Chondroitinase Ac A Therapeutic Enzyme - Prospects and Challenge-2024

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 17

Enzyme and Microbial Technology 172 (2024) 110348

Contents lists available at ScienceDirect

Enzyme and Microbial Technology


journal homepage: www.elsevier.com/locate/enzmictec

Chondroitinase as a therapeutic enzyme: Prospects and challenges


Asma Kheirollahi a, Solmaz Sadeghi b, Shirin Orandi c, Kiana Moayedi c, Khosro Khajeh d,
Mehdi Khoobi e, f, Abolfazl Golestani c, *
a
Department of Comparative Biosciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
b
Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
c
Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
d
Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran 14115-154, Iran
e
Department of Radiopharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
f
Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran

A R T I C L E I N F O A B S T R A C T

Keywords: The chondroitinases (Chase) are bacterial lyases that specifically digest chondroitin sulfate and/or dermatan
Chondroitinase sulfate glycosaminoglycans via a β-elimination reaction and generate unsaturated disaccharides. In recent de­
Combinational therapy cades, these enzymes have attracted the attention of many researchers due to their potential applications in
Drug delivery
various aspects of medicine from the treatment of spinal cord injury to use as an analytical tool. In spite of this
Enzyme engineering
Spinal cord injury
diverse spectrum, the application of Chase is faced with several limitations and challenges such as thermal
instability and lack of a suitable delivery system. In the current review, we address potential therapeutic ap­
plications of Chase with emphasis on the challenges ahead. Then, we summarize the latest achievements to
overcome the problems by considering the studies carried out in the field of enzyme engineering, drug delivery,
and combination-based therapy.

1. Introduction produces two distinct Chase enzymes (PvCS ABC I and II), which de­
grades ChS-A, ChS-B, ChS-C and hyaluronic acid, and F. heparinum
For the first time, bacterial degradation of chondroitin sulfate (ChS) (currently known as Pedobacter heparinus) produces two Chases with
was described by Neuberg and Rubin as early as 1914 [1], and in the different substrate specificity (FlavoAC and FlavoB) (Fig. 1) [4–6].
following, it was found that cell extract of Pseudomonas fluorescens, Chase can cleave chondroitin sulfate by a β-elimination reaction, which
Pseudomonas aeruginosa, and Proteus vulgaris break down ChS into a results in the formation of unsaturated oligosaccharides. This mecha­
product having reducing power and sulfuric acid [2]. Among glycos­ nism is based on proton acceptance and donation. Initially, a double
aminoglycans (GAGs) lyase-producing bacteria, P. vulagris was the first bond is formed between C-4 and C-5 as a result of the elimination of the
well-known organism to produce chondroitinase (Chase) [3]. Yamagata C-5 proton from the uronic acid moiety. This double bond formation is
et al. purified enzymes from the cells of P. vulgaris NCTC 4636 and Fla­ the basis for the enzyme activity assay at 232 nm. After stabilization of
vobacterium heparinum ATCC 13125 [4]. It is indicated that P. vulgaris the carbanion intermediate the anomeric oxygen protonates and leads to

Abbreviations: AAV, adeno-associated virus; ADSCs, adipose-derived mesenchymal stem cells; BDNF, brain-derived neurotrophic factor; bFGF, basic fibroblast
growth factor; BMSCs, bone marrow stromal cells; Chase, chondroitinases; ChS, chondroitin sulfate; CNS, central nervous system; CNTF, ciliary neurotrophic factor;
CSPGs, chondroitin sulfate glycosaminoglycans; db-cAMP, dibutyryl-cyclic adenosine monophosphate; DRG, dorsal root ganglia; ECM, extracellular matrix; EGF,
epidermal growth factor; ENSCs, enteric neural stem cells; GAP43, growth associated protein 43; FGF2, fibroblast growth factor 2; GDNF, glial cell-derived neu­
rotrophic factor; hUSCs, human urine stem cells; hADSCs, human adipose-derived stem cells; IGF, insulin-like growth factor; KLF7, Krüppel-like Factor 7; LV,
lentiviral virus; MSCs, mesenchymal stromal cells; EP1–40, Nogo-66 antagonist peptide; NESCs, neuroepithelial stem cells; NGF, nerve growth factor; NPs, Nano­
particles; NSCs, neural stem cells; NT-3, Neurotrophin-3; OECs, olfactory ensheathing cells; oNPCs, directly reprogrammed human NPCs biased toward an oligo­
dendrogenic fate; OPCs, Oligodendrocyte progenitor cells; OV, oncolytic virus; PDGF, platelet-derived growth factor; PEG, polyethylene glycol; PLGA, Poly(lactic-co-
glycolic acid); PLL, poly L-lysine; pMNs, ESC-derived progenitor motor neurons; PNGs, peripheral nerve grafts; PPN, pre-degenerated peripheral nerve; PVD, pos­
terior vitreous detachment; RHEB, Ras homolog enriched in brain; SCI, spinal cord injury; SH3, Src homology 3; TGFβ, transforming growth factor beta.
* Correspondence to: Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, P.O. Box: 14155-6447, Tehran, Iran.
E-mail address: golsetan@tums.ac.ir (A. Golestani).

https://doi.org/10.1016/j.enzmictec.2023.110348
Received 22 May 2023; Received in revised form 28 September 2023; Accepted 19 October 2023
Available online 21 October 2023
0141-0229/© 2023 Elsevier Inc. All rights reserved.
A. Kheirollahi et al. Enzyme and Microbial Technology 172 (2024) 110348

the breakage of the glycosidic bond [7]. Thses enzymes can be classified Chondroitinase B. Chase B is produced by F. heparinum bacteria
into two distinct categories based on their degradation patterns: (Fig. 1) [5]. The Cygler laboratory determined the structure of this lyase
endo-lyases and exo-lyases. Endo-lyases initially fragment GAG chains (Fig. 2B) [18]. Based on the crystal structure, Chase B is a member of the
into larger oligosaccharides and eventually into disaccharides, following β-helix family and has different folds from FlavoAC [18]. Moreover,
a random cleavage pattern. Conversely, exo-lyases systematically Chase B requires calcium for its activity and the crystal structure un­
remove disaccharide residues from the terminal end of the GAG chain, ravels its calcium-dependent catalytic machinery [19].
without generating larger oligosaccharides in the course of their enzy­ Chondroitinase ABC. The bacterium Proteus vulgaris generates two
matic action [8]. Nowadays, P. vulgaris, F. heparinum, and Arthrobacter Chases with wide substrate specificity, named Chase ABC I (PvCS ABC I)
aurescencs (currently known as Paenarthrobacter aurescens) are used for and Chase ABC II (PvCS ABC II) [6]. The catalytic mechanisms of these
commercial production of Chase [4,5,9,10]. Recently, an extracellular enzymes are different. PvCS ABC I is an endolytic enzyme with 997
Chase ABC with the highest activity at 42 ◦ C is purified form Acineto­ residues; whereas isoform PvCS ABC II has 990 residues and shows
bacter sp.C26. In spite of various Chase-producing bacteria strains, it is of exolytic activity [6]. While the crystal structure of PvCS ABC II has not
paramount importance to identify new microbial sources for obtaining yet been determined, the PvCS ABC I structure was crystallized and
an enzyme with improved catalytic activity and structural stability [11]. characterized by the Cygler laboratory (Fig. 2C) [16,20]. PvCS ABC I is a
Chase could have various medical and/or therapeutic applications 110 kD protein which is comprised of three domains with linear fashion
through the degradation of chondroitin sulfate glycosaminoglycans arrangement [16]. The N-terminal domain of PvCS ABC I (residues
(CSPGs) which increment in their level is related to various weakening 25–234) possesses a jellyroll fold and shows the highest structural
circumstances [12–14]. Nevertheless, the Chase application in medicine flexibility (Fig. 2D) [16]. The central domain of PvCS ABCI consists of
is faced with severe challenges. The current review addresses the ther­ residues 235–617 with an incomplete (α/α)5 toroid topology (Fig. 2D)
apeutic applications of Chase and the challenges ahead in this way and [16]. Mutagenesis studies have revealed that four structurally conserved
finally summarizes the latest findings to overcome the problems amino acid residues –including His-501, Tyr-508, Arg-560, and Glu-653-
regarding enzyme engineering, drug delivery, and combination-based have a critical role in Chase ABC I catalysis [7,16,21]. The C-terminal
therapy. domain of PvCS ABCI includes 618–1021 residues and is formed of the
stacked β-sheets (Fig. 2D) [16].
2. The structure of chondroitinase
3. Production of chondroitinase
GAG lyases are not found in vertebrates [15] and are divided into
three classes by substrate specificity: heparinases, chondroitinases, and Various bacterial strains, as shown in Fig. 1, are known for their
hyaluronidases [16]. Until now, three folds ( (α/α)5-toroid, right-handed ability to produce Chase. Nevertheless, P. vulgaris, F. heparinum, and
β-helix, and β-sandwich) have been recognized in these enzymes [15]. A. aurescencs have gained prominence as commonly utilized bacterial
Among several types of Chases, Chase ABC is the most important enzyme sources in the commercial production of Chase [4,5,9,10]. Researchers
for therapeutic use and research. The following sections introduce the have optimized fermentation conditions to enhance Chase yield, by
structure of Chases, briefly. controlling pH, optimizing induction media and adjusting temperature
Chondroitinase AC. As shown in Fig. 1, Chase AC is mainly pro­ [22–24]. Besides bacterial sources, there have been reports of fungal
duced by A. aurescens and F. heparinum bacteria [5,9]. Structurally, Chase production [25]. In this context, Kasinathan et al. optimized the
FlavoAC is a two-domain molecule consisting of several α-helices in the cultural conditions for the production of chondroitinase by Aspergillus
N-terminal domain and four β-sheets in the C-terminal domain (Fig. 2A) niger and documented a maximum chondroitinase yield of 22.5 U/ml
[17]. The N-terminal domain, which includes the catalytic site and a [26]. Challenges in Chase production include downstream processing
significant portion of the substrate-binding site, has an incomplete and purification to obtain highly active and pure enzymes for thera­
double-layered (α/α)5-toroid fold [17]. peutic use.

Fig. 1. Microbial sources of chondroitin lyase [4–6,9,32,252,253].

2
A. Kheirollahi et al. Enzyme and Microbial Technology 172 (2024) 110348

Fig. 2. 3D structure of chondroitinases. Protein structures of FlavoAC (A), FlavoB (B), and PvulABC I (C) are shown in cartoon representation and colored based on
the secondary structure elements. (D) Distinct domains of chondroitinase PvulABC I are illustrated in different colors, also, some mutated residues (in N- and C-
terminal domains) and involved residues in catalysis (in catalytic domain) are labeled. These residues are colored in red and displayed in stick representation.
Visualization is done by PyMOL software (PDB ID: 1CB8, 1DBG and 1HN0). Refer to the text for the details.

For the research objectives and commercial-scale production of 28]. The production process of recombinant Chase typically involves
Chase, genetic engineering techniques have been strategically employed several key steps, as depicted in Fig. 3 for the production of Chase ABC I
to introduce Chase genes into high-efficiency expression systems, facil­ from P. vulgaris. In the majority of studies, Escherichia coli has been
itating the production of recombinant Chase on a substantial scale [27, chosen as the preferred host for the production of recombinant Chase,

3
A. Kheirollahi et al. Enzyme and Microbial Technology 172 (2024) 110348

Fig. 3. Flowchart diagram of production process of recombinant chondroitinase ABC I from P. vulgaris in E.coli [27,122].

due to its well-understood genetics and ease of manipulation [27–30]. In and genetic modifications have been examined to enhance enzyme
this context, E. coli strains DH5α and BL21 (DE3) have been commonly expression [31,35,36]. Effectively regulating fermentation parameters
employed as appropriate strains for ligation reactions and enzyme and scaling up production processes are vital for achieving elevated
expression, respectively [27,28]. To date, cloning and recombinant yields of Chase enzyme. However, challenges in maintaining enzyme
expression of Chase from P. vulgaris, F. heparinum, Bacteroides stercoris stability and preventing proteolysis during production need to be
and A. aurescencs have been documented [21,28–31]. In 2001, Pojasek addressed to ensure the production of active and therapeutic-grade
et al. reported the cloning of Chase AC and B from F. heparinum in Chase.
pET15b and pCRT7/NT vectors. To facilitate the expression of Chase AC
and Chase B, an inducible T7 promoter was utilized in both the pET15b 4. Medical applications of chondroitinase
and pCRT7/NT expression systems, alongside the incorporation of an
N-terminal His6 tag to enhance purification efficiency. Their results In spite of important biological functions of CSPGs, increased levels
indicated that Chase AC, when produced from pET15b, achieved the of these molecules are related to various weakening circumstances.
highest yield, yielding 24.4 mg of protein and a total activity of 8820 U Therefore, chondroitinase can have direct medical applications in states
per liter of culture. In contrast, Chase B expressed in the pCRT7/NT where there is an up-regulation in the amount of chondroitin sulfates,
vector produced a higher amount of protein compared to the pET15b such as spinal cord injury (SCI) [12], cancer [37], vitreous attachment
clone, with a yield of 10.4 mg of enzyme and a total activity of 880 U [38] and keloid scars [39] (Fig. 4). Among various types of chon­
[28]. In the following, Prabhakar et al. cloned Chase ABC I from droitinases, Chase ABC is the most efficient one due to its broad sub­
P. vulgaris in pET28a vector and reported the expression of the enzyme strate spectrum, and over the last decade, numerous experimental
with an N-terminal His6 tag in E.coli. They indicated that the Chase ABC studies confirmed that this enzyme could be a promising drug candidate
I purification process resulted in the production of more than 35 mg of for the treatment of various pathological conditions. In this context,
protein from a 500 ml culture [27]. researches have focused on optimizing the beneficial effects of the
The production of Chase from bacteria is affected by several key enzyme in vivo.
factors. The choice of the bacterial strain is crucial, as different strains
may have varying levels of Chase production capability [32]. Opti­
mizing culture conditions, such as temperature, pH, aeration, and 4.1. Chondroitinase and the nervous system
agitation, is essential for maximizing enzyme yield [33]. Additionally,
the composition of the growth medium plays a significant role, with Several studies have indicated that Chase ABC improves nerve
carbon and nitrogen sources, as well as trace elements, affecting Chase regeneration in the central nervous system (CNS) [12,40]. Following
production [33]. The optimized cultural conditions for bacterial cells spinal cord injuries, regeneration of the axons would be inhibited in
containing a vector carrying the gene encoding Chase have been docu­ adult mammals due to a glial scar formation at the injury site [41]. The
mented, with minor variations observed among different studies [27,28, glial scar consists of extracellular matrix (ECM) molecules and is espe­
34]. Recently, Lu et al. conducted an optimization of the culture con­ cially rich in CSPGs [41]. In vitro and in vivo studies showed that CSPGs
ditions for the expression of Chase ABC I using response surface meth­ inhibit axonal growth and regeneration [42–45]. Numerous studies have
odology. Their findings revealed that the expression level achieved was demonstrated that Chase ABC which catalyzes the digestion of CSPGs,
1.65 times higher than that observed in the shake flask setup. Addi­ can reverse the repressive activity of CSPGs in vitro [46–49]. Moreover,
tionally, they incorporated a signal peptide at the N-terminal of the in a Lamprey model of SCI, Chase ABC caused not only digestion of
Chase ABC I gene and indicated that this signal peptide facilitates the CSPGs in the spinal cord but also, led to a significant reduction in
folding of Chase ABC I and prevents its formation as inclusion bodies retrograde apoptosis by reducing the number of caspase-3 positive
[35]. To date, the use of various inducible promoters (e.g. T7 and AOX1) neurons. On the other hand, Chase ABC treatment showed a great
reduction in PTPσ mRNA expression, a feasible receptor of CSPGs.

4
A. Kheirollahi et al. Enzyme and Microbial Technology 172 (2024) 110348

significant functional recovery of the deprived eye [66].


