You are on page 1of 16

Advanced Drug Delivery Reviews 192 (2023) 114671

Contents lists available at ScienceDirect

Advanced Drug Delivery Reviews


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

Role of wound microbiome, strategies of microbiota delivery system and


clinical management
Qinghan Tang a,1, Nannan Xue a,b,1, Xiaofeng Ding c,d, Kevin H.-Y. Tsai e, Jonathan J. Hew f, Ruihan Jiang a,
Rizhong Huang a, Xuxi Cheng a, Xiaotong Ding a, Yuen Yee Cheng g, Jun Chen a,b,⇑, Yiwei Wang a,b,e,⇑
a
Jiangsu Provincial Engineering Research Center of TCM External Medication Development and Application, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
b
Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
c
Department of Burns and Plastic Surgery, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine,
Nanjing 210023, PR China
d
Department of Plastic Surgery, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai 200434, PR China
e
Burns and Reconstructive Surgery Group, ANZAC Research Institute, Concord Hospital, The University of Sydney, Concord West, NSW 2137, Australia
f
General Surgery, Lesimore Hospital, NSW 2480, Australia
g
Institute for Biomedical Materials and Devices, School of Mathematical and Physical Sciences, University of Technology Sydney, NSW 2007 Australia

a r t i c l e i n f o a b s t r a c t

Article history: Delayed wound healing is one of the most global public health threats affecting nearly 100 million people
Received 27 September 2022 each year, particularly the chronic wounds. Many confounding factors such as aging, diabetic disease,
Revised 23 November 2022 medication, peripheral neuropathy, immunocompromises or arterial and venous insufficiency hypergly-
Accepted 14 December 2022
caemia are considered to inhibit wound healing. Therapeutic approaches for slow wound healing include
Available online 17 December 2022
anti-infection, debridement and the use of various wound dressings. However, the current clinical out-
comes are still unsatisfied. In this review, we discuss the role of skin and wound commensal microbiota
Keywords:
in the different healing stages, including inflammation, cell proliferation, re-epithelialization and remod-
Wound healing
Skin microbiome
elling phase, followed by multiple immune cell responses to commensal microbiota. Current clinical
Inflammation management in treating surgical wounds and chronic wounds was also reviewed together with potential
Clinical wound care controlled delivery systems which may be utilized in the future for the topical administration of probi-
Cell response to microbiome otics and microbiomes. This review aims to introduce advances, novel strategies, and pioneer ideas in
Probiotic delivery regulating the wound microbiome and the design of controlled delivery systems.
Ó 2022 Elsevier B.V. All rights reserved.

Abbreviations: 16S-rDNA, 16S ribosomal DNA identification; b2-AR, b2-adrenergic receptor; ADAM10, A disintegrin and metalloprotease 10; AHR, aryl hydrocarbon
receptor; AMP, adenosine monophosphate; B. subtilis, Bacillus subtilis; C. albicans, Candida albicans; CSF, colony-stimulating factor; CXCL2, chemokine (C-X-C motif) ligand 2;
CXCL10, chemokine (C-X-C motif) ligand 10; CXCL12, chemokine (C-X-C motif) ligand 12; DFU, diabetic foot ulcer; ECM, extracellular matrix; E. coli, Escherichia coli; EPB,
epidermal permeability barrier; EVs, extracellular vesicles; FDA, Food and Drug Administration; hBD, human b-defensin; HF, Hair follicle; HMP, Human Microbiome Project;
IL-1, interleukin-1; IL-1R, interleukin-1R; IL-1b, interleukin-1b; IL-6, interleukin-6; IL-23, interleukin-23; K. pneumoniae, Klebsiella pneumoniae; L. acidophilus, Lactobacillus
acidophilus; L. lactis, Lactococcus lactis; L. plantarum, Lactobacillus plantarum; L. reuteri, Lactobacillus reuteri; L. rhamnosus, Lactobacillus rhamnosus; M CSF, macrophage colony-
stimulating factor; MHCII, major histocompatibility complex II; MMP-9, matrix metalloproteinase-9; MRSA, methicillin-resistant Staphylococcus aureus; MYD88, myeloid
differentiation factor 88; NETs, neutrophil extracellular traps; NIH, National Institutes of Health; NPWT, negative pressure wound therapy; P. aeruginosa, Pseudomonas
aeruginosa; pDCs, plasmacytoid dendritic cells; PEO, polyethylene oxide; PLA, polylactic acid; ROS, reactive oxygen species; PRP, Plat-rich plasma; PVA, polyvinyl alcohol; PVP,
polyvinyl pyrrolidone; SadA, serum adenosine deaminase; S. aureus, Staphylococcus aureus; Sbi, staphylococcal immunoglobulin-binding protein; SCMC, sodium
carboxymethylcellulose; S. epidermidis, Staphylococcus epidermidis; SLO, streptolysin O; SpA, Staphylococcal protein A; SPF, specific pathogen-free; SSWI, surgical site wound
infection; TAs, trace amines; Tc17, cytotoxic T cells 17; TGF-b1, transforming growth factor-b1; TNF-a, tumor necrosis factor-a; VEGF, vascular endothelial growth factor;
WHO, World Health Organisation.
⇑ Corresponding authors at: Jiangsu Provincial Engineering Research Center of TCM External Medication Development and Application, Nanjing University of Chinese
Medicine, 138 Xianlin Avenue, Nanjing 210023, PR China.
E-mail addresses: chenjun75@163.com, chenjun75@njucm.edu.cn (J. Chen), yiweiwang@njucm.edu.cn (Y. Wang).
1
These authors contributed equally to these work.

https://doi.org/10.1016/j.addr.2022.114671
0169-409X/Ó 2022 Elsevier B.V. All rights reserved.
Q. Tang, N. Xue, X. Ding et al. Advanced Drug Delivery Reviews 192 (2023) 114671

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2. Skin microbiome and wound healing . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2.1. Skin microbiome . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2.2. Wound healing . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
2.3. Role of wound microbiome on the inflammation stage . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
2.4. Effect of wound microbiome on cell proliferation. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
2.5. Effect of wound microbiome on re-epithelialization. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
2.6. Wound microbiome and remodelling phase . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
3. Immune cell response to bacterial . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
3.1. Neutrophils . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
3.2. Monocytes and macrophages . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
3.3. Lymphocytes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
4. Clinical management in regulating wound microbiome . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
4.1. Role of commensal microbiota on surgical wounds . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
4.2. Clinical management in treating chronic wounds. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
5. Strategies in controlled delivery of probiotics and microbiome . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
5.1. Microencapsulation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10
5.2. Electrospun scaffolds. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11
5.3. Hydrogels and 3D bioprinting . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11
5.4. Microneedles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12
6. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12
Declaration of Competing Interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12
Acknowledgment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12

1. Introduction after born [5]. Approximately 1,000 species of known bacteria and
about 100 billion microbiomes are detectable on a person’s skin
Delayed wound healing has become a global public health prob- [6]. Bacteria (2 million/cm2) on human skin [5] can be divided into
lem, particularly in chronic wounds. According to the World Health four phyla: Actinobacteria (51.8 %), Firmicutes (24.4 %), Proteobac-
Organisation (WHO), over 6 million people are affected by delayed teria (16.5 %), and Bacteroidetes (6.3 %), including three most com-
wound repair every year in the United States with a cost of $25 bil- mon genera: Corynebacterium, Propionibacterium, and
lion (USD) in the health system [1]. About 100 million people need Staphylococcus [7]. This data suggests that bacterial colonies are
wound care every year, and about 30 million people with complex more abundant on the surface of the skin compared to any other
and refractory chronic skin wounds require further intervention epithelium [8]. Human skin can be seen as a culture medium,
[2]. All types of wounds or injuries initiate the healing process, and its composition is greatly influenced by genetics, diet, lifestyle
which consists of several highly integrated and overlapping and the living environment [6]. Therefore, each site of human skin
phases: inflammation, cell recruitment, matrix deposition, epithe- is unique in terms of the composition of the microbes, while resi-
lialization and tissue remodelling. Due to confounding factors such dent microbes in specific areas on the skin are also known to be
as aging, stage of diabetic disease, medication (or treatment), identical at the genus level [9–10]. The composition and abun-
peripheral neuropathy, immunocompromised status and/or arte- dance of different microbes vary widely across individuals and
rial and venous insufficiency hyperglycaemia, chronic wounds over time, forming a dynamic and highly variable microbiome
are at high risk of delayed healing [3]. At present, the therapeutic [5]. A Human Microbiome Project (HMP), led by the National Insti-
approach for the treatment of delayed wound healing is anti- tutes of Health (NIH), was conducted to characterize microbial
infection and debridement or the use of various wound dressing populations in five major body sites, including the oral cavity, skin,
and dermal templates, but the clinical outcomes are unsatisfactory. nasal cavity, gastrointestinal tract, and genitourinary tract [11].
Therefore, advanced approaches are urgently required by clini- Data from this project showed that microbiomes are able to control
cians. Recent studies demonstrated that regulation of the wound signalling pathways related to body metabolism, especially those
microbiome can promote diabetic wound healing. Few pioneer with diseases in different body sites. In addition, thousands of
studies reported that increased diversity of wound microbiome microbiomes (bacteria, viruses, fungi) were identified living on
and suppression of Staphylococcus aureus (S. aureus) can signifi- the skin surface, which has tremendous positive effects on human
cantly promote the wound repair [4]. In the present article, we dis- survival [11].
cuss the skin microbiome, the effect of the microbiome on different Skin is the first barrier of the human body while the formation
stages of the healing process, immune cell responses to the wound of a wound provides an opportunity for commensal microbiota on
microbiome and clinical treatment with a focus on advances and the skin surface to migrate to the underlying tissue with coloniza-
novel strategies in the future design of a controlled delivery system tion and growth [12–13]. During the healing process, cell interac-
for probiotics and microbiomes. tion with the wound microbiome is hypothesized to be beneficial
in regulating the innate immune response [13]. In contrast, patho-
2. Skin microbiome and wound healing genic microbiota is known to play a negative role in mediating the
wound healing process [14–15]. According to statistics, wound
2.1. Skin microbiome contains diverse microbiota, the main components of which are
Staphylococcus, Pseudomonas, Corynebacterium, Streptococcus,
Human skin is exposed to maternal and environmental bacteria Anaerobic Coccus, and Enterococcus, as well as other genera with
and other microbes (e.g. viruses, fungi and protozoa) immediately lower abundance [3]. Chronic wounds generally contain more

2
Q. Tang, N. Xue, X. Ding et al. Advanced Drug Delivery Reviews 192 (2023) 114671

microbes than acute wounds [3]. Over 2900 samples collected from of all microbial isolates. S. aureus is the most commonly found
various wounds were subjected to multiple microbiological analy- pathogen among them, followed by P. aeruginosa [3]. Wound bac-
ses, while Staphylococcus and Pseudomonas were found to be the teria, such as S. aureus, P. aeruginosa, Corynebacterium striatum, and
most common species, accounting for 63 % and 25 %, respectively Alcaligenes faecalis were detected in patients with diabetic foot
[16]. Moreover, a study proved that there is an association between ulcers [33]. In a study of 145 patients with pressure ulcers, S. aur-
microbiota composition and clinical factors [17]. Higher microbial eus and Gram-negative bacilli were detected in 112 wounds [34].
diversity was observed in deeper ulcers and ulcers of longer dura-
tion, as well as increased anaerobic and the relative abundance of
Gram-negative Proteobacteria [18]. In contrast, many Staphylococci,
mainly S. aureus, were found in superficial and short-term ulcers, 2.3. Role of wound microbiome on the inflammation stage
suggesting that bacterial species are correlated with the host
pathophysiology. Notably, commensal bacteria, including Immediately after injury, the wound initiates an inflammatory
Corynebacterium and Propionibacterium, as well as anaerobes were response that mediates pathogen clearance and stimulates restora-
prevalent in chronic wounds [19]. Taken together, a stable equilib- tion of the skin barrier integrity [35]. However, in a chronic wound,
rium between wound microbiotas and the host is believed to play these inflammatory responses are either absent or hyperactive and
an important role in wound repair and the healing process. in a state of dysregulated inflammation, resulting in abnormal tis-
Although the role of the skin microbiota on wound healing has sue formation and long-term defects in the epidermal barrier [14].
been comprehensively investigated [20], conclusions remain con- Despite the robust diversity of the skin microbiome, understanding
troversial. Few pioneer studies found that the wound microbiota how skin microbiota regulates the inflammatory response over the
may contribute to the composition and overall toxicity of the inflammation stage has important implications for the develop-
wound microbiome through forming biofilms in chronic wounds. ment of therapeutic and preventive strategies (Fig. 1).
Changes in force may lead to infection or hinder wound healing In 2020, a study reported for the first time the beneficial role of
[21]. In the past, scientists and clinicians all agreed that S. epider- the symbiotic skin microbiota and its correlation with the inflam-
midis positively affects healing, but S. aureus seems to be a villain matory response [36]. When the skin is injured, researchers found
[6]. Multiple studies have shown that modulating wound microbes that skin microbiota, consisting of commensal Staphylococci and
can improve wound healing, with significant positive implications Micrococci, migrated from the epidermis to the dermis of the
for human survival [20]. These new findings open a new avenue to injured skin, triggering the activation of neutrophils with the
explore microbes’ role and underlying mechanisms in the wound expression of CXC chemokine ligand-10 (CXCL10) (Fig. 1). There-
healing process. after, CXCL10 activates plasmacytoid dendritic cells (pDC) by form-
ing complexes with bacterial DNA, while activated pDC produce
2.2. Wound healing large amounts of type I interferon, which accelerate wound closure
via triggering inflammation and early T cell-independent
Wound healing is a dynamic and highly coordinated process, responses, mediated by macrophages and fibroblasts that produce
mainly including the haemostatic phase, inflammatory phase, cell major growth factors required for healing. In addition, the pro-
proliferation phase (formation of granulation tissue vasculariza- duced interferon induces early wound repair responses and accel-
tion and wound closure) and tissue remodelling phase [22–23]. A erates wound healing by stimulating the growth of fibroblasts [36].
variety of inflammatory cells, cell cytokines, inflammatory media- A recent study further revealed that the A20 clade of the skin com-
tors and extracellular matrix are involved in the healing process mensal S. epidermidis could induce adaptive immunity with both
[24–25]. Normally, wound healing can be divided into acute or anti-infection and tissue repair functions [37]. S. epidermidis was
chronic wound healing [26]. The primary difference between acute capable of stimulating the recruitment of a special CD8+ T cell
and chronic wounds is the length required for healing [27], in (Fig. 1), directly killing pathogens together with enhanced re-
which acute wounds can heal in 4 weeks, but chronic wounds have epithelialization and keratinocyte migration. The immune
significantly delayed healing over months or even years [28]. response elicited by S. epidermidis is found via the non-specific
The healing rate is highly related to injury types, severity, size of CD8 + MHCIb pathway, but not the CD4 + MHCII pathway [37]
the wounds aging, co-morbidities as well as post-injury care [3]. (Table 1).
Acute trauma has different wound microbiota composition. Studies By now, only a few specific microbiotas have been investigated
found that the most common pathogens in the acute phase post- in immunology or inflammatory processes [20]. In addition to Sta-
burns are S. aureus, followed by Escherichia coli (E. coli), Pseu- phylococcus, Corynebacterium, a commensal of mouse and human
domonas aeruginosa (P. aeruginosa), and coagulase-negative Staphy- skin microbiota, was reported to activate the skin immune system
lococci, etc [29–30]. Over surgery, microbiomes on sutures or in an IL-23-dependent manner [38]. Such a system depends on the
prostheses can induce wound infection and cause acute surgical expression of mycolic acid, a significant component of the coryne-
site wound infections (SSWIs). The most common pathogenic form cell wall. The inflammatory effect of microbiomes also
microbiotas involved in SSWIs are S. epidermidis and S. aureus, depends on the physiological state of the host, and the skin micro-
but E. coli, Clostridium difficile, and coagulase-negative Staphylococci biota is triggered by the host’s dietary changes, obesity, or meta-
also occur to a lesser extent [31]. bolic state to stimulate inflammation or immune responses [38].
Wounds that do not heal after 1 month of treatment are gener- Although S. aureus is the most prominent cause SSWIs worldwide,
ally considered chronic wounds. Chronic wound formation is com- studies have shown that S. aureus surface proteins A (SpA) and
plex and more likely to occur in the elderly patients with vascular staphylococcal immunoglobulin-binding protein (Sbi) induce IL-
disease, diabetes, or a combination of these factors compared to 1b and IL-6 at the initial stage of S. aureus infection and promote
acute wounds [13]. With the increased elderly population, chronic the expression of TNF-a and CXCL-1. Expression of IL-10 in wounds
wounds have become a major burden on healthcare worldwide induced by SpA and Sbi not only contributes to the recruitment of
[32]. The main cause of chronic wound infections and non- neutrophils but also extends the time of immune cells such as neu-
curative wounds is that the wound surface is colonized by polymi- trophils and macrophages recruited to the site of infection (Fig. 2).
crobial communities, leading to the formation of biofilms [17]. SpA is also known to aid the bacterial clearance by neutrophil
Generally, Gram-negative bacteria compose common colonizing extracellular traps (NET) (Table 1), thereby contributing to proper
microbiotas around the chronic wound area, accounting for 61 % abscess formation and inflammatory responses [39].
3
Q. Tang, N. Xue, X. Ding et al. Advanced Drug Delivery Reviews 192 (2023) 114671

Fig. 1. Microbiota regulates the inflammatory response and the related signalling pathways.

