluorescence - Cost effective method - Chances of false positive and
fluorimetry based method - Used to track both protein folding negative results are higher
state and thermal stability. - Thermal shift may interfere with
- Can provide reliable data even in the native state of protein sample
unpurified chemical reactions.
[128]
Microchannel dialysis - High concentration of protein can - Incorporation of dialysis membrane
be refolded into microchannel is challenging
- Low residence time for dialysis - Design and fabrication of
- Greater membrane surface area as microchannels is complex process
compared to conventional dialysis

Analytical characterization of solubilized IBs/ details of the structure and homogeneity of IBs/refolded
refolded proteins proteins are briefly discussed:
IBs are heterogeneous entities exhibiting significant diversity
Structural characterization
in their shape, architecture, organization, and function. In
view of these properties, a panel of analytical tools is often Circular dichroism
employed to derive the detailed fingerprint of the IBs. In the CD spectroscopy is a nondestructive technique that is usu-
following section, various analytical tools that can provide ally implemented to monitor IB refolding to detect
740 K. KACHHAWAHA ET AL.

conformational changes in the secondary (in the far-UV to monitor higher order structure (tertiary and quaternary)
region between 190 and 250 nm) and tertiary structure (in of a protein.[145] However, NMR can be used to characterize
the near-UV region between 260 and 300 nm) of the pro- the structural dynamics of only relatively smaller proteins
tein.[141,142] The absorption spectrum in the far-UV region (<40kDa) and are not suitable of proteins with larger sizes
is attributed to the amide bonds, while in the near-UV such as mAbs. Additionally, for NMR analysis, the protein
region, it is dominated by the absorption by the side chains samples should be highly pure and concentrated (0.5 mM
of the aromatic amino acids (buried within the core of the or higher), thereby limiting its application in protein refold-
protein).[143] While far- UV CD measures spectra of both a ing studies.[150,151] For instance, NMR analysis of IBs exhibit
helices and b-sheets, it is mostly applied for distinguishing additional resonance due to the presence of phospholipids
the refolded proteins rich in a helix from denatured protein. from the E. coli membrane, other proteins, or RNA necessi-
The major issue in successful implementation of CD spec- tating purification prior to NMR analysis.[152]
troscopy during monitoring of protein refolding is the need Another approach to decrease the NMR spectra complex-
of high sample concentration, i.e., 0.1 mg for far-UV and ity is isotope labeling. Utilization of 13C- and 15N-labeled
1 mg for near-UV.[143] Recent studies have also reported the amino acids corresponding to the residue pairs allows for
use of CD for real-time, in-situ analysis of the relationship the structural and thermodynamic characterization of inter-
between protein aggregation and the ensuing conformational mediately folded protein conformers since it allows us to
changes, if any.[144] However, the use of CD should be done examine a broad range of a protein’s conformational space
with caution as the resultant CD signal is a result of the sig- with a relatively neutral disturbance.[152] For instance,
nals emanating from different parts of the molecules, and Kamatari et al., investigated the atomic resolution structures
since these signals possess directionality, the overall signal and thermodynamic properties of several partially folded
may not reflect the actual conformational changes in the conformers under varying pressure using NMR spectros-
molecule.[145] Further, far-UV CD is also limited by intrinsic copy.[153] Water proton transverse relaxation rate R2(1H2O)
insolubility of proteins, which causes high level of light scat- measurement by NMR is a strong noninvasive method for
tering disturbances and low signal-to noise ratio.[146] the detection of inclusion bodies. The water proton NMR
(wNMR) technique is an indirect technique that draws its
conclusions from water–protein interactions and exhibits
Fluorescence spectroscopy excellent sensitivity toward protein aggregate formation.[154]
It is a nondestructive technique used for the real-time evalu-
ation of aggregation mechanisms, from monomers to the
formation of large aggregates. It also detects protein self- Dynamic light scattering
assembly.[147] It is a highly sensitive technique where the Dynamic light scattering (DLS) is a technique used for the
fluorophores used can be either intrinsic or extrinsic. In measurement of the surface charge of the proteins.[145]
intrinsic fluorophores, the fluorescent properties of aromatic Owing to this property, DLS can effectively measure the
amino acids are used for detection. In extrinsic fluorophore, magnitude of the electrostatic repulsion/attraction between
fluorescence is observed from the externally used fluorescent two molecules. Thus, it is a good technique to measure pro-
probes that can covalently bind with the hydrophobic tein aggregates in size range of 1 nm  5 mm.[155] In this
patches or chemically attach to the molecule.[148] Some of technique, monomers are not separated from the aggregate,
the commonly used extrinsic fluorescence dyes include and their average size is used for the estimation of protein
Thioflavin T, Congo Red, Nile Red.[149] These dyes give less aggregate. This is more effective in the analysis if the sample
fluorescence when present in an aqueous environment, and contains some visible particles.[155] For better detection, it is
their intensity is increased once they are bound with the very important to keep the working conditions constant
hydrophobic patch, which may be present in protein such as temperature, viscosity, and interaction between
aggregate.[147] aggregated particles.[156] This technique has been used for
The high sensitivity of the extrinsic fluorescence makes it the detection of protein aggregate formation by heat and the
ideal choice for high-throughput screening during protein suppression of aggregate formation using heat shock pro-
refolding. The variation in surface hydrophobicity is teins like a-crystallin. It has also been used for analysis of
reflected as increased fluorescence intensity during protein the nonspecific protein aggregation formation by the action
refolding.[145] However, the main limitation of using extrin- of detergents.[157]
sic dyes is that it could alter the protein structure and affect There are several advantages of using dynamic light scat-
the overall process of detection. On the other hand, quan- tering for aggregate characterization because it is highly sen-
tum yield of detection using intrinsic fluorescence method is sitive for the detection of even a small quantity of
significantly lower than the extrinsic fluores- aggregates, and it is a high throughput technique. In this
cence methods.[148] technique, both reversible and irreversible aggregates can be
easily characterized. The software used for the measurement
of aggregate size is Zetasizer which is very user-friendly. It
Nuclear magnetic resonance (NMR) spectroscopy helps in the extraction of maximum possible information
NMR is used to determine the time needed to establish the from the aggregate sample and a more accurate representa-
native state of the protein during refolding. The advantage tion of data. The procedures for measurement are highly
with NMR spectroscopy is that it can be used in real-time automated and thus do not require high degree of training
PREPARATIVE BIOCHEMISTRY & BIOTECHNOLOGY 741

to use the instrument.[156] Unlike extrinsic or intrinsic fluor- Homogeneity of IBs


escence methods, DLS being non-intrusive has minimal
Reversed-phase liquid chromatography
chances of assay induced aggregation, thereby significantly
The separation in reversed phase chromatography is based
reducing the chances of measurement artifacts.
on the differences in the hydrophobicity of various species
DLS also has some disadvantages, such as the data
obtained for analysis being semi-quantitative. As a result of in a sample. Hence, the method is capable of separating the
its sensitivity toward detecting even small quantities of product related impurities such as oxidized and reduced spe-
aggregate samples, it sometimes also detects dust particles cies formed during protein refolding.[163] However, since
and other contaminants present in the sample and gives RP-HPLC is carried out under denaturing conditions, no
false positive results.[158] This also has the significant draw- information is obtained about the secondary and tertiary
back of being unable to distinguish between scattering structure of the protein. Therefore, the information obtained
amounts from various aggregate molecules.[156] is merely chemical in nature. Further, the high column tem-
perature used during RP-HPLC might induce protein aggre-
gation. Nevertheless, RP-HPLC being very fast, robust, and
Cryo-electron microscopy high-resolution method is widely used to assess the product
It is a variant of transmission electron microscopy which homogeneity by measuring the levels of various product-
nowadays is widely used for the structure prediction of mac- related impurities.[151]
romolecules. This technique uses flash-freezing solutions of
proteins and samples are cooled up to their freezing temper-
atures.[159] Protein samples are kept in an environment of Size exclusion chromatography
vitreous water (ice with the properties of water) and are Size Exclusion Chromatography (SEC) is another method to
bombarded with electrons.[157] The image is reconstructed assess the levels of aggregation and fragmentation during
with the help of multiple individual microscopic images of protein refolding. Hence, the method is highly suitable to
molecules and is used for the elucidation of the 3D structure distinguish size- related variants such as dimers, oligomers,
and shape of the protein aggregate sample.[160] and fragments. The aggregation/misfolding is detected using
It offers several advantages, such as it does not require UV-based detectors/fluorescence/multi-angle light scattering
crystallization and yields near-atomic level resolution of the (MALS).[164] Recently, SEC coupled with mass spectrometry
molecular structures being examined. As there is practically under native conditions has become possible to enhance the
no upper limit of size for EM, it is an ideal tool for protein coverage and depth of information obtained.[165] The ubi-
aggregate analysis.[161] For example, cryo-EM has been used quitous use of the SEC in the characterization of protein
to elucidate the overall fibril structure showing the number aggregates is evident from the fact that it has been estab-
of protofilaments involved in aggregate formation and the lished as a quality control method for protein aggre-
degree of twist present in amyloid formation. It also gives gate analysis.
information about the atomic structure of fibrils. It is a Despite their widespread usage in the characterization of
next-generation method in which the electrons are bom- protein aggregates, it suffers from certain limitations. The
barded in such a way that they form a thin, sensitive layer biggest issue in the adoption of SEC as a method of choice
(usually a carbon film), thereby preventing signals from get- in industrial setup is attributed to low volumes of sample
ting scattered in the surroundings, which may result in poor loading (2–5% of a column volume) and large amounts of
image formation.[162] buffer requirement.[132] Further, secondary interactions on
While cryo-EM offers better resolution than other con- the column also limit the resolution and performance
temporary techniques like x-ray crystallography, however, it obtained while using SEC. The UV-based detectors also
has lower contrast. Several strategies, including ‘negative show issues of sensitivity while discriminating between big-
staining’ (the sample is immersed in a layer of heavy metal), ger vs. smaller aggregates.[132] These factors necessitate
are used to improve the contrast during cryo-EM measure- orthogonal analytical tools to be used for the analysis of IBs.
ment of the samples.[161] Further, the lower contrast results
in a low signal-to-noise ratio, necessitating advanced detec-
tion systems and image processing systems for accurate Capillary electrophoresis
detection of sample images.[161] Capillary electrophoresis, also called high-efficiency capillary
While recent advancements and automation techniques electrophoresis, is a class of liquid phase micro-separation
have solved many issues related to image resolution and analysis method. It is used for the separation of ions on the
sample preparation, major issues still exist. In particular, basis of their electrophoretic mobility under an applied volt-
cryo-EM instrumentation is very costly and reaches many age. The mobility of these ions depends upon the molecule
millions of dollars, exceeding the grasp of individual investi- charge, size and viscosity. The mobility of the molecules is
gators and universities.[162] Further, cryo-EM maintenance is directly proportional to the applied electric field. Capillary
very expensive and demanding.[162] One potential solution electrophoresis is utilized very commonly due to faster
for these issues is the establishment of national and inter- results and high-resolution separation.[166] Garza et al.
national facilities that operates on a cost-sharing basis. developed an IB solubilization method and determined the
These are starting to emerge in the US, UK, Germany, purity of recombinant proteins in inclusion bodies using
Sweden, and India. capillary gel electrophoresis. It is based on the utilization of
742 K. KACHHAWAHA ET AL.

a single-component solution which is completely compatible Novel applications of IBs


with capillary gel electrophoresis analysis. In this method,
IBs as a vehicle for drug delivery
IBs sample were prepared and separated from recombinant
proteins and other impurities. This allows capillary gel elec- Therapeutic proteins are susceptible to degradation via mul-
trophoresis to be regarded as a suitable analytical tool to tiple mechanisms during purification, formulation, and stor-
obtain the quantitative process information during IB age. The degraded protein product has attenuated efficacy
refolding.[167] and is even immunogenic, posing a serious threat to the
lives of the patients.[171] Protein aggregation is the most
common route causing protein degradation. Being immuno-
Peptide mapping genic, protein aggregates elicit an anti-drug antibody (ADA)
Peptide mapping is a useful method for the identification of
response upon the administration that may interfere with
product-related impurities formed during protein refolding. the absorption, distribution, metabolism, and excretion
The biggest advantage of peptide mapping is when used (ADME) profiles of the drug.[155]
with mass spectrometry to confidently identify site-specific The recombinant protein expression using E. coli inad-
modifications on the proteins.[168] In a recent application of vertently entails the formation of target proteins in the form
peptide mapping for studying protein refolding, the dynam- of inclusion bodies (IBs). Existing evidence about the archi-
ics of covalent and non-covalent interactions during the tecture and morphology of the IBs suggests the presence of
refolding of the light chain, heavy chain, and antibody frag- functional native-like structures within the overall IB net-
ment were studied. It was found that covalent interaction work.[172] The presence of these structures within the bacter-
(disulfide bond formation) was the major driving force for ial IBs has inspired the packaging of active protein-based
protein refolding.[169] Further, multi-attribute monitoring drugs in the form of IBs and is being used as a novel self-
(MAM) workflows are currently widely used for the discov- releasing drug delivery system.[173] However, the large
ery of unknown intermediates formed during the protein batch-to-batch variability in the morphological features of
refolding. This allows for better optimization of the protein IBs during protein biomanufacturing using microbial expres-
refolding process to enhance yields.[170] sion systems eludes their wider applicability as the potential
Table 4 lists other characterization techniques that are drug delivery platform. The presence of bacterial contamin-
used for the analysis of bacterial IBs and protein refolding. ation in such IBs poses regulatory challenges in terms of
The choice of the technique would depend on the detection accruing approvals for market authorization of such
limits, sensitivity, resolution, and reproducibility of the tool. novel platforms.
With the adoption of the QbD–approach in bio-manufactur- Despite these insurmountable challenges, innovations
ing, the tools that could yield real-time monitoring and ana- have been made by engineering artificial IBs for clinical use.
lysis would continue to be in demand and the focus of As opposed to the natural IBs formed from direct bacterial
further research. cell cultures, artificial IBs exhibit batch-to-batch consistency

Table 4. List of techniques.