Apart from several studies about Chase ABC’s well-known role in
spinal cord injury, its ability to create a favorable environment for
axonal regeneration has also been investigated in other scenarios. After
glaucomatous degeneration, an early feature of neuronal damage due to
Glaucoma, CSPGs restrict retinal ganglion cell regrowth by their ability
to inhibit neuronal plasticity. With the use of rat models of inducible
glaucoma, R. Tribble et al. observed that although injection of Chase
ABC couldn’t affect intraocular pressure (IOP) associated with injury, it
showed significant preservation of dendrites in injured eyes [67].
Given the beneficial effect of Chase on neural growth, it seems that
the enzyme has the potential for wider therapeutic application. In
addition to the CNS-related pathological states, using Chase has shown
improved outcomes following peripheral nerve injury [68,69]. More­
over, it has been found that CSPGs, which are present in the infarct after
ischemia-reperfusion, inhibit sympathetic regeneration axon outgrowth
in vitro [70]. Thus, Chase ABC can be used after myocardial infarction to
stimulate cardiac sympathetic nerve regeneration [70].
Also, the neuroprotective effect of Chase ABC treatment in partial 6-
hydroxydopamine (6-OHDA) lesion mouse models of Parkinson’s dis­
ease were reported by R. Fletcher et al. because of the enzyme’s ability
to preserve both substantia nigra pars compacta (SNc) cells and dopa­
minergic fibers. However, Chase ABC treatment in mice bearing full 6-
OHDA lesions failed to show such results. Also, despite the protective
Fig. 4. Potential applications of chondroitinase. function of Chase ABC treatment in partial 6-OHDA lesions, it didn’t
show any behavioral improvement in these animals [71].
Furthermore, after enzyme therapy, Akt activation, which is known to
be a mechanism to promote axonal regeneration, was increased, sug­ 4.2. Chondroitinase and cancer
gesting the possible role of Chase ABC in apoptosis inhibition through
the reduction of PTPσ expression and Akt activation [50] Lemon et al. Changes in proteoglycan levels are associated with different patho­
have reported the reducing level of CSPGs after SCI by delivery of Chase logical circumstances, including cancer [72]. Recent findings suggest
ABC in vivo [51]. A significant breakthrough came by Bradbury et al., that CSPGs have a significant role in tumor development, angiogenesis,
and they have observed a recovered locomotor function in the rat and metastasis by regulating cell-ECM, cell-cell, and cell-soluble factors
models of SCI after treating them with Chase [12]. In the following, interactions in cancer/host tissues [73–77]. Given the diverse functions
Tester et al. proved the therapeutic efficacy of Chase in higher animals of cell surface CSPGs in controlling cancer growth, migration, and in­
[52]. They demonstrated that Chase can recover the basic and skilled vasion, targeting these molecules can be considered as a therapeutic
locomotor function in hemisected cats [52]. Critically, the robustness of strategy. In this context, the efficacy of targeting cell surface PGs for
efficacy of Chase has been demonstrated in a rat model of chronic SCI inhibiting tumor growth and metastasis has been evaluated in some
[53], so potentially, numerous patients may make a profit from this studies [14,37]. In 1972, Takeuchi et al. inhibited tumor growth through
treatment. targeting cell surface CS by treating them with Chase AC in vitro and in
Although ischemic stroke induces sustained sensorimotor impair­ vivo [37]. After that, many researches demonstrated that hindering the
ments, several studies indicate that Chase may be a valuable treatment production of CSPGs with β-D-xyloside, and/or enzymatically digesting
to reduce stroke and brain injury inability [54–57]. It has been found them with Chase ABC, reduced the metastatic potency of tumor cells
that Chase ABC improves the recovery motor function of the affected [78–80]. In 2001, Denholm et al. showed that Chase suppresses invasion
limb after stroke and induces synaptic plasticity in rats [54,56,57]. and proliferation in melanoma cells [14]. Moreover, it has been
Based on these findings, Chase ABC can be considered as a novel ther­ demonstrated that enzymatic removal of tumor-cell surface CS-GAGs by
apeutic candidate in the acute and/or chronic phase of ischemic injury Chase ABC significantly reduces lung metastasis in the 4T1 murine
for aged individuals [54]. The effects of Chase ABC on the plasticity of mammary tumor model [81]. Since the nature of a CSPG varies with the
the axon, neuronal connections, cell adhesion, diffusion of the growth type of tumor [82–84], the efficacy of various kinds of Chases is related
factor, and inflammation have been investigated in different studies to the nature of the ECM surrounding the tumor. It seems that the
[58–61], even though the mechanisms underlying Chase ABC-induced relative amount of GAGs in various kinds of cells and their linkage to
improvement following stroke is not exactly defined. However, it different membrane receptors probably determines the level of CSPGs’
seems that the mechanisms of Chase ABC-induced improvement in the influence on many cellular activities. Thus, Chase possibly decreases cell
spinal cord and brain injuries may vary [62]. In addition to increasing proliferation by an alteration in binding between some ligands (e.g.
axonal regeneration in the brain, as shown in the mice model of trau­ hepatocyte growth factor, platelet factor 4, interferon-γ and so on) and
matic brain injury, Chase ABC can decrease post-traumatic brain edema their corresponding receptors [85–89]. Although Chase is considered to
[63]. inhibit tumor growth in a variety of ways, activation of caspase-3 and
Treatment of amblyopia, also called lazy eye, is another possible subsequent induction of apoptosis is expected to be one of the key action
therapeutic application of Chase. Amblyopia is decreased sight in an eye mechanisms of Chase in the treatment of cancer [14]. Despite promising
due to the loss of visual cortex plasticity [64]. To date, complete re­ results, the use of Chase in cancer treatment remains controversial due
covery of vision in adults with cortical visual impairment is therapeu­ to some conflicting results. In contrast to the mentioned studies, in a
tically infeasible. However, Pizzorusso et al. induced plasticity of the study of breast cancer in Balb/c mice, intra-tumor injection of Chase
visual cortex in adult rats by Chase ABC and attributed weak visual ABC caused higher lung metastases and the development of secondary
cortex plasticity to the presence of CSPGs [40,65]. In contrast, a study tumors [90].
the effect of Chase ABC on plasticity in the visual cortex of an adult cat Further application of Chase in cancer therapy is its usage as an
showed that the application of the enzyme was not associated with adjuvant. The efficiency of using an oncolytic virus (OV) in the

5
A. Kheirollahi et al. Enzyme and Microbial Technology 172 (2024) 110348

treatment of cancer is limited owing to the failure of the viruses to chymopapain [13]. In this context, studies in canines [102], goats [103],
efficiently diffuse and infect tumor cells in the presence of tumoral monkeys [104], and sheep [105] confirmed that Chase would be a good
CSPGs [91]. Dmitrieva et al. demonstrated that removal of the glioma candidate for nucleolysis.
ECM by using a Chase-expressing oncolytic virus enhances virus spread Since CSPGs are up-regulated in keloid matrixes [106], Chase may be
and anti-cancer effectiveness [92]. Recently, Jaime-Ramirez et al. pro­ helpful to treat keloid scars as shown in mice grafted with keloid im­
duced a humanized Chase enzyme-expressing OV and revealed that the plants and ex vivo models [39,107]. However, further investigations are
viral susceptibility of glioma cells and subsequently their sensitivity to needed for evaluating the effect of Chase alone or in combination with
temozolomide was improved [93]. The studies show that the use of other enzymes in vivo.
Chase-expressing OV in the treatment of gliomas is safer than other
adjuvants [92,93]. 5. Challenges in the medical application of chondroitinase
In addition to the therapeutic application of Chase in cancer, this
enzyme can serve as a laboratory diagnostic tool to precisely detect CS. Although the beneficial effects of Chase in treating the above-
For example, a significant increase in non-sulfated disaccharide levels in mentioned pathological conditions have been proved in experimental
the urine of patients with cancer of the head and neck has been reported and/or animal models, clinical translation remains challenging, partic­
so that this elevated level reduces after the removal of cancer [94]. Thus, ularly in the case of SCI treatment (Fig. 5). For example, the formation of
analysis of the presence of CS in urine samples by using various types of CSPGs continues over weeks after the SCI [108,109], whereas Chase
Chases may be useful in the diagnosis and follow-up of cancer. ABC quickly loses its enzymatic activity at body temperature [110]; So
Several studies have shown CSPGs’ role in the development of cancer that its activity remarkably diminished after 3 days and completely lost
since they play a significant role in metastasis and angiogenesis, there­ by 5 days at 37 ◦ C [110]. Therefore, thermal instability of currently
fore Chase ABC seems to be a promising approach in the treatment of available Chase and also diffusion of the enzyme far from the injury site,
cancerous cells [10]. Additionally, it is believed that the extracellular necessitate frequent intrathecal injection of the enzyme [111,112].
matrix’s (ECM) structural components such as proteoglycans create a However, repeated intrathecal injection is highly invasive, painful, and
barrier for the distribution of large therapeutic molecules. Having this in infection-prone, and increases the risk of physical injury [111,113].
mind, a study by Dmitrieva et al. [95] used Chase ABC to digest CS-GAGs Hence, finding a thermostable enzyme and/or a delivery procedure that
in cultured glioma cells to improve the oncolytic virus (OV) ability to the will ensure continuous catalytic activity of the enzyme at the injury site
destruction of cancerous cells. The results of this study showed that over long time intervals are big challenges of the therapeutic application
Chase ABC-mediated digestion of ECM improved OV diffusion in glioma of Chase. In this regard, the delivery methods tested in animal models,
grown in brain slices. Then, they administered OV expressing Chase ABC like local infusion pumps and intrathecal catheters, would be hard to
in intracranial xenografts and this resulted in longer survival of animals translate into the human, because of the need for highly skilled physi­
without affecting Glioma cell invasion and migration. Another study cians and enhanced the risk of infection [114]. In general, while the
demonstrated the effect of temozolomide combined with a mutant hu­ therapeutic power of Chase is promising, further experimental in­
manized version (Chase M) in order to improve the chemotherapeutic vestigations are needed to overcome the challenges facing the applica­
sensitivity of cultured glioma cells. This combinational treatment tion of the enzyme in vivo (Fig. 5) Researchers attempt to overcome
increased glioma cell death. Additionally, using Intracellular flow these obstacles through improving thermal stability of Chase, enzyme
cytometric analysis, it was revealed that this combinational treatment delivery by viral vectors, delivery within biomaterials, and generation of
resulted in a significant reduction in AKT phosphorylation, a Chase-secreting cells.
pro-survival protein, in cultured cells [93]. On the contrary, in a study of Although chronic delivery of Chase carries risks of immunogenicity,
breast cancer in Balb/c mice, intra-tumor injection of Chase ABC caused no study to date has addressed this issue. In addition to thermal insta­
higher lung metastases and the development of secondary tumors. bility, delivery and immunogenicity of chondroitinase, lack of speci­
Therefore, it can be concluded that chondroitin sulfate might act as an ficity is another potential issue facing the use of Chase ABC in SCI
inhibitor for cancerous cells spread and metastasis [90]. treatment, because it digests both inhibitory and growth-promoting
proteoglycans [115]. In this regard, delayed injection of growth fac­
4.3. Enzymatic vitrectomy, enzymatic nucleolysis and keloid scar tors after Chase ABC treatment and co-administration of Chase ABC with
cells that often express and release growth-promoting factors to ECM
In a few people, the vitreous cortex is connected to the internal may circumvent this problem. Recently, several studies have indicated
limiting lamina of the inner retina [96]. This adhesion between the that the delivery of Chase ABC in combination with other approaches is
vitreous and the retina participates in certain pathological conditions, significantly more efficient. The following sections discuss the applied
like diabetic retinopathy [96]. Vitrectomy which is a surgical technique strategies to make Chase ABC as a more impressive therapeutic agent
of removing the vitreous cortex entirely from the retina, decreases -particularly for SCI- by using protein engineering approaches, drug
macular edema and improves visual acuity in the eyes of diabetic pa­ delivery methods, and combinational therapies (Fig. 5).
tients [97]. In this context, many pharmacologic agents including en­
zymes e.g. hyaluronidase, plasmin, nattokinase, and chondroitinase 5.1. Thermal stabilization
have been investigated to induce a posterior vitreous detachment (PVD)
without harming the retina before vitrectomy [97,98]. Researchers Several approaches have been widely used to improve the structural
showed that intravitreal injection of Chase in monkeys and human stability of enzymes, such as enzyme engineering, immobilization,
donor eyes induced the separation of the vitreous from the retina [38, chemical modifications, and utilization of additives [116,117].
97]. However, some studies showed that the efficacy of Chase in vitre­ Site-directed mutagenesis is a method of protein engineering to create
oretinal separation is lower than the other enzymes. For example, Her­ specific, targeted changes in the protein that may produce a thermo­
mel and Schrage demonstrated that Chase is not able to induce PVD in stable variant [118]. Many researchers are trying to stabilize the Chase
pig eyes when compared to plasmin [99]. ABC enzyme using different strategies and approaches. So far, the Chase
Enzymatic nucleolysis is a non-surgical procedure for treating her­ ABC structure has been modified through amino acid substitutions to
niated disk of the lumbar spine that involves removing the gelatinous find out the structure-function relationship and subsequently to
cushioning material in an intervertebral disk by the injection of an generate thermostable forms of the enzyme (Table 1 and Fig. 2D). In
enzyme [100]. In 1990, Chase ABC was applied therapeutically to treat 2013, Nazari at al. showed that relieve of structural strain increases the
invertebral disc protrusion [101]. Investigations showed that using of thermal stability of the Chase ABC I so that Q140G and Q140A muta­
Chase in nucleolysis is more secure than the broadly examined tions improve both stability and catalytic efficiency of the enzyme

6
A. Kheirollahi et al. Enzyme and Microbial Technology 172 (2024) 110348

Fig. 5. Challenges in medical application of chondroitinase and applied strategies to overcome these issues.

Table 1
Impact of various mutations on activity and stability of the Chase ABC I.
Chase ABC I mutants Effects Ref

Q140A Improved both activity and thermostability of the enzyme [119]


H700N/L701T Improved thermal stability and decreased catalytic efficiency [120]
R692L/H700A/ Increased catalytic efficiency and decreased thermal stability [120]
Q787A
E138P Improved thermal stability and increased Km value with CS A as substrate [122]
E138D Total loss of the enzyme activity with CS A as substrate [229]
N806Y/Q810Y Improved both activity and thermostability of the enzyme [123]
R660A Total loss of the enzyme activity either with CS A or CS B as substrate [125]
N795A Improved both activity and thermostability of the enzyme; decreased Km value with CS A as substrate and increased Km value with CS B as substrate [125]
W818A Improved both activity and thermostability of the enzyme; decreased Km value either with CS A or CS B as substrate [125]
L679S Improved catalytic efficiency and enzyme half-life [121]
ChABC37-SH3 Improved stability and proteolytic resistance with slight decrease in catalytic efficiency [230]
NΔ5/E694P Improved enzyme half-life and specific activity [231]

[119]. By investigation of a network interaction around Asp689 at the E. tarda) Chase AC and Chase B from P. heparinus. They found that the
C-terminal domain of Chase ABC I, it was discovered that the H700N, thermostability was enhanced in two mutants, N795A and W818A,
L701T, and H700N/L701T mutations increase structural stability compared to the native protein [125].
although the catalytic efficiency of the mutants was decreased in com­ In addition to protein engineering, researchers developed other
parison with the wild type [120]. Besides, it was found that the substi­ ways, such as the utilization of stabilizing additives or co-solvents and
tution of Leu679 with Asp and Ser leads to the structural stability of deep eutectic solvents, to improve the stability of the Chase ABC. It is
Chase ABC [121]. In the following, Kheirollahi et al. examined the well known that co-solvents can improve the enzyme’s activity and
rigidifying flexible sites strategy to increase the thermal stability of the stability via alterations in the microenvironment of enzymes [126]. In
enzyme and showed that E138P mutation increases the half-life of the the case of Chase ABC I, it has been found that the enzyme stability
enzyme from 8 to 18 min [122]. Results of another study indicated that improves at physiological temperature by adding trehalose to the
the introduction of a cluster of aromatic residues on the surface of the enzyme solution [114]. In this regard, Lee et al. demonstrated that the
C-terminal domain increases the activity and stability of the enzyme enzymatic activity of trehalose-stabilized Chase ABC retains for up to 2
[123]. Despite the relative success of the above-mentioned strategies to weeks post-SCI in vivo, and CSPGs levels remain significantly reduced at
increase the thermal stability of the Chase ABC I, adding histidine pairs the SCI site for at least 6 weeks post-injury [114]. Based on our exper­
and creating His-His interactions could not increase the protein stability imental data, the Chase ABC I thermal stability increased by glycerol,
[124]. Chen et al. chose 13 residues for mutagenesis through multiple sorbitol, and trehalose, as a co-solvent, in vitro [34]. Moreover, it was
sequence alignment of Chase ABC I from P. vulgaris with the homologs found that the co-solvents decrease the rate of both Chase ABC I pro­
Chase ABC from three organisms (P. hauseri, P. mirabilisbb 2000 and teolytic and oxidative inactivation [127]. Among these co-solvents,