2.4. Effect of wound microbiome on cell proliferation epithelial cell differentiation via AHR and promoted the restoration
of skin barrier function compared with mice not colonized or trea-
The inflammatory phase is followed by a cell proliferation ted with antibiotics (Table 1).
phase, in which fibroblasts, keratinocytes, and endothelial cells
migrate and proliferate, promoting angiogenesis and synthesis of 2.5. Effect of wound microbiome on re-epithelialization
new extracellular matrix (ECM) [40]. The transition from the
inflammatory phase to the proliferative phase is a critical step in Re-epithelialization is the process of re-build/reformation of
the healing process. Thus, the poor transition is associated with the external surface of wound epithelium via keratinocyte migra-
impaired wound repair. tion from the wound edge to the centre area. Re-epithelialization
S. epidermidis is closely associated with keratinocytes as an mainly relies on keratinocyte migration, in which keratinocytes
antibacterial defender with increased wound healing rate [41]. S. are the most predominant cell type in the epidermis. Many bac-
epidermidis was reported to recruit IL-17A+ CD8+ T (Tc17) cells teria on human skin can convert aromatic amino acids into so-
infiltrating into the epidermis with enhanced innate barrier immu- called trace amines (TAs) [44] (Fig. 2). When the wound occurs,
nity via upregulating toll-like receptors and limiting the pathogen in response to stress, the damaged epithelial cells release epi-
invasion [41]. Upregulated TNF-a gene expression promotes rapid nephrine while b2-adrenergic receptor (b2-AR) in keratinocytes
cell proliferation of keratinocytes, thereby accelerating the healing is activated to hinder the migration of keratinocytes and reduce
rate (Table 1). Interestingly, S. epidermidis-induced Tc17 cells collagen production [45]. TAs are known as b2-AR antagonists
highly expressed a series of key factors involved in angiogenesis, that can attenuate the effects of epinephrine, resulting in acceler-
extracellular matrix production, and tissue remodelling [37] ated wound healing [44]. The serum adenosine deaminase (SadA)
(Fig. 2). Recent studies have shown that skin microbes can also gene encodes an aromatic amino acid decarboxylase that converts
act as a facilitator by activating biosensors on the skin, such as aromatic amino acids into TAs, such as tryptamine, phenethy-
the aryl hydrocarbon receptor (AHR) in keratinocytes to upregulate lamine, and tyramine [46]. Additionally, SadA homologs were
gene expressions that are involved in cell proliferation, differenti- found in at least 7 phyla in the human skin microbiota, including
ation, and inflammation [42–43]. AHR plays a crucial role in skin Firmicutes, Actinobacteria, Proteobacteria, Bacteroides Acidobac-
homeostasis and the skin development, function, and integrity of teria, Chloroflexi and Cyanobacteria [47]. Taking S. epidermidis
the epidermal permeability barrier. A comprehensive study with SadA gene as an example, it can increase the migration rate
showed that commensal flora is required for the formation of the of keratinocytes and ultimately accelerate wound healing
skin barrier and that skin epithelial cell development and differen- (Table 1).
tiation are impaired in germ-free mice [43]. When microbiota, The skin microbiome also plays a role in strengthening the
including S. epidermidis, Staphylococcus warneri, Staphylococcus physical barrier to skin integrity. Keratinocytes and the micro-
hemolyticus, Micrococcus luteus and Corynebacterium aurimucosum, biome produce sphingomyelinase, which processes lamellar lipids
isolated from normal human skin were transplanted into germ- into ceramides, which are important molecules in maintaining skin
free mouse skin, engraftment of commensal microbiota regulated structure and function [48]. Using a mouse model of impaired skin
4
Q. Tang, N. Xue, X. Ding et al. Advanced Drug Delivery Reviews 192 (2023) 114671

Table 1
Summary of wound microbiome effects in wound healing phases.

Wound healing phase Strains/proteins Wound model Mechanism References


Inflammatory Commensal Staphylococci and Tape stripping wound Activates neutrophils, produce type I interferons, and [36]
Micrococci (S. epidermidis, S. and full-thickness stimulates the growth of fibroblasts and macrophages.
xylosus, S. saprophiticus and wound of C57BL/6 mice
M. luteus,)
S. epidermidis Punch biopsy wound in Stimulate recruitment of CD8+ T cells and Up-regulated the [37]
the ear pinnae and back expression levels of various immune and tissue repair-
skin of C57BL/6 mice related genes.

Staphylococcal surface proteins BALB/c mice skin  Promotes the secretion of IL-1b, IL-6, TNF-a and CXCL-1, [39,52]
(SpA) and the Staphylococcal infected with and IL-10 in skin-infected tissues;
immunoglobulin binding protein Staphylococci  Contributes to the recruitment of neutrophils, extends the
(Sbi) time of neutrophils and macrophages recruiting to the site
of infection;
 Aid the clearance of bacterial contamination by NETs.

Cell proliferation S. epidermidis Ear pinnae of BALB/c  Recruit IL-17A+ CD8+ T cells and express key factors [41]
mice involved in angiogenesis, extracellular matrix production
and tissue remodelling, by upregulating toll-like
receptors;
 Up-regulated TNF-a gene promotes rapid cell
proliferation of keratinocytes.
S. epidermidis, S. warneri, S. Tape stripping wound of Activates aryl hydrocarbon receptors, promotes epithelial [42–43]
hemolyticus, M. luteus and C. germ-free C57BL6/J cell differentiation.
aurimucosum mice;
Epidermal keratinocytes
exposed to high calcium
media
Re-epithelialization Firmicutes, Actinobacteria, Full-thickness wounds Produces trace amines, antagonizes b2-adrenergic [44,46–47]
Proteobacteria, Bacteroides, of DDY mice receptors, increases keratinocyte migration.
Acidobacteria, Chloroflexi and
Cyanobacteria
A. faecalis and C. striatum Full-thickness Promote epithelial regeneration. [15]
excisional wounds of
db/db mice
Haemolytic exotoxin produced by Human skin organ  Stimulates keratinocyte migration; [49]
Streptococcus culture wound  Activate CD44 expression in keratinocytes in vitro and
promote the assembly of hyaluronan-rich ECMs.
S. epidermidis and low abundance Keratinocytes exposed Trigger keratinocytes to produce b-defensin and CAP18 [50,53]
of S. aureus to S. aureus against Staphylococcus aureus.
Tissue remodelling S. aureus Full thickness excisional Activates the IL-1b-IL-1R-MYD88 signalling pathway to [4]
wounds of C57BL/6 trigger hair follicle regeneration.
mice

barrier function, Staphylococcus colonization on the surface of 2.6. Wound microbiome and remodelling phase
mouse skin was reported to have less transdermal water loss com-
pared to the animals not treated with S. epidermidis [15,48]. This When the wound is completely closed, remodelling occurs
finding provides new insights into the underlying mechanisms of when the newly formed epithelial cells constantly divide, thicken-
how the skin microbiome interacts with the host skin. ing the epidermis. There is no direct evidence showing that the
In addition to the direct beneficial effects of wound microbiotas microbiome in wound healing regulates collagen deposition. How-
on wound healing, the bacterial toxins secreted also play a specific ever, skin bacteria were recently reported to have a direct correla-
role [49] (Table 1). Streptococcus pyogenes is known for causing tion with the hair follicle (HF) neogenesis [4] (Fig. 2). Scientists
superficial infections and invasive diseases, and streptolysin O found that germ-free mice have less ability to regenerate hair fol-
(SLO) is a hallmark toxin secreted by Streptococcus pyogenes at licles. In contrast, mice with skin commensal microbiota can trig-
low concentrations (0.02–0.2 U /mL). SLO promotes wound healing ger the regeneration of hair follicles via activation of the
in vitro by stimulating the migration of keratinocytes. Such a cellu- interleukin-1 receptor (IL-1R) - MYD88 signalling pathway [4,51].
lar mechanism via SLO may be explained via maintaining the Furthermore, recent data showed that the transmembrane
expression of the hyaluronic acid receptor, CD44, in keratinocytes, endopeptidase ADAM10-Notch signalling pathway in HF is an
which benefits the composition of the hyaluronic acid-rich ECM essential regulator of the microbiota in the hair follicle opening
through the formation of CD44-hyaluronic acid complexes wound region. Disruption of the DAM10-Notch signalling pathway leads
healing process [49]. Both S. epidermidis and typical low abundance to upregulation of inflammation-related genes’ expressions, and
S. aureus can stimulate human keratinocytes to express antimicro- inflammatory alopecia, resulting in dysbiosis of the Corynebac-
bial peptide (AMPs), protecting skin against the invasion of patho- terium-dominated flora, causing hair follicle cell death and irre-
genic microbiotas [50] (Fig. 2). Human b-defensin (hBD) is a small versible hair loss. ADAM10-Notch signalling can enhance innate
molecule cationic antimicrobial peptide, which is expressed and epithelial immunity by promoting type I interferon responses
uniformly distributed in all human epithelial cells, supporting ker- downstream of b-defensin-6. Therefore, the host-microbe symbio-
atinocyte differentiation together with recognition of microbiotas, sis balance mediated by the ADAM10-Notch signalling axis pro-
AMP has thus emerged as a new target for accelerating wound tects HF from inflammatory destruction and has clinical
healing. implications for maintaining the tissue integrity [4] (Table 1).

5
Q. Tang, N. Xue, X. Ding et al. Advanced Drug Delivery Reviews 192 (2023) 114671

Fig. 2. A summary of key bacteria and factors involved in different stages of the wound healing process.

3. Immune cell response to bacterial In-depth studies have found that the ability of bacteria to
induce NETs varies by bacterial species, while the most potent
Multiple immune cells are required to achieve tissue regenera- inducer of NETs is S. aureus [62–63]. However, S. aureus has mul-
tion and homeostasis post-injury, including innate and adaptive tiple evasion strategies against the human immune system, such
immune cells [54]. These cells are significantly heterogeneous, as the production of immunomodulators and secretion of nucle-
plastic, and can have different functions in response to different ases. It can also target neutrophils by blocking neutrophils roll-
environmental stimuli [55]. Wound microbes can also interact ing on activated endothelial cells antibodies and opsonin
dynamically with various types of cells to regulate wound repair necessary for pathogen recognition by cells to escape phagocytic
and regeneration (Fig. 3). neutrophils, allowing them to escape NETs [39]. Among the var-
ious virulence factors secreted by S. aureus, leukotoxins and
leukocidins can promote the production of NETs through a
3.1. Neutrophils non-ROS-dependent pathway [64]. In addition, the expression
of S. aureus protein SPA in subcutaneous skin infection can also
Following injuries, neutrophils, circulating inflammatory cells, assist neutrophils recruitment, and clearance of bacteria through
are the first responders recruited from the blood strain to the the NET capture [39,52].
wound site [56]. Neutrophils accumulate abundantly at the would In contrast, few studies have proved that NETs are a ‘‘double-
site, releasing chemokines and inflammatory cytokines that further edged sword”. NETs can eliminate pathogens and reduce inflam-
recruit more neutrophils and other immune cells [57]. Neutrophils mation, while they can reprogram macrophages to reach M2
also act as host defence by engulfing pathogens, secreting granules Phenotypic regulation for a faster wound healing [65]. However,
filled with cytotoxic enzymes, or expelling neutrophil extracellular over-promoted neutrophils by pathogens in the wound area stim-
traps (NETs) to trap and kill pathogens (a process is known as ulate severe expression of inflammatory mediators (such as IL-1,
NETosis) [58]. NETs are a DNA network structure formed by neu- TNF-a) and proteolytic enzymes, resulting in excessive and persis-
trophils activated by various stimuli and released to the outside tent inflammation that exacerbates tissue damage or even damage
of the cell [59]. With DNA as the backbone, the network structure surrounding healthy tissue [55]. For example, over-expressed NETs
is inlaid with a variety of proteins, including histones, matrix met- were found in diabetic foot wounds, leading to further tissue dam-
alloproteinase 9 (MMP-9), neutrophil elastase, myeloperoxidase, age and delayed wound healing. In an animal model of diabetic
protease 3, etc., aiming to kill pathogens or increase their perme- mice, inhibiting the formation of NETs or the cleavage of NETs
ability [60]. Currently, S. aureus, Streptococcus pneumoniae, E. coli, can significantly reduce inflammation and improve the wound
Helicobacter pylori, Pseudomonas aeruginosa, Candida albicans (C. healing [66]. The presence and activity of neutrophils mediated
albicans) and fungi were found to stimulate neutrophils to produce
NETs [61] (Fig. 3).
6
Q. Tang, N. Xue, X. Ding et al. Advanced Drug Delivery Reviews 192 (2023) 114671

Fig. 3. Response of immune cells, including neutrophils, macrophages, and T cells to wound microbiome.