Technique Peculiar feature Ref.
[232]
Fluorescence microscopy - Do not need prior sample filtration and can detect turbid (in vaccines) and concentrated
(subcutaneous injection) protein solution. Detect highly concentrated protein solutions without
dilution.
- Helps in stability testing of formulation by detecting small aggregates (seeds) formation even
before their fibrillar structure formation.
- Allows characterization in-situ even in presence of excipients.
[144]
Circular Dichroism - Quantitative analysis of secondary and tertiary structure of optically active protein aggregates.
- Study relationship between molecular aggregation and conformational changes.
[233]
Cryo electron microscopy - Atomic level structure prediction of macromolecules.
[234]
Atomic Force microscopy - It is capable of characterizing even a single molecule of protein aggregate.
- It supports the examination of untreated specimens even directly from tissue sample.
[235]
ATR-FTIR - Capable of detecting very low quantity of samples and also applicable for insoluble and
impure sample.
[236]
Confocal spectroscopy - Monitor protein misfolding and aggregates in living cells.
- Quantitatively measures protein oligomerization.
[237]
Native Mass spectrometry - Able to detect native quaternary structure of protein aggregate.
1 [238]
H- NMR - Monitors presence of coexisting classes of protein aggregates.
- Detects the self-assembly of both hydrophobic and hydrophilic peptides.
[238]
All Atom Molecular Dynamic Simulation - Provides information about the amino acid residues associated with peptide-peptide interaction in
(AA-MD) aggregate formation.
- Predicts the different aspects of protein aggregation behavior.
- Detects protein- excipients interaction.
[239]
SEC-ESI-MS - Differentiates between the oligomers present in protein samples and those formed during the
detection process.
[155]
Dynamic light scattering - Highly sensitive technique for small quantity of aggregate sample.
[164]
Size exclusion chromatography - Qualitative and quantitative evaluation of protein aggregates.
- Monitoring of modified protein such as protein-drug conjugates and PEGylated proteins.
PREPARATIVE BIOCHEMISTRY & BIOTECHNOLOGY 743

Figure 5. Artificial Inclusion Bodies (ArtIBs) production and brief characteristics of ArtIBs. (a) Showing the general procedure followed for Artificial Inclusion Bodies
(ArtIBs) production. Soluble proteins are linked with the divalent cations (ZnCl2, CaCl2, and MnCl2), followed by incubation for sufficient fabrication of proteins. The
fabricated protein contains both soluble and insoluble faction which are separated by centrifugation. The soluble fraction of protein was discarded and insoluble
fraction was used as ArtIBs. The ArtIBs will release the functional proteins as mimicking the action of secretory granules. (b) Characteristic properties of ArtIBs which
makes effective and suitable agents for therapeutics development.

and are devoid of any bacterial contaminations.[41] Further, carriers (resins, microparticles, silica gels, graphene oxide,
their synthesis routes are amenable to optimization of pro- carbon nanotubes, and nano-porous materials) via physical
tein-protein interactions that may assist in easily releasing (adsorption), ionic, or covalent bonding.[176] However, there
active proteins under physiological conditions are some challenges associated with carrier immobilization.
(Figure 5).[174] Over time, we envision the use of bacterial Firstly, the leakage of partially soluble IBs from carriers due
IBs as an attractive platform to package the glycoprotein to weak or improper binding. Secondly, nonspecific inter-
(for instance, IgGs) expressed using mammalian systems action between the active site of partially soluble enzymes
and being used on various delivery routes, including much and the carrier decreases the catalytic activity of immobi-
restricted oral routes of protein drug administration. lized IBs. Furthermore, carrier immobilization requires a
high amount of non-catalytic mass (carrier/support mater-
ial), which affects the yield and quality.[177]
IBs as immobilized catalysts
The above-mentioned drawbacks of carrier-based immo-
The ability of IBs to withstand harsh physicochemical condi- bilization are overcome by entrapment or encapsulation of
tions (pH, temperature, and solvents) renders them a useful the IBs in an insoluble polymer network made up of gelatin
candidate as immobilized catalysts with wide applications in beads, silica sol-gel, and hollow fiber or microcapsules. Since
the biotechnology, pharmaceutical, and cosmetic indus- the active site of the IB does not participate in any inter-
try.[42] The wider applicability of immobilized catalyzed ver- action with the encapsulating network, the structure and
sus the traditional soluble catalyst includes superior function of the IBs remain in toto.[176,178] However, due to
mechanical, thermal, and functional stability. In addition to mass transfer limitations owing to a bulky polymer network,
this, the former exhibits superior multi-enzyme functional- the catalytic efficiency of the immobilized IBs is reduced.
ity, enantioselectivity, protease resistance, re-usability, and Carrier-free immobilization is based on cross-linking of
easy downstream purification characteristics than IBs with the help of linkers such as glutaraldehyde.[179] In
the latter.[175] contrast to the entrapment and carrier-based immobilization
The IBs immobilization is carried out by primarily three methods, this technique does not need any bulky polymer
methods: carrier immobilization, entrapment of enzymes to matrix, thereby overcoming the mass-transfer-induced
polymer network, and cross-linking of enzyme aggregates reduction in the catalytic efficiency of the immobilized IBs.
with linker agents (carrier-free immobilization). In addition, the carrier-free method is a low-cost technique
Carrier immobilization involves the attachment of par- for the immobilization of IBs yielding high purity catalyst
tially soluble enzymes to an insoluble solid support or systems (due to the absence of the carrier support that
744 K. KACHHAWAHA ET AL.

potentially ends up as a process impurity), enhancing its were used as an adjuvant for an immunogenic response dur-
prospects of industrial applicability multi-fold.[180] The ing vaccine development as IBs have potential dendritic cell
cross-linking of IBs enhances their mechanical and thermal maturation capacity.[194] However, optimal cell colonization
stability and allows them to withstand extremely harsh pro- is achieved by multi-factorial process optimization of param-
cess conditions without affecting their catalytic activity.[176] eters such as surface topography, size, wettability, and cellu-
For instance, the carrier-free immobilization of the hyaluro- lar migration.[195]
nidase active IBs enhanced the cell permeability for drug
absorption, enhancing its pharmacokinetic/pharmacody-
namic (PK/PD) profile.[181] In addition, a plethora of Current status and future prospects
enzymes, including kinases, lyases, aldoses, oxidases, reduc- The current breakthrough in the enhancement of the overall
tases, and phosphorylases, have been used as carrier-free or efficacy of recovery of the active protein from bacterial IBs
self-immobilized formats for enhanced catalytic could, to a large part, be attributed to the use of computa-
activity.[182,183] tional methods to analyze protein structure and identify the
Another version of the carrier-free immobilized IBs that potential product-related variants/impurities that could be
are gaining prominence is the catalytically active IBs formed during refolding. Further, the availability of struc-
(CatIBs). The CatIBs are solid and amorphous in nature and ture-function relationship data has enabled the identification
contain a specific amount of catalytic activity.[184] This class of critical quality attributes (CQAs) for the target prod-
of IBs is heterogeneously produced inside the bacterial cell
uct.[196] The emphasis on the development of analytical
(E. coli) and formed by the fusion of aggregation-prone
methods with improved precision, accuracy, robustness, and
domain (fusion tags) to the N/C-terminus of the target pro-
sensitivity has opened avenues for a deeper understanding
tein.[185] The fusion tags do not interfere with the native
of bacterial IBs and their bioprocessing.[197] For instance,
structure of the target protein (immobilized IBs). The
researchers have employed a combination of statistical
CatIBs have been widely used for the biosynthesis of cadav-
design of experiments (DOE) and high throughput process
erine from the CatIBs of lysine decarboxylase (LDC).[186] A
development (HTPD) to explore a wide characterization
recent application also suggests the co-immobilization of dif-
space for various process parameters and raw material
ferent enzymes within the same CatIBs to catalyze multistep
reactions.[187] Similarly, co-immobilization of CatIBs was attributes under consideration.[198] In addition, process
carried out in E. coli via co-expression of two fluorescent understanding gleaned through these tools issued to create
proteins, which catalyzes a two-step reaction for the produc- appropriate real-time monitoring and control tools that
tion of (1 R,2R)-1-phenylpropane-1,2-diol (PPD).[188] potentially serve as process analytical technology (PAT)
tool.[199] Further, use of these advanced methods of moni-
toring and analysis of bacterial IBs, the current endeavors
IBs as biocompatible and mechanically stable materials are to integrate the bioprocessing along with other estab-
IBs extoll the virtues of high mechanical stability due to lished unit operations during bio-manufacturing for con-
their ability to withstand cell lysis conditions during isola- tinuous bio-processing to yield a highly productive
tion, proteolytic stability, and bio-adhesiveness property. IBs, manufacturing process.
unlike active proteins, are not susceptible to changes in their
size, geometry, and biological activity during long-term stor- Disclosure statement
age or incubation.[189] In addition, the porous nature of the
IBs renders them an excellent candidate for the development No potential conflict of interest was reported by the author(s).
of novel biomaterials as they hardly offer any mass-transfer
resistance to the ensuing flow regimes.[11] These characteris- Funding
tics make them an excellent choice for developing novel
This work was supported by the Indian Institute of Technology
materials possessing superior elasticity, adhesives, and self-
(Banaras Hindu University) Varanasi’s Seed Grant Ref. No IIT(BHU)/
assembling peptides spanning a wide range of Budget/19-(14)/2021-22/0975 dated 31-November-2021 to SKS.
applications.[190]
One of the well-recognized uses of IBs as biomaterials is
their ability to be utilized as a drug delivery agent for sus- References
tained drug release.[55] The ability emanates from its prop- [1] Moremen, K. W.; Ramiah, A.; Stuart, M.; Steel, J.; Meng, L.;
erty to mimic the endocrine release action of hormones[191] Forouhar, F.; Moniz, H. A.; Gahlay, G.; Gao, Z.; Chapla, D.;
and the ability to cross the membrane with minimal tox- et al. Expression System for Structural and Functional Studies
icity.[174] An example of such a system is proteinase-K- of Human Glycosylation Enzymes. Nat. Chem. Biol. 2018, 14,
resistant amyloid IBs.[192] These are self-releasing chemically 156–162. DOI: 10.1038/nchembio.2539.
homogenous IBs, and themselves act as both the scaffold [2] Gileadi, O. Recombinant Protein Expression in E. Coli : A
Historical Perspective. Methods Mol. Biol. 2017, 1586, 3–10.
material and the target drug substance. Another application [3] Walsh, G. Biopharmaceutical Benchmarks 2018. Nat.
of IBs as biomaterials is for eliciting stimuli for cell prolifer- Biotechnol. 2018, 36, 1136–1145. DOI: 10.1038/nbt.4305.
ation and differentiation by providing adequate support for [4] Buscajoni, L.; Martinetz, M. C.; Berkemeyer, M.; Brocard, C.
cell adhesion during cell culture.[193] Ovalbumin-derived IBs Refolding in the Modern Biopharmaceutical Industry.
PREPARATIVE BIOCHEMISTRY & BIOTECHNOLOGY 745