7
A. Kheirollahi et al. Enzyme and Microbial Technology 172 (2024) 110348

trehalose has FDA approval, thus, it can be utilized as a safe stabilizer of which is an indicator of structural plasticity was increased confirming
Chase ABC I in vivo. Furthermore, it has been shown that sucrose so­ the efficiency of this inducible vector [142]. In a further study by Car­
lution has a considerable effect on Chase ABC stability for about 2 weeks wardine et al. lentiviral vectors encoding active Chase ABC were used to
[128]. Despite the co-solvent mentioned above adding Ca (2 +), aden­ modify canine olfactory ensheathing cells (OECs) in order to develop a
osine, ATP, TNFα, Glu, Gln, Asp, and a combination of them with chase combination-based therapy that not only has the beneficial effects of a
ABC have no beneficial impact on chase ABC stabilization [129]. In cell transplant but also allows for sustained delivery of Chase ABC at the
addition to co-solvents, the effects of natural deep eutectic solvents on injured siteChase ABC[143]. Since the LV- or AAV-Chase ABC vectors
the structure and function of Chase ABCI enzyme was examined and it have a longer-lasting effect and only have to be administered once, they
was observed that deep eutectic solvents were better media for reduce risks of inflammation, trauma, and infection at the lesion site
improving the thermal stability of Chase ABCI in comparison with the [134]. Due to these advantages of the viral delivery, LV- or AAV-Chase
buffer [130]. ABC vectors may be considered as a substitute for Chase ABC in
combinational studies. However, it should be noted that the viral de­
5.2. Enzyme delivery livery of Chase might cause new problems since the effects of long-term
or persistent CSPG digestion are unknown [144].
Delivery of the Chase ABC -especially in the case of SCI treatment- is
one of the most important challenges with using it, because this protein 5.2.2. Biomaterials
is extremely sensitive and fragile. This enzyme should be used for a long The use of drug-releasing biomaterials can help to resolve the above-
period and delivered locally; however, a satisfactory system has not mentioned challenges in the therapeutic application of Chase. These
been yet introduced to accomplish this purpose [131,132]. A polymer-based biomaterials can be either injectable or implantable
well-designed and stable delivery method that could provide the enzyme [145]. The local and tunable delivery of drug-releasing biomaterials can
at a sustained rate would increase the half-life of the enzyme and sup­ prevent adverse repercussions of the systemic delivery of drugs
port it against temperature-accelerated degradation, which subse­ including a compromised immune system [145]. Biomaterials should be
quently would decrease the need of frequent usage and consecutive safe and degrade over time to non-toxic products with minimal immune
intrathecal injection [112,133]. response [145]. Hydrogels, particles/tubes, and fibers/conduits are the
most common biomaterial types used in the CNS [146–148]. The
5.2.1. Viral vectors following section describes these biomaterials used in Chase delivery to
Viral vectors with an insertion of the Chase gene can be injected the CNS.
directly into the lesion site or transfected into cells that will be trans­
planted into a CNS lesion [113,133–136]. Researchers have modified 5.2.2.1. Hydrogels. Hydrogels, three-dimensional hydrophilic poly­
the Chase ABC gene of bacterial origin in order to ablate cryptic meric networks, are interesting and trendy bio-compatible composites
N-glycosylation sites and produce active enzymes by mammalian cells for tissue engineering, diagnostics, immobilization, and drug delivery
[134,137,138]. Lentiviral (LV) and adeno-associated virus (AAV) vec­ due to their porosity and compatibility with aqueous environments. The
tors have been widely employed to carry the Chase ABC gene into the porosity of these composites can be adjusted by controlling the density
CNS [134,137]. Results obtained from the viral delivery of Chase ABC of the cross-linked matrix and this provides a lot of advantages for
showed that injection of LV-Chase ABC and AAV-Chase ABC into adult loaded molecules such as sustained drug delivery [149]. Having this in
rat cerebral cortex led to the secretion of active Chase ABC locally as mind, Rossi et al. applied an agarose-carbomer (AC1) hydrogel as a
well as from long-distance axons. Furthermore, it has been observed that delivery system for Chase ABC and examined the digestion of decorin, a
the activity persisted for more than one month [134,137]. Moreover, scar-like molecule, to investigate entrapped enzyme activity. Using this
direct injection of LV-Chase ABC nearby a spinal cord lesion has been delivery system showed enzyme activity for up to 7 days through the
shown to prevent axonal dieback in corticospinal axons and induce determination of decorin-digested products. Also, this hydrogel didn’t
axonal sprouting in tested animals [134]. In 2015, James et al. demon­ induce any stable interaction with the enzyme and enzyme denaturation
strated that Chase ABC gene therapy –using a lentiviral transfer vector- [150]. Despite these data, designing another delivery system based on
could considerably augment the function of the upper limb after cervical methylcellulose (MC) hydrogel for affinity-based release of Chase ABC
contusion injury [139]. To evade T cell recognition, R. Burnside et al. by M. Pakulska et al. [132]. showed better results. Indeed, they modified
designed a novel strategy using a dual lentiviral immune-evasive vector the hydrogel by Src homology domain 3 (SH3) binding peptide and also
system for Chase ABC expression based on controlling by a an SH3 protein to express with Chase ABC protein to investigate whether
doxycycline-inducible regulatory switch and this dual vector was or not this strategy can control the enzyme release. The results of this
injected into a rat model of contusion SCI [140]. Using this regulable study showed that this affinity-based MC-hydrogel resulted in sustained
system, it was reported that following the sustained release of release of active Chase ABC for up to 7 weeks in vitro. Additionally, this
dox-i-Chase ABC both the horizontal ladder walking task and projection delivery system allows for a controlled release pattern by adjusting the
of dorsal column neurotransmission were improved. However, although SH3 peptide to SH3 protein ratio, for example. Similarly, in another
short-term delivery of the enzyme didn’t suffice to improve motor study, a degradable MC hydrogel was synthesized based on an inverse
function, an investigation of the density of vGlut1 + innervation in the electron-demand Diels− Alder (IEDDA) reaction, and its potential for
injured spinal cord showed that its long-term delivery improved skilled controlled release of Chase ABC was investigated. It was reported that
hand function due to its ability of long-term CS-GAG digestion [140]. Chase ABC showed a release pattern for up to 4 days. Additionally, the
Generally, perineuronal nets (PNNs) are an aggregation form of ECM, cytocompatibility of these hydrogels was confirmed by assessing the
enriched with chondroitin sulfate proteoglycans, hyaluronan, and survival and proliferation of neuronal progenitor cells, suggesting a new
tenascin-R, in the brain [141]. Several studies have suggested that the strategy for the co-delivery of therapeutic molecules to modulate the
postnatal maturation of PNNs plays a significant role in neuronal plas­ microenvironment of the injured site effectively [151].
ticity and neurological disorders [141]. In light of these findings, Cars­ Interestingly, H. Hettiaratchi et al. designed a two-pronged approach
tens KE et al. injected a Cre-dependant AAV vector encoding Chase ABC for improving Chase ABC stability, as such, they applied site-directed
into the hippocampus of mice to selectively target tamoxifen-inducible mutagenesis and PEGylation, which is known to inhibit protein dena­
Cre-expressing CA2 neurons. After a 2-week period of daily treatment turation and aggregation, in order to improve enzyme activity and sta­
with tamoxifen, PNN loss was observed in the targeted region mean­ bility. They reported that N1000G mutation showed better activity and
while PNNs showed a normal expression in mice receiving the vehicle. half-life and additionally, PEGylation of this mutant resulted in further
In addition, using this controlled delivery system, mossy fiber sprouting

8
A. Kheirollahi et al. Enzyme and Microbial Technology 172 (2024) 110348

improvement of enzyme activity, about 10-fold when compared to un­ combination in injured animals led to improved axonal regeneration by
modified proteins. Moreover, they also described a methylcellulose the increase of GAP-43 positive neurons. This result was also associated
hydrogel as a vehicle for sustained delivery of this modified enzyme in with the reduction of glial scar thickness and improved BBB score,
rat models of stroke and they found a significant decrease in CSPG levels suggesting a higher functional recovery in rats [158].
up to 3 weeks post-injury [152].
Also recently, a study by Raspa et al. [153] used a self-assembling 5.2.2.5. Chitosan. Chitosan, the second largest biopolymer after cellu­
peptide hydrogel to investigate its potential for immobilization and lose, is a natural polysaccharide produced from chitin by a deacetylation
sustained release of Chase ABC. Using this delivery system showed a reaction. In recent years, due to some unique properties such as being
sustained release pattern of the enzyme for up to 42 days. Additionally, biodegradable and biocompatible, chitosan and its derivatives have
hydrogels containing the enzyme were injected in chronic SCI lesions attracted a lot of attention for biomedical applications [159]. Addi­
and this resulted in greater BBB (The Basso, Beattie, and Bresnahan) tionally, some studies have shown that owing to having almost similar
scores and improved hindlimb locomotor recovery in injured rats. composition and structure to GAGs, it shows a minimal immune
Overall, despite showing effective function in several studies, there’s a response after implantation into the human body, a property that has
demand for more studies to confirm hydrogel’s effectiveness in the proven its non-toxicity [160]. Chitosan has also attracted significant
repair and regeneration of soft tissues. attention for enzyme immobilization because of having amino and hy­
droxyl groups as its functional groups that improve the interaction be­
5.2.2.2. Collagen. Collagen, the most abundant protein in mammals, is tween enzymes and support, a situation that allows for simple
an attractive polymer due to its low immunogenicity, biocompatibility, immobilization techniques namely adsorption [161]. A study conducted
and high degradability. In fact, the hydrophilic nature of this polymer by Ni et al. investigated the effect of sustained delivery of Chase ABC
allows for its high dissolution and release of loaded drugs. Additionally, using polypropylene carbonate- chitosan (PPC-CS) micron fibers when
as this polymer plays a significant role in ECM, several studies have implanted into hemisected spinal cord injury models. The results of this
investigated its function in the repair of the central and peripheral study revealed that the enzyme showed a ten-day period release profile
nervous systems as well [154]. Therefore, due to its beneficial charac­ from micron fibers in PBS at 37 ◦ C and 5% CO2. Additionally, the
teristics, a study conducted by Betül Ahi et al. designed a multilayer enzyme released from this system resulted in improved functional re­
scaffold of collagen/PLGA/Laminin for sustained release of NT-3 and covery, axonal regeneration, and reduced scar thickness [162]. Another
Chase ABC and investigated its effect on adhesion, proliferation, and study by L. Francis et al. assessed the effect of Chitosan-Alginate (C-A)
axonal outgrowth in human adipose tissue-derived stem cells (ACSs) and scaffolds containing chitosan and alginate microspheres for controlled
dorsal root ganglia (DRG) cultures. The results of this study showed that delivery of Chase ABC and neurotrophin-3 (NT-3). The in vitro release
although the collagen and PLGA layers of this system were able to pattern of these molecules from C-A scaffolds showed an 8-week and a
improve cell adhesion, proliferation, and outgrowth, in the meantime, 7-week release profile for NT-3 and Chase ABC, respectively. Also,
loaded molecules were able to improve their effectiveness [155]. To sum molecules released from this delivery system retained their bioactivity,
up, although countless studies about collagen efficacy for tissue regen­ and culturing DRG neurons in the presence of C-A scaffold containing
eration are available, its potential for delivering active Chase ABC in SCI NT-3 and Chase ABC improved neurite outgrowth through a growth
lesions is still a challenge. inhibitory barrier of CSPGs [163]. However, in vivo, studies need to be
conducted in order to evaluate the ability of these systems for nerve
5.2.2.3. Silk fibroin films. Silk fibrin (SF), a natural fibrous protein, that regeneration.
can be extracted from silkworms and spiders is a relatively new strategy
in sustained drug delivery systems for a variety of bioactive molecules 5.2.2.6. PLGA. Polylactic-co-glycolic acid (PLGA) is an FDA-approved
such as drugs, genes, and so on. Its popularity is due to its slow rate of nanoparticle renowned for biocompatibility, low toxicity, and low-cost
degradation, structural stiffness, and biocompatibility. In addition, and controllable drug delivery features [164]. Due to these advan­
multiple studies have reported its ability to repair nervous tissues [156]. tages, PLGA is an outstanding polymer-based nanoparticle that has
Meanwhile, a study by N. Sivak et al. investigated the potential of a silk sparked a lot of attention for its application in various diseases. PLGA
fibroin-based conduit in the delivery of Chase ABC and glial cell synthesized thorough interactions between glycolic acid and lactic acid
line-derived neurotrophic factor (GDNF) in a rat model of sciatic nerve monomers that link together via ester bonds and eventually generate a
gap injury and animals were followed for 6 weeks. The results of this negative-charge aliphatic polymer. These nanoparticles integrate into
study demonstrated that rats receiving conduits releasing both Chase cells through clathrin-mediated endocytosis or pinocytosis and ulti­
ABC and GDNF had higher Schwann cell migration to the injured site. mately enter the cytoplasm. Since PLGA degrades into carbon dioxide
Additionally, axonal regeneration was seen in these rats however there and water via the Krebs cycle it has non-detrimental effects on cells
wasn’t almost a significant difference between this group and those who [165,166]. In this regard, Azizi et al. have loaded Chase ABC I on PLGA
were implanted with Chase ABC or GDNF alone [69]. Therefore, more nanoparticles (NPs) and subsequently administered it into SCI-model
studies should be conducted to introduce convincing evidence about the rats. According to this study, the BBB score and CSPG degradation
efficacy of these materials in axonal regeneration. concentration in the immobilized-enzyme-treated group were signifi­
cantly higher compared with the control groups. Additionally, the MTT
5.2.2.4. Poly (propylene carbonate). Poly (propylene carbonate) (PPC), assay on OECs confirmed that PLGA NPs are non-toxic [167].
a linear aliphatic polymer, can be synthesized from carbon dioxide
(CO2) and propylene. In addition to ease of fabrication, being biode­ 5.2.3. Inorganic nano-materials
gradable and biocompatible, the degradation products of PPC, water, Inorganic nano-materials –comprised of noble metals, metal-oxides,
and CO2, are non-toxic thus making it more attractive for being applied silica-based nanoparticles, and quantum dots have significant physico­
in a broad range of applications namely drug delivery [157]. Recently, a chemical, electromagnetic, and optical features that make them appro­
study by Xia et al. designed a PPC microfiber for sustained delivery of priate for medical imaging and therapeutic applications. However,
dibutyryl cyclic adenosine monophosphate (db-cAMP) and Chase ABC despite their favorable characteristics, their usage is restricted due to
and investigated not only the in vitro release pattern but also the their toxicity and low solubility [168,169].
implementation of these microfibers in rat models of SCI. The results of
this study showed a stable release profile for db-cAMP and Chase ABC 5.2.3.1. Noble metals. Since noble metallic nanoparticles, like gold
over a 204-h and 252-h period, respectively. Additionally, applying this (Au), silver, and platinum. possess remarkable properties, they have

9
A. Kheirollahi et al. Enzyme and Microbial Technology 172 (2024) 110348

Table 2
Combinatorial strategies focused on chondroitinase.
Combination therapies with Delivery of Chase Models Comments Ref.
chondroitinase

Schwann cells and Osmotic pump In vivo/rat Significant improvements in BBB score. [182]
OECs Increased numbers of both myelinated axons and serotonergic fibers.
OECs OECs transduced In vivo/rat The Chase-generating canine OECs persisted at 4 weeks following [232]
with the Chase ABC-LV vector transplantation.
Increased the number of corticospinal axons caudal.
OECs OECs transduced In vivo/rat A partial return of voluntary movement function. [233]
with the Chase ABC-LV vector Forepaw reaching recovery improvement.
ENSCs LV vector transduction In vivo/rat Injured spinal cords’ cavity area reduction. [234]
The amount of cells projecting axons via the damage site increase.
PPN, BMSCs Direct injection In vivo/rat Considerable axonal regeneration. [235]
Locomotor performance improvement.
oNPCs, methylcellulose biomaterial Direct injection In vitro and in Differentiation of grafted cells to oligodendrocytes increased. [236]
vivo/rat Remyelination of the spared axons increased.
Functional synapse enhancement.
Motor functional recovery improvement.
NESCs, methylcellulose hydrogel Direct injection In vivo/rat Lesion volume reduction. [237]
Chase ABC provides a condition to maintain a greater number of
neurons from transplanted NESCs.
No improvement in functional repair.
hUSCs Direct injection In vivo/rat Functional recovery enhancement. [238]
Increase in NGF and BDNF expression.
ADSCs LV vector transduction In vitro Chase ABC express constantly from ADSCs. [239]
Chase ABC expression rise the chance of ADSCs migration.
AAV-L1 Direct injection In vivo/mice Voluntary movements and functional recovery improvement. [240]
Synaptic plasticity increase.
insulin Direct injection In vivo/rat Neuronal growth and regeneration. [241]
Locomotor and sensor function promotion.
hADSCs Direct injection In vivo/rat Locomotor function improvement. [242]
Lesion area cavity size reduction.
DRG microtransplants, neurotrophin- Direct injection In vivo/rat Axon regeneration [243]
3-expressing lentivirus
PNG Direct injection In vivo/ Adult Axonal regrowth [244]
female cats Synapse formation
OMPCs [245]
MSCs, BDNF Direct injection In vivo/dogs TNF-α, IL-6, COX-2 reduction, BDNF, [246]
β3-tubulin neurofilament augmentation,
BBB score improvement
Lithium Chloride Direct injection In vivo/rat Fortifies Chase ABC function via a Bcl-2-dependent mechanism [247]
NSCs Osmotic pump In vivo/mice Diminished chronic-injury scar and increased iPSC-NSC [206]
differentiation and survival.
pMNs, NT-3, PDGF and NEP1–40 Lipid microtubes embedded in In vivo/rat The combination of pMNs with Chase and NEP1–4- resulted in [248]
fibrin scaffold decreased cell survival and enhanced macrophage infiltration.
PNGs and GDNF Direct injection In vivo/rat Functional axonal regeneration by chronically injured neurons [184]
PNGs and Rheb In vivo/rat Increased the construction of synapses in the spinal cord and [249]
improvement of forelimb function.
Human adipose-derived stem cells Direct injection In vivo/rat Improvement in functional recovery. [205]
NSCs or NT-3 overexpressing NSCs Osmotic pump In vivo/rat Higher neuronal and oligodendroglial [193]
differentiation of NT-3 NSCs.
Enhancement in transplanted cells migration, promoted axonal
remodeling and as well as increased locomotor recovery.
EGF, FGF2, PDGF-AA and treadmill Direct injection In vivo/rat Increased chronic neuroanatomical plasticity with no additional long- [223]
training term improvement in locomotor function.
NGF Alginate In vivo/rat Improved locomotor recovery. [217]
microspheres embedded in
polydioxanone electrospun
filaments
NT-3 Lipid microtubes embedded in In vivo/rat Improvement in locomotor function. [217]
agarose gel
db-cAMP Poly(propylene carbonate) (PPC) In vivo/rat More axonal plasticity, locomotor recovery, and decreased glial scar [114]
electrospun microfibres formation.
KLF7 Viral (LV) transduction In vivo/rat Reduced the net retraction of axons from sites of spinal injury. [158]
Poly(glycerol sebacate) Direct injection In vivo/rat Augmented nerve regeneration and [250]
functional recovery.
Low-level laser (660 nm) Direct injection In vivo/rat Decreased cavity size, increment of myelination and axonal [251]
regeneration.
α-Nogo-A antibody and rehabilitation Direct injection In vivo/rat Increased in sprouting and axon regeneration and enhanced [189]
functional recovery.
Zymosan Direct injection In vitro and in Robust and functional regeneration of sensory axons. [187]
vivo/rat
Oncomodulin and cAMP Direct injection In vitro and in Limited regeneration to injured sensory nerves. [218]
vivo/rat
Clenbuterol Direct injection In vivo/rat Axonal regeneration around the injury site and improved locomotor [220]
function.
(continued on next page)