by microbiomes is therefore a challenge in wound care which To date, the effect of probiotics on macrophage polarization has
request close monitoring of inflammation or infection [66]. become a new research trend. The lactic acid secreted by Lactococ-
cus lactis (L. lactis) was proved to reverse the inflammatory and
proteolytic characteristics of diabetic wounds via the M2 polariza-
3.2. Monocytes and macrophages tion [75–76]. Scientists also attempted to programme L. lactis by
transforming a plasmid encoding VEGF, to maximize VEGF produc-
Over the inflammatory phase of wound healing, neutrophils are tion, and reduce intrinsic deactivation of the VEGF [77]. The genet-
cleared by macrophages [67], further removing pathogens and cel- ically engineered L. lactis can continuously secrete VEGF and
lular debris directly or indirectly via secreting transforming growth lactate, with enhanced angiogenesis and induction of M2 polariza-
factor-b (TGF-b1) and vascular endothelial growth factor (VEGF) tion [77]. Such pioneer ideas are based on live engineered bacteria,
[68–69]. Macrophages include tissue-resident macrophages which can promote wound healing by exogenous supplementation
located in the skin and monocytes recruited from bone marrow of probiotics in the future.
through blood circulation. Macrophages are the key orchestrators Similar to mammalian cells, gram-negative and gram-positive
of the wound healing process against infection [70], while pro- bacteria constitutively release lipid bilayer-enclosed, nanosized
inflammatory macrophages are often described as M1 macro- extracellular vehicles (EVs) or membrane vesicles [78]. As EVs con-
phages, and anti-inflammatory and repairing macrophages are tain bioactive molecules, including nucleic acids, proteins, lipids,
M2 macrophages [71]. M1 macrophages are usually detected early toxins and various virulence factors, they were reported to have
in the repair process, secreting cytokines such as IL-6, TNF-a and diverse functions in cell-to-cell communication between bacteria
nitric oxide synthase that regulate the early stages of healing [79]. Bacterial EVs are proven more effective compared to bacterial
[72]. Approximately 80–85 % of the M1 pro-inflammatory pheno- extracts or purified toxins in evoking cellular responses in the
type are converted to anti-inflammatory M2 macrophages on days recipient cells [56]. A pioneer study proved that EVs derived from
5–7 of acute wounding [73]. M2 macrophages normally lead to Lactobacillus plantarum (L. plantarum), LEV, could successfully
increased secretion of anti-inflammatory cytokines (e.g., IL-10, induce monocyte differentiation into M2 macrophages in human
Arginase-1) that suppress inflammation and increase growth fac- THP1 cells, with increased secretion of the anti-inflammatory cyto-
tors required for proliferation, migration, and repair processes kine IL-10, along with immunomodulatory cytokines IL-1b, GM-
[71]. In diabetic patients, M1 macrophages drive the elevated CSF and a higher population of M2b macrophages [60]. Such
and prolonged non-resolving inflammation seen in diabetic foot promising results showed that LEVs can be used as anti-
ulcers (DFU) [74] with around 80 % of macrophages at the edge inflammatory and immunomodulatory substances by regulating
of chronic wounds being the pro-inflammatory M1 phenotype, the conversion of M1 and M2 macrophages, thereby improving
and the transition to an M2 phenotype is a hindrance. Therefore, hyperinflammatory skin conditions and the phenotypes of inflam-
the polarization of macrophages is crucial for faster wound healing. matory skin disorders. Therefore, determining which uptake path-

7
Q. Tang, N. Xue, X. Ding et al. Advanced Drug Delivery Reviews 192 (2023) 114671

ways and signalling molecules are involved in LEV-evoked cellular isms, including bacteria, fungi and viruses [99]. Reservoirs of resi-
responses may further assist in interpreting bacterial EV-mediated dent organisms, which form the neomicrobiome, are found on the
phenomena in human cells and designing alternative methods surrounding superficial skin, in deeper skin layers and the pilose-
using bacterial EVs as immunomodulatory materials. baceous unit [12]. Other organisms may enter the wound from
environmental contamination before an operation at the time of
wounding, or during the operation e.g. respiratory, alimentary,
3.3. Lymphocytes
genital or urinary tract organisms or after in the hospital or home
environment [103].
Lymphocytes are immune cells that are recruited to the wound
Repopulating the wound neomicrobiome is a dynamic process
at a late stage aiming for fibrous tissue regeneration [80]. B lym-
and changes with time. Immediately after wounding, there is a
phocytes are relatively rare in the skin at a steady state. Elevation
gram-negative shift with resident commensals such as Proteobacte-
of B lymphocytes can be observed in the skin disease, such as
ria and Bacteroidetes and gram-negative environmental contami-
human atopic eczema, cutaneous leishmaniasis, and skin sclerosis
nants such as Pseudomonas colonising wounds [104–105]. Gram-
[81]. Recent studies have found that B cells can act as a regulator of
positive bacteria are still present but with a reduced number, such
the tissue regeneration [82]. ECM component can induce CD19-
as the facultative anaerobe Propionibacterium [12]. This is in-part
dependent Toll-like receptor (TLR) signalling in wound B cells,
due to the suppression of gram-positive Staphylococcus and Strep-
thereby inducing the production of cytokines, IL-10 and TGF-b,
tococcus populations with the SSWI antibiotic prophylaxis [105].
directly reducing the inflammatory response [83]. When mature
With wound healing, reservoirs of resident commensal organisms
B cells were applied locally to acute or chronic wounds, increased
repopulate the wound area while a gram-positive shift occurs with
fibroblast proliferation is noted with a reduced level of apoptosis
a return to the normal skin flora [12,104]. Total microbial load is
and an accelerated healing rate [84]. However, to our knowledge,
also variable but critical to wound healing and infection. Overpop-
no data or report was seen on the role of wound microbiome-
ulation of commensal organisms or environmental pathogens is
mediated B cells.
causative of SSWI and delays wound healing. Once bacterial levels
T lymphocytes are mainly categorized into ab T cells and cd T
reach a threshold of > 105 colony-forming units, tissue invasion
cells according to their composition [85]. cd T cells have a high pro-
and wound infection are common [100,106]. The dynamic process
portion in the mouse skin tissue, accounting for more than 90 % of
of the surgical wound microbiome is a novel area of research. At
T cells in the epidermis as a skin immune defensor [86–87]. Human
this point, analysis has focused on the bacterial neomicrobiome
skin contains ab T cells and cd T cells with an expression ratio of
[107].
approximately 5:1, distributed in both the epidermis and dermis
Repopulation of the wound with resident commensal organ-
[88–89]. cd T cells are highly involved in psoriasis [90], tumours
isms accelerates wound healing and prevents wound infection by
[91–92], viral infection [93] and wound healing [94]. Burn injury
opportunist pathogens with evidence suggesting that this process
has been found to activate cdT cells to produce a large amount of
occurs over a period of 2–6 weeks [4,12,108]. Commensal micro-
insulin-like growth factor 1 (IGF-1) (Fig. 3). However, the presence
biota is beneficial through indirect competition, occupying the skin
of P. aeruginosa in burn wounds was reported to reduce the synthe-
niche and utilising available nutrients [109]. The diversity of the
sis of IGF-1 in burn patients with peripheral blood cdT cells,
resident commensal microbes prevents domination by a single
adversely affecting wound repair [95]. When cd T cells are acti-
pathogen. Evidence of this has been demonstrated in numerous sit-
vated by an infection, a large amount of IL-17 can be produced to
uations. For example, low levels of commensal Pseudomonas accel-
resist infection [55] Hence, IL-17 also plays a crucial role in the
erate wound healing. However, overpopulation results in tissue
immune response to pathogens, including Klebsiella pneumoniae
invasion and SSWI’s [100,110]. Resident microbes also directly
(K. pneumoniae), Citrobacter rodentium, C. albicans [96–97]. In addi-
support wound healing by producing anti-microbial molecules,
tion, cd T can also regulate and control ab T cell-mediated inflam-
metabolites and immune-enhancing actions [36,109,111–112].
mation, to reduce the activity of lymphocytes and the production
Surgical wound management, therefore, aims to encourage the
of IFN-c in delayed hypersensitivity reactions [98].
repopulation of a neomicrobiome with the diverse commensal res-
ident flora restoring skin homeostasis and preventing wound infec-
4. Clinical management in regulating wound microbiome tion. Various surgical techniques are directed at controlling the
number of microbes and resorting balance to the neomicrobiome
4.1. Role of commensal microbiota on surgical wounds [13,113]. This includes surgical techniques to aid wound closure,
control of contamination, antiseptic and antibiotics.
Surgical incisions breach the skin barrier, disrupting the com- Surgical wounds may be closed primarily where healing occurs
mensal resident microbiota, triggering a host immune response from approximation of tissue using sutures, staples or glue.
and wound healing processes [99]. All surgical wounds are re- Wounds heal by primary intention, minimising exposure to
colonised by a patient’s resident microbiome and environmental endogenous and exogenous contaminants [114]. Healing by sec-
organisms. Some wounds become infected when organisms invade ondary intention is the process of an open wound healing through
tissue or cause cellular injury. Depletion of the resident skin micro- granulation, contracture and epithelialisation. An open wound is
biome in favour of opportunistic pathological organisms capable of exposed to environmental contaminants, and healing occurs in
invasion results in SSWIs [100]. SSWIs impose a significant burden the presence of a complex relationship between microbial coloni-
on the health care system and are a leading cause of morbidity sation and the immune system [115–116]. The success and failure
after surgery occurring in up to 5 % of patients post-operatively of tertiary healing largely depend on the wound bioburden with
[101]. This accounts for approximately 20 % of all healthcare- graft or primary closure failure, highly likely with an elevated bac-
associated infections and costs $5000-$13,000 per incident [102]. terial load [100,117].
Methods for the prevention of wound infection are largely The degree of wound contamination influences the inoculum of
instrumental in the management of surgical patients. These inter- a wound with foreign microbes and depends on pre- and intra-
ventions support the return of a commensal wound neomicrobiota operative events determining whether a wound is clean, clean-
and suppress domination by opportunistic pathogens. The neomi- contaminated, contaminated, or dirty. In clean wounds, the respi-
crobiome describes the process of re-colonisation of the wound ratory, alimentary, genital or urinary tract is not entered. Clean
after surgery with resident and transmitted non-resident organ- contaminated wounds reflect a controlled entry into the respira-
8
Q. Tang, N. Xue, X. Ding et al. Advanced Drug Delivery Reviews 192 (2023) 114671

tory, alimentary, genital or urinary tract. Contaminated wounds quate glycemic control, decompression, wound coverage with
are open, fresh, accidental wounds or occur in operations with appropriate dressings, revascularization performed if necessary
major breaks in sterile technique, gross spillage of the gastroin- and appropriate antimicrobial therapy for clinically infected
testinal tract or acute non-purulent inflammation encountered. [128–129]. A recent clinical study showed that when wounds have
In both clean-contaminated and contaminated wounds, the neomi- no infection and no antibiotic treatment, debridement can signifi-
crobiome is exposed to foreign microbes e.g., gastrointestinal bac- cantly reduce the pathogenic anaerobes, such as Anaerococcus lac-
teria. Dirty-infected wounds include old traumatic wounds and tolyticus, Porphyromonas Somerae, Prevotella melaninogenica, and
wounds with existing infection. Contamination of a wound during Veillonella dispar. However, debridement was found to have no
or before an operation inoculates the wound with potential patho- effect on the commensal wound microbiota [15,130]. Under antibi-
gens which may dominate the neomicrobiome [106]. This is sup- otic treatment, effects of debridement on the diversity of micro-
ported by evidence which shows the SSWI rates following clean, biomes seem to be overcome by antibacterial therapy, resulting
clean-contaminated, contaminated, and dirty procedures are in no changes in the wound microbiota [131]. These data suggest
1.76 %, 3.94 %, 4.75 %, and 5.16 %, respectively [101]. that debridement may be beneficial for controlling wound infec-
In treating acute wounds, ethanol, povidone-iodine and/or tion without antibiotic treatment, but its role in mediating wound
chlorhexidine are widely applied clinically as the pre-operative microbiome is limited [131].
topical antiseptics. The latest report showed that exposure to Antibiotic treatment is well known for treating chronic wound
antiseptic treatments can elicit rapid but temporary depletion of infection, and it also leads to major changes in the composition
microbial community diversity and membership [118], while the of the wound commensal microbiota [18]. A clinical study on DFUs
wound microbiome was found highly dependant on personalized revealed that at 0, 4 and 8 weeks post-debridement and antibiotic
and body site-specific colonization signature. Moreover, another treatment, patients with healed wounds had a larger abundance of
clinical study compared the efficacy of povidone-iodine-alcohol Actinomycetales and Staphylococcaceae [132]. However, in the same
and chlorhexidine-alcohol for surgical wounds, showing skin study, another 12 unhealed patients were detectable with Bac-
microflora was more affected by povidone-iodine in short time teroidales and Streptococcaceae, suggesting the presence of Actino-
but not in prolonged contact times over 3 h [119]. These studies mycetales and Staphylococcaceae is a good prognostic indicator
confirmed the short-time stability and resilience of the skin micro- but appears that Bacteroides and Streptococcaceae seem to have a
biome to antiseptic and its effect on the incidence of SSWI poor outcome [132]. Moreover, an increase in the Bacteroides fam-
remained unknown. ily may be a sign of changing antibacterial or requiring additional
The use of antibiotics before, during and after surgery alters the debridement. Another study further showed that antibiotics could
structure and function of wound microbial communities. Systemic result in a significantly decreased diversity of microbiota in
and topical antibiotics suppress common pathogens, and lower 3 weeks, following an unexpected increase thereafter [133]. Swab
wound tissue inoculum from intra-operative contamination but samples taken before debridement showed greater changes in bac-
also eliminate commensal resident bacteria [120–121]. It has been terial composition associated with antibiotics (doxycycline) [134],
proven definitively that systemic antibiotics given at the time of while samples taken from wound centres had higher bacterial
induction reduce SSWI for wound healing by primary and sec- loads compared to the wound edges [135]. To date, Imipenem
ondary intention [122]. Topical antibiotics after primary closure was found to be the most efficient antibiotic against Gram-
also reduce SSWI [101]. It is unclear whether the antibiotics’ mech- positive and Gram-negative bacteria for patients with chronic
anism of SSWI reduction after surgery is from eliminating environ- wounds [136–137].
mental pathogens or through suppressing the overgrowth of a Negative pressure wound therapy (NPWT) is also commonly
commensal opportunist, it is likely a combination of both [103]. used in treating chronic wounds [138]. A study found that addi-
Overall healing of surgical wounds is intrinsically linked to the tional oxygen supply to diabetic wounds could promote wound
health of the wound neomicrobiome. A complex interplay exists repair, while wound microbiota shifted to a diverse bacteria dom-
between the neomicrobiome composed of commensal residential inated by aerobic bacteria and facultative anaerobes [139].
and environmental organisms, wound healing processes, the Platelet-rich plasma (PRP), as an additional therapeutic option for
wound environment and the immune system. Surgical techniques infected wounds, was reported to significantly inhibit the growth
are aimed at preventing wound infection and restoring the resi- of methicillin-resistant S. aureus (MRSA), K. pneumoniae and P.
dential wound microbiome. aeruginosa [140]. It is important to emphasize that few chronic
wounds do not heal despite various treatments. Therefore, clinical
4.2. Clinical management in treating chronic wounds management aims to create a balanced microbiome environment
to achieve normal wound healing.
Chronic wounds are different from surgical wounds. Studies
have found that chronic wounds are colonized by bacteria living
in biofilms, and are more likely to be home to bacterial infection 5. Strategies in controlled delivery of probiotics and
[32]. Colonization of skin bacteria is now considered a major cause microbiome
of delayed healing and ineffective treatment response [123]. Gram-
positive (GP) bacteria were sensitive to linezolid, vancomycin and Although topical antibiotics for wound sterilization are still
teicoplanin [124–125]. However, over 50 % of Gram-negative (GN) widely used clinically, studies have pointed out that local antimi-
bacteria were resistant to third-generation cephalosporins, while crobial therapy is difficult to achieve effective antibacterial concen-
the resistance rates of piperacillin/tazobactam, amikacin, merope- trations and is prone to antibiotic abuse and to cause bacterial
nem and imipenem were low [126]. A study found that the most resistance [141]. In addition, removal of beneficial microbiota from
common pathogens in 216 diabetic foot patients were: S. aureus wounds via topical administration of antibiotics can further result
(28 %), E. coli (19 %), and S. epidermidis (14 %) [127]. Antibiotics in an imbalance of the wound microbiota [4,142]. As discussed
were therefore prescribed from ceftriaxone (49.4 %), metronidazole above, more recent studies confirmed that microbiomes are closely
(21 %) to ciprofloxacin (7.5 %) for skin and soft tissue infections. related to wound healing [143]. However, to our knowledge, there
Currently, therapeutic interventions for chronic wound man- is no report of using microbiomes in the treatment of wound heal-
agement are aiming to prevent the infection or chronicity. The ing, but probiotics are widely developed, aiming to achieve optimal
standard protocols include debridement of necrotic tissue, ade- wound microbiome and promote wound healing [144–147]. Such a
9
Q. Tang, N. Xue, X. Ding et al. Advanced Drug Delivery Reviews 192 (2023) 114671