Biotechnol. Adv. 2022, 61, 108050–108050. DOI: 10.1016/j.bio- Functions. Nat. Rev. Mol. Cell Biol. 2020, 21, 729–749. DOI:
techadv.2022.108050. 10.1038/s41580-020-00294-x.
[5] Hwang, P. M.; Pan, J. S.; Sykes, B. D. Targeted Expression, [22] Clausen, H.; Wandall, H. H.; DeLisa, M. P.; Stanley, P.;
Purification, and Cleavage of Fusion Proteins from Inclusion Schnaar, R. L. Chapter 56: Glycosylation Engineering. In
Bodies in Escherichia Coli. FEBS Lett. 2014, 588, 247–252. Essentials of Glycobiology: Varki, A., Cummings, R. D., Esko, J.
DOI: 10.1016/j.febslet.2013.09.028. D., Stanley, P., Hart, G. W., Aebi, M., Mohnen, D., Kinoshita,
[6] Hirata, F.; Sugita, M.; Yoshida, M.; Akasaka, K. Perspective: T., Packer, N. H., Prestegard, J. H., Schnaar, R. L., Seeberger,
Structural Fluctuation of Protein and Anfinsen’s P. H., Eds.; Cold Spring Harbor Laboratory Press: New York,
Thermodynamic Hypothesis. J. Chem. Phys. 2018, 148, 2022; pp 1–16.
020901. DOI: 10.1063/1.5013104. [23] Strutton, B.; Jaffe, S. R. P.; Pandhal, J.; Wright, P. C.
[7] Mirhosseini, S. A.; Latifi, A. M.; Hosseini, H. M.; Seidmoradi, Producing a Glycosylating Escherichia Coli Cell Factory: The
R.; Aghamollaei, H.; Farnoosh, G. The Efficient Solubilization Placement of the Bacterial Oligosaccharyl Transferase pglB
and Refolding of Recombinant Organophosphorus Hydrolyses onto the Genome. Biochem. Biophys. Res. Commun. 2018, 495,
Inclusion Bodies Produced in Escherichia Coli. J. Apple 686–692. DOI: 10.1016/j.bbrc.2017.11.023.
Biotechnol. Rep. 2019, 6, 20–25. DOI: 10.29252/JABR.06.01.04. [24] Yates, L. E.; Natarajan, A.; Li, M.; Hale, M. E.; Mills, D. C.;
[8] Sarker, A.; Rathore, A. S.; Gupta, R. D. Evaluation of ScFv DeLisa, M. P. Glyco-Recoded Escherichia Coli:
Protein Recovery from E. coli by in Vitro Refolding and Mild Recombineering-Based Genome Editing of Native
Solubilization Process. Microb. Cell Fact. 2019, 18, 1–12. DOI: Polysaccharide Biosynthesis Gene Clusters. Metab. Eng. 2019,
10.1186/s12934-019-1053-9. 53, 59–68. DOI: 10.1016/j.ymben.2019.02.002.
[9] Halan, V.; Maity, S.; Bhambure, R.; Rathore, A. S. Multimodal [25] Benz, I.; Schmidt, M. A. Never Say Never Again: Protein
Chromatography for Purification of Biotherapeutics – a Glycosylation in Pathogenic Bacteria. Mol. Microbiol. 2002, 45,
Review. Curr. Protein Pept. Sci. 2019, 20, 4–13. DOI: 10.2174/ 267–276. DOI: 10.1046/j.1365-2958.2002.03030.x.
1389203718666171020103559. [26] Carrio, M. M.; Villaverde, A. Protein Aggregation as Bacterial
[10] Rosano, G. L.; Ceccarelli, E. A. Recombinant Protein Inclusion Bodies is Reversible. FEBS Lett. 2001, 489, 29–33.
Expression in Escherichia Coli: Advances and Challenges. DOI: 10.1016/s0014-5793(01)02073-7.
Front Microbiol. 2014, 1–17. [27] Sanchez, J. M.; Carratala, J. V.; Gifre-Renom, L.; Arıs, A.;
[11] Corchero, J. L.; Vazquez, E.; Garcıa-Fruit os, E.; Ferrer- Garcia-Fruitos, E.; Ferrer-Miralles, N. Quality Control of
Miralles, N.; Villaverde, A. Recombinant Protein Materials for Proteins Solubilized from Inclusion Bodies. Methods Mol. Biol.
Bioengineering and Nanomedicine. Nanomedicine (Lond) 2022, 2406, 469–477. DOI: 10.1007/978-1-0716-1859-2_28.
[28] Wang, L. Towards Revealing the Structure of Bacterial
2014, 9, 2817–2828. DOI: 10.2217/nnm.14.153.
Inclusion Bodies. Prion 2009, 3, 139–145. DOI: 10.4161/pri.3.
[12] Singhvi, P.; Saneja, A.; Srichandan, S.; Panda, A. K. Bacterial
3.9922.
Inclusion Bodies: A Treasure Trove of Bioactive Proteins.
[29] J€
urgen, B.; Breitenstein, A.; Urlacher, V.; B€ uttner, K.; Lin, H.;
Trends Biotechnol. 2020, 38, 474–486. DOI: 10.1016/j.tibtech.
Hecker, M.; Schweder, T.; Neubauer, P. Quality Control of
2019.12.011.
Inclusion Bodies in Escherichia Coli. Microb. Cell Fact. 2010,
[13] Huang, C. J.; Lin, H.; Yang, X. Industrial Production of
9, 13. DOI: 10.1186/1475-2859-9-41.
Recombinant Therapeutics in Escherichia Coli and Its Recent
[30] Siew, Y. Y.; Zhang, W. Downstream Processing of
Advancements. J. Ind. Microbiol. Biotechnol. 2012, 39,
Recombinant Human Insulin and Its Analogues Production
383–399. DOI: 10.1007/s10295-011-1082-9.
from E. coli Inclusion Bodies. Bioresour. Bioprocess 2021, 8,
[14] Margreiter, G.; Messner, P.; Caldwell, K. D.; Bayer, K. Size
1–27.
Characterization of Inclusion Bodies by Sedimentation Field-
[31] Rinas, U.; Garcia-Fruit os, E.; Corchero, J. L.; Vazquez, E.;
Flow Fractionation. J. Biotechnol. 2008, 138, 67–73. DOI: 10. Seras-Franzoso, J.; Villaverde, A. Bacterial Inclusion Bodies:
1016/j.jbiotec.2008.07.1995. Discovering Their Better Half. Trends Biochem. Sci. 2017, 42,
[15] Bhatwa, A.; Wang, W.; Hassan, Y. I.; Abraham, N.; Li, X. Z.; 726–737. DOI: 10.1016/j.tibs.2017.01.005.
Zhou, T. Challenges Associated with the Formation of [32] Coquel, A. S.; Jacob, J. P.; Primet, M.; Demarez, A.; Dimiccoli,
Recombinant Protein Inclusion Bodies in Escherichia Coli and M.; Julou, T.; Moisan, L.; Lindner, A. B.; Berry, H.
Strategies to Address Them for Industrial Applications. Front Localization of Protein Aggregation in Escherichia Coli is
Bioeng. Biotechnol. 2021, 1–18. Governed by Diffusion and Nucleoid Macromolecular
[16] Taylor, G.; Hoare, M.; Gray, D. R.; Marston, F. A. O. Size and Crowding Effect. PLoS Comput. Biol. 2013, 9, e1003038. DOI:
Density of Protein Inclusion Bodies. Bio/Technology 1986, 4, 10.1371/journal.pcbi.1003038.
553–557. [33] Oliveira, S. M. D.; Neeli-Venkata, R.; Goncalves, N. S. M.;
[17] Humer, D.; Spadiut, O. Wanted: More Monitoring and Santinha, J. A.; Martins, L.; Tran, H.; M€akel€a, J.; Gupta, A.;
Control During Inclusion Body Processing. World J. Microbiol. Barandas, M.; H€akkinen, A.; et al. Increased Cytoplasm
Biotechnol. 2018, 34, 1–9. DOI: 10.1007/s11274-018-2541-5. Viscosity Hampers Aggregate Polar Segregation in Escherichia
[18] Singh, A.; Upadhyay, V.; Upadhyay, A. K.; Singh, S. M.; Coli. Mol. Microbiol. 2016, 99, 686–699. DOI: 10.1111/mmi.
Panda, A. K. Protein Recovery from Inclusion Bodies of 13257.
Escherichia Coli Using Mild Solubilization Process. Microb. [34] Laloux, G.; Jacobs-Wagner, C. How Do Bacteria Localize
Cell Fact. 2015, 14, 10. DOI: 10.1186/s12934-015-0222-8. Proteins to the Cell Pole? J. Cell Sci. 2014, 127, 11–19.
[19] Khodabakhsh, F.; Zia, M. F.; Moazen, F.; Rabbani, M.; [35] Pu, Y.; Li, Y.; Jin, X.; Tian, T.; Ma, Q.; Zhao, Z.; Lin, S.;
Sadeghi, H. M. M. Comparison of the Cytoplasmic and Yuan , Chen, Z.; Li, B.; Yao, G.; et al. ATP-Dependent
Periplasmic Production of Reteplase in Escherichia coli. Prep. Dynamic Protein Aggregation Regulates Bacterial Dormancy
Biochem. Biotechnol. 2013, 43, 613–623. DOI: 10.1080/ Depth Critical for Antibiotic Tolerance. Mol. Cell 2019, 73,
10826068.2013.764896. 143–156.e4. DOI: 10.1016/j.molcel.2018.10.022.
[20] Jaroentomeechai, T.; Stark, J. C.; Natarajan, A.; Glasscock, [36] Rokney, A.; Shagan, M.; Kessel, M.; Smith, Y.; Rosenshine, I.;
C. J.; Yates, L. E.; Hsu, K. J.; Mrksich, M.; Jewett, M. C.; Oppenheim, AB. E. coli Transports Aggregated Proteins to the
Delisa, M. P. Single-Pot Glycoprotein Biosynthesis Using a Poles by a Specific and Energy-Dependent Process. J. Mol.
Cell-Free Transcription-Translation System Enriched With Biol. 2009, 392, 589–601. DOI: 10.1016/j.jmb.2009.07.009.
Glycosylation Machinery. Nat. Commun. 2018, 9, 1–11. [37] Govers, S. K.; Dutre, P.; Aertsen, A. In Vivo Disassembly and
[21] Schjoldager, K. T.; Narimatsu, Y.; Joshi, H. J.; Clausen, H. Reassembly of Protein Aggregates in Escherichia Coli. J.
Global View of Human Protein Glycosylation Pathways and Bacteriol. 2014, 196, 2325–2332. DOI: 10.1128/JB.01549-14.
746 K. KACHHAWAHA ET AL.