10
A. Kheirollahi et al. Enzyme and Microbial Technology 172 (2024) 110348

Table 2 (continued )
Combination therapies with Delivery of Chase Models Comments Ref.
chondroitinase

NT3 and NR2D Direct injection In vivo/rat Improved axonal sprouting and behavioral recovery. [219]
Reestablished electrical conductivity across the lesion.
Liposomal clodronate and Rolipram Direct injection In vivo/rat Reduced axonal injury and enhanced neuroprotection, plasticity, and [186]
hindlimb functional recovery.

been widely used in various research. Among them, gold nanoparticles photoluminescence qualities [176]. These features allow them to be an
have gained a lot of attention. They are, stable, non-toxic, have simple efficient platform for drug delivery or imaging [177].
manufacturing procedures, and exhibit proper optical behavior. Gold In this aspect, green pSi nanoparticles were chosen to entrap Chase
nanoparticles have shown to be a promising choice for delivering ABC I through electrostatic interaction. The Km value was roughly the
diverse biomolecules and drugs to the target location [170,171]. same; nevertheless, the pSi had an adverse effect on the specific activity
In this context, Naderi et al. decided to investigate the impact of and Vmax. These data suggest that the enzyme active sites may engage
different concentrations of Au nanorods on the activity and stability of during the immobilization processes that also suppress the Chase ABC I
Chase ABC I in comparison with the free enzyme. Analysis of circular production rate. However, pSi enhanced enzyme thermal stability at
dichroism Spectro polarimetry and Fluorescence data showed that the − 20, 4, 25, and 37 ◦ C [176].
rigidity of the enzyme raises with a lower concentration of Au nanorods In a separate investigation, immobilized Chase ABC I on red pSi ki­
while it declines at higher concentrations. This gives rise to the theory netic parameters yield different results. The study has shown that the
that gold nanorods make more chromophores like Tyr and Trp residues Km rate for the immobilized enzyme was relatively higher than the free
available on the surface and cause the instability of the enzyme. In one. Additionally, the activity of the immobilized enzyme on red pSi was
addition, this study showed that the presence of gold nanorods can reduced more than the immobilized enzyme on green pSi. The results
improve both the substrate affinity and storage stability of the enzyme; also revealed that the red pSi enhanced the Chase ABC I durability at
however, it has a negative impact on the enzyme activity that may be a diverse temperatures. For instance, 70% of the initial activity remained
consequence of gold nanorod inhibition [172]. after 50 min at 37ºC for the immobilized Chase ABC I on red pSi
meanwhile the enzyme activity drops to 60% and 15% for the green pSi
5.2.3.2. Metal oxide. Metal oxide NPs are multifunctional, stable, less and free respectively. The MTT test found no evidence of cytotoxicity for
toxic, and biocompatible drug delivery systems that have widespread red pSi [178].
use in medical research. The most common metal oxide nanoparticles In consistence with these data, Rezaei et al. carry out research on the
such as Fe3O4, ZnO, CuO, and TiO2 gain a fair amount of attention due effect of immobilized Chase ABC I on red pSi (Chase ABC@PSi) in an
to their magnetic and anti-bacterial effects [173]. animal model of MS. Two weeks after the administration of the drug
In an investigation, Fe3O4 (magnetic) NPs were used as a carrier for delivery system they found that CSPGs concertation reduced in the
Chase ABC I. The enzyme-linked via ionic, hydrogen, and van der Waals treatment group in comparison with vehicle and PSi treated groups.
bonds to the magnetic NPs and their physical connection has been Chase ABC@PSi also increased the number of oligodendrocyte cells in
confirmed with FTIR. The results of the kinetic parameters revealed that this study, which helped to promote myelin production. Furthermore,
the enzyme affinity to the substrate increased while the specific activity the reactivity of astrocytes decreased in the treatment group [179].
decreased in the immobilized enzyme.
In another study dextran-coated Fe3O4 was used to evaluate the
5.3. Combination-based therapy
Chase ABC I features. The reduced immobilized enzyme’s Vmax and the
increased Km indicate the favorable steric orientation of enzyme active
Due to the complex nature of SCI pathology, a single-treatment
sites by means of dextran. The nanoparticle also enhanced the stability
strategy seems to be insufficient for complete functional recovery. For
of Chase ABC I by 2.5-fold; although, it was inimical to the enzyme
this purpose, it may be necessary to apply Chase ABC along with other
activity. Furthermore, the release pattern of the enzyme reports
effective therapeutic approaches to agitate neuronal regeneration. In
demonstrated that 70% of the enzyme was released initially in 9 h
recent years, different methods such as immunomodulation [180],
[174].
transplantation of cell and peripheral nerve [181–184], neurotrophic
Additionally, Gao et al. had been seeking a proper approach to
factors administration [135,185,186], anti-myelin-based inhibitors an­
induce the migration of the Schwann cells to the region of the SCI-
tibodies (anti-Nogo A) [187], rehabilitation [188], laser therapy and use
induced glial scars. In this scenario, Schwann cells were equipped
of hyperbaric oxygen (HBO) have been combined with delivery of Chase
with immobilized Chase ABC I gene on polyethyleneimine super­
ABC and revealed more desirable effects [189,190](Table 2).
paramagnetic iron oxide nanoparticles (Chase ABC I/PEISPIONs). The
In addition to increasing axon regeneration, Chase improves migra­
Chase ABC I degrades CSPGs to make a pathway through the damaged
tion of the transplanted cells and their integration into the brain and/or
area. Thereupon, Schwann cells and the resident astrocytes can fuse
CSI lesion through degrading the inhibitory action of CSPGs [191–193].
together that potentially improve axonal regeneration. Noteworthy that
For example, when the axonal growth inhibitory factors were eliminated
SPIONs magnetic characteristics were used to control the direction of
by Chase ABC, dopaminergic graft integration in Parkinsonian mice was
Schwann cells toward the desirable region under a magnetic field and
remarkably improved [194]. Furthermore, it has also shown that Chase
also preserve the Chase ABC I gene against nuclease degradation.
ABC facilitate the adhesion of chondrocyte and synovial
Moreover, they were covered by polyethyleneimine to provide a cationic
membrane-derived mesenchymal stem cells to the cartilage disc surface
surface that is more conducive to the carrier’s cell uptake. Since the
and partial-thickness chondral defects, respectively [183,195–197]. A
toxicity results point out the safety of the nano-system below 4 µg/ml,
range of cells including neural stem cells [198,199] and glial cells -such
the Chase ABC I/PEISPIONs are regarded as an auspicious candidate for
as OECs [200,201], Schwann cells [202,203], and oligodendrocyte
SCI treatment [175].
precursor cells [204]- have been investigated for transplantation in the
site of SCI. Recently, the combinatorial effects of Chase and several cell
5.2.3.3. Silica-based Nanoparticles. Porous silicon (pSi) NPs seem
types were examined in SCI models (Table 2) [182,205–207]. In a study,
appealing to scientists owing to their desirable attributes including
the implantation of OECs or Schwann cells into the site of the lesion was
biodegradability, biostability, non-toxicity, high loading capacity, and
investigated in combination with the prolonged Chase ABC

11
A. Kheirollahi et al. Enzyme and Microbial Technology 172 (2024) 110348

administration for four weeks. By employing this combination therapy weeks. Subsequently, the study showed functional recovery, axonal
graft integration was enhanced and regeneration of axons was improved projections, and myelination improvement in the treatment group. It
across the lesion [208]. In 2015, Lee et al. evaluated the efficacy of the also demonstrated the decrease in cavity size in the damaged lesion.
simultaneous administration of mesenchymal stromal cells and Chase Furthermore, aquaporin 4 (AQP4) expression was reduced which in­
ABC on chronic SCI and showed that this combination is more effective dicates a reduction in edema. Glycogen synthase kinase-3β (GSk3β), a
in improving clinical signs and neural regeneration [209]. The trans­ demyelination factor, was also decreased [189]. It has also been
planted cells can increase axonal regeneration and promote injured revealed that the combination of LLL and Chase ABC alleviates allody­
tissue repair by refilling formed cavities in the site of lesions and nia, thermal hyperalgesia, and pain-related cytokines such as IL-6 and
expression of growth factors such as neurotrophins [210,211]. So far, BDNF; meanwhile, increased analgesic factors like the glutamic acid
numerous studies have shown the beneficial effect of various growth decarboxylase 65-kilodalton isoform (Gad65) and GDNF [228].
factors such as NT-3, GDNF, IGF, bFGF, TGF-β, BDNF, PDGF, and CNTF The combination of HBO and Chase ABC also appeared to be
on spinal cord injury [212–216]. The combination of Chase ABC and contributive in SCI treatment. Following injury significant amount of
neurotrophins omit the inhibitory effect of CSPGs and boost axonal free radicals are generated and lead to lipid peroxidation which even­
growth in the new environment with excessive freedom [207]. Hence, tually cause neuronal damage. It has also been revealed that the GSK3β
Chase ABC was combined with various growth factors like NT-3 and and AQP4 expression increase during SCI which are in charge of CSPG
NGF and showed significantly improved functional recovery [114,217]. production and edema respectively. HBO treatment reduces free radical
Furthermore, the triple-combination of neural precursor cells, Chase production and turn the GSK3β and AQP4 expression back to a normal
ABC, and a mixture of PDGF, EGF, and FGF in a clip-compression SCI amount. This study showed that HBO plus Chase ABC treatment would
model successfully promoted the neural precursor cells’ integration and be effective in neuronal functional recovery [190].
differentiation, as well as their migration. Also, the level of GAP43 was
elevated and the plasticity of the corticospinal tract and serotonergic 6. Conclusion
axons was enhanced, which finally led to improved functional recovery
[191]. In a similar study Hwang et al. combined the poly (ε-capro­ According to the observed results in vitro and preclinical studies,
lactone) scaffolds seeded with NT-3 overexpressing neural precursor Chase is a promising enzyme in treating various pathological conditions.
cells and Chase ABC in a rat hemisection model and demonstrated that For clinical applications, achieving a biochemically stable enzyme is
cell migration, axonal remodeling, and locomotor recovery increase in important because of its thermal instability. In this context, several
the combination group [193]. studies have shown that the stability of Chase is increased in vitro by
In some studies, the effect of neuroprotective agents such as zymosan using different approaches; however, it should be investigated in vivo. In
[218], clenbuterol (a β2-adrenoceptor agonist) [219] and oncomodulin addition, the need for invasive pumps or direct injections can be
(Ca2+-binding protein) [220] in combination with Chase ABC is evalu­ removed by delivery of the enzyme through natural, synthetic, and viral
ated and their beneficial impacts are proved. In addition, it has been drug delivery systems. Therefore, it seems necessary to focus on the
shown that rehabilitation along with using Chase ABC, in both delayed development of an appropriate delivery system for Chase with the least
and acute stages in the SCI model is more effective [188,221,222]. Be­ side effects. Generally, numerous studies in the fields of enzyme engi­
sides, specific rehabilitation strategies probably help to the formation of neering and delivery have been conducted to overcome the Chase
appropriate connections and adjust the increased plasticity caused by problems in clinical translation. Furthermore, combination therapies
Chase treatment in the CNS [221]. With an eye toward whether regular with Chase have also shown promising results, but limited research has
treadmill exercise has a synergistic impact when combined with Chase been carried out on Chase drug delivery systems coupled with other
ABC and growth factors like EGF, FGF2, and PDGF-AA. Alluin and his inspiring therapies.
colleagues conducted a study on SCI rat models. This study showed that
the mentioned combinatorial strategy induced considerable structural Funding acknowledgments
plasticity improvement, even so, had no further long-lasting benefits on
the locomotor metrics such as the hindlimb-forelimb connection after This research did not receive any specific grant from funding
the seven-week recuperation period [223]. In 2013, the impact of agencies in the public, commercial, or not-for-profit sectors.
combining rehabilitation, α-Nogo-A antibody, and Chase ABC was
examined in a rat cervical partial SCI model [187]. The N-terminal re­ CRediT authorship contribution statement
gion of Nogo-A, called Amino-Nogo-A, is a myelin-associated molecule
and one of the most potent inhibitors of regeneration [224–226]. The Asma Kheirollahi: Investigation, Writing – original draft prepara­
results showed that the combination treatment is more effective; so the tion, Visualization; Solmaz Sadeghi: Investigation, Writing – original
population receiving combination treatment displayed a great recovery draft preparation, Visualization; Shirin Orandi: Investigation, Writing –
of 80% [187]. One more approach that has been assessed to gain a original draft preparation; Kiana Moayedi: Investigation, Writing –
beneficial effect of combination therapy was to use GDNF, Chase ABC, original draft preparation; Khosro Khajeh: Writing – review & editing;
Nogo-A antibody, and PLGA together. GDNF has been utilized to support Mehdi Khoobi: Writing – review & editing; Abolfazl Golestani:
neural recovery following SCI since it is a crucial neurotrophic factor. Conceptualization, Supervision, Writing – review & editing.
However, its prompt degradation gave rise to using it with PLGA. It
comes to be more engaging when the research group decides to look into Data Availability
their synergistic impact with Nogo-A antibody, a corticospinal tract
axonal sprouting accelerative, and Chase ABC. This study showed No data was used for the research described in the article.
PLGA-GDNF, as opposed to GDNF administered directly, clearly
enhanced functional recovery in SCI rats. The combination of Acknowledgments
PLGA-GDNF, PLGA-Chase ABC, and PLGA-Nogo worked best to promote
the functional recovery of SCI rats, and the rats in the PLGA-Chase ABC We thank Tehran University of Medical Sciences, Tarbiat Modares
and PLGA-Nogo groups also had enhanced motor function [227]. University and the University of Tehran for the support of this
Recently, the combination of Chase ABC and laser therapy was manuscript.
evaluated. The low-level laser (LLL) 660 nm was used with Chase ABC to
investigate their treatment potential together. SCI model rats received
LLL and intraspinal injection of 1 μg Chase ABC treatment for two