delivery system may also be utilized in the future for the topical Chitosan which is produced by N-deacetylation of the polysac-
administration of microbiomes. charide chitin demonstrates non-toxicity and satisfying biocom-
Two strategies are currently utilized in probiotic development. patibility and biodegradability in the food and pharmaceutical
The first is to directly use natural probiotic strains or engineered field [159]. These unique biological properties have enabled chi-
probiotics in wound healing. The second strategy is to use a probi- tosan to develop into a variety of wound dressings, which have
otic strain loaded in a delivery carrier with controlled delivery antimicrobial, biologically adhesive and hemostatic effects, and
capacity to enhance wound healing. The topical application of L. provide an ideal moist wound environment, infection control and
plantarum showed great potential in a mouse model for the treat- removal of the wound exudate [160–162]. Moreover, positively
ment of burn wound sepsis infected by P. aeruginosa [124]. Topical charged chitosan has strong adhesive properties with negatively
use of Kefirs gel (a mixture containing Lactobacillus and Yeast) on charged polymers [163]. Thus, recent studies showed that chitosan
the burn injury wound was further confirmed with the faster can act as a coating material for microcapsules of probiotics to pre-
regeneration of epidermis and more collagen deposition, promot- vent direct contact between probiotics microcapsules and harsh
ing wound healing rate [148]. Genetically engineered bacteria, environments, providing protective effects [164]. As a coating
AUP-16, is the first-in-class that has entered the clinical stage for material, chitosan has been found to increase probiotics survival
the treatment of diabetic foot ulcers, venous ulcers of the lower rate and probiotics stability during cold storage [165–166]. How-
extremities, and pressure ulcers [149]. AUP-16 is a genetically ever, the solublity of chitosan is pH-dependent. Chitosan has poor
engineered Lactococcus lactate, a non-pathogenic probiotic that solubility when the pH value exceeds 5.4 [167]. In general, healthy
carries genes encoding multiple regenerative factors, including skin has a pH value ranging from 4.5 to 6.5. A wound pH value can
human basic fibroblast growth factors (FGF2, bFGF), interleukin 4 increase to a range of 7.4 to 9 due to the by-products of proliferat-
(IL-4) and macrophage colony-stimulating factor (CSF1, mCSF) ing bacterial colonies [3]. Thus, the application of chitosan as a
[149]. AUP-16 can continuously secrete lactic acid, reverse the delivery system of probiotics on wounds might be limited. Chem-
alkaline environment of the wound and induce M2 macrophage ical modification of chitosan through derivatization by acylation,
polarization to promote angiogenesis and granulation tissue for- alkylation and quaternization reactions has been proven to signif-
mation with rapid wound healing [149]. Furthermore, studies have icantly increase the water solubility of chitosan, further improving
shown that the chemokine CXCL12 can increase the density of its drug delivery application on skin and wounds [168]. Alginate,
macrophages and TGF-b expressing macrophages in the dermis extracted from seaweeds, is another common anionic biomaterial
and epidermis at the wound edge. Lactobacillus Reuteri were trans- for probiotics encapsulation [156,163]. Alginate demonstrates
fected with a plasmid encoding the chemokines CXCL12 and deliv- extraordinary physicochemical and mechanical properties, favour-
ered directly to the damaged skin of mice, which were proved to able hydrogel formability, great hydrophilicity and excellent cell
release large amounts of CXCL12. The lactate production can fur- adhesion compatibility [169]. Alginate-formulated microcapsules
ther reduce the local wound pH, improve the bioavailability of have a highly porous structure [170]. Therefore, most studies used
CXCL12, and strongly promote wound healing [150]. Thereafter, a composite of alginate with other materials to facilitate its encap-
varying drug delivery system was developed and investigated for sulation effects. For instance, the composite of sodium alginate and
the controlled release of probiotics which may be an alternative calcium chloride can form probiotics capsules having better per-
used for delivering microbiome in the future study (Table 2). meability, which is favourable for air and nutrient exchange, and
metabolite release [171–173]. Alginate has also been broadly
employed as a wound dressing for specifically exuding wounds
5.1. Microencapsulation
by generating a slightly wet wound healing environment [174].
Some alginate-based dressings have been demonstrated to stimu-
Microencapsulation is a widely used technology for probiotics
late human macrophage activities in chronic wound beds to gener-
delivery [156–157]. According to biomedical applications with
ate a pro-inflammatory signal and initiate a resolving
desired outcomes, materials utilised to encapsulate probiotics
inflammation characteristic of healing wounds [175]. Carrageenan
should meet the following characteristics, such as non-toxicity,
extracted from red seaweeds, known as Irish moss, is natural linear
the viability of probiotics, bioactivity maintenance, fine biocom-
sulphated polysaccharides [176]. It is a hydrophilic biopolymer and
patibility and biodegradability, permselectivity, protection of pro-
has been widely used in the food industry and recently drew a lot
biotics from harsh environments, controlled/delayed release,
of attention to its uses in tissue engineering, wound coverage and
good formability, low cost and easy accessibility [156]. In recent
drug delivery [177]. Carrageenan involves 15–40 % ester-sulphate
decades, varying polysaccharides (biomaterials), such as alginate,
content, making them anionic polysaccharides. They can be classi-
carrageenan, gums, and chitosan, have been utilised for probiotics
fied into three different types according to their sulphate content,
delivery aiming to maintain the viability and stability of probiotics
including kappa-carrageenan (one sulphate group per disaccha-
viability over drug delivery [158].

Table 2
Overview of different probiotic strains, methods of delivery, materials used for skin-related application.

Strain Method Material Application Reference


L. reuteri; Microencapsulation Chitosan Wound healing [151]
Lactobacillus fermentum;
Bacillus subtilis sp. natto
Hydrogel fabrication Hyaluronate-dipic dihydrazide/ Superbacteria-induced skin [147]
L. rhamnosus Pluronic F127/ Fucoidan wound infections
Enterococcus mundtii Electrospinning PVA/PVP/Glycerol Infection control [152]
Dermal burn healing
Lactobacillus acidophilus; Microencapsulation Alginate Wound infection [153]
Lactobacillus casei;
L. rhamnosus
L. plantarum Microencapsulation Sodium alginate /Calcium chloride Atopic dermatitis [154]
Bacillus coagulans Microencapsulation Gelatin emulgel Wound healing [155]

10
Q. Tang, N. Xue, X. Ding et al. Advanced Drug Delivery Reviews 192 (2023) 114671

ride), iota-carrageenan (two sulphate groups), and lambda- can be released through the kidneys [200]. PLA can be fabricated
carrageenan (three sulphate group), whereas only kappa- and as a variety of biodegradable devices, such as absorbable surgical
iota-carrageenan have gelling ability [178]. Kappa-carrageenan is sutures, injection capsules, orthopedic fixation materials, micro-
thermal-responsive and is able to undergo reversible volume tran- spheres for drug release, and tissue-engineered stents. In a recent
sitions in response to thermal environments [163] with the capa- study, coaxial electrospun PLA nanofibers have been proved to
bility to be used for delivery systems based on the thermal increase probiotics viability through encapsulation [200]. Pluro-
control [179]. Kappa-carrageenan encapsulating probiotics have nicÒ F127 is a copolymer synthesized using dicyclohexylcarbodi-
shown great acid and bile tolerance. Carrageenan blended with imide and dimethylaminopyridine. PluronicÒ F127 has also been
other materials has also been used to improve encapsulation widely used as it has decent cytocompatibility and temperature-
effects [180]. Carrageen-based hydrogels can create a damp envi- sensitive sol gel properties. However, its incompetent mechani-
ronment to accelerate the wound healing [181]. It has been cal strength and stability have minimised its medical application
reported composite cyclic glucan/carrageenan hydrogels could [147]. Therefore, the composites of PluronicÒ F127 with other bio-
induce fibroblast cell migration, facilitating wound healing attrib- materials, such as chitosan, collagen, and hyaluronic acid, have
uted to their immunomodulatory properties [181]. Xanthan gum been employed to improve the aforesaid weakness [147].
is a polysaccharide, produced from simple sugars by Xanthomonas Whilst these synthetic polymers have favourable characteristics
campestris fermentation [182]. In tissue engineering, xanthan gum as materials of electrospun fibres for probiotics delivery purposes,
has exhibited superb biocompatibility, gelation ability and stability a couple of challenges such as poor cell affinity and adverse foreign
in a wide range of pH, temperature, and ionic strength, remaining body response still limited the clinical application. Therefore, most
activity/effectivity in acid and heat conditions [183]. Xanthan gum studies investigated to use the synthetic polymers combined with
was also combined with alginate to encapsulate probiotics, which biopolymers, such as chitosan, alginate, and cellulose to improve
has shown improved encapsulation efficiency, survival rate under biodegradability, bioactivity, and biocompatibility.
simulated gastric conditions, heat resistance, storage stability and
target release to the intestine [184].
5.3. Hydrogels and 3D bioprinting
5.2. Electrospun scaffolds
Hydrogel, within hydrophilic 3D network structures, can mimic
Electrospun scaffolds with micro- or nano-fibres, can be ulized the extracellular matrix and support cell growth [201–202]. The
to encapsulate probiotics bacterial for a controlled delivery [185]. therapeutic potential of hydrogel-encapsulated bacteria has been
Polymers, such as polyethylene oxide (PEO), polyvinyl alcohol validated in many studies [203], as hydrogel can protect bacteria
(PVA), copolymers, polyvinyl pyrrolidone (PVP), polylactic acid from environmental damage over recycling together via providing
(PLA), and PluronicÒ F127 were genuinely favoured and safe mate- ingredients for bacterial growth. Bacillus subtilis was encapsulated
rials for entrapping probiotics and wound healing [156,186–188]. in a thermosensitive hydrogel, and the formed hydrogel not only
PEO is a non-immunogenic, non-toxic and hydrophilic polymer effectively accumulate in the epidermis, but serve as a small fac-
[189]. PEO has excellent biocompatibility and mucoadhesive activ- tory for the efficient the production of antifungal ingredients (e.g.
ity and it appears not to impact the bioactivity of delivered sub- surfactin), showing a promising antifungal effect in the skin model
stances. A number of studies have reported the addition of of C. albicans infection [204]. A heparin-poloxamer heat-responsive
lyoprotectants into PEO can significantly enhance the viability of hydrogel was investigated in loading Lactobacillus to treat diabetic
probiotics during the drying process [190]. PVA is also a widely wounds. The results showed that when the thermosensitive hydro-
accepted synthetic polymer for probiotics delivery. It is generated gel is applied to wounds, it can rapidly form a gel to limit the
from polyvinyl acetate and possesses numerous hydroxyl groups. spread of bacteria, while maintaining bacterial activity and pro-
PVA has shown good stability in a wide range of pH from 1 to longing its residential time on the wound area [77]. The use of pro-
13, and the great gelling ability by chemical or physical crosslink- biotics in hydrogels for open wounds can partially reduce the
ing. It has been reported that the combination of PVA and occurrence of local inflammation, and the strong lactic acid pro-
fructooligo-saccharides considerably improved the thermal stabil- duction capacity of Lactobacillus can promote the transformation
ity and viability of entrapped probiotics [191]. PVA-based hydro- of macrophages to M2 phenotype, improve tissue regeneration
gels have also been used as wound dressings to enhance chronic and recovery potential, and promote rapid healing of the wound
wound healing attributed to their outstanding biological properties environment [149]. The hydrogel system confines bacterial popu-
and drug delivery capability [192–193]. The in-vitro cell study con- lations within the wound, thereby minimizing the risk of systemic
firmed that PVA-based dressings have either low or no cytotoxicity toxicity [77]. However, the composition of the hydrogel may influ-
to human dermal fibroblasts, human epidermal keratinocytes and ence the viability and function of the loaded bacteria [205], so it
mouse fibroblast cells [194–195]. PVP has long been used in tissue needs to be further investigated for safety prior to clinical
engineering, drug delivery or probiotics delivery via electrospin- application.
ning. The previous study designed the composite PVP/PCL nanofi- 3D bioprinting is considered as a powerful and advanced
bers loaded with a bioactive agent, pioglitazone, with non- biomanufacturing technique in skin tissue engineering [206–
toxicity and sustained drug release for treating diabetic wounds 207], due to its precisely controlled distribution of a bioink consist-
[196]. ing of biomaterials, bioactive factors and incorporated living cells
Nanofibers of PVP have been evidenced to ameliorate the meta- via the computer-aided design [208]. Currently, embryonic stem
bolic activity of probiotics, L. acidophilus [197]. The other study cells, and mesenchymal stem cells can be successfully bio-
exhibited that the viability of various lactobacilli species entrapped printed as the artificial skin [209], suggesting the possibility of
in electrospun fibres varied depending on the ratio of PVP incorpo- using bioprinting technology and bioink for delivering probiotics
ration during delivery [198]. PLA is a polymer of organic acid and and microbiota in the future. However, there is no study on the
can be produced by polycondensation of lactic acid and/or ring- delivery system of probiotics prepared by 3D bioprinting which
opening polymerization of lactide [199]. PLA has long been may be explained by the low viability of probiotics post-
approved by The US Food and Drug Administration (FDA) due to fabrication and the complexity of commensal microbiota on the
its attractive biocompatibility and biodegradability. In humans, wound area. As discussed above, the environmental factors should
PLA is metabolised into harmless products, CO2 and H2O, which not be overlooked when scientists attempt to design 3D printing
11
Q. Tang, N. Xue, X. Ding et al. Advanced Drug Delivery Reviews 192 (2023) 114671