[38] Sun, X.; Shen, W.; Gao, Y.; Cai, M.; Zhou, M.; Zhang, Y. Garcıa-Fruitos, E.; Villaverde, A. A Nanostructured Bacterial
Heterologous Expression and Purification of a Marine Bioscaffold for the Sustained Bottom-Up Delivery of Protein
Alginate Lyase in Escherichia Coli. Protein Expr. Purif. 2019, Drugs. Nanomedicine (Lond). 2013, 8, 1587–1599. DOI: 10.
153, 97–104. DOI: 10.1016/j.pep.2018.09.002. 2217/nnm.12.188.
[39] Upadhyay, A. K.; Murmu, A.; Singh, A.; Panda, A. K. Kinetics [56] Berlec, A.; Strukelj, B. Current State and Recent Advances in
of Inclusion Body Formation and Its Correlation With the Biopharmaceutical Production in Escherichia Coli, Yeasts and
Characteristics of Protein Aggregates in Escherichia Coli. PLoS Mammalian Cells. J. Ind. Microbiol. Biotechnol. 2013, 40,
One 2012, 7, e33951. DOI: 10.1371/journal.pone.0033951. 257–274. DOI: 10.1007/s10295-013-1235-0.
[40] Hashemzadeh, M. S.; Mohammadi, M.; Ghaleh, H. E. G.; [57] Carneiro, S.; Ferreira, E. C.; Rocha, I. Metabolic Responses to
Sharti, M.; Choopani, A.; Panda, A. K. Expression, Recombinant Bioprocesses in Escherichia Coli. J. Biotechnol.
Solubilization, Refolding and Final Purification of 2013, 164, 396–408. DOI: 10.1016/j.jbiotec.2012.08.026.
Recombinant Proteins as Expressed in the Form of “Classical [58] Fakruddin, M.; Mazumdar, R. M.; Mannan, K. S. b.;
Inclusion Bodies” in E. Coli. Protein Pept. Lett. 2021, 28, Chowdhury, A.; Hossain, M. N. Critical Factors Affecting the
122–130. DOI: 10.2174/0929866527999200729182831. Success of Cloning, Expression, and Mass Production of
[41] Slouka, C.; Kopp, J.; Spadiut, O.; Herwig, C. Perspectives of Enzymes by Recombinant E. coli. ISRN Biotechnol. 2013,
Inclusion Bodies for Bio-Based Products: Curse or Blessing? 590587, 1–7.
Appl. Microbiol. Biotechnol. 2019, 103, 1143–1153. DOI: 10. [59] Huleani, S.; Roberts, M. R.; Beales, L.; Papaioannou, E. H.
1007/s00253-018-9569-1. Escherichia Coli as an Antibody Expression Host for the
[42] Belkova, M.; K€oszagova, R.; Nahalka, J.; Belkova, M. M. Active Production of Diagnostic Proteins: Significance and
Inclusion Bodies: The Unexpected Journey. J. Microb. Biotech. Expression. Crit. Rev. Biotechnol. 2022, 42, 756–773. DOI: 10.
Food Sci. 2022, 12, e5951. DOI: 10.55251/jmbfs.5951. 1080/07388551.2021.1967871.
[43] Peternel, S.; Bele, M.; Gaberc-Porekar, V.; Menart, V. [60] Joseph, B. C.; Pichaimuthu, S.; Srimeenakshi, S. An Overview
Nonclassical Inclusion Bodies in Escherichia Coli. Microb. Cell of the Parameters for Recombinant Protein Expression in
Fact. 2006, 5, P23–2. DOI: 10.1186/1475-2859-5-S1-P23. Escherichia Coli. J. Cell. Sci. Ther 2015, 6, 1–7. DOI: 10.4172/
[44] Trinh, N. T. M.; Thuoc, T. L.; Thao, D. T. P. Production of 2157-7013.1000221.
PEGylated GCSF from Non-Classical Inclusion Bodies [61] Korkmaz, G.; Holm, M.; Wiens, T.; Sanyal, S. Comprehensive
Expressed in Escherichia Coli. Avicenna J. Med. Biotechnol. Analysis of Stop Codon Usage in Bacteria and Its Correlation
2021, 13, 192–200. with Release Factor Abundance. J. Biol. Chem. 2014, 289,
[45] Peternel, S. Designing Non-Classical Inclusion Bodies. In 30334–30342. DOI: 10.1074/jbc.M114.606632.
Ribosomal Proteins and Protein Engineering; Ortendhal V., [62] Liu, L.; Yang, H.; Shin, H.; Chen, R. R.; Li, J.; Du, G.; Chen, J.
Salchow H., Eds; Nova Science Publishers: New York, 2010; How to Achieve High-Level Expression of Microbial Enzymes.
pp. 1–20. Bioengineered 2013, 4, 212–223. DOI: 10.4161/bioe.24761.
[46] Villaverde, A.; Garcıa-Fruitos, E.; Rinas, U.; Seras-Franzoso, J.; [64] Gopal, G. J.; Kumar, A. Strategies for the Production of
Kosoy, A.; Corchero, J. L.; Vazquez, E. Packaging Protein Recombinant Protein in Escherichia Coli. Protein. J. 2013, 32,
Drugs as Bacterial Inclusion Bodies for Therapeutic 419–425. DOI: 10.1007/s10930-013-9502-5.
Applications. Microb. Cell Fact. 2012, 11, 76. DOI: 10.1186/ [64] Baeshen, M. N.; Al-Hejin, A. M.; Bora, R. S.; Ahmed,
1475-2859-11-76. M. M. M.; Ramadan, H. A. I.; Saini, K. S.; Baeshen, N. A.;
[47] Hannig, G.; Makrides, S. C. Strategies for Optimizing Redwan, E. M. Production of Biopharmaceuticals in E. Coli:
Heterologous Protein Expression in Escherichia Coli. Trends Current Scenario and Future Perspectives. J. Microbiol.
Biotechnol. 1998, 16, 54–60. DOI: 10.1016/s0167- Biotechnol. 2015, 25, 953–962. DOI: 10.4014/jmb.1412.12079.
7799(97)01155-4. [65] Sørensen, H. P.; Mortensen, K. K. Advanced Genetic Strategies
[48] Ventura, S.; Villaverde, A. Protein Quality in Bacterial for Recombinant Protein Expression in Escherichia Coli. J.
Inclusion Bodies. Trends Biotechnol. 2006, 24, 179–185. DOI: Biotechnol. 2005, 115, 113–128. DOI: 10.1016/j.jbiotec.2004.08.
10.1016/j.tibtech.2006.02.007. 004.
[49] Singhvi, P.; Panda, A. K. Solubilization and Refolding of [66] Mauro, V. P.; Chappell, S. A. A Critical Analysis of Codon
Inclusion Body Proteins. Methods Mol. Biol. 2022, 2406, Optimization in Human Therapeutics Optimizing Codon
371–387. DOI: 10.1007/978-1-0716-1859-2_22. Usage for Increased Protein Expression. Trends. Mol. Med.
[50] Rosano, G. L.; Morales, E. S.; Ceccarelli, E. A. New Tools for 2014, 20, 604–613. DOI: 10.1016/j.molmed.2014.09.003.
Recombinant Protein Production in Escherichia Coli: A 5-Year [67] M€uhlmann, M.; Forsten, E.; Noack, S.; B€ uchs, J. Optimizing
Update. Protein Sci. 2019, 28, 1412–1422. DOI: 10.1002/pro. Recombinant Protein Expression via Automated Induction
3668. Profiling in Microtiter Plates at Different Temperatures.
[51] Phoeurk, C.; Mushtaq, A. U.; Rogne, P.; Wolf-Watz, M. Microb. Cell. Fact 2017, 16, 1–12. DOI: 10.1186/s12934-017-
Milligram Scale Expression, Refolding, and Purification of 0832-4.
Bombyx mori Cocoonase Using a Recombinant E. coli System. [68] Hayat, S. M. G.; Farahani, N.; Golichenari, B.; Sahebkar, A.
Protein Expr Purif 2021, 186, 105919. DOI: 10.1016/j.pep.2021. Recombinant Protein Expression in Escherichia Coli (E.Coli):
105919. What We Need to Know. Curr. Pharm. Des. 2018, 24,
[52] Sanchez-Garcia, L.; Martın, L.; Mangues, R.; Ferrer-Miralles, 718–725. DOI: 10.2174/1381612824666180131121940.
N.; Vazquez, E.; Villaverde, A. Recombinant Pharmaceuticals [69] Papaneophytou, C. P.; Kontopidis, G. Statistical Approaches to
from Microbial Cells: A 2015 Update. Microb. Cell Fact 2016, Maximize Recombinant Protein Expression in Escherichia Coli:
15, 1–7. DOI: 10.1186/s12934-016-0437-3. A General Review. Protein. Expr. Purif. 2014, 94, 22–32. DOI:
[53] de Marco, A.; Ferrer-Miralles, N.; Garcia-Fruit os, E.; Mitraki, 10.1016/j.pep.2013.10.016.
A.; Peternel, S.; Rinas, U.; Trujillo-Roldan, M. A.; Valdez- [70] Tsai, W. C.; Wu, T. C.; Chiang, B. L.; Wen, H. W. Cloning,
Cruz, N. A.; Vazquez, E.; Villaverde, A. Bacterial Inclusion Expression, and Purification of Recombinant Major Mango
Bodies Are Industrially Exploitable Amyloids. FEMS Microbiol. Allergen Man i 1 in Escherichia Coli. Protein. Expr. Purif.
Rev. 2019, 43, 53–72. DOI: 10.1093/femsre/fuy038. 2017, 130, 35–43. DOI: 10.1016/j.pep.2016.06.009.
[54] Schumann, W.; Ferreira, L. C. S. Production of Recombinant [71] Rosano, G. L.; Ceccarelli, E. A. Recombinant Protein
Proteins in Escherichia Coli. Genet. Mol. Biol 2004, 27, Expression in Escherichia Coli: Advances and Challenges.
442–453. DOI: 10.1590/S1415-47572004000300022. Front. Microbiol. 2014, 5, 172.
[55] Seras-Franzoso, J.; Peebo, K.; Luis Corchero, J.; Tsimbouri, [72] Khow, O.; Suntrarachun, S. Strategies for Production of Active
P. M.; Unzueta, U.; Rinas, U.; Dalby, M. J.; Vazquez, E.; Eukaryotic Proteins in Bacterial Expression System. Asian.
PREPARATIVE BIOCHEMISTRY & BIOTECHNOLOGY 747