12
A. Kheirollahi et al. Enzyme and Microbial Technology 172 (2024) 110348

Conflict of interest [27] V. Prabhakar, I. Capila, C.J. Bosques, K. Pojasek, R. Sasisekharan, Chondroitinase
ABC I from Proteus vulgaris: cloning, recombinant expression and active site
identification, Biochem. J. 386 (1) (2005) 103–112.
The authors declare that they have no conflict of interest. [28] K. Pojasek, Z. Shriver, P. Kiley, G. Venkataraman, R. Sasisekharan, Recombinant
expression, purification, and kinetic characterization of chondroitinase AC and
References chondroitinase B from Flavobacterium heparinum, Biochem. Biophys. Res.
Commun. 286 (2) (2001) 343–351.
[29] A.L. Tkalec, D. Fink, Fo Blain, G. Zhang-Sun, M. Laliberte, D.C. Bennett, K. Gu, J.
[1] C. Neuberg, O. Rubin, Über die Bildung von Thioschwefelsäure aus J. Zimmermann, H. Su, Isolation and expression in Escherichia coli of cslA and
Ätherschwefelsäuren und Sulfonsäuren, Biochem Z. 67 (1914) 82–89. cslB, genes coding for the chondroitin sulfate-degrading enzymes chondroitinase
[2] C. Neuberg, H. Hofmann, Enzymatischer Abbau von Chondroitinschwefelsäure, AC and chondroitinase B, respectively, from Flavobacterium heparinum, Appl.
Biochem Z. 234 (1931) 345–346. Environ. Microbiol. 66 (1) (2000) 29–35.
[3] K.S. Dodgson, A.G. Lloyd, Studies on sulphatases. 18. Preparation of [30] A. Williams, W. He, B.F. Cress, X. Liu, J. Alexandria, H. Yoshizawa, K. Nishimura,
chondroitinase-free chondrosulphatase from extracts of Proteus vulgaris, T. Toida, M. Koffas, R.J. Linhardt, Cloning and expression of recombinant
Biochem. J. 66 (3) (1957) 532. chondroitinase ACII and its comparison to the Arthrobacter aurescens enzyme,
[4] T. Yamagata, H. Saito, O. Habuchi, S. Suzuki, Purification and properties of Biotechnol. J. 12 (10) (2017), 1700239.
bacterial chondroitinases and chondrosulfatases, J. Biol. Chem. 243 (7) (1968) [31] K.-W. Shim, D.-H. Kim, Cloning and expression of chondroitinase AC from
1523–1535. Bacteroides stercoris HJ-15, Protein Expr. Purif. 58 (2) (2008) 222–228.
[5] K. Gu, R.J. Linhardt, M. Laliberte, J. Zimmermann, Purification, characterization [32] M. Kitamikado, Y.-Z. Lee, Chondroitinase-producing bacteria in natural habitats,
and specificity of chondroitin lyases and glycuronidase from Flavobacterium Appl. Microbiol. 29 (3) (1975) 414–421.
heparinum, Biochem. J. 312 (2) (1995) 569–577. [33] N.K. Tripathi, Production and purification of recombinant proteins from
[6] A. Hamai, N. Hashimoto, H. Mochizuki, F. Kato, Y. Makiguchi, K. Horie, Escherichia coli, ChemBioEng Rev. 3 (3) (2016) 116–133.
S. Suzuki, Two distinct chondroitin sulfate ABC lyases an endoeliminase yielding [34] M. Nazari-Robati, K. Khajeh, M. Aminian, M. Fathi-Roudsari, A. Golestani, Co-
tetrasaccharides and an exoeliminase preferentially acting on oligosaccharides, solvent mediated thermal stabilization of chondroitinase ABC I form Proteus
J. Biol. Chem. 272 (14) (1997) 9123–9130. vulgaris, Int. J. Biol. Macromol. 50 (3) (2012) 487–492.
[7] V. Prabhakar, R. Raman, I. Capila, C.J. Bosques, K. Pojasek, R. Sasisekharan, [35] X. Lu, Q. Zhong, J. Liu, F. Yang, C. Lu, H. Xiong, S. Li, Y. Zhu, L. Wu, Efficient
Biochemical characterization of the chondroitinase ABC I active site, Biochem. J. expression of chondroitinase ABC I for specific disaccharides detection of
390 (2) (2005) 395–405. chondroitin sulfate, Int. J. Biol. Macromol. 143 (2020) 41–48.
[8] W. Wang, J. Wang, F. Li, Hyaluronidase and chondroitinase, Protein Rev. Volume [36] Y. Fang, S. Yang, X. Fu, W. Xie, L. Li, Z. Liu, H. Mou, C. Zhu, Expression,
17 (2017) 75–87. purification and characterization of chondroitinase AC II from marine bacterium
[9] K. Hiyama, S. Okada, Crystallization and some properties of chondroitinase from Arthrobacter sp. CS01, Mar. Drugs 17 (3) (2019) 185.
Arthrobacter aurescens, J. Biol. Chem. 250 (5) (1975) 1824–1828. [37] J. Takeuchi, Effect of chondroitinases on the growth of solid Ehrlich ascites
[10] N. Kasinathan, S.M. Volety, V.R. Josyula, Chondroitinase: a promising tumour, Br. J. Cancer 26 (2) (1972) 115–119.
therapeutic enzyme, Crit. Rev. Microbiol. 42 (3) (2016) 474–484. [38] G.S. Hageman, S.R. Russell, Chondroitinase-mediated disinsertion of the primate
[11] C. Zhu, J. Zhang, J. Zhang, Y. Jiang, Z. Shen, H. Guan, X. Jiang, Purification and vitreous body, Invest. Ophthalmol. Vis. Sci. 33 (1994), 1260-1260.
characterization of chondroitinase ABC from Acinetobacter sp. C26, Int. J. Biol. [39] Y. Yagi, E. Muroga, M. Naitoh, Z. Isogai, S. Matsui, S. Ikehara, S. Suzuki,
Macromol. 95 (2017) 80–86. Y. Miyachi, A. Utani, An ex vivo model employing keloid-derived cell-seeded
[12] E.J. Bradbury, L.D.F. Moon, R.J. Popat, V.R. King, G.S. Bennett, P.N. Patel, J. collagen sponges for therapy development, J. Invest. Dermatol. 133 (2) (2013)
W. Fawcett, S.B. McMahon, Chondroitinase ABC promotes functional recovery 386–393.
after spinal cord injury, Nature 416 (6881) (2002) 636–640. [40] T. Pizzorusso, P. Medini, N. Berardi, S. Chierzi, J.W. Fawcett, L. Maffei,
[13] M.D. Brown, The rationale for and pre-clinical results of chondroitinase ABC in Reactivation of ocular dominance plasticity in the adult visual cortex, Science 298
chemonucleolysis. International Congress Series, Elsevier,, 2001, pp. 171–176. (5596) (2002) 1248–1251.
[14] E.M. Denholm, Y.-Q. Lin, P.J. Silver, Anti-tumor activities of chondroitinase AC [41] J.W. Fawcett, R.A. Asher, The glial scar and central nervous system repair, Brain
and chondroitinase B: inhibition of angiogenesis, proliferation and invasion, Eur. Res. Bull. 49 (6) (1999) 377–391.
J. Pharmacol. 416 (3) (2001) 213–221. [42] R.J. McKeon, R.C. Schreiber, J.S. Rudge, J. Silver, Reduction of neurite outgrowth
[15] R.J. Linhardt, F.Y. Avci, T. Toida, Y.S. Kim, M. Cygler, CS lyases: structure, in a model of glial scarring following CNS injury is correlated with the expression
activity, and applications in analysis and the treatment of diseases, Adv. of inhibitory molecules on reactive astrocytes, J. Neurosci. 11 (11) (1991)
Pharmacol. 53 (2006) 187–215. 3398–3411.
[16] W. Huang, V. Lunin, Y. Li, S. Suzuki, N. Sugiura, H. Miyazono, M. Cygler, Crystal [43] B.P. Niederöst, D.R. Zimmermann, M.E. Schwab, C.E. Bandtlow, Bovine CNS
structure of Proteus vulgaris chondroitin sulfate ABC lyase I at 1.9 A˚resolution, myelin contains neurite growth-inhibitory activity associated with chondroitin
J. Mol. Biol. 328 (3) (2003) 623–634. sulfate proteoglycans, J. Neurosci. 19 (20) (1999) 8979–8989.
[17] J. Féthière, B. Eggimann, M. Cygler, Crystal structure of chondroitin AC lyase, a [44] L.C. Smith-Thomas, J. Fok-Seang, J. Stevens, J.-S. Du, E. Muir, A. Faissner, H.
representative of a family of glycosaminoglycan degrading enzymes, J. Mol. Biol. M. Geller, J.H. Rogers, J.W. Fawcett, An inhibitor of neurite outgrowth produced
288 (4) (1999) 635–647. by astrocytes, J. Cell Sci. 107 (6) (1994) 1687–1695.
[18] W. Huang, A. Matte, Y. Li, Y.S. Kim, R.J. Linhardt, H. Su, M. Cygler, Crystal [45] S.J.A. Davies, D.R. Goucher, C. Doller, J. Silver, Robust regeneration of adult
structure of chondroitinase B from Flavobacterium heparinum and its complex sensory axons in degenerating white matter of the adult rat spinal cord,
with a disaccharide product at 1.7 Å resolution, J. Mol. Biol. 294 (5) (1999) J. Neurosci. 19 (14) (1999) 5810–5822.
1257–1269. [46] P.S. Fidler, K. Schuette, R.A. Asher, A. Dobbertin, S.R. Thornton, Y. Calle-Patino,
[19] G. Michel, K. Pojasek, Y. Li, T. Sulea, R.J. Linhardt, R. Raman, V. Prabhakar, E. Muir, J.M. Levine, H.M. Geller, J.H. Rogers, Comparing astrocytic cell lines
R. Sasisekharan, M. Cygler, The structure of chondroitin B lyase complexed with that are inhibitory or permissive for axon growth: the major axon-inhibitory
glycosaminoglycan oligosaccharides unravels a calcium-dependent catalytic proteoglycan is NG2, J. Neurosci. 19 (20) (1999) 8778–8788.
machinery, J. Biol. Chem. 279 (31) (2004) 32882–32896. [47] R.J. McKeon, A. Höke, J. Silver, Injury-induced proteoglycans inhibit the
[20] W. Huang, A. Matte, S. Suzuki, N. Sugiura, H. Miyazono, M. Cygler, potential for laminin-mediated axon growth on astrocytic scars, Exp. Neurol. 136
Crystallization and preliminary X-ray analysis of chondroitin sulfate ABC lyases I (1) (1995) 32–43.
and II from Proteus vulgaris, Acta Crystallogr. Sect. D: Biol. Crystallogr. 56 (7) [48] L.D.F. Moon, R.A. Asher, K.E. Rhodes, J.W. Fawcett, Regeneration of CNS axons
(2000) 904–906. back to their target following treatment of adult rat brain with chondroitinase
[21] V. Prabhakar, I. Capila, J. Carlos, K. Pojasek, R. Sasisekharan, Chondroitinase ABC, Nat. Neurosci. 4 (5) (2001) 465–466.
ABC I from Proteus vulgaris: cloning, recombinant expression and active site [49] J. Zuo, D. Neubauer, K. Dyess, T.A. Ferguson, D. Muir, Degradation of chondroitin
identification, Biochem. J. 386 (1) (2005) 103–112. sulfate proteoglycan enhances the neurite-promoting potential of spinal cord
[22] L.A. Vijay, K. Krishnamoorthy, Optimization of induction media for production of tissue, Exp. Neurol. 154 (2) (1998) 654–662.
chondroitinase from proteus penneri SN5 using response surface methodology, [50] J. Hu, W. Rodemer, G. Zhang, L.Q. Jin, S. Li, M.E. Selzer, Chondroitinase ABC
J. Pharm. Sci. Res. 10 (11) (2018) 2702–2708. promotes axon regeneration and reduces retrograde apoptosis signaling in
[23] J. Fu, Z. Jiang, J. Chang, B. Han, W. Liu, Y. Peng, Purification, characterization of lamprey, Front Cell Dev. Biol. 9 (2021), 653638.
Chondroitinase ABC from Sphingomonas paucimobilis and in vitro [51] M.L. Lemons, D.R. Howland, D.K. Anderson, Chondroitin sulfate proteoglycan
cardiocytoprotection of the enzymatically degraded CS-A, Int. J. Biol. Macromol. immunoreactivity increases following spinal cord injury and transplantation, Exp.
115 (2018) 737–745. Neurol. 160 (1) (1999) 51–65.
[24] N. Sato, K. Murata, A. Kimura, Cultural conditions for the induction of [52] N.J. Tester, D.R. Howland, Chondroitinase ABC improves basic and skilled
chondroitinase ABC in Proteus vulgaris cells, J. Ferment. Technol. 64 (2) (1986) locomotion in spinal cord injured cats, Exp. Neurol. 209 (2) (2008) 483–496.
155–159. [53] G. García-Alías, R. Lin, S.F. Akrimi, D. Story, E.J. Bradbury, J.W. Fawcett,
[25] K. Narayanan, N. Sivagurunathan, V.M. Subrahmanyam, J. Venkata Rao, Fungal Therapeutic time window for the application of chondroitinase ABC after spinal
chondroitinase: production and prospects for therapeutic application, Recent cord injury, Exp. Neurol. 210 (2) (2008) 331–338.
Adv. Appl. Microbiol. (2017) 275–290. [54] S. Soleman, P.K. Yip, D.A. Duricki, L.D.F. Moon, Delayed treatment with
[26] N. Kasinathan, V. Subrahmanyam, V.J. Rao, Design of cultural conditions for chondroitinase ABC promotes sensorimotor recovery and plasticity after stroke in
production of chondroitinase by Aspergillus niger using design of experiments, aged rats, Brain 135 (4) (2012) 1210–1223.
Indian Drugs 52 (6) (2015) 24–32.