materials for probiotics delivery together with internal structure However, our knowledge about wound microbiota remains lim-
and varying fabrication approaches. ited, and further research is needed. In particular, investigation
on how to control wound microbiome with essential inflammatory
response, development of a controlled delivery system for sus-
5.4. Microneedles
tained releasing of beneficial microbiome and design of monitoring
system for health diversity of wound microbiome, should be prior-
Microneedle is a non-invasive or minimally invasive route
itized. Further research will continue to extend our understanding
developed for transdermal drug delivery. Microneedle drug deliv-
of microbiomes on skin and identify novel targets in wound care to
ery can penetrate the stratum corneum of the skin and release
ultimately enhance the clinical outcome for patients.
the drug directly into the subcutaneous space without irritating
nerves and causing pain [210], and additionally microneedles can
also be utilized in the treatment of chronic wounds [211–212]. Dis- Declaration of Competing Interest
solvable microneedles filled with cell-laden GelMA cores were
investigated for the delivery of mesenchymal stem cells with the The authors declare that they have no known competing finan-
intention to modulate the wound microenvironment via secreting cial interests or personal relationships that could have appeared
growth factors, exosomes, and cytokines to enhance angiogenesis to influence the work reported in this paper.
and tissue remodelling [213]. Bioactive probiotics Lactobacillus
was loaded into soluble sodium carboxymethylcellulose (SCMC)
microneedle arrays and successfully released into the dermis Acknowledgment
[214]. Transdermal administration of Lactobacillus was further
proved to enhance skin conditioning and immunity [214]. Soluble This study was supported by National Science Foundation of
microneedles can be used as a painless release tool for probiotics China (82172217), Fundamental Science (Natural Science)
and achieve highly stable encapsulation of probiotics after skin Research Project of the Jiangsu Higher Education Institutions of
penetration and their rapid release without causing any adverse China (No. 21KJB360016) and Natural Science Foundation of Nan-
skin irritation. jing University of Chinese Medicine (No. XZR2020069). Professor
Emerging antimicrobial transdermal and ocular microneedle Yiwei Wang was supported by Jiangsu Distinguish Professor fel-
patches have become promising medical devices for the delivery lowship, Nanjing University of Chinese Medicine.
of antibacterial, antifungal, and antiviral therapeutics, including
nano-silver, nanoparticles, gentamicin and zinc oxide [215–216]
to treat bacterial biofilms [217–218]. Scientists developed a flexi- References
ble polymer composite microneedle array that can overcome the
[1] M. Parani, G. Lokhande, A. Singh, A.K. Gaharwar, Engineered Nanomaterials
bacterial biofilm present in chronic wounds, and co-deliver oxygen for Infection Control and Healing Acute and Chronic Wounds, ACS Appl.
and bactericidal agents [217]. Such microneedles effectively ele- Mater. Interfaces 8 (2016) 10049–10069.
vated the oxygen level from 8 to 12 ppm over 2 h and also provided [2] X. Fu, State policy for managing chronic skin wounds in China, Wound Repair
Regen. 28 (2020) 576–577.
strong anti-bactericidal effects on both liquid and biofilm bacteria [3] X. Ding, Q. Tang, Z. Xu, Y. Xu, H. Zhang, D. Zheng, S. Wang, Q. Tan, J. Maitz, P.K.
found in dermal wounds. Self-dissolvable microneedles and needle Maitz, S. Yin, Y. Wang, J. Chen, Challenges and innovations in treating chronic
tips loaded with chloramphenicol and gelatinase-sensitive and acute wound infections: from basic science to clinical practice, Burns &
Trauma 10 (2022) tkac014.
nanoparticles were further fabricated and proved to simultane-
[4] G. Wang, E. Sweren, H. Liu, E. Wier, M.P. Alphonse, R. Chen, N. Islam, A. Li, Y.
ously penetrate the biofilm matrix with controlled delivery of chlo- Xue, J. Chen, S. Park, Y. Chen, S. Lee, Y. Wang, S. Wang, N.K. Archer, W.
ramphenicol into the active regions of the biofilm [218]. More Andrews, M.A. Kane, E. Dare, S.K. Reddy, Z. Hu, E.A. Grice, L.S. Miller, L.A.
Garza, Bacteria induce skin regeneration via IL-1b signaling, Cell Host Microbe
recently, novel ice microneedles made from versatile soft materials
29 (2021) 777–791.e776.
were successfully developed aiming to carry and deliver small [5] V.W. Wong, R.G. Martindale, M.T. Longaker, G.C. Gurtner, From germ theory
molecules and biosafe Bacillus subtilis (B. subtilis) in the treatment to germ therapy: skin microbiota, chronic wounds, and probiotics, Plast.
of fungal infection in the wound area [219]. Results from this study Reconstr. Surg. 132 (2013) 854e–861e.
[6] E.A. Grice, J.A. Segre, The skin microbiome, Nat. Rev. Microbiol. 9 (2011) 244–
showed live B. subtilis can be incorporated in the microneedles 253.
with remaining high viability over 12 h and continue to divide [7] E.A. Grice, H.H. Kong, S. Conlan, C.B. Deming, J. Davis, A.C. Young, N.C.S.
and proliferate, significantly inhibiting fungal infection in a mouse Program, G.G. Bouffard, R.W. Blakesley, P.R. Murray, E.D. Green, M.L. Turner, J.
A. Segre, Topographical and temporal diversity of the human skin
model [219–220]. While there are many advantages over tradi- microbiome, Science 324 (2009) 1190–1192.
tional delivery systems, microbiome-based therapies face various [8] J.A. Sanford, R.L. Gallo, Functions of the skin microbiota in health and disease,
challenges. Understanding the microbiome and its functions may Semin. Immunol. 25 (2013) 370–377.
[9] A.A. Ross, K.M. Muller, J.S. Weese, J.D. Neufeld, Comprehensive skin
help prevent or treat personalized medicine in the setting. At pre- microbiome analysis reveals the uniqueness of human skin and evidence
sent, the specific mechanism of action of scavenging microbial for phylosymbiosis within the class Mammalia, PNAS 115 (2018) E5786–
components on wound healing is not yet known, but there is a E5795.
[10] E. Sohn, Skin microbiota’s community effort, Nature 563 (2018) S91–S93.
deep connection between the two. Targeting or repairing the
[11] P.J. Turnbaugh, R.E. Ley, M. Hamady, C.M. Fraser-Liggett, R. Knight, J.I. Gordon,
microbiota may be an effective therapeutic strategy in the future. The human microbiome project, Nature 449 (2007) 804–810.
[12] P.L. Zeeuwen, J. Boekhorst, E.H.v.d. Bogaard, H.D.d. Koning, P.M.v.d. Kerkhof,
D.M. Saulnier, I.I.v. Swam, S.A.v. Hijum, M. Kleerebezem, J. Schalkwijk, H.M.
6. Conclusion Timmerman, Microbiome dynamics of human epidermis following skin
barrier disruption, Genome Biol. 13 (2012) 1–18.
[13] M. Tomic-Canic, J.L. Burgess, K.E. O’Neill, N. Strbo, I. Pastar, Skin Microbiota
Pioneer investigations have been made in studying the role of and its Interplay with Wound Healing, Am. J. Clin. Dermatol. 21 (2020) 36–43.
wound microbiome in various wound healing, including [14] S.A. Eming, P. Martin, M. Tomic-Canic, Wound repair and regeneration:
mechanisms, signaling, and translation, Sci. Transl. Med. 6 (2014) 265sr266.
microbiome-regulated inflammatory, proliferation and remod-
[15] L.R. Kalan, J.S. Meisel, M.A. Loesche, J. Horwinski, I. Soaita, X. Chen, A. Uberoi,
elling together with the skin appendage regeneration, immune cell S.E. Gardner, E.A. Grice, Strain- and Species-Level Variation in the Microbiome
response and enhanced systemic drug delivery for wound infec- of Diabetic Wounds Is Associated with Clinical Outcomes and Therapeutic
tion. As a result, mediation of wound microbiome is expected to Efficacy, Cell Host Microbe 25 (2019) 641–655 e645.
[16] R.D. Wolcott, J.D. Hanson, E.J. Rees, L.D. Koenig, C.D. Phillips, R.A. Wolcott, S.B.
be a new target for finding novel pharmacological interventions Cox, J.S. White, Analysis of the chronic wound microbiota of 2,963 patients by
or therapeutical approaches in the treatment of wound repair. 16S rDNA pyrosequencing, Wound Repair Regen. 24 (2016) 163–174.