Pac. J. Trop. Biomed. 2012, 2, 159–162. DOI: 10.1016/S2221- Protein Synthesis for Extending the Abilities of Biological
1691(11)60213-X. Systems. Front. Bioeng. Biotechnol. 2019, 7, 1–16.
[73] Coskun, K. A.; Yurekli, N.; Abay, E. C.; Tutar, M.; Al, M.; [91] Guzman-Chavez, F.; Arce, A.; Adhikari, A.; Vadhin, S.;
Tutar, Y.; Coskun, K. A.; Yurekli, N.; Abay, E. C.; Tutar, M., Pedroza-Garcia, J. A.; Gandini, C.; Ajioka, J. W.; Molloy, J.;
Chapter 1: Structure- and Design-Based Difficulties in Sanchez-Nieto, S.; Varner, J. D.; et al. Constructing Cell-Free
Recombinant Protein Purification in Bacterial Expression. In Expression Systems for Low-Cost Access. ACS. Synth. Biol.
Protein Detection; Tutar, Y., Tutar, L., Eds.; IntechOpen 2022, 11, 1114–1128. DOI: 10.1021/acssynbio.1c00342.
Limited: London, 2022; pp 1–19. [92] Islam, M. S.; Aryasomayajula, A.; Selvaganapathy, P. R. A
[74] Huang, C. J.; Peng, H. L.; Patel, A. K.; Singhania, R. R.; Dong, Review on Macroscale and Microscale Cell Lysis Methods.
C. d.; Cheng, C. Y. Effects of Lower Temperature on Micromachines 2017, 8, 83. DOI: 10.3390/mi8030083.
Expression and Biochemical Characteristics of HCV NS3 [93] Gomes, T. A.; Zanette, C. M.; Spier, M. R. An Overview of
Antigen Recombinant Protein. Catalysts 2021, 11, 1297. DOI: Cell Disruption Methods for Intracellular Biomolecules
10.3390/catal11111297. Recovery. Prep. Biochem. Biotechnol. 2020, 50, 635–654. DOI:
[75] Farshdari, F.; Ahmadzadeh, M.; Nematollahi, L.; Mohit, E. The 10.1080/10826068.2020.1728696.
Improvement of Anti-HER2 ScFv Soluble Expression in [94] Pyatkovskyy, T. I.; Shynkaryk, M. v.; Mohamed, H. M.;
Escherichia Coli. Braz. J. Pharm. Sci. 2020, 56, 17861. DOI: 10. Yousef, A. E.; Sastry, S. K. Effects of Combined High Pressure
1590/s2175-97902019000317861. (HPP), Pulsed Electric Field (PEF) and Sonication Treatments
[76] Li, R. Y.; Cheng, C. Y. Investigation of Inclusion Body on Inactivation of Listeria Innocua. J. Food. Eng. 2018, 233,
Formation in Recombinant Escherichia Coli with a Bioimaging 49–56. DOI: 10.1016/j.jfoodeng.2018.04.002.
System. J. Biosci. Bioeng. 2009, 107, 512–515. DOI: 10.1016/j. [95] Harrison, S. T. L. Bacterial Cell Disruption: A Key Unit
jbiosc.2009.01.025. Operation in the Recovery of Intracellular Products.
[77] Sugimoto, S.; Yokoo, Y.; Hatakeyama, N.; Yotsuji, A.; Teshiba, Biotechnol. Adv 1991, 9, 217–240. DOI: 10.1016/0734-
S.; Hagino, H. Higher Culture PH is Preferable for Inclusion 9750(91)90005-G.
Body Formation of Recombinant Salmon Growth Hormone in [96] Asenjo, J. A. Product Release and Recovery. Sep. Processes
Esherichia Coli. Biotechnol. Lett. 1991, 13, 385–388. DOI: 10. Biotechnol. Part II 2020, 1, 142.
1007/BF01030987. [97] Haddad, L.; Babaeipour, V.; Mofid, M. R. The Effect of Cell
[78] Wittung-Stafshede, P. Role of Cofactors in Protein Folding. Disruption Techniques and Chaotropic Agents on the
Acc. Chem. Res. 2002, 35, 201–208. DOI: 10.1021/ar010106e. Downstream Purification Process of Mecasermin Produced as
[79] Bae, S. W.; Eom, D.; Mai, N. L.; Koo, Y. M. Refolding of Inclusion Body in E. coli. Res. Pharm. Sci. 2015, 10, 553–561.
Horseradish Peroxidase is Enhanced in Presence of Metal [98] Primo, E. D.; Otero, L. H.; Ruiz, F.; Klinke, S.; Giordano, W.
Cofactors and Ionic Liquids. Biotechnol. J. 2016, 11, 464–472. The Disruptive Effect of Lysozyme on the Bacterial Cell Wall
DOI: 10.1002/biot.201500142. Explored by an In-Silico Structural Outlook. Biochem. Mol.
[80] Graumann, K.; Premstaller, A. Manufacturing of Recombinant Biol. Educ. 2018, 46, 83–90. DOI: 10.1002/bmb.21092.
Therapeutic Proteins in Microbial Systems. Biotechnol. J. 2006, [99] Martins, M.; Ooi, C. W.; Neves, M. C.; Pereira, J. F. B.;
1, 164–186. DOI: 10.1002/biot.200500051. Coutinho, J. A. P.; Ventura, S. P. M. Extraction of
[81] Sandomenico, A.; Sivaccumar, J. P.; Ruvo, M. Evolution of Recombinant Proteins from Escherichia Coli by Cell
Escherichia Coli Expression System in Producing Antibody Disruption With Aqueous Solutions of Surface-Active
Recombinant Fragments. Int. J. Mol. Sci. 2020, 21, 1–39. Compounds. J. Chem. Technol. Biotechnol. 2018, 93,
[82] Kastenhofer, J.; Rajamanickam, V.; Libiseller-Egger, J.; Spadiut, 1864–1870. DOI: 10.1002/jctb.5596.
O. Monitoring and Control of E. coli Cell Integrity. J. [100] Pekarsky, A.; Spadiut, O.; Rajamanickam, V.; Wurm, D. J. A
Biotechnol. 2021, 329, 1–12. DOI: 10.1016/j.jbiotec.2021.01. Fast and Simple Approach to Optimize the Unit Operation
009. High Pressure Homogenization - A Case Study for a Soluble
[83] Tripathi, N. K.; Shrivastava, A. Recent Developments in Therapeutic Protein in E. Coli. Prep. Biochem. Biotechnol.
Bioprocessing of Recombinant Proteins: Expression Hosts and 2019, 49, 74–81. DOI: 10.1080/10826068.2018.1536988.
Process Development. Front. Bioeng. Biotechnol. 2019, 7, 1–35. [101] Sharma, M.; Tyagi, J. L.; Poluri, K. M. Quantifying Bacterial
DOI: 10.3389/fbioe.2019.00420. Cell Lysis Using GFP Based Fluorimetric Assay. Int. J. Biol.
[84] Bellani, C. F.; Ajeian, J.; Duffy, L.; Miotto, M.; Groenewegen, Macromol. 2019, 138, 881–889. DOI: 10.1016/j.ijbiomac.2019.
L.; Connon, C. J. Scale-Up Technologies for the Manufacture 07.172.
of Adherent Cells. Front. Nutr. 2020, 7, 1–14. [102] Baumann, P.; Hubbuch, J. Downstream Process Development
[85] Neurohr, G. E.; Amon, A. Relevance and Regulation of Cell Strategies for Effective Bioprocesses: Trends, Progress, and
Density. Trends. Cell. Biol. 2020, 30, 213–225. DOI: 10.1016/j. Combinatorial Approaches. Eng. Life. Sci. 2017, 17,
tcb.2019.12.006. 1142–1158. DOI: 10.1002/elsc.201600033.
[86] Subramaniam, R.; Thirumal, V.; Chistoserdov, A.; Bajpai, R.; [103] Rodrıguez-Carmona, E.; Cano-Garrido, O.; Seras-Franzoso, J.;
Bader, J.; Popovic, M. High-Density Cultivation in the Villaverde, A.; Garcıa-Fruit os, E. Isolation of Cell-Free
Production of Microbial Products. Chem. Biochem. Eng. Q Bacterial Inclusion Bodies. Microb. Cell. Fact 2010, 9, 1–9.
2019, 32, 451–464. DOI: 10.15255/CABEQ.2018.1394. DOI: 10.1186/1475-2859-9-71.
[87] Gupta, S. K.; Shukla, P. Advanced Technologies for Improved [104] Rueda, F.; Cano-Garrido, O.; Mamat, U.; Wilke, K.; Seras-
Expression of Recombinant Proteins in Bacteria: Perspectives Franzoso, J.; Garcıa-Fruit
os, E.; Villaverde, A. Production of
and Applications. Crit. Rev. Biotechnol. 2016, 36, 1089–1098. Functional Inclusion Bodies in Endotoxin-Free Escherichia
DOI: 10.3109/07388551.2015.1084264. Coli. Appl. Microbiol. Biotechnol. 2014, 98, 9229–9238. DOI:
[88] Panda, A. K. Bioprocessing of Therapeutic Proteins from the 10.1007/s00253-014-6008-9.
Inclusion Bodies of Escherichia Coli. Adv. Biochem. Eng. [105] Singh, A.; Upadhyay, V.; Panda, A. K. Solubilization and
Biotechnol. 2003, 85, 43–93. DOI: 10.1007/3-540-36466-8_3. Refolding of Inclusion Body Proteins. Methods. Mol. Biol.
[89] Chiba, C. H.; Knirsch, M. C.; Azzoni, A. R.; Moreira, A. R.; 2015, 1258, 283–291. DOI: 10.1007/978-1-4939-2205-5_15.
Stephano, M. A. Cell-Free Protein Synthesis: Advances on [106] Walther, C.; Mayer, S.; Sekot, G.; Antos, D.; Hahn, R.;
Production Process for Biopharmaceuticals and Jungbauer, A.; D€ urauer, A. Mechanism and Model for
Immunobiological Products. Biotechniques 2021, 70, 126–133. Solubilization of Inclusion Bodies. Chem. Eng. Sci. 2013, 101,
DOI: 10.2144/btn-2020-0155. 631–641. DOI: 10.1016/j.ces.2013.07.026.
[90] Khambhati, K.; Bhattacharjee, G.; Gohil, N.; Braddick, D.; [107] Fischer, B.; Sumner, I.; Goodenough, P. Isolation,
Kulkarni, V.; Singh, V. Exploring the Potential of Cell-Free Renaturation, and Formation of Disulfide Bonds of Eukaryotic
748 K. KACHHAWAHA ET AL.

Proteins Expressed in Escherichia Coli as Inclusion Bodies. Temperature and Pressure on the Sub-Nanosecond Dynamics
Biotechnol. Bioeng. 1993, 41, 3–13. DOI: 10.1002/bit. and Folding Stability of Lysozyme. Phys. Chem. Chem. Phys.
260410103. 2017, 19, 14230–14237. DOI: 10.1039/C7CP00705A.
[108] Datta, I.; Gautam, S.; Gupta, M. N. Microwave Assisted [124] Phelps, D. J.; Hesterberg, L. K. Protein Disaggregation and
Solubilization of Inclusion Bodies. Sustain. Chem. Processes. Refolding Using High Hydrostatic Pressure. J. Chem. Technol.
2013, 1, 1–7. Biotechnol. 2007, 82, 610–613. DOI: 10.1002/jctb.1707.
[109] Mukherjee, J.; Nath Gupta, M. Paradigm Shifts in Our Views [125] Mizutani, H.; Sugawara, H.; Buckle, A. M.; Sangawa, T.;
on Inclusion Bodies. CBE 2015, 3, 47–55. DOI: 10.2174/ Miyazono, K. I.; Ohtsuka, J.; Nagata, K.; Shojima, T.; Nosaki,
2212711902666150302212051. S.; Xu, Y.; et al. REFOLDdb: A New and Sustainable Gateway
[110] Tsumoto, K.; Abe, R.; Ejima, D.; Arakawa, T. Non-Denaturing to Experimental Protocols for Protein Refolding. BMC. Struct.
Solubilization of Inclusion Bodies. Curr. Pharm. Biotechnol. Biol. 2018, 17, 8. DOI: 10.1186/s12900-017-0074-z.
2010, 11, 309–312. DOI: 10.2174/138920110791111924. [126] Chow, M. K. M.; Amin, A. A.; Fulton, K. F.; Fernando, T.;
[111] Mohammadian, A.; Kaghazian, H.; Kavianpour, A.; Jalalirad, Kamau, L.; Batty, C.; Louca, M.; Ho, S.; Whisstock, J. C.;
R. Solubilization of Inclusion Body Proteins Using Low and Bottomley, S. P.; et al. The REFOLD Database: A Tool for the
Very Low Concentrations of Chemicals: Implications of Novel Optimization of Protein Expression and Refolding. Nucleic.
Combined Chemical Treatment Designs in Enhancement of Acids Res. 2006, 34, D207–D212. DOI: 10.1093/nar/gkj080.
Post-Solubilization Target Protein Purity and Biological [127] Yamaguchi, H.; Miyazaki, M. Refolding Techniques for
Activity. J. Chem. Technol. Biotechnol. 2018, 93, 1579–1587. Recovering Biologically Active Recombinant Proteins from
DOI: 10.1002/jctb.5525. Inclusion Bodies. Biomolecules 2014, 4, 235–251. DOI: 10.
[112] Padhiar, A. A.; Chanda, W.; Joseph, T. P.; Guo, X.; Liu, M.; 3390/biom4010235.
Sha, L.; Batool, S.; Gao, Y.; Zhang, W.; Huang, M.; et al. [128] Kato, A.; Ohashi, H. Quick Refolding of High-Concentration
Comparative Study to Develop a Single Method for Retrieving Proteins Via Microchannel Dialysis. Ind. Eng. Chem. Res.
Wide Class of Recombinant Proteins from Classical Inclusion 2021, 60, 10076–10082. DOI: 10.1021/acs.iecr.1c00410.
Bodies. Appl. Microbiol. Biotechnol. 2018, 102, 2363–2377. [129] Kashanian, F.; Masoudi, M. M.; Shamloo, A.; Habibi-Rezaei,
DOI: 10.1007/s00253-018-8754-6. M.; Moosavi-Movahedi, A. A. Modeling, Simulation, and
[113] Rosa Da Silva, C. M.; Chura-Chambi, R. M.; Ramos Pereira, Employing Dilution–Dialysis Microfluidic Chip (DDMC) for
L.; Cordeiro, Y.; de Souza Ferreira, L. C.; Morganti, L. Heightening Proteins Refolding Efficiency. Bioprocess. Biosyst.
Association of High Pressure and Alkaline Condition for Eng. 2018, 41, 707–714. DOI: 10.1007/s00449-018-1904-5.
Solubilization of Inclusion Bodies and Refolding of the NS1 [130] Nishinami, S.; Yoshizawa, S.; Arakawa, T.; Shiraki, K.
Protein from Zika Virus. BMC. Biotechnol. 2018, 18, 1–10. Allantoin and Hydantoin as New Protein Aggregation
DOI: 10.1186/s12896-018-0486-2. Suppressors. Int. J. Biol. Macromol. 2018, 114, 497–503. DOI:
[114] Chura-Chambi, R. M.; de Brand~ao Prieto-Da-Silva, A. R.; di 10.1016/j.ijbiomac.2018.03.011.
Lela, M. M.; Oliveira, J. E.; Abreu, P. E. A.; Meireles, L. R.; de [131] Brusotti, G.; Calleri, E.; Colombo, R.; Massolini, G.; Rinaldi,
Andrade Junior, H. F.; Morganti, L. High Level SARS-CoV-2 F.; Temporini, C. Advances on Size Exclusion
Nucleocapsid Refolding Using Mild Condition for Inclusion Chromatography and Applications on the Analysis of Protein
Bodies Solubilization: Application of High Pressure at PH 9.0. Biopharmaceuticals and Protein Aggregates: A Mini Review.
PLoS. One 2022, 17, e0262591. DOI: 10.1371/journal.pone. Chromatographia 2018, 81, 3–23. DOI: 10.1007/s10337-017-
0262591. 3380-5.
[115] Maksum, I. P.; Yosua, Y.; Nabiel, A.; Pratiwi, R. D.; Sriwidodo, [132] Burgess, R. R. A Brief Practical Review of Size Exclusion
S.; Soedjanaatmadja, U. M. S. Refolding of Bioactive Human Chromatography: Rules of Thumb, Limitations, and
Epidermal Growth Factor from E. coli BL21(DE3) Inclusion Troubleshooting. Protein. Expr. Purif. 2018, 150, 81–85. DOI:
Bodies & Evaluations on Its In Vitro & In Vivo Bioactivity. 10.1016/j.pep.2018.05.007.
Heliyon 2022, 8, e09306. DOI: 10.1016/j.heliyon.2022.e09306. [133] Zhang, K.; Liu, X. Reprint of “Mixed-Mode Chromatography
[116] Costa, S.; Almeida, A.; Castro, A.; Domingues, L. Fusion Tags in Pharmaceutical and Biopharmaceutical Applications”. J.
for Protein Solubility, Purification, and Immunogenicity in Pharm. Biomed. Anal. 2016, 130, 19–34. DOI: 10.1016/j.jpba.
Escherichia Coli: The Novel Fh8 System. Front. Microbiol. 2016.09.013.
2014, 5, 63. [134] Eriksson, K. O., Chapter 19: Hydrophobic Interaction
[117] Fatima, K.; Naqvi, F.; Younas, H. A Review: Molecular Chromatography. In Biopharmaceutical Processing:
Chaperone-Mediated Folding, Unfolding and Disaggregation Development, Design, and Implementation of Manufacturing
of Expressed Recombinant Proteins. Cell. Biochem. Biophys. Processes; Jagschies, G., Lindskog, E., Ła˛cki, K., Galliher, P.,
2021, 79, 153–174. DOI: 10.1007/s12013-021-00970-5. Eds.; Elsevier: Amsterdam, 2017; pp. 401–408.
[118] Xie, X.; Wu, P.; Huang, X.; Bai, W. F.; Li, B.; Shi, N. Retro- [135] Kaur, J.; Singh, A.; Panda, A. K.; Lal, R. Protocol for In-Vitro
Protein XXA is a Remarkable Solubilizing Fusion Tag for Purification and Refolding of Hexachlorocyclohexane
Inclusion Bodies. Microb. Cell. Fact 2022, 21, 15. DOI: 10. Degrading Enzyme Haloalkane Dehalogenase LinB from
1186/s12934-022-01776-7. Inclusion Bodies. Enzyme. Microb. Technol. 2021, 146, 109760.
[119] Ki, M. R.; Pack, S. P. Fusion Tags to Enhance Heterologous DOI: 10.1016/j.enzmictec.2021.109760.
Protein Expression. Appl. Microbiol. Biotechnol. 2020, 104, [136] Ma, S.; Li, Y.; Ma, C.; Wang, Y.; Ou, J.; Ye, M. Challenges and
2411–2425. DOI: 10.1007/s00253-020-10402-8. Advances in the Fabrication of Monolithic Bioseparation
[120] Schramm, F. D.; Schroeder, K.; Jonas, K. Protein Aggregation Materials and Their Applications in Proteomics Research. Adv.
in Bacteria. FEMS. Microbiol. Rev. 2020, 44, 54–72. DOI: 10. Mater. 2019, 31, 1902023–1902027. DOI: 10.1002/adma.
1093/femsre/fuz026. 201902023.
[121] Hirota, N.; Edskes, H.; Hall, D. Unified Theoretical [137] Hoffmann, D.; Leber, J.; Loewe, D.; Lothert, K.; Oppermann,
Description of the Kinetics of Protein Aggregation. Biophys. T.; Zitzmann, J.; Weidner, T.; Salzig, D.; Wolff, M.; Czermak,
Rev. 2019, 11, 191–208. DOI: 10.1007/s12551-019-00506-5. P., Chapter 5: Purification of New Biologicals Using
[122] Akiba, H.; Tsumoto, K. Expression in Bacteria and Refolding. Membrane-Based Processes. In Current Trends and Future
In Advance Methods in Structural Biology; Senda, T., Maenaka, Developments on (Bio-) Membranes: Membrane Processes in the
K., Eds.; Springer Protocols Handbooks: Tokyo, 2016; pp. Pharmaceutical and Biotechnological Field; Basile, A.,
3–23. Charcosset, C., Eds.; Elsevier: Amsterdam, 2019; pp. 123–150.
[123] Al-Ayoubi, S. R.; Schummel, P. H.; Golub, M.; Peters, J.; [138] Poddar, S.; Sharmeen, S.; Hage, D. S. Affinity Monolith
Winter, R. Influence of Cosolvents, Self-Crowding, Chromatography: A Review of General Principles and Recent
PREPARATIVE BIOCHEMISTRY & BIOTECHNOLOGY 749