13
A. Kheirollahi et al. Enzyme and Microbial Technology 172 (2024) 110348

[55] J.J. Hill, K. Jin, X.O. Mao, L. Xie, D.A. Greenberg, Intracerebral chondroitinase [81] C.A. Cooney, F. Jousheghany, A. Yao-Borengasser, B. Phanavanh, T. Gomes, A.
ABC and heparan sulfate proteoglycan glypican improve outcome from chronic M. Kieber-Emmons, E.R. Siegel, L.J. Suva, S. Ferrone, T. Kieber-Emmons,
stroke in rats, Proc. Natl. Acad. Sci. 109 (23) (2012) 9155–9160. Chondroitin sulfates play a major role in breast cancer metastasis: a role for
[56] L. Gherardini, M. Gennaro, T. Pizzorusso, Perilesional treatment with CSPG4 and CHST11gene expression in forming surface P-selectin ligands in
chondroitinase ABC and motor training promote functional recovery after stroke aggressive breast cancer cells, Breast Cancer Res. 13 (3) (2011), R58.
in rats, Cereb. cortex 25 (1) (2013) 202–212. [82] Z. Isogai, T. Shinomura, N. Yamakawa, J. Takeuchi, T. Tsuji, D. Heinegård,
[57] X.-r Chen, S.-j Liao, L.-x Ye, Q. Gong, Q. Ding, J.-s Zeng, J. Yu, Neuroprotective K. Kimata, 2B1 antigen characteristically expressed on extracellular matrices of
effect of chondroitinase ABC on primary and secondary brain injury after stroke human malignant tumors is a large chondroitin sulfate proteoglycan, PG-M/
in hypertensive rats, Brain Res. 1543 (2014) 324–333. versican, Cancer Res. 56 (17) (1996) 3902–3908.
[58] A. Rolls, R. Shechter, M. Schwartz, The bright side of the glial scar in CNS repair, [83] S. Suwiwat, C. Ricciardelli, R. Tammi, M. Tammi, P. Auvinen, V.-M. Kosma, R.
Nat. Rev. Neurosci. 10 (3) (2009) 235–241. G. LeBaron, W.A. Raymond, W.D. Tilley, D.J. Horsfall, Expression of extracellular
[59] C.M. Galtrey, J.W. Fawcett, The role of chondroitin sulfate proteoglycans in matrix components versican, chondroitin sulfate, tenascin, and hyaluronan, and
regeneration and plasticity in the central nervous system, Brain Res. Rev. 54 (1) their association with disease outcome in node-negative breast cancer, Clin.
(2007) 1–18. Cancer Res. 10 (7) (2004) 2491–2498.
[60] A.S. Hunanyan, G. García-Alías, V. Alessi, J.M. Levine, J.W. Fawcett, L. [84] T. Aigner, S. Loos, C. Inwards, R. Perris, D. Perissinotto, K.K. Unni, T. Kirchner,
M. Mendell, V.L. Arvanian, Role of chondroitin sulfate proteoglycans in axonal Chondroblastoma is an osteoid-forming, but not cartilage-forming neoplasm,
conduction in Mammalian spinal cord, J. Neurosci. 30 (23) (2010) 7761–7769. J. Pathol. 189 (4) (1999) 463–469.
[61] J.M. Massey, C.H. Hubscher, M.R. Wagoner, J.A. Decker, J. Amps, J. Silver, S. [85] E.M. Denholm, E. Cauchon, C. Poulin, P.J. Silver, Inhibition of human dermal
M. Onifer, Chondroitinase ABC digestion of the perineuronal net promotes fibroblast proliferation by removal of dermatan sulfate, Eur. J. Pharmacol. 400
functional collateral sprouting in the cuneate nucleus after cervical spinal cord (2) (2000) 145–153.
injury, J. Neurosci. 26 (16) (2006) 4406–4414. [86] E. Hurt-Camejo, B. Rosengren, P. Sartipy, K. Elfsberg, Gn Camejo, L. Svensson,
[62] R.J. Nudo, Postinfarct cortical plasticity and behavioral recovery, Stroke 38 (2) CD44, a cell surface chondroitin sulfate proteoglycan, mediates binding of
(2007) 840–845. interferon-γ and some of its biological effects on human vascular smooth muscle
[63] J.D. Finan, F.S. Cho, S.G. Kernie, B. Morrison, Intracerebroventricular cells, J. Biol. Chem. 274 (27) (1999) 18957–18964.
administration of chondroitinase ABC reduces acute edema after traumatic brain [87] M. Lyon, J.A. Deakin, H. Rahmoune, D.G. Fernig, T. Nakamura, J.T. Gallagher,
injury in mice, BMC Res. Notes 9 (1) (2016), 160. Hepatocyte growth factor/scatter factor binds with high affinity to dermatan
[64] E.E. Birch, Amblyopia and binocular vision, Prog. Retin. eye Res. 33 (2013) sulfate, J. Biol. Chem. 273 (1) (1998) 271–278.
67–84. [88] N. Maeda, T. Nishiwaki, T. Shintani, H. Hamanaka, M. Noda, 6B4 proteoglycan/
[65] T. Pizzorusso, P. Medini, S. Landi, S. Baldini, N. Berardi, L. Maffei, Structural and phosphacan, an extracellular variant of receptor-like protein-tyrosine
functional recovery from early monocular deprivation in adult rats, Proc. Natl. phosphatase ζ/RPTPβ, binds pleiotrophin/heparin-binding growth-associated
Acad. Sci. 103 (22) (2006) 8517–8522. molecule (HB-GAM), J. Biol. Chem. 271 (35) (1996) 21446–21452.
[66] V. Vorobyov, J.C.F. Kwok, J.W. Fawcett, F. Sengpiel, Effects of digesting [89] F. Petersen, L. Bock, H.-D. Flad, E. Brandt, A chondroitin sulfate proteoglycan on
chondroitin sulfate proteoglycans on plasticity in cat primary visual cortex, human neutrophils specifically binds platelet factor 4 and is involved in cell
J. Neurosci. 33 (1) (2013) 234–243. activation, J. Immunol. 161 (8) (1998) 4347–4355.
[67] J.R. Tribble, P.A. Williams, B. Caterson, F. Sengpiel, J.E. Morgan, Digestion of the [90] R. Prinz, C. Willis, A. Viloria-Petit, M. Klüppel, Elimination of breast tumor-
glycosaminoglycan extracellular matrix by chondroitinase ABC supports retinal associated chondroitin sulfate promotes metastasis, Genet Mol. Res 10 (4) (2011)
ganglion cell dendritic preservation in a rodent model of experimental glaucoma, 3901–3913.
Mol. Brain 11 (1) (2018), 69. [91] B. Kaur, E. Antonio Chiocca, T.P. Cripe, Oncolytic HSV-1 virotherapy: clinical
[68] S. Tuffaha, M. Quigley, T. Ng, V.S. Gorantla, J.T. Shores, B. Pulikkottil, experience and opportunities for progress, Curr. Pharm. Biotechnol. 13 (9) (2012)
C. Shestak, G. Brandacher, W.P.A. Lee, The effect of chondroitinase on nerve 1842–1851.
regeneration following composite tissue allotransplantation, J. hand Surg. 36 (9) [92] N. Dmitrieva, L. Yu, M. Viapiano, T.P. Cripe, E.A. Chiocca, J.C. Glorioso, B. Kaur,
(2011) 1447–1452. Chondroitinase ABC I-mediated enhancement of oncolytic virus spread and
[69] W.N. Sivak, J.D. White, J.M. Bliley, L.W. Tien, H.T. Liao, D.L. Kaplan, K.G. Marra, antitumor efficacy, Clin. Cancer Res. 17 (6) (2011) 1362–1372.
Delivery of chondroitinase ABC and glial cell line-derived neurotrophic factor [93] A.C. Jaime-Ramirez, N. Dmitrieva, J.Y. Yoo, Y. Banasavadi-Siddegowda,
from silk fibroin conduits enhances peripheral nerve regeneration, J. Tissue Eng. J. Zhang, T. Relation, C. Bolyard, J. Wojton, B. Kaur, Humanized chondroitinase
Regen. Med. 11 (3) (2017) 733–742. ABC sensitizes glioblastoma cells to temozolomide, J. gene Med. 19 (3) (2017),
[70] R.T. Gardner, B.A. Habecker, Infarct-derived chondroitin sulfate proteoglycans e2942.
prevent sympathetic reinnervation after cardiac ischemia-reperfusion injury, [94] J.R.M. Martins, M.E.C. Gadelha, S.M. Fonseca, L.O. Sampaio, P.A.D.L. Pontes, C.
J. Neurosci. 33 (17) (2013) 7175–7183. P. Dietrich, H.B. Nader, Patients with head and neck tumors excrete a chondroitin
[71] E.J.R. Fletcher, L.D.F. Moon, S. Duty, Chondroitinase ABC reduces dopaminergic sulfate with a low degree of sulfation: a new tool for diagnosis and follow-up of
nigral cell death and striatal terminal loss in a 6-hydroxydopamine partial lesion cancer therapy, Otolaryngol. -Head. neck Surg. 122 (1) (2000) 115–118.
mouse model of Parkinson’s disease, BMC Neurosci. 20 (1) (2019), 61. [95] N. Dmitrieva, L. Yu, M. Viapiano, T.P. Cripe, E.A. Chiocca, J.C. Glorioso, B. Kaur,
[72] N. Afratis, C. Gialeli, D. Nikitovic, T. Tsegenidis, E. Karousou, A.D. Theocharis, M. Chondroitinase ABC I-mediated enhancement of oncolytic virus spread and
S. Pavão, G.N. Tzanakakis, N.K. Karamanos, Glycosaminoglycans: key players in antitumor efficacy, Clin. Cancer Res 17 (6) (2011) 1362–1372.
cancer cell biology and treatment, FEBS J. 279 (7) (2012) 1177–1197. [96] F. Lopez-Lopez, M. Rodriguez-Blanco, F. Gomez-Ulla, J. Marticorena, Enzymatic
[73] G. Pluschke, M. Vanek, A. Evans, T. Dittmar, P. Schmid, P. Itin, E.J. Filardo, R. vitreolysis, Curr. Diabetes Rev. 5 (1) (2009) 57–62.
A. Reisfeld, Molecular cloning of a human melanoma-associated chondroitin [97] A. Gandorfer, Enzymatic vitreous disruption, Eye 22 (10) (2008) 1273–1277.
sulfate proteoglycan, Proc. Natl. Acad. Sci. 93 (18) (1996) 9710–9715. [98] F. Staubach, V. Nober, P. Janknecht, Enzyme-assisted vitrectomy in enucleated
[74] J. Iida, K.L. Wilhelmson, J. Ng, P. Lee, C. Morrison, E. Tam, C.M. Overall, J. pig eyes: a comparison of hyaluronidase, chondroitinase, and plasmin, Curr. eye
B. McCarthy, Cell surface chondroitin sulfate glycosaminoglycan in melanoma: Res. 29(4-5) (2004) 261–268.
role in the activation of pro-MMP-2 (pro-gelatinase A), Biochem. J. 403 (3) [99] M. Hermel, N.F. Schrage, Efficacy of plasmin enzymes and chondroitinase ABC in
(2007) 553–563. creating posterior vitreous separation in the pig: a masked, placebo-controlled in
[75] K.M. Eisenmann, J.B. McCarthy, M.A. Simpson, P.J. Keely, J.-L. Guan, vivo study, Graefe’S. Arch. Clin. Exp. Ophthalmol. 245 (3) (2007) 399–406.
K. Tachibana, L. Lim, E. Manser, L.T. Furcht, J. Iida, Melanoma chondroitin [100] L. Smith, Chemonucleolysis, Clin. Orthop. Relat. Res. 67 (1969) 72–80.
sulphate proteoglycan regulates cell spreading through Cdc42, Ack-1 and p130, [101] F. Kato, H. Iwata, K. Mimatsu, T. Miura, Experimental chemonucleolysis with
Cas., Biochem. –Fac. Publ. 1 (8) (1999) 507–5013. chondroitinase ABC, Clin. Orthop. Relat. Res. 253 (1990) 301–308.
[76] J. Iida, J. Dorchak, R. Clancy, J. Slavik, R. Ellsworth, Y. Katagiri, E.N. Pugacheva, [102] D.-S. Lü, Y. Shono, I. Oda, K. Abumi, K. Kaneda, Effects of chondroitinase ABC and
T.H. van Kuppevelt, R.J. Mural, M.L. Cutler, Role for chondroitin sulfate chymopapain on spinal motion segment biomechanics: an in vivo biomechanical,
glycosaminoglycan in NEDD9-mediated breast cancer cell growth, Exp. Cell Res. radiologic, and histologic canine study, Spine 22 (16) (1997) 1828–1834.
330 (2) (2015) 358–370. [103] R.J. Hoogendoorn, P.I. Wuisman, T.H. Smit, V.E. Everts, M.N. Helder,
[77] J.D. Esko, K.S. Rostand, J.L. Weinke, Tumor formation dependent on Experimental intervertebral disc degeneration induced by chondroitinase ABC in
proteoglycan biosynthesis, Science 241 (4869) (1988) 1092–1096. the goat, Spine 32 (17) (2007) 1816–1825.
[78] C.A. Henke, U. Roongta, D.J. Mickelson, J.R. Knutson, J.B. McCarthy, CD44- [104] T. Sugimura, F. Kato, K. Mimatsu, O. Takenaka, H. Iwata, Experimental
related chondroitin sulfate proteoglycan, a cell surface receptor implicated with chemonucleolysis with chondroitinase ABC in monkeys, Spine 21 (2) (1996)
tumor cell invasion, mediates endothelial cell migration on fibrinogen and 161–165.
invasion into a fibrin matrix, J. Clin. Investig. 97 (11) (1996) 2541–2552. [105] M. Sasaki, T. Takahashi, K. Miyahara, T. Hirose, Effects of chondroitinase ABC on
[79] A.E. Faassen, J.A. Schrager, D.J. Klein, T.R. Oegema, J.R. Couchman, J. intradiscal pressure in sheep: an in vivo study, Spine 26 (5) (2001) 463–468.
B. McCarthy, A cell surface chondroitin sulfate proteoglycan, immunologically [106] M. Ikeda, M. Naitoh, H. Kubota, T. Ishiko, K. Yoshikawa, S. Yamawaki,
related to CD44, is involved in type I collagen-mediated melanoma cell motility M. Kurokawa, A. Utani, T. Nakamura, K. Nagata, Elastic fiber assembly is
and invasion, J. Cell Biol. 116 (2) (1992) 521–531. disrupted by excessive accumulation of chondroitin sulfate in the human dermal
[80] A.E. Faassen, D.L. Mooradian, R.T. Tranquillo, R.B. Dickinson, P.C. Letourneau, T. fibrotic disease, keloid, Biochem. Biophys. Res. Commun. 390 (4) (2009)
R. Oegema, J.B. McCarthy, Cell surface CD44-related chondroitin sulfate 1221–1228.
proteoglycan is required for transforming growth factor-beta-stimulated mouse [107] T. Ishiko, M. Naitoh, H. Kubota, S. Yamawaki, M. Ikeda, K. Yoshikawa, H. Fujita,
melanoma cell motility and invasive behavior on type I collagen, J. Cell Sci. 105 H. Yamaguchi, Y. Kurahashi, S. Suzuki, Chondroitinase injection improves keloid
(2) (1993) 501–511. pathology by reorganizing the extracellular matrix with regenerated elastic fibers,
J. Dermatol. 40 (5) (2013) 380–383.

14
A. Kheirollahi et al. Enzyme and Microbial Technology 172 (2024) 110348

[108] L.L. Jones, R.U. Margolis, M.H. Tuszynski, The chondroitin sulfate proteoglycans to secrete neurotrophin and chondroitinase promotes axonal regeneration and
neurocan, brevican, phosphacan, and versican are differentially regulated locomotion after spinal cord injury, J. Neurosci. 34 (5) (2014) 1838–1855.
following spinal cord injury, Exp. Neurol. 182 (2) (2003) 399–411. [136] K. Bartus, N.D. James, A. Didangelos, K.D. Bosch, J. Verhaagen, R.J. Yáñez-
[109] Dm Crespo, R.A. Asher, R. Lin, K.E. Rhodes, J.W. Fawcett, How does Muñoz, J.H. Rogers, B.L. Schneider, E.M. Muir, E.J. Bradbury, Large-scale
chondroitinase promote functional recovery in the damaged CNS? Exp. Neurol. chondroitin sulfate proteoglycan digestion with chondroitinase gene therapy
206 (2) (2007) 159–171. leads to reduced pathology and modulates macrophage phenotype following
[110] N.J. Tester, A.H. Plaas, D.R. Howland, Effect of body temperature on spinal cord contusion injury, J. Neurosci. 34 (14) (2014) 4822–4836.
chondroitinase ABC’s ability to cleave chondroitin sulfate glycosaminoglycans, [137] J.N. Alves, E.M. Muir, M.R. Andrews, A. Ward, N. Michelmore, D. Dasgupta,
J. Neurosci. Res. 85 (5) (2007) 1110–1118. J. Verhaagen, E.B. Moloney, R.J. Keynes, J.W. Fawcett, AAV vector-mediated
[111] Y.-C. Huang, S.-H. Hsu, M.-T. Chen, C.-H. Hsieh, W.-C. Kuo, H. Cheng, Y.- secretion of chondroitinase provides a sensitive tracer for axonal arborisations,
Y. Huang, Controlled release of chondroitinase ABC in chitosan-based scaffolds J. Neurosci. Methods 227 (2014) 107–120.
and PDLLA microspheres, Carbohydr. Polym. 84 (2) (2011) 788–793. [138] E. Muir, M. Raza, C. Ellis, E. Burnside, F. Love, S. Heller, M. Elliot, E. Daniell,
[112] A.J.T. Hyatt, D. Wang, J.C. Kwok, J.W. Fawcett, K.R. Martin, Controlled release of D. Dasgupta, N. Alves, Trafficking and processing of bacterial proteins by
chondroitinase ABC from fibrin gel reduces the level of inhibitory mammalian cells: Insights from chondroitinase ABC, PloS One 12 (11) (2017),
glycosaminoglycan chains in lesioned spinal cord, J. Control. Release 147 (1) e0186759.
(2010) 24–29. [139] N.D. James, J. Shea, E.M. Muir, J. Verhaagen, B.L. Schneider, E.J. Bradbury,
[113] G.M. Curinga, D.M. Snow, C. Mashburn, K. Kohler, R. Thobaben, A.O. Caggiano, Chondroitinase gene therapy improves upper limb function following cervical
G.M. Smith, Mammalian-produced chondroitinase AC mitigates axon inhibition contusion injury, Exp. Neurol. 271 (2015) 131–135.
by chondroitin sulfate proteoglycans, J. Neurochem. 102 (1) (2007) 275–288. [140] E.R. Burnside, F. De Winter, A. Didangelos, N.D. James, E.C. Andreica, H. Layard-
[114] H. Lee, R.J. McKeon, R.V. Bellamkonda, Sustained delivery of thermostabilized Horsfall, E.M. Muir, J. Verhaagen, E.J. Bradbury, Immune-evasive gene switch
chABC enhances axonal sprouting and functional recovery after spinal cord enables regulated delivery of chondroitinase after spinal cord injury, Brain 141
injury, Proc. Natl. Acad. Sci. 107 (8) (2010) 3340–3345. (8) (2018) 2362–2381.
[115] M.A. Anderson, J.E. Burda, Y. Ren, Y. Ao, T.M. O’Shea, R. Kawaguchi, [141] J.W. Fawcett, T. Oohashi, T. Pizzorusso, The roles of perineuronal nets and the
G. Coppola, B.S. Khakh, T.J. Deming, M.V. Sofroniew, Astrocyte scar formation perinodal extracellular matrix in neuronal function, Nat. Rev. Neurosci. 20 (8)
aids central nervous system axon regeneration, Nature 532 (7598) (2016) (2019) 451–465.
195–200. [142] K.E. Carstens, B.R. Gloss, G.M. Alexander, S.M. Dudek, Modified adeno-associated
[116] G. DeSantis, J.B. Jones, Chemical modification of enzymes for enhanced virus targets the bacterial enzyme chondroitinase ABC to select mouse neuronal
functionality, Curr. Opin. Biotechnol. 10 (4) (1999) 324–330. populations in vivo using the Cre-LoxP system, Eur. J. Neurosci. 53 (12) (2021)
[117] C.Ó. Fágáin, Understanding and increasing protein stability, Biochim. Et. Biophys. 4005–4015.
Acta (BBA)-Protein Struct. Mol. Enzymol. 1252 (1) (1995) 1–14. [143] D. Carwardine, L.F. Wong, J.W. Fawcett, E.M. Muir, N. Granger, Canine olfactory
[118] H. Yang, J. Li, G. Du, L. Liu, Microbial Production and Molecular Engineering of ensheathing cells from the olfactory mucosa can be engineered to produce active
Industrial Enzymes: Challenges and Strategies. Biotechnology of Microbial chondroitinase ABC, J. Neurol. Sci. 367 (2016) 311–318.
Enzymes, Elsevier, 2017, pp. 151–165. [144] N. Weidner, R. Rupp, K.E. Tansey, Neurological Aspects of Spinal Cord Injury,
[119] M. Nazari-Robati, K. Khajeh, M. Aminian, N. Mollania, A. Golestani, Springer, 2017.
Enhancement of thermal stability of chondroitinase ABC I by site-directed [145] A.M. Ziemba, R.J. Gilbert, Biomaterials for Local, Controlled Drug Delivery to the
mutagenesis: an insight from Ramachandran plot, Biochim. Et. Biophys. Acta Injured Spinal Cord, Front. Pharmacol. 8 (2017).
(BBA)-Proteins Proteom. 1834 (2) (2013) 479–486. [146] K.S. Straley, C.W.P. Foo, S.C. Heilshorn, Biomaterial design strategies for the
[120] S.A. Shirdel, K. Khalifeh, A. Golestani, B. Ranjbar, K. Khajeh, Critical role of a treatment of spinal cord injuries, J. Neurotrauma 27 (1) (2010) 1–19.
loop at C-terminal domain on the conformational stability and catalytic efficiency [147] A.K. Varma, A. Das, G. Wallace, J. Barry, A.A. Vertegel, S.K. Ray, N.L. Banik,
of chondroitinase ABC I, Mol. Biotechnol. 57 (8) (2015) 727–734. Spinal cord injury: a review of current therapy, future treatments, and basic
[121] M. Shamsi, S.A. Shirdel, V. Jafarian, S.S. Jafari, K. Khalifeh, A. Golestani, science frontiers, Neurochem. Res. 38 (5) (2013) 895–905.
Optimization of conformational stability and catalytic efficiency in [148] R.C. Assunção-Silva, E.D. Gomes, N. Sousa, N.A. Silva, A.J. Salgado, Hydrogels
chondroitinase ABC І by protein engineering methods, Eng. Life Sci. 16 (8) (2016) and cell based therapies in spinal cord injury regeneration, Stem Cells Int. 2015
690–696. (2015).
[122] A. Kheirollahi, K. Khajeh, A. Golestani, Rigidifying flexible sites: an approach to [149] R. Narayanaswamy, V.P. Torchilin, Hydrogels and their applications in targeted
improve stability of chondroitinase ABC I, Int. J. Biol. Macromol. 97 (2017) drug delivery, Molecules 24 (3) (2019).
270–278. [150] F. Rossi, P. Veglianese, M. Santoro, S. Papa, C. Rogora, V. Dell’Oro, G. Forloni,
[123] M.E. Shahaboddin, K. Khajeh, M. Maleki, A. Golestani, Improvement of activity M. Masi, G. Perale, Sustained delivery of chondroitinase ABC from hydrogel
and stability of Chondroitinase ABC I by introducing an aromatic cluster at the system, J. Funct. Biomater. 3 (1) (2012) 199–208.
surface of protein, Enzym. Microb. Technol. 105 (2017) 38–44. [151] V. Delplace, A.J. Pickering, M.H. Hettiaratchi, S. Zhao, T. Kivijärvi, M.S. Shoichet,
[124] M. Maleki, K. Khajeh, M. Amanlou, A. Golestani, Role of His-His interaction in Ser Inverse electron-demand diels-alder methylcellulose hydrogels enable the co-
474-His 475-Tyr 476 sequence of Chondroitinase ABC I in the enzyme activity delivery of chondroitinase ABC and neural progenitor cells, Biomacromolecules
and stability, Int. J. Biol. Macromol. (2017). 21 (6) (2020) 2421–2431.
[125] Z. Chen, Y. Li, Y. Feng, L. Chen, Q. Yuan, Enzyme activity enhancement of [152] M.H. Hettiaratchi, M.J. O’Meara, C.J. Teal, S.L. Payne, A.J. Pickering, M.
chondroitinase ABC I from Proteus vulgaris by site-directed mutagenesis, RSC S. Shoichet, Local delivery of stabilized chondroitinase ABC degrades chondroitin
Adv. 5 (93) (2015) 76040–76047. sulfate proteoglycans in stroke-injured rat brains, J. Control Release 297 (2019)
[126] T. Arakawa, S.N. Timasheff, Stabilization of protein structure by sugars, 14–25.
Biochemistry 21 (25) (1982) 6536–6544. [153] A. Raspa, L. Carminati, R. Pugliese, F. Fontana, F. Gelain, Self-assembling peptide
[127] M. Nazari-Robati, A. Golestani, G. Asadikaram, Improvement of proteolytic and hydrogels for the stabilization and sustained release of active Chondroitinase ABC
oxidative stability of Chondroitinase ABC I by cosolvents, Int. J. Biol. Macromol. in vitro and in spinal cord injuries, J. Control Release 330 (2021) 1208–1219.
91 (2016) 812–817. [154] A.T. Mneimneh, M.M. Mehanna, Collagen-based scaffolds: an auspicious tool to
[128] A. Raspa, E. Bolla, C. Cuscona, F. Gelain, Feasible stabilization of chondroitinase support repair, recovery, and regeneration post spinal cord injury, Int J. Pharm.
abc enables reduced astrogliosis in a chronic model of spinal cord injury, CNS 601 (2021), 120559.
Neurosci. Ther. 25 (1) (2019) 86–100. [155] Z.B. Ahi, R.C. Assunção-Silva, A.J. Salgado, K. Tuzlakoglu, A combinatorial
[129] M. Bagherieh, A. Kheirollahi, M.E. Shahaboddin, K. Khajeh, A. Golestani, Calcium approach for spinal cord injury repair using multifunctional collagen-based
and TNFα additively affect the chondroitinase ABC I activity, Int J. Biol. matrices: development, characterization and impact on cell adhesion and axonal
Macromol. 103 (2017) 1201–1206. growth, Biomed. Mater. 15 (5) (2020), 055024.
[130] S. Daneshjou, S. Khodaverdian, B. Dabirmanesh, F. Rahimi, S. Daneshjoo, [156] W. Huang, S. Ling, C. Li, F.G. Omenetto, D.L. Kaplan, Silkworm silk-based
F. Ghazi, K. Khajeh, Improvement of chondroitinases ABCI stability in natural materials and devices generated using bio-nanotechnology, Chem. Soc. Rev. 47
deep eutectic solvents, J. Mol. Liq. 227 (2017) 21–25. (17) (2018) 6486–6504.
[131] M.M. Pakulska, K. Vulic, R.Y. Tam, M.S. Shoichet, Hybrid crosslinked [157] P.K. Gupta, R. Gahtori, K. Govarthanan, V. Sharma, S. Pappuru, S. Pandit, A.
methylcellulose hydrogel: a predictable and tunable platform for local drug S. Mathuriya, S. Dholpuria, D.K. Bishi, Recent trends in biodegradable polyester
delivery, Adv. Mater. 27 (34) (2015) 5002–5008. nanomaterials for cancer therapy, Mater. Sci. Eng. C. Mater. Biol. Appl. 127
[132] M.M. Pakulska, K. Vulic, M.S. Shoichet, Affinity-based release of chondroitinase (2021), 112198.
ABC from a modified methylcellulose hydrogel, J. Control Release 171 (1) (2013) [158] T. Xia, B. Huang, S. Ni, L. Gao, J. Wang, J. Wang, A. Chen, S. Zhu, B. Wang, G. Li,
11–16. S. Zhu, X. Li, The combination of db-cAMP and ChABC with poly(propylene
[133] Y. Jin, A. Ketschek, Z. Jiang, G. Smith, I. Fischer, Chondroitinase activity can be carbonate) microfibers promote axonal regenerative sprouting and functional
transduced by a lentiviral vector in vitro and in vivo, J. Neurosci. Methods 199 (2) recovery after spinal cord hemisection injury, Biomed. Pharm. 86 (2017)
(2011) 208–213. 354–362.
[134] R.-R. Zhao, E.M. Muir, Jo.N. Alves, H. Rickman, A.Y. Allan, J.C. Kwok, K.C. [159] G. Kravanja, M. Primožič, Ž. Knez, M. Leitgeb, Chitosan-based (Nano)materiALS
D. Roet, J. Verhaagen, B.L. Schneider, J.-C. Bensadoun, Lentiviral vectors express FOR NOVEL BIOMEDICAL APPLications, Molecules 24 (10) (2019).
chondroitinase ABC in cortical projections and promote sprouting of injured [160] Y. Pang, A. Qin, X. Lin, L. Yang, Q. Wang, Z. Wang, Z. Shan, S. Li, J. Wang, S. Fan,
corticospinal axons, J. Neurosci. Methods 201 (1) (2011) 228–238. Q. Hu, Biodegradable and biocompatible high elastic chitosan scaffold is cell-
[135] H. Kanno, Y. Pressman, A. Moody, R. Berg, E.M. Muir, J.H. Rogers, H. Ozawa, friendly both in vitro and in vivo, Oncotarget 8 (22) (2017) 35583–35591.
E. Itoi, D.D. Pearse, M.B. Bunge, Combination of engineered Schwann cell grafts