12
Q. Tang, N. Xue, X. Ding et al. Advanced Drug Delivery Reviews 192 (2023) 114671

[17] L. Kalan, M. Loesche, B.P. Hodkinson, K. Heilmann, G. Ruthel, S.E. Gardner, E.A. [42] C. Esser, I. Bargen, H. Weighardt, T. Haarmann-Stemmann, J. Krutmann,
Grice, Redefining the Chronic-Wound Microbiome: Fungal Communities Are Functions of the aryl hydrocarbon receptor in the skin, Semin. Immunopathol.
Prevalent, Dynamic, and Associated with Delayed Healing, mBio 7 (2016). 35 (2013) 677–691.
[18] S.E. Gardner, S.L. Hillis, K. Heilmann, J.A. Segre, E.A. Grice, The neuropathic [43] A. Uberoi, C. Bartow-McKenney, Q. Zheng, L. Flowers, A. Campbell, S.A.B.
diabetic foot ulcer microbiome is associated with clinical factors, Diabetes 62 Knight, N. Chan, M. Wei, V. Lovins, J. Bugayev, J. Horwinski, C. Bradley, J.
(2013) 923–930. Meyer, D. Crumrine, C.H. Sutter, P. Elias, E. Mauldin, T.R. Sutter, E.A. Grice,
[19] Y. Choi, A. Banerjee, S. McNish, K.S. Couch, M.G. Torralba, S. Lucas, A. Commensal microbiota regulates skin barrier function and repair via
Tovchigrechko, R. Madupu, S. Yooseph, K.E. Nelson, V.K. Shanmugam, A.P. signaling through the aryl hydrocarbon receptor, Cell Host Microbe 29
Chan, Co-occurrence of Anaerobes in Human Chronic Wounds, Microb. Ecol. (2021) 1235–1248 e1238.
77 (2019) 808–820. [44] A. Luqman, M.Z. Muttaqin, S. Yulaipi, P. Ebner, M. Matsuo, S. Zabel, P.M.
[20] E.K. White, E.A. Grice, The Wound Microbiome, Cold Spring Harb Perspect Tribelli, K. Nieselt, D. Hidayati, F. Götz, Trace amines produced by skin
Biol, 2022. bacteria accelerate wound healing in mice, Commun Biol 3 (2020) 277.
[21] M. Jamal, W. Ahmad, S. Andleeb, F. Jalil, M. Imran, M.A. Nawaz, T. Hussain, M. [45] B. Romana-Souza, L.C. Porto, A. Monte-Alto-Costa, Cutaneous wound healing
Ali, M. Rafiq, M.A. Kamil, Bacterial biofilm and associated infections, J. Chin. of chronically stressed mice is improved through catecholamines blockade,
Med. Assoc. 81 (2018) 7–11. Exp. Dermatol. 19 (2010) 821–829.
[22] H. Ren, F. Zhao, Q. Zhang, X. Huang, Z. Wang, Autophagy and skin wound [46] A. Luqman, F. Gotz, The Ambivalent Role of Skin Microbiota and Adrenaline in
healing, Burns & Trauma 10 (2022) tkac003. Wound Healing and the Interplay between Them, Int. J. Mol. Sci. 22 (2021).
[23] H. Shi, K. Cheer, U. Simanainen, B. Lesmana, D. Ma, J.J. Hew, R.J. Parungao, Z. Li, [47] A. Luqman, S. Zabel, S. Rahmdel, B. Merz, N. Gruenheit, J. Harter, K. Nieselt, F.
M.S. Cooper, D.J. Handelsman, P.K. Maitz, Y. Wang, The contradictory role of Götz, The Neuromodulator-Encoding sadA Gene Is Widely Distributed in the
androgens in cutaneous and major burn wound healing, Burns & Trauma 9 Human Skin Microbiome, Front. Microbiol. 11 (2020).
(2021) tkaa046. [48] Y. Zheng, R.L. Hunt, A.E. Villaruz, E.L. Fisher, R. Liu, Q. Liu, G.Y.C. Cheung, M. Li,
[24] H. Sorg, D.J. Tilkorn, S. Hager, J. Hauser, U. Mirastschijski, Skin Wound M. Otto, Commensal Staphylococcus epidermidis contributes to skin barrier
Healing: An Update on the Current Knowledge and Concepts, Eur. Surg. Res. homeostasis by generating protective ceramides, Cell Host Microbe 30 (2022)
58 (2017) 81–94. 301–313 e309.
[25] Z. Wang, C. Shi, Cellular senescence is a promising target for chronic wounds: [49] M. Tomic-Canic, S.W. Mamber, O. Stojadinovic, B. Lee, N. Radoja, J. McMichael,
a comprehensive review, Burns & Trauma 8 (2020) tkaa021. Streptolysin O enhances keratinocyte migration and proliferation and
[26] P. Hu, Q. Yang, Q. Wang, C. Shi, D. Wang, U. Armato, I.D. Prà, A. Chiarini, promotes skin organ culture wound healing in vitro, Wound Repair Regen.
Mesenchymal stromal cells-exosomes: a promising cell-free therapeutic 15 (2007) 71–79.
tool for wound healing and cutaneous regeneration, Burns & Trauma 7 (2019) [50] K. Midorikawa, K. Ouhara, H. Komatsuzawa, T. Kawai, S. Yamada, T. Fujiwara,
38. K. Yamazaki, K. Sayama, M.A. Taubman, H. Kurihara, K. Hashimoto, M. Sugai,
[27] A.J. Singer, Healing Mechanisms in Cutaneous Wounds: Tipping the Balance, Staphylococcus aureus susceptibility to innate antimicrobial peptides, beta-
Tissue Eng. B Rev. (2022). defensins and CAP18, expressed by human keratinocytes, Infect. Immun. 71
[28] X. Wei, M. Li, Z. Zheng, J. Ma, Y. Gao, L. Chen, Y. Peng, S. Yu, L. Yang, (2003) 3730–3739.
Senescence in chronic wounds and potential targeted therapies, Burns & [51] X. Wang, Y. Liu, J. He, J. Wang, X. Chen, R. Yang, Regulation of signaling
Trauma 10 (2022) tkab045. pathways in hair follicle stem cells, Burns & Trauma 10 (2022) tkac022.
[29] M. Guggenheim, R. Zbinden, A.E. Handschin, A. Gohritz, M.A. Altintas, P. [52] C.D. Gonzalez, C. Ledo, E. Cela, I. Stella, C. Xu, D.S. Ojeda, P.S. Frenette, M.I.
Giovanoli, Changes in bacterial isolates from burn wounds and their Gómez, The good side of inflammation: Staphylococcus aureus proteins SpA
antibiograms: a 20-year study (1986–2005), Burns 35 (2009) 553–560. and Sbi contribute to proper abscess formation and wound healing during
[30] R.S. Howell-Jones, M.J. Wilson, K.E. Hill, A.J. Howard, P.E. Price, D.W. Thomas, skin and soft tissue infections, Biochim. Biophys. Acta Mol. basis Dis. 2019
A review of the microbiology, antibiotic usage and resistance in chronic skin (1865) 2657–2670.
wounds, J. Antimicrob. Chemother. 55 (2005) 143–149. [53] R.L. Gallo, S. epidermidis influence on host immunity: more than skin deep,
[31] W. Branch-Elliman, J.E. Ripollone, W.J. O’Brien, K.M.F. Itani, M.L. Schweizer, E. Cell Host Microbe, 17 (2015) 143-144.
Perencevich, J. Strymish, K. Gupta, Risk of surgical site infection, acute kidney [54] K. Raziyeva, Y. Kim, Z. Zharkinbekov, K. Kassymbek, S. Jimi, A. Saparov,
injury, and Clostridium difficile infection following antibiotic prophylaxis Immunology of Acute and Chronic Wound Healing, Biomolecules 11 (2021).
with vancomycin plus a beta-lactam versus either drug alone: A national [55] R. Mu, S. Campos de Souza, Z. Liao, L. Dong, C. Wang, Reprograming the
propensity-score-adjusted retrospective cohort study, PLoS Med. 14 (2017) immune niche for skin tissue regeneration - From cellular mechanisms to
e1002340. biomaterials applications, Adv. Drug Deliv. Rev. 185 (2022).
[32] Y.K. Wu, N.C. Cheng, C.M. Cheng, Biofilms in Chronic Wounds: Pathogenesis [56] W. Kim, E.J. Lee, I.H. Bae, K. Myoung, S.T. Kim, P.J. Park, K.H. Lee, A.V.Q. Pham,
and Diagnosis, Trends Biotechnol. 37 (2019) 505–517. J. Ko, S.H. Oh, E.G. Cho, Lactobacillus plantarum-derived extracellular vesicles
[33] L.R. Kalan, J.S. Meisel, M.A. Loesche, J. Horwinski, I. Soaita, X. Chen, A. Uberoi, induce anti-inflammatory M2 macrophage polarization in vitro, J Extracell
S.E. Gardner, E.A. Grice, Strain-and species-level variation in the microbiome Vesicles 9 (2020) 1793514.
of diabetic wounds is associated with clinical outcomes and therapeutic [57] T.A. Wilgus, S. Roy, J.C. McDaniel, Neutrophils and Wound Repair: Positive
efficacy, Cell Host Microbe 25 (2019) 641–655, e645. Actions and Negative Reactions, Adv Wound Care (New Rochelle) 2 (2013)
[34] G. Norman, J.C. Dumville, Z.E. Moore, J. Tanner, J. Christie, S. Goto, Antibiotics 379–388.
and antiseptics for pressure ulcers, Cochrane Database Syst Rev, 4 (2016) [58] H.R. Thiam, S.L. Wong, D.D. Wagner, C.M. Waterman, Cellular Mechanisms of
Cd011586. NETosis, Annu. Rev. Cell Dev. Biol. 36 (2020) 191–218.
[35] M. Azam, H. Ghufran, H. Butt, A. Mehmood, R. Ashfaq, A.M. Ilyas, M.R. Ahmad, [59] N. Sorvillo, D. Cherpokova, K. Martinod, D.D. Wagner, Extracellular DNA NET-
S. Riazuddin, Curcumin preconditioning enhances the efficacy of adipose- Works With Dire Consequences for Health, Circ. Res. 125 (2019) 470–488.
derived mesenchymal stem cells to accelerate healing of burn wounds, Burns [60] V. Brinkmann, U. Reichard, C. Goosmann, B. Fauler, Y. Uhlemann, D.S. Weiss,
& Trauma 9 (2021) tkab021. Y. Weinrauch, A. Zychlinsky, Neutrophil extracellular traps kill bacteria,
[36] J. Di Domizio, C. Belkhodja, P. Chenuet, A. Fries, T. Murray, P.M. Mondéjar, O. Science 303 (2004) 1532–1535.
Demaria, C. Conrad, B. Homey, S. Werner, D.E. Speiser, B. Ryffel, M. Gilliet, The [61] M. Honda, P. Kubes, Neutrophils and neutrophil extracellular traps in the liver
commensal skin microbiota triggers type I IFN-dependent innate repair and gastrointestinal system, Nat. Rev. Gastroenterol. Hepatol. 15 (2018) 206–
responses in injured skin, Nat. Immunol. 21 (2020) 1034–1045. 221.
[37] J.L. Linehan, O.J. Harrison, S.J. Han, A.L. Byrd, I. Vujkovic-Cvijin, A.V. Villarino, [62] F.H. Pilsczek, D. Salina, K.K. Poon, C. Fahey, B.G. Yipp, C.D. Sibley, S.M. Robbins,
S.K. Sen, J. Shaik, M. Smelkinson, S. Tamoutounour, N. Collins, N. Bouladoux, F.H. Green, M.G. Surette, M. Sugai, M.G. Bowden, M. Hussain, K. Zhang, P.
A. Dzutsev, S.P. Rosshart, J.H. Arbuckle, C.R. Wang, T.M. Kristie, B. Rehermann, Kubes, A novel mechanism of rapid nuclear neutrophil extracellular trap
G. Trinchieri, J.M. Brenchley, J.J. O’Shea, Y. Belkaid, Non-classical Immunity formation in response to Staphylococcus aureus, J. Immunol. 185 (2010)
Controls Microbiota Impact on Skin Immunity and Tissue Repair, Cell, 172 7413–7425.
(2018) 784-796 e718. [63] H. Parker, M. Dragunow, M.B. Hampton, A.J. Kettle, C.C. Winterbourn,
[38] V.K. Ridaura, N. Bouladoux, J. Claesen, Y.E. Chen, A.L. Byrd, M.G. Requirements for NADPH oxidase and myeloperoxidase in neutrophil
Constantinides, E.D. Merrill, S. Tamoutounour, M.A. Fischbach, Y. Belkaid, extracellular trap formation differ depending on the stimulus, J. Leukoc.
Contextual control of skin immunity and inflammation by Corynebacterium, Biol. 92 (2012) 841–849.
J. Exp. Med. 215 (2018) 785–799. [64] N. Malachowa, S.D. Kobayashi, K.R. Braughton, A.R. Whitney, M.J. Parnell, D.J.
[39] T. Hoppenbrouwers, A.R. Sultan, T.E. Abraham, N.A. Lemmens-den Toom, S. Gardner, F.R. Deleo, Staphylococcus aureus leukotoxin GH promotes
Hansenova Manaskova, W.A. van Cappellen, A.B. Houtsmuller, W.J.B. van inflammation, J Infect Dis 206 (2012) 1185–1193.
Wamel, M.P.M. de Maat, J.W. van Neck, Staphylococcal Protein A Is a Key [65] E. Kolaczkowska, P. Kubes, Neutrophil recruitment and function in health and
Factor in Neutrophil Extracellular Traps Formation, Front. Immunol. 9 (2018) inflammation, Nat. Rev. Immunol. 13 (2013) 159–175.
165. [66] S.L. Wong, M. Demers, K. Martinod, M. Gallant, Y. Wang, A.B. Goldfine, C.R.
[40] R.Z. Murray, Z.E. West, A.J. Cowin, B.L. Farrugia, Development and use of Kahn, D.D. Wagner, Diabetes primes neutrophils to undergo NETosis, which
biomaterials as wound healing therapies, Burns & Trauma 7 (2019) 2. impairs wound healing, Nat. Med. 21 (2015) 815–819.
[41] S. Naik, N. Bouladoux, J.L. Linehan, S.J. Han, O.J. Harrison, C. Wilhelm, S. [67] M. Hesketh, K.B. Sahin, Z.E. West, R.Z. Murray, Macrophage Phenotypes
Conlan, S. Himmelfarb, A.L. Byrd, C. Deming, M. Quinones, J.M. Brenchley, H. Regulate Scar Formation and Chronic Wound Healing, Int. J. Mol. Sci. 18
H. Kong, R. Tussiwand, K.M. Murphy, M. Merad, J.A. Segre, Y. Belkaid, (2017).
Commensal-dendritic-cell interaction specifies a unique protective skin [68] S.K. Brancato, J.E. Albina, Wound macrophages as key regulators of repair:
immune signature, Nature 520 (2015) 104–108. origin, phenotype, and function, Am. J. Pathol. 178 (2011) 19–25.