Developments. Electrophoresis 2021, 42, 2577–2598. DOI: 10. [154] Taraban, M. B.; Depaz, R. A.; Lobo, B.; Yu, Y. B. Use of
1002/elps.202100163. Water Proton NMR to Characterize Protein Aggregates:
[139] Dods, S. R.; Hardick, O.; Stevens, B.; Bracewell, D. G. Gauging the Response and Sensitivity. Anal. Chem. 2019, 91,
Fabricating Electrospun Cellulose Nanofibre Adsorbents for 4107–4115. DOI: 10.1021/acs.analchem.8b05733.
Ion-Exchange Chromatography. J. Chromatogr. A 2015, 1376, [155] Bansal, R.; Gupta, S.; Rathore, A. S. Analytical Platform for
74–83. DOI: 10.1016/j.chroma.2014.12.010. Monitoring Aggregation of Monoclonal Antibody
[140] Fu, Q.; Duan, C.; Yan, Z.; Si, Y.; Liu, L.; Yu, J.; Ding, B. Therapeutics. Pharm. Res. 2019, 36, 1–11. DOI: 10.1007/
Electrospun Nanofibrous Composite Materials: A Versatile s11095-019-2690-8.
Platform for High Efficiency Protein Adsorption and [156] Stetefeld, J.; McKenna, S. A.; Patel, T. R. Dynamic Light
Separation. Compos. Commun. 2018, 8, 92–100. DOI: 10.1016/ Scattering: A Practical Guide and Applications in Biomedical
j.coco.2017.11.007. Sciences. Biophys. Rev. 2016, 8, 409–427. DOI: 10.1007/
[141] Miles, A. J.; Janes, R. W.; Wallace, B. A. Tools and Methods s12551-016-0218-6.
for Circular Dichroism Spectroscopy of Proteins: A Tutorial [157] Aggregates, P. Protein Aggregates. Biophys. J. 2008, 94,
Review. Chem. Soc. Rev. 2021, 50, 8400–8413. DOI: 10.1039/ 933–946.
d0cs00558d. [158] Philo, J. S. Is Any Measurement Method Optimal for All
[142] Pathak, M.; Dixit, S.; Muthukumar, S.; Rathore, A. S. Aggregate Sizes and Types? AAPS J. 2006, 8, E564–E571. DOI:
Analytical Characterization of In Vitro Refolding in the 10.1208/aapsj080365.
Quality by Design Paradigm: Refolding of Recombinant [159] Tatkiewicz, W.; Elizondo, E.; Moreno, E.; Dıez-Gil, C.;
Human Granulocyte Colony Stimulating Factor. J. Pharm. Ventosa, N.; Veciana, J.; Ratera, I. Methods for
Biomed. Anal. 2016, 126, 124–131. DOI: 10.1016/j.jpba.2016. Characterization of Protein Aggregates. Methods. Mol. Biol.
05.001. 2015, 1258, 387–401. DOI: 10.1007/978-1-4939-2205-5_22.
[143] Ling, C.; Zhang, J.; Lin, D.; Tao, A. Approaches for the [160] Kumar, M.; Pant, A.; Bansal, R.; Pandey, A.; Gomes, J.; Khare,
Generation of Active Papain-like Cysteine Proteases K.; Singh Rathore, A.; Banerjee, M. Electron Microscopy-Based
from Inclusion Bodies of Escherichia Coli. World. J. Semi-Automated Characterization of Aggregation in
Microbiol. Biotechnol. 2015, 31, 681–690. DOI: 10.1007/ Monoclonal Antibody Products. Comput. Struct. Biotechnol. J.
s11274-015-1804-7. 2020, 18, 1458–1465. DOI: 10.1016/j.csbj.2020.06.009.
[144] Zhang, H.; Zheng, X.; Kwok, R. T. K.; Wang, J.; Leung, [161] Hurtley, S. M. Continuing the Resolution Revolution. Science
N. L. C.; Shi, L.; Sun, J. Z.; Tang, Z.; Lam, J. W. Y.; Qin, A.; 2018, 360, 280.11–282. DOI: 10.1126/science.360.6386.280-k.
et al. In Situ Monitoring of Molecular Aggregation Using [162] Almeida, Z. L.; Brito, R. M. M. Structure and Aggregation
Circular Dichroism. Nat. Commun. 2018, 9, 1–9. DOI: 10. Mechanisms in Amyloids. Molecules 2020, 25, 1195–1130.
1038/s41467-018-07299-3.
DOI: 10.3390/molecules25051195.
[145] Kelly, S. M.; Price, N. C. The Use of Circular Dichroism in [163] Schluter, H. Reversed-Phase Chromatography. J. Chromatogr.
the Investigation of Protein Structure and Function. Curr.
A 2000, 61, 147–234.
Protein. Pept. Sci. 2000, 1, 349–384. DOI: 10.2174/
[164] Hong, P.; Koza, S.; Bouvier, E. S. P. A Review Size-Exclusion
1389203003381315.
Chromatography for the Analysis of Protein Biotherapeutics
[146] Gatti-Lafranconi, P.; Natalello, A.; Ami, D.; Doglia, S. M.;
and Their Aggregates. J. Liq. Chromatogr. Relat. Technol.
Lotti, M. Concepts and Tools to Exploit the Potential of
2012, 35, 2923–2950. DOI: 10.1080/10826076.2012.743724.
Bacterial Inclusion Bodies in Protein Science and
[165] Haberger, M.; Leiss, M.; Heidenreich, A.-K.; Pester, O.;
Biotechnology. FEBS. J. 2011, 278, 2408–2418. DOI: 10.1111/j.
Hafenmair, G.; Hook, M.; Bonnington, L.; Wegele, H.; Haindl,
1742-4658.2011.08163.x.
M.; Reusch, D.; Bulau, P. Rapid Characterization of
[147] Pignataro, M. F.; Herrera, M. G.; Dodero, V. I. Evaluation of
Biotherapeutic Proteins by Size-Exclusion Chromatography
Peptide/Protein Self-Assembly and Aggregation by
Spectroscopic Methods. Molecules 2020, 25, 4854–4140. DOI: Coupled to Native Mass Spectrometry. MABS 2016, 8,
10.3390/molecules25204854. 331–339. DOI: 10.1080/19420862.2015.1122150.
[148] Dodero, V. I.; Messina, P. V., Chapter 4: Analyzing the [166] Castagnola, M. Capillary Electrophoresis: Principles, Practice
Solution State of Protein Structure, Interactions, and Ligands and Applications. J. Chromatogr. A 1992, 52, 118.
by Spectroscopic Methods. In Proteins in Solution and at [167] Espinosa-de la Garza, C. E.; Perdomo-Ab undez, F. C.;
Interfaces: Methods and Applications in Biotechnology and Campos-Garcıa, V. R.; Perez, N. O.; Flores-Ortiz, L. F.;
Materials Science; Ruso, J. M., Pi~ neiro, A., Eds.; Wiley: New Medina-Rivero, E. Capillary Gel Electrophoresis for the
Jersey, 2013; pp. 73–98. Quantification and Purity Determination of Recombinant
[149] Hawe, A.; Sutter, M.; Jiskoot, W. Extrinsic Fluorescent Dyes as Proteins in Inclusion Bodies. Electrophoresis 2013, 34,
Tools for Protein Characterization. Pharm. Res. 2008, 25, 2754–2759. DOI: 10.1002/elps.201300232.
1487–1499. DOI: 10.1007/s11095-007-9516-9. [168] Formolo, T.; Ly, M.; Levy, M.; Kilpatrick, L.; Lute, S.; Phinney,
[150] Tokunaga, Y.; Takeuchi, K. Role of NMR in High Ordered K.; Marzilli, L.; Brorson, K.; Boyne, M.; Davis, D.
Structure Characterization of Monoclonal Antibodies. Int. J. Determination of the NISTmAb Primary Structure. ACS Symp.
Mol. Sci. 2021, 22, 1–12. Ser. 2015, 1201, 1–62.
[151] Humer, D.; Spadiut, O. Wanted: More Monitoring and [169] Gani, K.; Chirmade, T.; Ughade, S.; Thulasiram, H.;
Control During Inclusion Body Processing. World. J. Bhambure, R. Understanding Unfolding and Refolding of the
Microbiol. Biotechnol. 2018, 34, 1–9. DOI: 10.1007/s11274-018- Antibody Fragment (Fab) III: Mapping Covalent and Non-
2541-5. Covalent Interactions During In-Vitro Refolding of Light
[152] Schaefer, A.; Naser, D.; Siebeneichler, B.; Tarasca, M. V.; Chain, Heavy Chain, and Fab. Biochem. Eng. J. 2022, 187,
Meiering, E. M. Methodological Advances and Strategies for 108644. DOI: 10.1016/j.bej.2022.108644.
High Resolution Structure Determination of Cellular Protein [170] Puranik, A.; Rasam, P.; Dandekar, P.; Jain, R. Development
Aggregates. J. Biol. Chem. 2022, 298, 102197. DOI: 10.1016/j. and Optimization of a LC-MS Based Multi-Attribute Method
jbc.2022.102197. (MAM) Workflow for Characterization of Therapeutic Fc-
[153] Kamatari, Y. O.; Kitahara, R.; Yamada, H.; Yokoyama, S.; Fusion Protein. Anal. Biochem. 2023, 660, 114969. DOI: 10.
Akasaka, K. High-Pressure NMR Spectroscopy for 1016/j.ab.2022.114969.
Characterizing Folding Intermediates and Denatured States of [171] Rosenberg, A. S. Effects of Protein Aggregates: An
Proteins. Methods 2004, 34, 133–143. DOI: 10.1016/j.ymeth. Immunologic Perspective. AAPS. J 2006, 8, E501–E507. DOI:
2004.03.010. 10.1208/aapsj080359.
750 K. KACHHAWAHA ET AL.