15
A. Kheirollahi et al. Enzyme and Microbial Technology 172 (2024) 110348

[161] E.S. Ribeiro, B.S. de Farias, T.R. Sant’Anna Cadaval Junior, L.A. de Almeida Pinto, neurotrophin NT-3 secretion and NR2D expression promotes axonal plasticity and
P.S. Diaz, Chitosan-based nanofibers for enzyme immobilization, Int J. Biol. functional recovery in rats with lateral hemisection of the spinal cord, J. Neurosci.
Macromol. 183 (2021) 1959–1970. 31 (49) (2011) 17788–17799.
[162] S. Ni, T. Xia, X. Li, X. Zhu, H. Qi, S. Huang, J. Wang, Sustained delivery of [187] R.-R. Zhao, M.R. Andrews, D. Wang, P. Warren, M. Gullo, L. Schnell, M.
chondroitinase ABC by poly(propylene carbonate)-chitosan micron fibers E. Schwab, J.W. Fawcett, Combination treatment with anti-Nogo-A and
promotes axon regeneration and functional recovery after spinal cord chondroitinase ABC is more effective than single treatments at enhancing
hemisection, Brain Res. 1624 (2015) 469–478. functional recovery after spinal cord injury, Eur. J. Neurosci. 38 (6) (2013)
[163] N.L. Francis, P.M. Hunger, A.E. Donius, U.G. Wegst, M.A. Wheatley, Strategies for 2946–2961.
neurotrophin-3 and chondroitinase ABC release from freeze-cast chitosan-alginate [188] D. Wang, R.M. Ichiyama, R. Zhao, M.R. Andrews, J.W. Fawcett, Chondroitinase
nerve-guidance scaffolds, J. Tissue Eng. Regen. Med 11 (1) (2017) 285–294. combined with rehabilitation promotes recovery of forelimb function in rats with
[164] H.I. Chiu, N.A. Samad, L. Fang, V. Lim, Cytotoxicity of targeted PLGA chronic spinal cord injury, J. Neurosci. 31 (25) (2011) 9332–9344.
nanoparticles: a systematic review, RSC Adv. 11 (16) (2021) 9433–9449. [189] A. Janzadeh, A. Sarveazad, M. Yousefifard, S. Dameni, F.S. Samani,
[165] F.Sadat Tabatabaei Mirakabad, K. Nejati-Koshki, A. Akbarzadeh, M.R. Yamchi, K. Mokhtarian, F. Nasirinezhad, Combine effect of Chondroitinase ABC and low
M. Milani, N. Zarghami, V. Zeighamian, A. Rahimzadeh, S. Alimohammadi, level laser (660nm) on spinal cord injury model in adult male rats, Neuropeptides
Y. Hanifehpour, S.W. Joo, PLGA-based nanoparticles as cancer drug delivery 65 (2017) 90–99.
systems, Asian Pac. J. Cancer Prev. 15 (2) (2014) 517–535. [190] X. Liu, J. Wang, G. Li, H. Lv, Effect of combined chondroitinase ABC and
[166] F. Danhier, E. Ansorena, J.M. Silva, R. Coco, A. Le Breton, V. Préat, PLGA-based hyperbaric oxygen therapy in a rat model of spinal cord injury, Mol. Med Rep. 18
nanoparticles: an overview of biomedical applications, J. Control. Release 161 (2) (1 (2018) 25–30.
(2012) 505–522. [191] S. Karimi-Abdolrezaee, E. Eftekharpour, J. Wang, D. Schut, M.G. Fehlings,
[167] M. Azizi, F. Farahmandghavi, M.T. Joghataei, M. Zandi, M. Imani, M. Bakhtiari, Synergistic effects of transplanted adult neural stem/progenitor cells,
H. Omidian, ChABC-loaded PLGA nanoparticles: a comprehensive study on chondroitinase, and growth factors promote functional repair and plasticity of the
biocompatibility, functional recovery, and axonal regeneration in animal model chronically injured spinal cord, J. Neurosci. 30 (5) (2010) 1657–1676.
of spinal cord injury, Int J. Pharm. 577 (2020), 119037. [192] T. Ikegami, M. Nakamura, J. Yamane, H. Katoh, S. Okada, A. Iwanami,
[168] M.J. Mitchell, M.M. Billingsley, R.M. Haley, M.E. Wechsler, N.A. Peppas, K. Watanabe, K. Ishii, F. Kato, H. Fujita, Chondroitinase ABC combined with
R. Langer, Engineering precision nanoparticles for drug delivery, Nat. Rev. Drug neural stem/progenitor cell transplantation enhances graft cell migration and
Discov. 20 (2) (2021) 101–124. outgrowth of growth-associated protein-43-positive fibers after rat spinal cord
[169] J.J. Giner-Casares, M. Henriksen-Lacey, M. Coronado-Puchau, L.M. Liz-Marzán, injury, Eur. J. Neurosci. 22 (12) (2005) 3036–3046.
Inorganic nanoparticles for biomedicine: where materials scientists meet medical [193] D.H. Hwang, H.M. Kim, Y.M. Kang, I.S. Joo, C.-S. Cho, B.-W. Yoon, S.U. Kim, B.
research, Mater. Today 19 (1) (2016) 19–28. G. Kim, Combination of multifaceted strategies to maximize the therapeutic
[170] V. Neuschmelting, S. Harmsen, N. Beziere, H. Lockau, H.T. Hsu, R. Huang, benefits of neural stem cell transplantation for spinal cord repair, Cell Transplant.
D. Razansky, V. Ntziachristos, M.F. Kircher, Dual-modality surface-enhanced 20 (9) (2011) 1361–1379.
resonance Raman scattering and multispectral optoacoustic tomography [194] J.A. Kauhausen, L.H. Thompson, C.L. Parish, Chondroitinase improves midbrain
nanoparticle approach for brain tumor delineation, Small 14 (23) (2018), pathway reconstruction by transplanted dopamine progenitors in parkinsonian
1800740. mice, Mol. Cell. Neurosci. 69 (2015) 22–29.
[171] S.J. Amina, B. Guo, A REVIEW ON THE SYNTHESIS AND FUNCTIONALIZATION [195] M.C. Lee, K.L.P. Sung, M.S. Kurtis, W.H. Akeson, R.L. Sah, Adhesive force of
OF GOLD NANOPARTICLES AS A DRUG DELIVERY VEhicle, Int. J. Nanomed. 15 chondrocytes to cartilage: effects of chondroitinase ABC, Clin. Orthop. Relat. Res.
(2020) 9823–9857. 370 (2000) 286–294.
[172] M.S. Naderi, T.T. Moghadam, K. Khajeh, B. Ranjbar, Improving the stability of [196] C.H. Jo, E.M. Kim, H.J. Ahn, H.J. Kim, S.C. Seong, M.C. Lee, Degree of
chondroitinase ABC I via interaction with gold nanorods, Int J. Biol. Macromol. degeneration and chondroitinase ABC treatment of human articular cartilage
107 (Pt A) (2018) 297–304. affect adhesion of chondrocytes, Tissue Eng. 12 (1) (2006) 167–176.
[173] S. Stankic, S. Suman, F. Haque, J. Vidic, Pure and multi metal oxide nanoparticles: [197] J.-C. Lee, H.J. Min, S. Lee, S.C. Seong, M.C. Lee, Effect of chondroitinase ABC on
synthesis, antibacterial and cytotoxic properties, J. Nanobiotechnology 14 (1) adhesion and behavior of synovial membrane-derived mesenchymal stem cells in
(2016) 73. rabbit partial-thickness chondral defects, J. Orthop. Res. 31 (8) (2013)
[174] H. Askaripour, M. Vossoughi, K. Khajeh, I. Alemzadeh, Examination of 1293–1301.
chondroitinase ABC I immobilization onto dextran-coated Fe(3)O(4) [198] G.M. Ghobrial, K.D. Anderson, M. Dididze, J. Martinez-Barrizonte, G.H. Sunn, K.
nanoparticles and its in-vitro release, J. Biotechnol. 309 (2020) 131–141. L. Gant, A.D. Levi, Human Neural Stem Cell Transplantation in Chronic Cervical
[175] J. Gao, B. Xia, S. Li, L. Huang, T. Ma, X. Shi, K. Luo, Y. Yang, L. Zhao, H. Zhang, Spinal Cord Injury: Functional Outcomes at 12 Months in a Phase II Clinical Trial,
B. Luo, J. Huang, Magnetic field promotes migration of schwann cells with Neurosurg. 64(CN_suppl_1) (2017) 87–91.
chondroitinase ABC (ChABC)-loaded superparamagnetic nanoparticles across [199] A.J. Mothe, C.H. Tator, Review of transplantation of neural stem/progenitor cells
astrocyte boundary in vitro, Int J. Nanomed. 15 (2020) 315–332. for spinal cord injury, Int. J. Dev. Neurosci. 31 (7) (2013) 701–713.
[176] S. Daneshjou, B. Dabirmanesh, F. Rahimi, K. Khajeh, Porous silicon nanoparticle [200] R.J.M. Franklin, S.C. Barnett, Olfactory ensheathing cells and CNS regeneration:
as a stabilizing support for chondroitinase, Int J. Biol. Macromol. 94 (Pt B) (2017) the sweet smell of success? Neuron 28 (1) (2000) 15–18.
852–858. [201] G. Raisman, Olfactory ensheathing cells-another miracle cure for spinal cord
[177] H.A. Santos, E. Mäkilä, A.J. Airaksinen, L.M. Bimbo, J. Hirvonen, Porous silicon injury? Nat. Rev. Neurosci. 2 (5) (2001) 369–375.
nanoparticles for nanomedicine: preparation and biomedical applications, [202] I.D. Duncan, A.J. Aguayo, R.P. Bunge, P.M. Wood, Transplantation of rat
Nanomed. (Lond. ) 9 (4) (2014) 535–554. Schwann cells grown in tissue culture into the mouse spinal cord, J. Neurol. Sci.
[178] S. Daneshjou, B. Dabirmanesh, F. Rahimi, S. Jabbari, K. Khajeh, Catalytic 49 (2) (1981) 241–252.
parameters and thermal stability of chondroitinase ABCI on red porous silicon [203] M.H. Tuszynski, N. Weidner, M. McCormack, I. Miller, H. Powell, J. Conner,
nanoparticles, J. Biotechnol. 324 (2020) 83–90. Grafts of genetically modified Schwann cells to the spinal cord: survival, axon
[179] S. Rezaei, B. Dabirmanesh, L. Zare, A. Golestani, M. Javan, K. Khajeh, Enhancing growth, and myelination, Cell Transplant. 7 (2) (1998) 187–196.
myelin repair in experimental model of multiple sclerosis using immobilized [204] S.C. Barnett, A.-M. Hutchins, M. Noble, Purification of olfactory nerve
chondroitinase ABC I on porous silicon nanoparticles, Int J. Biol. Macromol. 146 ensheathing cells from the olfactory bulb, Dev. Biol. 155 (2) (1993) 337–350.
(2020) 162–170. [205] A. Sarveazad, A. Babahajian, M. Bakhtiari, M. Soleimani, B. Behnam, A. Yari,
[180] M.J. Grosso, V. Matheus, M. Clark, N. van Rooijen, C.A. Iannotti, M.P. Steinmetz, A. Akbari, M. Yousefifard, A. Janzadeh, N. Amini, The combined application of
Effects of an immunomodulatory therapy and chondroitinase after spinal cord human adipose derived stem cells and Chondroitinase ABC in treatment of a
hemisection injury, Neurosurgery 75 (4) (2014) 461–471. spinal cord injury model, Neuropeptides 61 (2017) 39–47.
[181] C.H. Chau, D.K.Y. Shum, H. Li, J. Pei, Y.Y. Lui, L. Wirthlin, Y.S. Chan, X.M. Xu, [206] H. Suzuki, C.S. Ahuja, R.P. Salewski, L. Li, K. Satkunendrarajah, N. Nagoshi,
Chondroitinase ABC enhances axonal regrowth through Schwann cell-seeded S. Shibata, M.G. Fehlings, Neural stem cell mediated recovery is enhanced by
guidance channels after spinal cord injury, FASEB J. 18 (1) (2004) 194–196. Chondroitinase ABC pretreatment in chronic cervical spinal cord injury, PloS One
[182] K. Fouad, L. Schnell, M.B. Bunge, M.E. Schwab, T. Liebscher, D.D. Pearse, 12 (8) (2017), e0182339.
Combining Schwann cell bridges and olfactory-ensheathing glia grafts with [207] T. Führmann, P.N. Anandakumaran, M.S. Shoichet, Combinatorial therapies after
chondroitinase promotes locomotor recovery after complete transection of the spinal cord injury: how can biomaterials help? Adv. Healthc. Mater. (2017).
spinal cord, J. Neurosci. 25 (5) (2005) 1169–1178. [208] M.B. Bunge, Novel combination strategies to repair the injured mammalian spinal
[183] Y.-S. Lee, C.-Y. Lin, H.-H. Jiang, M. DePaul, V.W. Lin, J. Silver, Nerve cord, J. Spinal Cord. Med. 31 (3) (2008) 262–269.
regeneration restores supraspinal control of bladder function after complete [209] S.H. Lee, Y. Kim, D. Rhew, M. Kuk, M. Kim, W.H. Kim, O.-K. Kweon, Effect of the
spinal cord injury, J. Neurosci. 33 (26) (2013) 10591–10606. combination of mesenchymal stromal cells and chondroitinase ABC on chronic
[184] V.J. Tom, H.R. Sandrow-Feinberg, K. Miller, L. Santi, T. Connors, M.A. Lemay, J. spinal cord injury, Cytotherapy 17 (10) (2015) 1374–1383.
D. Houlé, Combining peripheral nerve grafts and chondroitinase promotes [210] X. Chen, M. Katakowski, Y. Li, D. Lu, L. Wang, L. Zhang, J. Chen, Y. Xu,
functional axonal regeneration in the chronically injured spinal cord, J. Neurosci. S. Gautam, A. Mahmood, Human bone marrow stromal cell cultures conditioned
29 (47) (2009) 14881–14890. by traumatic brain tissue extracts: growth factor production, J. Neurosci. Res. 69
[185] A.S. Hunanyan, H.A. Petrosyan, V. Alessi, V.L. Arvanian, Combination of (5) (2002) 687–691.
chondroitinase ABC and AAV-NT3 promotes neural plasticity at descending spinal [211] S. Wu, Y. Suzuki, Y. Ejiri, T. Noda, H. Bai, M. Kitada, K. Kataoka, M. Ohta,
pathways after thoracic contusion in rats, J. Neurophysiol. 110 (8) (2013) H. Chou, C. Ide, Bone marrow stromal cells enhance differentiation of cocultured
1782–1792. neurosphere cells and promote regeneration of injured spinal cord, J. Neurosci.
[186] G. García-Alías, H.A. Petrosyan, L. Schnell, P.J. Horner, W.J. Bowers, L. Res. 72 (3) (2003) 343–351.
M. Mendell, J.W. Fawcett, V.L. Arvanian, Chondroitinase ABC combined with