13
Q. Tang, N. Xue, X. Ding et al. Advanced Drug Delivery Reviews 192 (2023) 114671

[69] S. Yamakawa, K. Hayashida, Advances in surgical applications of growth on the synthesis of insulin-like growth factor-1 by cdT cells Chin, J Injury and
factors for wound healing, Burns & Trauma 7 (2019) 10. Repair 11 (2016) 260–266.
[70] M. Sharifiaghdam, E. Shaabani, R. Faridi-Majidi, S.C. De Smedt, K. Braeckmans, [96] E.E. Gray, K. Suzuki, J.G. Cyster, Cutting edge: Identification of a motile IL-17-
J.C. Fraire, Macrophages as a therapeutic target to promote diabetic wound producing gammadelta T cell population in the dermis, J. Immunol. 186
healing, Mol. Ther. 30 (2022) 2891–2908. (2011) 6091–6095.
[71] M.L. Novak, T.J. Koh, Macrophage phenotypes during tissue repair, J. Leukoc. [97] J.Z. Liu, M. Pezeshki, M. Raffatellu, Th17 cytokines and host-pathogen
Biol. 93 (2013) 875–881. interactions at the mucosa: dichotomies of help and harm, Cytokine 48
[72] K.L. Spiller, S. Nassiri, C.E. Witherel, R.R. Anfang, J. Ng, K.R. Nakazawa, T. Yu, G. (2009) 156–160.
Vunjak-Novakovic, Sequential delivery of immunomodulatory cytokines to [98] A.V. Nguyen, A.M. Soulika, The Dynamics of the Skin’s Immune System, Int. J.
facilitate the M1-to-M2 transition of macrophages and enhance Mol. Sci. 20 (2019).
vascularization of bone scaffolds, Biomaterials 37 (2015) 194–207. [99] T.A. Harris-Tryon, E.A. Grice, Microbiota and maintenance of skin barrier
[73] S.M. Aitcheson, F.D. Frentiu, S.E. Hurn, K. Edwards, R.Z. Murray, Skin Wound function, Science 376 (2022) 940–9456.
Healing: Normal Macrophage Function and Macrophage Dysfunction in [100] M.D. Caldwell, Bacteria and Antibiotics in Wound Healing, Surg. Clin. North
Diabetic Wounds, Molecules 26 (2021). Am. 100 (2020) 757–776.
[74] A. Sindrilaru, T. Peters, S. Wieschalka, C. Baican, A. Baican, H. Peter, A. Hainzl, [101] C.F. Heal, J.L. Banks, P.D. Lepper, E. Kontopantelis, M.L. van Driel, Topical
S. Schatz, Y. Qi, A. Schlecht, J.M. Weiss, M. Wlaschek, C. Sunderkötter, K. antibiotics for preventing surgical site infection in wounds healing by
Scharffetter-Kochanek, An unrestrained proinflammatory M1 macrophage primary intention, Cochrane Database Syst. Rev. 11 (2016) CD011426.
population induced by iron impairs wound healing in humans and mice, J. [102] C.A. Umscheid, M.D. Mitchell, J.A. Doshi, R. Agarwal, K. Williams, P.J. Brennan,
Clin. Invest. 121 (2011) 985–997. Estimating the proportion of healthcare-associated infections that are
[75] D. Zhang, Z. Tang, H. Huang, G. Zhou, C. Cui, Y. Weng, W. Liu, S. Kim, S. Lee, M. reasonably preventable and the related mortality and costs, Infect. Control
Perez-Neut, J. Ding, D. Czyz, R. Hu, Z. Ye, M. He, Y.G. Zheng, H.A. Shuman, L. Hosp. Epidemiol. 32 (2011) 101–114.
Dai, B. Ren, R.G. Roeder, L. Becker, Y. Zhao, Metabolic regulation of gene [103] J.C. Alverdy, N. Hyman, J. Gilbert, Re-examining causes of surgical site
expression by histone lactylation, Nature 574 (2019) 575–580. infections following elective surgery in the era of asepsis, Lancet Infect. Dis.
[76] J. Zhang, J. Muri, G. Fitzgerald, T. Gorski, R. Gianni-Barrera, E. Masschelein, G. 20 (2020) e38–e43.
D’Hulst, P. Gilardoni, G. Turiel, Z. Fan, T. Wang, M. Planque, P. Carmeliet, L. [104] G.D. Hannigan, B.P. Hodkinson, K. McGinnis, A.S. Tyldsley, J.B. Anari, A.D.
Pellerin, C. Wolfrum, S.M. Fendt, A. Banfi, C. Stockmann, I. Soro-Arnáiz, M. Horan, E.A. Grice, S. Mehta, Culture-independent pilot study of microbiota
Kopf, K. De Bock, Endothelial Lactate Controls Muscle Regeneration from colonizing open fractures and association with severity, mechanism, location,
Ischemia by Inducing M2-like Macrophage Polarization, Cell Metab. 31 and complication from presentation to early outpatient follow-up, J. Orthop.
(2020) 1136–1153.e1137. Res. 32 (2014) 597–605.
[77] Y. Lu, H. Li, J. Wang, M. Yao, Y. Peng, T. Liu, Z. Li, G. Luo, J. Deng, Engineering [105] S. Romano-Bertrand, J.M. Frapier, B. Calvet, P. Colson, B. Albat, S. Parer, E.
Bacteria-Activated Multifunctionalized Hydrogel for Promoting Diabetic Jumas-Bilak, Dynamics of the surgical microbiota along the cardiothoracic
Wound Healing, Adv. Funct. Mater. 31 (2021). surgery pathway, Front. Microbiol. 5 (2014) 787.
[78] J.H. Kim, J. Lee, J. Park, Y.S. Gho, Gram-negative and Gram-positive bacterial [106] M.C. Robson, Wound infection: A failure of wound healing caused by an
extracellular vesicles, Semin. Cell Dev. Biol. 40 (2015) 97–104. imbalance of bacteria, Surg. Clin. N. Am. 77 (1997) 637–650.
[79] S. Bose, S. Aggarwal, D.V. Singh, N. Acharya, Extracellular vesicles: An [107] Z. Xu, H.C. Hsia, The Impact of Microbial Communities on Wound Healing: A
emerging platform in gram-positive bacteria, Microb, Cell 7 (2020) 312–322. Review, Ann. Plast. Surg. 81 (2018) 113–123.
[80] S.N. Mueller, A. Zaid, F.R. Carbone, Tissue-resident T cells: dynamic players in [108] C. Bartow-McKenney, G.D. Hannigan, J. Horwinski, P. Hesketh, A.D. Horan, S.
skin immunity, Front. Immunol. 5 (2014) 332. Mehta, E.A. Grice, The microbiota of traumatic, open fracture wounds is
[81] I. Lerman, D.C. Mitchell, C.T. Richardson, Human cutaneous B cells: what do associated with mechanism of injury, Wound Repair Regen. 26 (2018) 127–
we really know?, Ann Transl Med 9 (2021) 440 135.
[82] W. Hoffman, F.G. Lakkis, G. Chalasani, B. Cells, Antibodies, and More, Clin. J. [109] M.H. Swaney, L.R. Kalan, Living in Your Skin: Microbes, Molecules, and
Am. Soc. Nephrol. 11 (2016) 137–154. Mechanisms, Infect. Immun. 89 (2021).
[83] Y. Iwata, A. Yoshizaki, K. Komura, K. Shimizu, F. Ogawa, T. Hara, E. Muroi, S. [110] T.R. Johnson, B.I. Gómez, M.K. McIntyre, M.A. Dubick, R.J. Christy, S.E.
Bae, M. Takenaka, T. Yukami, M. Hasegawa, M. Fujimoto, Y. Tomita, T.F. Nicholson, D.M. Burmeister, The Cutaneous Microbiome and Wounds: New
Tedder, S. Sato, CD19, a response regulator of B lymphocytes, regulates Molecular Targets to Promote Wound Healing, Int. J. Mol. Sci. 19 (2018).
wound healing through hyaluronan-induced TLR4 signaling, Am. J. Pathol. [111] C. Leonel, I.F.G. Sena, W.N. Silva, P. Prazeres, G.R. Fernandes, P. Mancha
175 (2009) 649–660. Agresti, M. Martins Drumond, A. Mintz, V.A.C. Azevedo, A. Birbrair,
[84] R.F. Sîrbulescu, C.K. Boehm, E. Soon, M.Q. Wilks, I. Iliesß, H. Yuan, B. Maxner, N. Staphylococcus epidermidis role in the skin microenvironment, J. Cell Mol.
Chronos, C. Kaittanis, M.D. Normandin, G. El Fakhri, D.P. Orgill, A.E. Sluder, M. Med. 23 (2019) 5949–5955.
C. Poznansky, Mature B cells accelerate wound healing after acute and [112] S. Kirchner, V. Lei, A.S. MacLeod, The Cutaneous Wound Innate
chronic diabetic skin lesions, Wound Repair Regen. 25 (2017) 774–791. Immunological Microenvironment, Int. J. Mol. Sci. 21 (2020).
[85] C. Chen, Z. Meng, H. Ren, N. Zhao, R. Shang, W. He, J. Hao, The molecular [113] R. Knackstedt, T. Knackstedt, J. Gatherwright, The role of topical probiotics on
mechanisms supporting the homeostasis and activation of dendritic wound healing: A review of animal and human studies, Int. Wound J. 17
epidermal T cell and its role in promoting wound healing, Burns & Trauma (2020) 1687–1694.
9 (2021) tkab009. [114] P.G. Bowler, B.I. Duerden, D.G. Armstrong, Wound microbiology and
[86] Y. Cai, X. Shen, C. Ding, C. Qi, K. Li, X. Li, V.R. Jala, H.G. Zhang, T. Wang, J. Zheng, associated approaches to wound management, Clin. Microbiol. Rev. 14
J. Yan, Pivotal role of dermal IL-17-producing cd T cells in skin inflammation, (2001) 244–269.
Immunity 35 (2011) 596–610. [115] G. Norman, C. Shi, M.J. Westby, B.L. Price, A.J. McBain, J.C. Dumville, N.
[87] A.S. Marshall, J.R. Silva, C.A. Bannerman, I. Gilron, N. Ghasemlou, Skin- Cullum, Bacteria and bioburden and healing in complex wounds: A
Resident cd T Cells Exhibit Site-Specific Morphology and Activation States, J. prognostic systematic review, Wound Repair Regen. 29 (2021) 466–477.
Immunol. Res. 2019 (2019) 9020234. [116] I.C. Chetter, A.V. Oswald, E. McGinnis, N. Stubbs, C. Arundel, H. Buckley, K.
[88] J.D. Bos, M.B. Teunissen, I. Cairo, S.R. Krieg, M.L. Kapsenberg, P.K. Das, J. Borst, Bell, J.C. Dumville, N.A. Cullum, M.O. Soares, P. Saramago, Patients with
T-cell receptor gamma delta bearing cells in normal human skin, J, Invest. surgical wounds healing by secondary intention: A prospective, cohort study,
Dermatol. 94 (1990) 37–42. Int. J. Nurs. Stud. 89 (2019) 62–71.
[89] A. Toulon, L. Breton, K.R. Taylor, M. Tenenhaus, D. Bhavsar, C. Lanigan, R. [117] J.K. Plichta, X. Gao, H. Lin, Q. Dong, E. Toh, D.E. Nelson, R.L. Gamelli, E.A. Grice,
Rudolph, J. Jameson, W.L. Havran, A role for human skin-resident T cells in K.A. Radek, Cutaneous Burn Injury Promotes Shifts in the Bacterial
wound healing, J. Exp. Med. 206 (2009) 743–750. Microbiome in Autologous Donor Skin: Implications for Skin Grafting
[90] S. Shibata, Y. Tada, C.S. Hau, A. Mitsui, M. Kamata, Y. Asano, M. Sugaya, T. Outcomes, Shock 48 (2017) 441–448.
Kadono, Y. Masamoto, M. Kurokawa, T. Yamauchi, N. Kubota, T. Kadowaki, S. [118] A.J. SanMiguel, J.S. Meisel, J. Horwinski, Q. Zheng, C.W. Bradley, E.A. Grice,
Sato, Adiponectin regulates psoriasiform skin inflammation by suppressing Antiseptic Agents Elicit Short-Term, Personalized, and Body Site-Specific
IL-17 production from cd-T cells, Nat. Commun. 6 (2015) 7687. Shifts in Resident Skin Bacterial Communities, J, Invest. Dermatol. 138 (2018)
[91] G. Crawford, M.D. Hayes, R.C. Seoane, S. Ward, T. Dalessandri, C. Lai, E. Healy, 2234–2243.
D. Kipling, C. Proby, C. Moyes, K. Green, K. Best, M. Haniffa, M. Botto, D. Dunn- [119] D. Dörfel, M. Maiwald, G. Daeschlein, G. Müller, R. Hudek, O. Assadian, G.
Walters, J. Strid, Epithelial damage and tissue cd T cells promote a unique Kampf, T. Kohlmann, J.C. Harnoss, A. Kramer, Comparison of the antimicrobial
tumor-protective IgE response, Nat. Immunol. 19 (2018) 859–870. efficacy of povidone-iodine-alcohol versus chlorhexidine-alcohol for surgical
[92] M. Girardi, D.E. Oppenheim, C.R. Steele, J.M. Lewis, E. Glusac, R. Filler, P. skin preparation on the aerobic and anaerobic skin flora of the shoulder
Hobby, B. Sutton, R.E. Tigelaar, A.C. Hayday, Regulation of cutaneous region, Antimicrob. Resist. Infect. Control 10 (2021) 17.
malignancy by gammadelta T cells, Science 294 (2001) 605–609. [120] M. Zhang, Z. Jiang, D. Li, D. Jiang, Y. Wu, H. Ren, H. Peng, Y. Lai, Oral antibiotic
[93] C.K. Mantri, A.L. St John, Immune synapses between mast cells and cd T cells treatment induces skin microbiota dysbiosis and influences wound healing,
limit viral infection, J. Clin. Invest. 129 (2019) 1094–1108. Microb. Ecol. 69 (2015) 415–421.
[94] H.K. Komori, D.A. Witherden, R. Kelly, K. Sendaydiego, J.M. Jameson, L. Teyton, [121] A.A. Chan, E.A. Flores, M. Navarrete, D. Phan Tran, D.J. Lee, L.G. Miller, The
W.L. Havran, Cutting edge: dendritic epidermal cd T cell ligands are rapidly Effect of Systemic Antibiotics for Suppurative Skin and Soft Tissue Infections
and locally expressed by keratinocytes following cutaneous wounding, J. on the Skin Microbiome, Open Forum, Infect. Dis. 9 (2022) ofac141.
Immunol. 188 (2012) 2972–2976. [122] G. Norman, J.C. Dumville, D.P. Mohapatra, G.L. Owens, E.J. Crosbie, Antibiotics
[95] X.D. Deng, J. Wang, M.M. Zhang, P.P. Wang, B.X. Su, Q.W. Cui, M.Q. Xu, C.Y. Jia, and antiseptics for surgical wounds healing by secondary intention, Cochrane
Effect of Pseudomonas aeruginosa density-sensing signaling molecule OdDHL Database Syst. Rev. 3 (2016) CD011712.

14
Q. Tang, N. Xue, X. Ding et al. Advanced Drug Delivery Reviews 192 (2023) 114671

[123] J.W. Costerton, P.S. Stewart, E.P. Greenberg, Bacterial biofilms: a common [151] Y. Ashoori, M. Mohkam, R. Heidari, S.N. Abootalebi, S.M. Mousavi, S.A.
cause of persistent infections, Science 284 (1999) 1318–1322. Hashemi, N. Golkar, A. Gholami, Development and in vivo characterization of
[124] L. Satish, P.H. Gallo, S. Johnson, C.C. Yates, S. Kathju, Local Probiotic Therapy probiotic lysate-treated chitosan nanogel as a novel biocompatible
with Lactobacillus plantarum Mitigates Scar Formation in Rabbits after Burn formulation for wound healing, Biomed Res. Int. 2020 (2020).
Injury and Infection, Surg. Infect. (Larchmt) 18 (2017) 119–127. [152] M.A. Khan, Z. Hussain, S. Ali, Z. Qamar, M. Imran, F.Y. Hafeez, Fabrication of
[125] K. Credito, G. Lin, P.C. Appelbaum, Antistaphylococcal activity of DX-619 electrospun probiotic functionalized nanocomposite scaffolds for infection
alone and in combination with vancomycin, teicoplanin, and linezolid control and dermal burn healing in a mice model, ACS Biomater Sci. Eng. 5
assessed by time-kill synergy testing, Antimicrob. Agents Chemother. 51 (2019) 6109–6116.
(2007) 1508–1511. [153] Z. Li, A.M. Behrens, N. Ginat, S.Y. Tzeng, X. Lu, S. Sivan, R. Langer, A. Jaklenec,
[126] F. Du, J. Ma, H. Gong, R. Bista, P. Zha, Y. Ren, Y. Gao, D. Chen, X. Ran, C. Wang, Biofilm-inspired encapsulation of probiotics for the treatment of complex
Microbial Infection and Antibiotic Susceptibility of Diabetic Foot Ulcer in infections, Adv. Mater. 30 (2018) 1803925.
China: Literature Review, Front Endocrinol (Lausanne) 13 (2022). [154] C. Prakoeswa, N. Herwanto, R. Prameswari, L. Astari, S. Sawitri, A. Hidayati, D.
[127] O.I. Butranova, T.N. Razdrogina, Antibiotics for skin and soft tissues infections Indramaya, E. Kusumowidagdo, I. Surono, Lactobacillus plantarum IS-10506
in type 2 diabetes mellitus, Int. J. Risk Saf. Med. 27 (Suppl 1) (2015) S57–S58. supplementation reduced SCORAD in children with atopic dermatitis,
[128] B.A. Lipsky, É. Senneville, Z.G. Abbas, J. Aragón-Sánchez, M. Diggle, J.M. Embil, Benefic. Microbes 8 (2017) 833–840.
S. Kono, L.A. Lavery, M. Malone, S.A. van Asten, V. Urbančič-Rovan, E.J.G. [155] G. Sharma, M. Sharma, R. Sood, J. Neelamraju, S.G. Lakshmi, R.S. Madempudi,
Peters, Guidelines on the diagnosis and treatment of foot infection in persons P. Rishi, I.P. Kaur, Self-preserving gelatin emulgel containing whole cell
with diabetes (IWGDF 2019 update), Diabetes Metab. Res. Rev. 36 (Suppl 1) probiotic for topical use: Preclinical safety, efficacy, and germination studies,
(2020) e3280. Expert Opin. Drug Deliv. 18 (2021) 1777–1789.
[129] D.G. Armstrong, A.J.M. Boulton, S.A. Bus, Diabetic Foot Ulcers and Their [156] Y. Zhu, Z. Wang, L. Bai, J. Deng, Q. Zhou, Biomaterial-based encapsulated
Recurrence, N. Engl. J. Med. 376 (2017) 2367–2375. probiotics for biomedical applications: Current status and future
[130] C. Dunyach-Remy, A. Cadière, J.L. Richard, S. Schuldiner, S. Bayle, B. Roig, A. perspectives, Mater. Des. 210 (2021).
Sotto, J.P. Lavigne, Polymerase chain reaction-denaturing gradient gel [157] A. Das, S. Ray, U. Raychaudhuri, R. Chakraborty, Microencapsulation of
electrophoresis (PCR-DGGE): a promising tool to diagnose bacterial probiotic bacteria and its potential application in food technology,
infections in diabetic foot ulcers, Diabetes Metab. 40 (2014) 476–480. International Journal of Agriculture, Environment and Biotechnology 7
[131] S. Verbanic, Y. Shen, J. Lee, J.M. Deacon, I.A. Chen, Microbial predictors of (2014) 47–53.
healing and short-term effect of debridement on the microbiome of chronic [158] L.P. Ta, E. Bujna, O. Antal, M. Ladányi, R. Juhász, A. Szécsi, S. Kun, S. Sudheer, V.
wounds, npj Biofilms Microbiomes 6 (2020) 21. K. Gupta, Q.D. Nguyen, Effects of various polysaccharides (alginate,
[132] A. MacDonald, J.D. Brodell Jr., J.L. Daiss, E.M. Schwarz, I. Oh, Evidence of carrageenan, gums, chitosan) and their combination with prebiotic
differential microbiomes in healing versus non-healing diabetic foot ulcers saccharides (resistant starch, lactosucrose, lactulose) on the encapsulation
prior to and following foot salvage therapy, J. Orthop. Res. 37 (2019) 1596– of probiotic bacteria Lactobacillus casei 01 strain, Int. J. Biol. Macromol. 183
1603. (2021) 1136–1144.
[133] I.M. Michalek, K. Mitura, A. Krechowska, F.L. Caetano Dos Santos, Microbiota [159] A.K. Anal, H. Singh, Recent advances in microencapsulation of probiotics for
and Its Antibiotic Susceptibility in Diabetic Foot Infections: Observations industrial applications and targeted delivery, Trends Food Sci. Technol. 18
From Polish Nonmetropolitan Hospital, 2015–2016, Int. J. Low. Extrem. (2007) 240–251.
Wounds 1534734620953686 (2020). [160] M.A. Matica, F.L. Aachmann, A. Tøndervik, H. Sletta, V. Ostafe, Chitosan as a
[134] D.D. Sprockett, C.G. Ammons, M.S. Tuttle, Use of 16S rRNA sequencing and Wound Dressing Starting Material: Antimicrobial Properties and Mode of
quantitative PCR to correlate venous leg ulcer bacterial bioburden dynamics Action, Int. J. Mol. Sci. 20 (2019).
with wound expansion, antibiotic therapy, and healing, Wound Repair Regen. [161] S. Alven, B.A. Aderibigbe, Chitosan and Cellulose-Based Hydrogels for Wound
23 (2015) 765–771. Management, Int. J. Mol. Sci. 21 (2020).
[135] L. Bay, K.N. Kragh, S.R. Eickhardt, S.S. Poulsen, L.M.R. Gjerdrum, K. Ghathian, [162] R. Singh, K. Shitiz, A. Singh, Chitin and chitosan: biopolymers for wound
H. Calum, M.S. Ågren, T. Bjarnsholt, Bacterial Aggregates Establish at the management, Int. Wound J. 14 (2017) 1276–1289.
Edges of Acute Epidermal Wounds, Adv Wound Care (New Rochelle) 7 (2018) [163] I. Kwiecień, M. Kwiecień, Application of polysaccharide-based hydrogels as
105–113. probiotic delivery systems, Gels 4 (2018) 47.
[136] F. Naeem, F.R. Anjum, M.A. Arshad, A.A. Bukhari, B. Aslam, J.A. Khan, M.I. [164] M. Yao, J. Xie, H. Du, D.J. McClements, H. Xiao, L. Li, Progress in
Arshad, Isolation and antibiotic sensitivity pattern of drug resistant bacteria microencapsulation of probiotics: A review, Compr. Rev. Food Sci. Food Saf.
in ulcerative foot of type 2 diabetic patients, Pak. J. Pharm. Sci. 32 (2019) 19 (2020) 857–874.
1843–1848. [165] P. Ebrahimnezhad, M. Khavarpour, S. Khalili, Survival of Lactobacillus
[137] M.S. Bader, Diabetic foot infection, Am. Fam. Physician 78 (2008) 71–79. acidophilus as probiotic bacteria using chitosan nanoparticles, Int. J. Eng.
[138] M.V. Schintler, Negative pressure therapy: theory and practice, Diabetes 30 (2017) 456–463.
Metab. Res. Rev. 28 (Suppl 1) (2012) 72–77. [166] L.-F. Călinoiu, B.E. S ß tefănescu, I.D. Pop, L. Muntean, D.C. Vodnar,
[139] P. Hunter, E. Greco, K. Cross, J. Perry, Topical Oxygen Therapy Shifts Chitosan coating applications in probiotic microencapsulation, Coatings 9
Microbiome Dynamics in Chronic Diabetic Foot Ulcers, Wounds 32 (2020) (2019) 194.
81–85. [167] I. Hassan, A. Gani, Z.U. Ashraf, Simulated Gastrointestinal System to Assess
ß imsek, T. Kula
[140] R.A. Çetinkaya, E. Yenilmez, P. Petrone, S. Yılmaz, B. Bektöre, B. S the Probiotic Properties Modified to Encapsulation of Probiotics and Their
Atik, M. Özyurt, A. Ünlü, Platelet-rich plasma as an additional therapeutic Survival Under Simulated Gastrointestinal System, Advances in Probiotics
option for infected wounds with multi-drug resistant bacteria: in vitro (Elsevier 2021,) 37–44.
antibacterial activity study, Eur. J. Trauma Emerg. Surg. 45 (2019) 555–565. [168] I.A. Duceac, S. Coseri, Biopolymers and their derivatives: Key components of
[141] D. Davies, Understanding biofilm resistance to antibacterial agents, Nat. Rev. advanced biomedical technologies, Biotechnol. Adv. 61 (2022).
Drug Discov. 2 (2003) 114–122. [169] D.R. Sahoo, T. Biswal, Alginate and its application to tissue engineering, SN
[142] A.J. SanMiguel, J.S. Meisel, J. Horwinski, Q. Zheng, E.A. Grice, Topical Applied Sciences 3 (2021) 1–19.
Antimicrobial Treatments Can Elicit Shifts to Resident Skin Bacterial [170] J. Kim, S.P. Hlaing, J. Lee, A. Saparbayeva, S. Kim, D.S. Hwang, E.H. Lee, I.-S.
Communities and Reduce Colonization by Staphylococcus aureus Yoon, H. Yun, M.-S. Kim, Exfoliated bentonite/alginate nanocomposite
Competitors, Antimicrob. Agents Chemother. 61 (2017). hydrogel enhances intestinal delivery of probiotics by resistance
[143] L. Morelli, L. Capurso, FAO/WHO guidelines on probiotics: 10 years later, J. to gastric pH and on-demand disintegration, Carbohydr. Polym.
Clin. Gastroenterol. 46 (Suppl) (2012) S1–S2. 272 (2021).
[144] P. Srivastava, T. Sondak, K. Sivashanmugam, K.S. Kim, A Review of [171] G. Alharaty, H.S. Ramaswamy, The effect of sodium alginate-calcium chloride
Immunomodulatory Reprogramming by Probiotics in Combating Chronic coating on the quality parameters and shelf life of strawberry cut fruits,
and Acute Diabetic Foot Ulcers (DFUs), Pharmaceutics 14 (2022). Journal of Composites Science 4 (2020) 123.
[145] Y. Yu, S. Dunaway, J. Champer, J. Kim, A. Alikhan, Changing our microbiome: [172] M. Hashino, T. Katagiri, N. Kubota, Y. Ohmukai, T. Maruyama, H. Matsuyama,
probiotics in dermatology, Br. J. Dermatol. 182 (2020) 39–46. Effect of membrane surface morphology on membrane fouling with sodium
[146] M. Habeebuddin, R.K. Karnati, P.N. Shiroorkar, S. Nagaraja, S.M.B. Asdaq, M. alginate, J. Membr. Sci. 366 (2011) 258–265.
Khalid Anwer, S. Fattepur, Topical Probiotics: More Than a Skin Deep, [173] S. Gantait, S. Kundu, L. Yeasmin, M.N. Ali, Impact of differential levels of
Pharmaceutics 14 (2022). sodium alginate, calcium chloride and basal media on germination frequency
[147] L. Mei, D. Zhang, H. Shao, Y. Hao, T. Zhang, W. Zheng, Y. Ji, P. Ling, Y. Lu, Q. of genetically true artificial seeds of Rauvolfia serpentina (L.) Benth. ex Kurz,
Zhou, Injectable and Self-Healing Probiotics-Loaded Hydrogel for Promoting J. Appl. Res. Med. Aromat. Plants 4 (2017) 75–81.
Superbacteria-Infected Wound Healing, ACS Appl. Mater. Interfaces (2022). [174] P. Yu, W. Zhong, Hemostatic materials in wound care, Burns & Trauma 9
[148] H.F. Huseini, G. Rahimzadeh, M.R. Fazeli, M. Mehrazma, M. Salehi, Evaluation (2021).
of wound healing activities of kefir products, Burns 38 (2012) 719–723. [175] A. Thomas, K.G. Harding, K. Moore, Alginates from wound dressings activate
[149] Wirth, Thomas, Yrjnheikki, Juha, Samaranayake, Haritha., Recombinant human macrophages to secrete tumour necrosis factor-a, Biomaterials 21
probiotic bacteria for use in the treatment of askindys function, (2000) 1797–1802.
WO2016124239, (2016) 08-11. [176] J. Necas, L. Bartosikova, Carrageenan: a review, Vet. Med. 58 (2013).
[150] E. Vågesjö, E. Öhnstedt, A. Mortier, H. Lofton, F. Huss, P. Proost, S. Roos, M. [177] A. Noreen, J. Akram, I. Rasul, A. Mansha, N. Yaqoob, R. Iqbal, S. Tabasum, M.
Phillipson, Accelerated wound healing in mice by on-site production and Zuber, K.M. Zia, Pectins functionalized biomaterials; a new viable approach
delivery of CXCL12 by transformed lactic acid bacteria, PNAS 115 (2018) for biomedical applications: A review, Int. J. Biol. Macromol. 101 (2017) 254–
1895–1900. 272.