[172] Garcıa-Fruitos, E.; Vazquez, E.; Dıez-Gil, C.; Corchero, J. L.; Escherichia Coli for Enhanced Production of 1-Butanol. Metab.
Seras-Franzoso, J.; Ratera, I.; Veciana, J.; Villaverde, A. Eng. 2017, 40, 41–49. DOI: 10.1016/j.ymben.2016.12.012.
Bacterial Inclusion Bodies: Making Gold from Waste. Trends [188] J€ager, V. D.; Lamm, R.; Kloß, R.; Kaganovitch, E.; Gr€ unberger,
Biotechnol 2012, 30, 65–70. DOI: 10.1016/j.tibtech.2011.09.003. A.; Pohl, M.; B€ uchs, J.; Jaeger, K. E.; Krauss, U. A Synthetic
[173] Serna, N.; Cano-Garrido, O.; Sanchez, J. M.; Sanchez-Chardi, Reaction Cascade Implemented by Colocalization of Two
A.; Sanchez-Garcıa, L.; L opez-Laguna, H.; Fernandez, E.; Proteins Within Catalytically Active Inclusion Bodies. ACS.
Vazquez, E.; Villaverde, A. Release of Functional Fibroblast Synth. Biol. 2018, 7, 2282–2295. DOI: 10.1021/acssynbio.
Growth Factor-2 from Artificial Inclusion Bodies. J. Control 8b00274.
Release 2020, 327, 61–69. DOI: 10.1016/j.jconrel.2020.08.007. [189] Martınez-Miguel, M.; Tatkiewicz, W.; K€ ober, M.; Ventosa, N.;
[174] Sanchez, J. M.; L 
opez-Laguna, H.; Alamo, P.; Serna, N.; Veciana, J.; Guasch, J.; Ratera, I. Methods for Processing
Sanchez-Chardi, A.; Nolan, V.; Cano-Garrido, O.; Casanova, Protein Aggregates Into Surfaces. Methods. Mol. Biol. 2022,
I.; Unzueta, U.; Vazquez, E.; et al. Artificial Inclusion Bodies 2406, 517–530. DOI: 10.1007/978-1-0716-1859-2_31.
for Clinical Development. Adv. Sci. (Weinh) 2020, 7, 1902420. [190] Gifre-Renom, L.; Seras-Franzoso, J.; Rafael, D.; Andrade, F.;
DOI: 10.1002/advs.201902420. Cano-Garrido, O.; Martinez-Trucharte, F.; Ugarte-Berzal, E.;
[175] Liese, A.; Hilterhaus, L. Evaluation of Immobilized Enzymes Martens, E.; Boon, L.; Villaverde, A.; et al. The Biological
for Industrial Applications. Chem. Soc. Rev. 2013, 42, Potential Hidden in Inclusion Bodies. Pharmaceutics 2020, 12,
6236–6249. DOI: 10.1039/c3cs35511j. 157. DOI: 10.3390/pharmaceutics12020157.
[176] Rehm, F. B. H.; Chen, S.; Rehm, B. H. A. Enzyme Engineering [191] Cespedes, M. V.; Fernandez, Y.; Unzueta, U.; Mendoza, R.;
for In Situ Immobilization. Molecules 2016, 21, 1370. DOI: 10. 
Seras-Franzoso, J.; Sanchez-Chardi, A.; Alamo, P.; Toledo-
3390/molecules21101370. Rubio, V.; Ferrer-Miralles, N.; Vazquez, E.; et al. Bacterial
[177] Santos, J. C. S. D.; Barbosa, O.; Ortiz, C.; Berenguer-Murcia, Mimetics of Endocrine Secretory Granules as Immobilized
A.; Rodrigues, R. C.; Fernandez-Lafuente, R. Importance of In Vivo Depots for Functional Protein Drugs. Sci. Rep. 2016,
the Support Properties for Immobilization or Purification of 6, 1–10.
Enzymes. ChemCatChem 2015, 7, 2413–2432. DOI: 10.1002/ [192] Cano-Garrido, O.; Rodrıguez-Carmona, E.; Dıez-Gil, C.;
cctc.201500310. Vazquez, E.; Elizondo, E.; Cubarsi, R.; Seras-Franzoso, J.;
[178] Mestrom, L.; Marsden, S. R.; McMillan, D. G. G.; Schoevaart, Corchero, J. L.; Rinas, U.; Ratera, I.; et al. Supramolecular
R.; Hagedoorn, P. L.; Hanefeld, U. Comparison of Enzymes Organization of Protein-Releasing Functional Amyloids Solved
Immobilised on Immobeads and Inclusion Bodies: A Case in Bacterial Inclusion Bodies. Acta. Biomater. 2013, 9,
Study of a Trehalose Transferase. ChemCatChem 2020, 12,
6134–6142. DOI: 10.1016/j.actbio.2012.11.033.
3249–3256. DOI: 10.1002/cctc.202000241. [193] Seras-Franzoso, J.; Dıez-Gil, C.; Vazquez, E.; Garcıa-Fruit os,
[179] Migneault, I.; Dartiguenave, C.; Bertrand, M. J.; Waldron,
E.; Cubarsi, R.; Ratera, I.; Veciana, J.; Villaverde, A.
K. C. Glutaraldehyde: Behavior in Aqueous Solution, Reaction
Bioadhesiveness and Efficient Mechanotransduction Stimuli
with Proteins, and Application to Enzyme Crosslinking.
Synergistically Provided by Bacterial Inclusion Bodies as
Biotechniques 2004, 37, 790–802. DOI: 10.2144/04375RV01.
Scaffolds for Tissue Engineering. Nanomedicine (Lond) 2012,
[180] Hrabarova, E.; Achbergerova, L.; Nahalka, J. Insoluble Protein
7, 79–93. DOI: 10.2217/nnm.11.83.
Applications: The Use of Bacterial Inclusion Bodies as
[194] Schetters, S. T. T.; Jong, W. S. P.; Kruijssen, L. J. W.; van den
Biocatalysts. Methods. Mol. Biol. 2015, 1258, 411–422. DOI:
Berg van Saparoea, H. B.; Engels, S.; Unger, W. W. J.;
10.1007/978-1-4939-2205-5_24.
Houben, D.; den Haan, J. M. M.; Luirink, J.; Kooyk, Y. V.
[181] Guo, X.; Liu, F.; Zhu, X.; Su, Y.; Ling, P. Expression of a
Bacterial Inclusion Bodies Function as Vehicles for Dendritic
Novel Hyaluronidase from Streptococcus Zooepidemicus in
Escherichia Coli and Its Application for the Preparation of HA Cell-Mediated T Cell Responses. jCell. Mol. Immunol 2020, 17,
Oligosaccharides. Carbohydr. Polym. 2009, 77, 254–260. DOI: 415–417. DOI: 10.1038/s41423-019-0298-x.
10.1016/j.carbpol.2008.12.036. [195] Martınez-Miguel, M.; Kyvik, A. R.; M Ernst, L.; Martınez-
[182] Kloss, R.; Karmainski, T.; J€ager, V. D.; Hahn, D.; Gr€ unberger, Moreno, A.; Cano-Garrido, O.; Garcia-Fruit os, E.; Vazquez, E.;
A.; Baumgart, M.; Krauss, U.; Jaeger, K. E.; Wiechert, W.; Ventosa, N.; Guasch, J.; Veciana, J.; et al. Stable Anchoring of
Pohl, M. Tailor-Made Catalytically Active Inclusion Bodies for Bacteria-Based Protein Nanoparticles for Surface Enhanced
Different Applications in Biocatalysis. Catal. Sci. Technol. Cell Guidance. J. Mater. Chem. B 2020, 8, 5080–5088. DOI:
2018, 8, 5816–5826. DOI: 10.1039/C8CY01891J. 10.1039/d0tb00702a.
[183] Han, H.; Zeng, W.; Zhang, G.; Zhou, J. Active Tyrosine [196] Kontoravdi, C.; Jimenez del Val, I. Computational Tools for
Phenol-Lyase Aggregates Induced by Terminally Attached Predicting and Controlling the Glycosylation of
Functional Peptides in Escherichia Coli. J. Ind. Microbiol. Biopharmaceuticals. Curr. Opin. Chem. Eng 2018, 22, 89–97.
Biotechnol. 2020, 47, 563–571. DOI: 10.1007/s10295-020- DOI: 10.1016/j.coche.2018.08.007.
02294-4. [197] Smiatek, J.; Jung, A.; Bluhmki, E. Towards a Digital
[184] J€ager, V. D.; Lamm, R.; K€ € uc€
usters, K.; Olç€ u, G.; Oldiges, M.; Bioprocess Replica: Computational Approaches in
Jaeger, K. E.; B€ uchs, J.; Krauss, U. Catalytically-Active Biopharmaceutical Development and Manufacturing. Trends.
Inclusion Bodies for Biotechnology—General Concepts, Biotechnol 2020, 38, 1141–1153. DOI: 10.1016/j.tibtech.2020.
Optimization, and Application. Appl. Microbiol. Biotechnol. 05.008.
2020, 104, 7313–7329. DOI: 10.1007/s00253-020-10760-3. [198] Keulen, D.; Geldhof, G.; Bussy, O. l.; Pabst, M.; Ottens, M.
[185] € uc€
Olç€ u, G.; Baumer, B.; K€ usters, K.; M€ollenhoff, K.; Oldiges, Recent Advances to Accelerate Purification Process
M.; Pietruszka, J.; Jaeger, K. E.; Krauss, U. Catalytically Active Development: A Review with a Focus on Vaccines. J.
Inclusion Bodies-Benchmarking and Application in Flow Chromatogr. A 2022, 1676, 463195. DOI: 10.1016/j.chroma.
Chemistry. ACS. Synth. Biol. 2022, 11, 1881–1896. DOI: 10. 2022.463195.
1021/acssynbio.2c00035. [199] von Stosch, M.; Hamelink, J.-M.; Oliveira, R. Hybrid Modeling
[186] Ma, W.; Chen, K.; Li, Y.; Hao, N.; Wang, X.; Ouyang, P. as a QbD/PAT Tool in Process Development: An Industrial E.
Advances in Cadaverine Bacterial Production and Its coli Case Study. Bioprocess. Biosyst. Eng 2016, 39, 773–784.
Applications. Engineering 2017, 3, 308–317. DOI: 10.1016/J. DOI: 10.1007/s00449-016-1557-1.
ENG.2017.03.012. [200] Geciova, J.; Bury, D.; Jelen, P. Methods for Disruption of
[187] Han, G. H.; Seong, W.; Fu, Y.; Yoon, P. K.; Kim, S. K.; Yeom, Microbial Cells for Potential Use in the Dairy Industry—a
S. J.; Lee, D. H.; Lee, S. G. Leucine Zipper-Mediated Targeting Review. Int. Dairy J 2002, 12, 541–553. DOI: 10.1016/S0958-
of Multi-Enzyme Cascade Reactions to Inclusion Bodies in 6946(02)00038-9.
PREPARATIVE BIOCHEMISTRY & BIOTECHNOLOGY 751