16
A. Kheirollahi et al. Enzyme and Microbial Technology 172 (2024) 110348

[212] J.D. Houle, J.H. Ye, C.J.M. Kane, Axonal regeneration by chronically injured alongside rehabilitation enhances recovery after spinal cord injury, Exp. Neurol.
supraspinal neurons can be enhanced by exposure to insulin-like growth factor, 340 (2021), 113660.
basic fibroblast growth factor or transforming growth factor beta, Restor. Neurol. [234] B. Jevans, N.D. James, E. Burnside, C.J. McCann, N. Thapar, E.J. Bradbury, A.
Neurosci. 10 (4) (1996) 205–215. J. Burns, Combined treatment with enteric neural stem cells and chondroitinase
[213] J.D. Houle, J.H. Ye, Changes occur in the ability to promote axonal regeneration ABC reduces spinal cord lesion pathology, Stem Cell Res Ther. 12 (1) (2021), 10.
as the postâ€‫ﮒ‬injury period increases, Neuroreport 8 (3) (1997) 749–755. [235] V. Buzoianu-Anguiano, J. Rivera-Osorio, S. Orozco-Suárez, A. Vega-García,
[214] J.-H. Ye, J.D. Houle, Treatment of the chronically injured spinal cord with E. García-Vences, S. Sánchez-Torres, I. Jiménez-Estrada, G. Guizar-Sahagún,
neurotrophic factors can promote axonal regeneration from supraspinal neurons, J. Mondragon-Caso, F. Fernández-Valverde, I. Madrazo, I. Grijalva, Single vs.
Exp. Neurol. 143 (1) (1997) 70–81. Combined Therapeutic Approaches in Rats With Chronic Spinal Cord Injury, Front
[215] B.S. Bregman, E. Broude, M. McAtee, M.S. Kelley, Transplants and neurotrophic Neurol. 11 (2020) 136.
factors prevent atrophy of mature CNS neurons after spinal cord injury, Exp. [236] S. Nori, M. Khazaei, C.S. Ahuja, K. Yokota, J.E. Ahlfors, Y. Liu, J. Wang,
Neurol. 149 (1) (1998) 13–27. S. Shibata, J. Chio, M.H. Hettiaratchi, T. Führmann, M.S. Shoichet, M.G. Fehlings,
[216] D. Dolbeare, J.D. Houle, Restriction of axonal retraction and promotion of axonal Human Oligodendrogenic Neural Progenitor Cells Delivered with Chondroitinase
regeneration by chronically injured neurons after intraspinal treatment with glial ABC Facilitate Functional Repair of Chronic Spinal Cord Injury, Stem Cell Rep. 11
cell line-derived neurotrophic factor (GDNF), J. Neurotrauma 20 (11) (2003) (6) (2018) 1433–1448.
1251–1261. [237] T. Führmann, P.N. Anandakumaran, S.L. Payne, M.M. Pakulska, B.V. Varga,
[217] R.J. Colello, W.N. Chow, J.W. Bigbee, C. Lin, D. Dalton, D. Brown, B.S. Jha, B. A. Nagy, C. Tator, M.S. Shoichet, Combined delivery of chondroitinase ABC and
E. Mathern, K.D. Lee, D.G. Simpson, The incorporation of growth factor and human induced pluripotent stem cell-derived neuroepithelial cells promote tissue
chondroitinase ABC into an electrospun scaffold to promote axon regrowth repair in an animal model of spinal cord injury, Biomed. Mater. 13 (2) (2018),
following spinal cord injury, J. Tissue Eng. Regen. Med. 10 (8) (2016) 656–668. 024103.
[218] M.P. Steinmetz, K.P. Horn, V.J. Tom, J.H. Miller, S.A. Busch, D. Nair, D.J. Silver, [238] H. Chen, J. Li, H. Yan, The transplantation of human urine stem cells combined
J. Silver, Chronic enhancement of the intrinsic growth capacity of sensory with chondroitinase ABC promotes brain-derived neurotrophic factor and nerve
neurons combined with the degradation of inhibitory proteoglycans allows growth factor following spinal cord injury in rats, Int J. Clin. Exp. Pathol. 11 (8)
functional regeneration of sensory axons through the dorsal root entry zone in the (2018) 3858–3866.
mammalian spinal cord, J. Neurosci. 25 (35) (2005) 8066–8076. [239] J.H. Wu, M. Li, Y. Liang, T. Lu, C.Y. Duan, Migration of Adipose-derived
[219] F. Bai, H. Peng, J.D. Etlinger, R.J. Zeman, Partial functional recovery after Mesenchymal Stem Cells Stably Expressing Chondroitinase ABC In vitro, Chin.
complete spinal cord transection by combined chondroitinase and clenbuterol Med J. (Engl. ) 129 (13) (2016) 1592–1599.
treatment, Pfl‫¼ﺃ‬gers Arch. -Eur. J. Physiol. 460 (3) (2010) 657–666. [240] H.J. Lee, S. Bian, I. Jakovcevski, B. Wu, A. Irintchev, M. Schachner, Delayed
[220] R. Harel, C.A. Iannotti, D. Hoh, M. Clark, J. Silver, M.P. Steinmetz, Oncomodulin applications of L1 and chondroitinase ABC promote recovery after spinal cord
affords limited regeneration to injured sensory axons in vitro and in vivo, Exp. injury, J. Neurotrauma 29 (10) (2012) 1850–1863.
Neurol. 233 (2) (2012) 708–716. [241] Y.G. Yang, D.M. Jiang, Z.X. Quan, Y.S. Ou, Insulin with chondroitinase ABC treats
[221] G. García-Alías, S. Barkhuysen, M. Buckle, J.W. Fawcett, Chondroitinase ABC the rat model of acute spinal cord injury, J. Int Med Res 37 (4) (2009) 1097–1107.
treatment opens a window of opportunity for task-specific rehabilitation, Nat. [242] A. Sarveazad, M. Bakhtiari, A. Babahajian, A. Janzade, A. Fallah, F. Moradi,
Neurosci. 12 (9) (2009) 1145–1151. M. Soleimani, M. Younesi, F. Goudarzi, J. Mohammad, Taghi, Comparison of
[222] M. Shinozaki, A. Iwanami, K. Fujiyoshi, S. Tashiro, K. Kitamura, S. Shibata, human adipose-derived stem cells and chondroitinase ABC transplantation on
H. Fujita, M. Nakamura, H. Okano, Combined treatment with chondroitinase ABC locomotor recovery in the contusion model of spinal cord injury in rats, Iran. J.
and treadmill rehabilitation for chronic severe spinal cord injury in adult rats, Basic Med Sci. 17 (9) (2014) 685–693.
Neurosci. Res. 113 (2016) 37–47. [243] J.M. Massey, J. Amps, M.S. Viapiano, R.T. Matthews, M.R. Wagoner, C.
[223] O. Alluin, H. Delivet-Mongrain, M.K. Gauthier, M.G. Fehlings, S. Rossignol, M. Whitaker, W. Alilain, A.L. Yonkof, A. Khalyfa, N.G. Cooper, J. Silver, S.
S. Karimi-Abdolrezaee, Examination of the combined effects of chondroitinase M. Onifer, Increased chondroitin sulfate proteoglycan expression in denervated
ABC, growth factors and locomotor training following compressive spinal cord brainstem targets following spinal cord injury creates a barrier to axonal
injury on neuroanatomical plasticity and kinematics, PLoS One 9 (10) (2014), regeneration overcome by chondroitinase ABC and neurotrophin-3, Exp. Neurol.
e111072. 209 (2) (2008) 426–445.
[224] T. Oertle, M.E. van der Haar, C.E. Bandtlow, A. Robeva, P. Burfeind, A. Buss, A. [244] M.P. Côté, A. Hanna, M.A. Lemay, K. Ollivier-Lanvin, L. Santi, K. Miller,
B. Huber, M. Simonen, L. Schnell, C. Brösamle, Nogo-A inhibits neurite outgrowth R. Monaghan, J.D. Houlé, Peripheral nerve grafts after cervical spinal cord injury
and cell spreading with three discrete regions, J. Neurosci. 23 (13) (2003) in adult cats, Exp. Neurol. 225 (1) (2010) 173–182.
5393–5406. [245] W.C. Huang, W.C. Kuo, S.H. Hsu, C.H. Cheng, J.C. Liu, H. Cheng, Gait analysis of
[225] M.E. Schwab, Nogo and axon regeneration, Curr. Opin. Neurobiol. 14 (1) (2004) spinal cord injured rats after delivery of chondroitinase ABC and adult olfactory
118–124. mucosa progenitor cell transplantation, Neurosci. Lett. 472 (2) (2010) 79–84.
[226] M.S. Chen, A.B. Huber, M.E. van der Haar, M. Frank, L. Schnell, A.A. Spillmann, [246] S.H. Lee, Y. Kim, D. Rhew, A. Kim, K.R. Jo, Y. Yoon, K.U. Choi, T. Jung, W.H. Kim,
F. Christ, M.E. Schwab, Nogo-A is a myelin-associated neurite outgrowth inhibitor O.K. Kweon, Impact of local injection of brain-derived neurotrophic factor-
and an antigen for monoclonal antibody IN-1, Nature 403 (6768) (2000) expressing mesenchymal stromal cells (MSCs) combined with intravenous MSC
434–439. delivery in a canine model of chronic spinal cord injury, Cytotherapy (2016).
[227] Y. Zhang, Z. Gu, G. Qiu, Y. Song, Combination of chondroitinase ABC, glial cell [247] L.W. Yick, K.F. So, P.T. Cheung, W.T. Wu, Lithium chloride reinforces the
line-derived neurotrophic factor and Nogo A antibody delayed-release regeneration-promoting effect of chondroitinase ABC on rubrospinal neurons
microspheres promotes the functional recovery of spinal cord injury, J. Craniofac after spinal cord injury, J. Neurotrauma 21 (7) (2004) 932–943.
Surg. 24 (6) (2013) 2153–2157. [248] T.S. Wilems, J. Pardieck, N. Iyer, S.E. Sakiyama-Elbert, Combination therapy of
[228] A. Janzadeh, A. Sarveazad, M.R. Hamblin, G. Teheripak, K. Kookli, stem cell derived neural progenitors and drug delivery of anti-inhibitory
F. Nasirinezhad, The effect of chondroitinase ABC and photobiomodulation molecules for spinal cord injury, Acta Biomater. 28 (2015) 23–32.
therapy on neuropathic pain after spinal cord injury in adult male rats, Physiol. [249] D. Wu, M.C. Klaw, T. Connors, N. Kholodilov, R.E. Burke, M.-P. Côté, V.J. Tom,
Behav. 227 (2020), 113141. Combining constitutively active rheb expression and chondroitinase promotes
[229] A. Kheirollahi, K. Khajeh, A. Golestani, Investigating the role of loop 131-140 in functional axonal regeneration after cervical spinal cord injury, Mol. Ther. 25
activity and thermal stability of chondroitinase ABC I, Elsevier,, 2018, (12) (2017) 2715–2726.
pp. 811–816. [250] Z. Wang, K. Winsor, C. Nienhaus, E. Hess, M.G. Blackmore, Combined
[230] M.H. Hettiaratchi, M.J. O’Meara, T.R. O’Meara, A.J. Pickering, N. Letko-Khait, M. chondroitinase and KLF7 expression reduce net retraction of sensory and CST
S. Shoichet, Reengineering biocatalysts: Computational redesign of axons from sites of spinal injury, Neurobiol. Dis. 99 (2017) 24–35.
chondroitinase ABC improves efficacy and stability, Sci. Adv. 6 (34) (2020), [251] Q. Pan, Y. Guo, F. Kong, Poly (glycerol sebacate) combined with chondroitinase
eabc6378. ABC promotes spinal cord repair in rats, J. Biomed. Mater. Res. Part B: Appl.
[231] H. Wang, L. Zhang, Y. Wang, J. Li, G. Du, Z. Kang, Engineering a thermostable Biomater. (2017).
chondroitinase for production of specifically distributed low-molecular-weight [252] S.-W. Hong, B.-T. Kim, H.-Y. Shin, W.-S. Kim, K.-S. Lee, Y.-S. Kim, D.-H. Kim,
chondroitin sulfate, Biotechnol. J. 16 (5) (2021), e2000321. Purification and characterization of novel chondroitin ABC and AC lyases from
[232] D. Carwardine, J. Prager, J. Neeves, E.M. Muir, J. Uney, N. Granger, Bacteroides stercoris HJ-15, a human intestinal anaerobic bacterium, FEBS J. 269
Transplantation of canine olfactory ensheathing cells producing chondroitinase (12) (2002) 2934–2940.
ABC promotes chondroitin sulphate proteoglycan digestion and axonal sprouting [253] A.A. Salyers, M. O’Brien, Cellular location of enzymes involved in chondroitin
following spinal cord injury, PLoS ONE 12 (12) (2017), e0188967. sulfate breakdown by Bacteroides thetaiotaomicron, J. Bacteriol. 143 (2) (1980)
[233] J. Prager, D. Ito, D.R. Carwardine, P. Jiju, D.M. Chari, N. Granger, L.F. Wong, 772–780.
Delivery of chondroitinase by canine mucosal olfactory ensheathing cells

17

You might also like