[201] Walther, C.; Kellner, M.; Berkemeyer, M.; Brocard, C.; [217] Yamamoto, K.; Ogasawara, H.; Ishihama, A. Involvement of
D€urauer, A. A Microscale Bacterial Cell Disruption Technique Multiple Transcription Factors for Metal-Induced Spy Gene
as First Step for Automated and Miniaturized Process Expression in Escherichia Coli. J. Biotechnol. 2008, 133,
Development. Process. Biochem. 2017, 59, 207–215. DOI: 10. 196–200. DOI: 10.1016/j.jbiotec.2007.08.002.
1016/j.procbio.2017.05.013. [218] Chen, Q. Chi; Liu, L.; Yu, T. Y.; Tang, L.; Yin, M. Li; Zhu,
[202] Starke, R.; Jehmlich, N.; Alfaro, T.; Dohnalkova, A.; Capek, P.; W. H.; Jiang, X. Yun.; Wang, H. Y. High-Level Expression and
Bell, S. L.; Hofmockel, K. S. Incomplete Cell Disruption of Purification of Melittin in Escherichia Coli Using SUMO
Resistant Microbes. Sci. Rep 2019, 9, 1–5. DOI: 10.1038/ Fusion Partner. Int. J. Pept. Res. Ther. 2021, 27, 9–15. DOI:
s41598-019-42188-9. 10.1007/s10989-020-10060-4.
[203] Haryati, T.; Widhiastuty, M. P.; Warganegara, F. M.; [219] Kim, Y. S.; Lee, H. J.; Han, M. Ho; Yoon, N. Kyung; Kim,
Akhmaloka . Response Surface Methodology for Optimization Y. Chun; Ahn, J. Effective Production of Human Growth
Membrane Disruption Using Thermolysis in Lipase Lk2 and Factors in Escherichia Coli by Fusing With Small Protein
Lk3. J. Pure Appl. Microbiol 2022, 16, 1274–1283. DOI: 10. 6HFh8. Microb. Cell Fact. 2021, 20, 16. DOI: 10.1186/s12934-
22207/JPAM.16.2.56. 020-01502-1.
[204] Berrill, A.; Biddlecombe, J.; Bracewell, D., Chapter 13: Product [220] Liu, M.; Chen, Z.; Huang, S.; Chu, S.; Issaro, N.; Tian, H.; Hu,
Quality during Manufacture and Supply. In Peptide and H.; Jiang, C. Effective Protection on Acute Liver Injury by
Protein Delivery; Walle, C. V. D., Ed.; Elsevier: Amsterdam, Halo Tag-Flanked Recombinant Fibroblast Growth Factor 7.
2011; pp. 313–339. Biotechnol. J. 2018, 13, 1700411. DOI: 10.1002/biot.201700411.
[205] Grabski, A. C. Advances in Preparation of Biological Extracts [221] Hemmati, S.; Ranjbari, J. Soluble Form Production of
for Protein Purification. Methods Enzymol 2009, 463, 285–303. Recombinant Human Insulin-Like Growth Factor-1 by NusA
DOI: 10.1016/S0076-6879(09)63018-4. Fusion Partner in E. coli. Trends. Peptide Protein Sci. 2019, 4,
[206] Stanbury, P. F.; Whitaker, A.; Hall, S. J. Chapter 10: The 1–5.
Recovery and Purification of Fermentation Products. In [222] Paal, M.; Heel, T.; Schneider, R.; Auer, B. A Novel Ecotin-
Principles of Fermentation Technology; Butterworth- Ubiquitin-Tag (ECUT) for Efficient, Soluble Peptide
Heimeman; Elsevier Science: Burlington MA, 2017; pp. Production in the Periplasm of Escherichia Coli. Microb. Cell
619–686. Fact. 2009, 8, 1–9. DOI: 10.1186/1475-2859-8-7.
[207] Peternel, S. Bacterial Cell Disruption: A Crucial Step in [223] Zheng, X.; Wu, X.; Fu, X.; Dai, D.; Wang, F. Expression and
Protein Production. N Biotechnol 2013, 30, 250–254. DOI: 10. Purification of Human Epidermal Growth Factor (HEGF)
1016/j.nbt.2011.09.005. Fused with GB1. Biotechnol. Biotechnol. Equip. 2016, 30,
[208] Stanbury, P. F.; Whitaker, A.; Hall, S. J., Chapter 3: The 813–818. DOI: 10.1080/13102818.2016.1166984.
Isolation and Improvement of Industrially Important [224] Chamachi, N.; Hartmann, A.; Ma, M. Q.; Krainer, G.; Schlierf,
Microorganisms. In Principles of Fermentation M. Chaperones Skp and SurA Dynamically Expand Unfolded
Technology;Third Edition; Butterworth-Heimeman; Elsevier Outer Membrane Protein X and Synergistically Disassemble
Science: Burlington MA, 2016; pp. 75–211. Oligomeric Aggregates. Proc. Natl. Acad. Sci. U S A 2021, 119,
[209] Hoffmann, D.; Ebrahimi, M.; Gerlach, D.; Salzig, D.; Czermak, 1–11.
P. Reassessment of Inclusion Body-Based Production as a [225] Ban, B.; Sharma, M.; Shetty, J. Optimization of Methods for
Versatile Opportunity for Difficult-to-Express Recombinant the Production and Refolding of Biologically Active Disulfide
Proteins. Crit. Rev. Biotechnol 2018, 38, 729–744. DOI: 10. Bond-Rich Antibody Fragments in Microbial Hosts. Antibodies
1080/07388551.2017.1398134. 2020, 9, 39. DOI: 10.3390/antib9030039.
[210] Dodd, C. E. R. Characteristics of Foodborne Hazard and [226] Sørensen, H. P.; Sperling-Petersen, H. U.; Mortensen, K. K.
Diseases: Microbial Stress Response. Encyclopedia Food Saf. Dialysis Strategies for Protein Refolding: Preparative
2014, 1, 183–187. Streptavidin Production. Protein Expr. Purif. 2003, 31,
[211] Salazar, O.; Asenjo, J. A. Enzymatic Lysis of Microbial Cells. 149–154. DOI: 10.1016/s1046-5928(03)00133-5.
Biotechnol. Lett. 2007, 29, 985–994. DOI: 10.1007/s10529-007- [227] Yamaguchi, S.; Yamamoto, E.; Mannen, T.; Nagamune, T.;
9345-2. Nagamune, T. Protein Refolding Using Chemical Refolding
[212] Ehgartner, D.; Sagmeister, P.; Langemann, T.; Meitz, A.; Additives. Biotechnol. J. 2013, 8, 17–31. DOI: 10.1002/biot.
Lubitz, W.; Herwig, C. A Novel Method to Recover Inclusion 201200025.
Body Protein from Recombinant E. coli Fed-Batch Processes [228] Yamaguchi, H.; Miyazaki, M. Microfluidic Chips with Multi-
Based on Phage UX174-Derived Lysis Protein E. Appl. Junctions: An Advanced Tool in Recovering Proteins from
Microbiol. Biotechnol. 2017, 101, 5603–5614. DOI: 10.1007/ Inclusion Bodies. Bioeng. Bugs 2015, 6, 1–4. DOI: 10.4161/
s00253-017-8281-x. 21655979.2014.987022.
[213] Xie, L.; Terada, A.; Hosomi, M. Disentangling the Multiple [229] Godawat, R.; Brower, K.; Jain, S.; Konstantinov, K.; Riske, F.;
Effects of a Novel High Pressure Jet Device Upon Bacterial Warikoo, V. Periodic Counter-Current Chromatography –
Cell Disruption. Chem. Eng. J. 2017, 323, 105–113. DOI: 10. Design and Operational Considerations for Integrated and
1016/j.cej.2017.04.067. Continuous Purification of Proteins. Biotechnol. J. 2012, 7,
[214] Cheong, D. E.; Choi, H. J.; Yoo, S. K.; Lee, H. D.; Kim, G. J. A 1496–1508. DOI: 10.1002/biot.201200068.
Designed Fusion Tag for Soluble Expression and Selective [230] Wang, Q.; Zhang, C.; Li, Z.; Guo, F.; Zhang, J.; Liu, Y.; Su, Z.
Separation of Extracellular Domains of Fibroblast Growth High Hydrostatic Pressure Refolding of Highly Hydrophobic
Factor Receptors. Sci. Rep. 2021, 11, 10. DOI: 10.1038/s41598- Protein: A Case Study of Recombinant Human Interferon
021-01029-4. b-1b from Inclusion Bodies. Biochem. Eng. J. 2021, 172,
[215] Zhou, L.; Liu, Z.; Xu, G.; Li, L.; Xuan, K.; Xu, Y.; Zhang, R. 108055. DOI: 10.1016/j.bej.2021.108055.
Expression of Melittin in Fusion with GST in Escherichia Coli [231] Gao, K.; Oerlemans, R.; Groves, M. R. Theory and
and Its Purification as a Pure Peptide with Good Bacteriostatic Applications of Differential Scanning Fluorimetry in Early-
Efficacy. ACS Omega 2020, 5, 9251–9258. DOI: 10.1021/acso- Stage Drug Discovery. Biophys. Rev. 2020, 12, 85–104. DOI:
mega.0c00085. 10.1007/s12551-020-00619-2.
[216] Rasooli, F.; Hashemi, A. Efficient Expression of EpEX in the [232] Demeule, B.; Gurny, R.; Arvinte, T. Detection and
Cytoplasm of Escherichia Coli Using Thioredoxin Fusion Characterization of Protein Aggregates by Fluorescence
Protein. Res. Pharm. Sci. 2019, 14, 554–565. DOI: 10.4103/ Microscopy. Int. J. Pharm. 2007, 329, 37–45. DOI: 10.1016/j.
1735-5362.272564. ijpharm.2006.08.024.
752 K. KACHHAWAHA ET AL.

[233] Fitzpatrick, A. W.; Saibil, H. R. Cryo-EM of Amyloid Fibrils [237] Tong, W.; Wang, G. How Can Native Mass Spectrometry
and Cellular Aggregates. Curr. Opin. Struct. Biol. 2019, 58, Contribute to Characterization of Biomacromolecular Higher-
34–42. DOI: 10.1016/j.sbi.2019.05.003. Order Structure and Interactions? Methods 2018, 144, 3–13.
[234] Ruggeri, F. S.; Sneideris, T.; Vendruscolo, M.; Knowles, T. P. J. DOI: 10.1016/j.ymeth.2018.04.025.
Atomic Force Microscopy for Single Molecule Characterisation [238] Hjalte, J.; Hossain, S.; Hugerth, A.; Sj€
ogren, H.; Wahlgren, M.;
of Protein Aggregation. Arch. Biochem. Biophys. 2019, 664, Larsson, P.; Lundberg, D. Aggregation Behavior of Structurally
134–148. DOI: 10.1016/j.abb.2019.02.001.
Similar Therapeutic Peptides Investigated by 1H NMR and
[235] Tiernan, H.; Byrne, B.; Kazarian, S. G. ATR-FTIR
All-Atom Molecular Dynamics Simulations. Mol. Pharm. 2022,
Spectroscopy and Spectroscopic Imaging for the Analysis of
Biopharmaceuticals. Spectrochim. Acta. A Mol. Biomol. 19, 904–917. DOI: 10.1021/acs.molpharmaceut.1c00883.
Spectrosc 2020, 241, 118636. DOI: 10.1016/j.saa.2020.118636. [239] Desligniere, E.; Ley, M.; Bourguet, M.; Ehkirch, A.;
[236] Gambin, Y.; Polinkovsky, M.; Francois, B.; Giles, N.; Bhumkar, Botzanowski, T.; Erb, S.; Hernandez-Alba, O.; Cianferani, S.
A.; Sierecki, E. Confocal Spectroscopy to Study Dimerization, Pushing the Limits of Native MS: Online SEC-Native MS for
Oligomerization and Aggregation of Proteins: A Practical Structural Biology Applications. Int. J. Mass Spectrom. 2021,
Guide. Int. J. Mol. Sci. 2016, 17, 1–20. 461, 116502. DOI: 10.1016/j.ijms.2020.116502.

You might also like