15
Q. Tang, N. Xue, X. Ding et al. Advanced Drug Delivery Reviews 192 (2023) 114671

[178] K.M. Zia, S. Tabasum, M. Nasif, N. Sultan, N. Aslam, A. Noreen, M. Zuber, A [199] E. Castro-Aguirre, F. Iñiguez-Franco, H. Samsudin, X. Fang, R. Auras, Poly
review on synthesis, properties and applications of natural polymer based (lactic acid)-Mass production, processing, industrial applications, and end of
carrageenan blends and composites, Int. J. Biol. Macromol. 96 (2017) 282– life, Adv. Drug Deliv. Rev. 107 (2016) 333–366.
301. [200] H. Yu, W. Liu, D. Li, C. Liu, Z. Feng, B. Jiang, Targeting delivery system for
[179] A.L. Daniel-da-Silva, S. Fateixa, A.J. Guiomar, B.F. Costa, N.J. Silva, T. Trindade, Lactobacillus plantarum based on functionalized electrospun nanofibers,
B.J. Goodfellow, A.M. Gil, Biofunctionalized magnetic hydrogel nanospheres Polymers 12 (2020) 1565.
of magnetite and j-carrageenan, Nanotechnology 20 (2009). [201] R.S. Stilhano, J.L. Madrigal, K. Wong, P.A. Williams, P.K. Martin, F.S.
[180] S. Doherty, M. Auty, C. Stanton, R. Ross, G. Fitzgerald, A. Brodkorb, Application Yamaguchi, V.Y. Samoto, S.W. Han, E.A. Silva, Injectable alginate hydrogel
of whey protein micro-bead coatings for enhanced strength and probiotic for enhanced spatiotemporal control of lentivector delivery in murine
protection during fruit juice storage and gastric incubation, J. Microencapsul. skeletal muscle, J. Control. Release 237 (2016) 42–49.
29 (2012) 713–728. [202] H. Lu, L. Yuan, X. Yu, C. Wu, D. He, J. Deng, Recent advances of on-demand
[181] R. Yegappan, V. Selvaprithiviraj, S. Amirthalingam, R. Jayakumar, Carrageenan dissolution of hydrogel dressings, Burns & Trauma 6 (2018) 35.
based hydrogels for drug delivery, tissue engineering and wound healing, [203] L. Yang, Z. Han, C. Chen, Z. Li, S. Yu, Y. Qu, R. Zeng, Novel probiotic-bound
Carbohydr. Polym. 198 (2018) 385–400. oxidized Bletilla striata polysaccharide-chitosan composite hydrogel, Mater
[182] A. Palaniraj, V. Jayaraman, Production, recovery and applications of xanthan Sci Eng C Mater Biol Appl 117 (2020).
gum by Xanthomonas campestris, J. Food Eng. 106 (2011) 1–12. [204] M. Lufton, O. Bustan, B.H. Eylon, E. Shtifman-Segal, T. Croitoru-Sadger, A.
[183] G. Singhvi, N. Hans, N. Shiva, S.K. Dubey, Xanthan gum in drug delivery Shagan, A. Shabtay-Orbach, E. Corem-Salkmon, J. Berman, A. Nyska,
applications, Natural polysaccharides in drug delivery and biomedical Antifungal Hydrogels: Living Bacteria in Thermoresponsive Gel for Treating
applications, Elsevier (2019) 121–144. Fungal Infections (Adv. Funct. Mater. 40/2018), Adv. Funct. Mater. 28 (2018).
[184] K. Oberoi, A. Tolun, Z. Altintas, S. Sharma, Effect of alginate- [205] D.-W. Zheng, K.-W. Chen, J.-H. Yan, Z.-Y. Rao, C.-H. Yang, R.-L. Li, Y. Tang, H.
microencapsulated hydrogels on the survival of lactobacillus rhamnosus Cheng, X.-Z. Zhang, A seed-like hydrogel with metabolic cascade microbiota
under simulated gastrointestinal conditions, Foods 10 (2021) 1999. for oral treatment of liver failure, Mater. Today (2022).
[185] L.D.H. Zhang, Recent Advances in Probiotics Encapsulation by [206] R.D. Pedde, B. Mirani, A. Navaei, T. Styan, S. Wong, M. Mehrali, A. Thakur, N.K.
Electrospinning, ES Food & Agroforestry 2 (2020) 3–12. Mohtaram, A. Bayati, A. Dolatshahi-Pirouz, M. Nikkhah, S.M. Willerth, M.
[186] N. Amiri, S. Ajami, A. Shahroodi, N. Jannatabadi, S. Amiri Darban, B.S. Fazly Akbari, Emerging Biofabrication Strategies for Engineering Complex Tissue
Bazzaz, E. Pishavar, F. Kalalinia, J. Movaffagh, Teicoplanin-loaded chitosan- Constructs, Adv. Mater. 29 (2017).
PEO nanofibers for local antibiotic delivery and wound healing, Int. J. Biol. [207] J. Wang, Y. Zhang, N.H. Aghda, A.R. Pillai, R. Thakkar, A. Nokhodchi, M.
Macromol. 162 (2020) 645–656. Maniruzzaman, Emerging 3D printing technologies for drug delivery devices:
[187] J. Zhang, K. Chen, C. Ding, S. Sun, Y. Zheng, Q. Ding, B. Hong, W. Liu, Current status and future perspective, Adv. Drug Deliv. Rev. 174 (2021) 294–
Fabrication of chitosan/PVP/dihydroquercetin nanocomposite film for in vitro 316.
and in vivo evaluation of wound healing, Int. J. Biol. Macromol. 206 (2022) [208] S.V. Murphy, A. Atala, 3D bioprinting of tissues and organs, Nat. Biotechnol.
591–604. 32 (2014) 773–785.
[188] M. Hajikhani, Z. Emam-Djomeh, G. Askari, Fabrication and characterization of [209] A.S. Moghaddam, H.A. Khonakdar, M. Arjmand, S.H. Jafari, Z. Bagher, Z.S.
mucoadhesive bioplastic patch via coaxial polylactic acid (PLA) based Moghaddam, M. Chimerad, M.M. Sisakht, S. Shojaei, Review of Bioprinting in
electrospun nanofibers with antimicrobial and wound healing application, Regenerative Medicine: Naturally Derived Bioinks and Stem Cells, ACS Appl
Int. J. Biol. Macromol. 172 (2021) 143–153. Bio Mater 4 (2021) 4049–4070.
[189] P. Basu, A. Repanas, A. Chatterjee, B. Glasmacher, U. NarendraKumar, I. [210] Z. Sartawi, C. Blackshields, W. Faisal, Dissolving microneedles: Applications
Manjubala, PEO–CMC blend nanofibers fabrication by electrospinning for soft and growing therapeutic potential, J. Control. Release 348 (2022) 186–205.
tissue engineering applications, Mater. Lett. 195 (2017) 10–13. [211] L. Barnum, M. Samandari, T.A. Schmidt, A. Tamayol, Microneedle arrays for
[190] K. Škrlec, Š. Zupančič, S.P. Mihevc, P. Kocbek, J. Kristl, A. Berlec, Development the treatment of chronic wounds, Expert Opin. Drug Deliv. 17 (2020) 1767–
of electrospun nanofibers that enable high loading and long-term viability of 1780.
probiotics, Eur. J. Pharm. Biopharm. 136 (2019) 108–119. [212] Y. Wang, H. Lu, M. Guo, J. Chu, B. Gao, B. He, Personalized and Programmable
[191] S. Razavi, S. Janfaza, N. Tasnim, D.L. Gibson, M. Hoorfar, Microencapsulating Microneedle Dressing for Promoting Wound Healing, Adv. Healthc. Mater. 11
polymers for probiotics delivery systems: Preparation, characterization, and (2022) e2101659.
applications, Food Hydrocoll. 120 (2021). [213] K. Lee, Y. Xue, J. Lee, H.J. Kim, Y. Liu, P. Tebon, E. Sarikhani, W. Sun, S. Zhang, R.
[192] K.S. Masters, S.J. Leibovich, P. Belem, J.L. West, L.A. Poole-Warren, Effects of Haghniaz, B. Çelebi-Saltik, X. Zhou, S. Ostrovidov, S. Ahadian, N. Ashammakhi,
nitric oxide releasing poly(vinyl alcohol) hydrogel dressings on dermal M.R. Dokmeci, A. Khademhosseini, A Patch of Detachable Hybrid Microneedle
wound healing in diabetic mice, Wound Repair Regen. 10 (2002) 286–294. Depot for Localized Delivery of Mesenchymal Stem Cells in Regeneration
[193] L. Zhao, L. Niu, H. Liang, H. Tan, C. Liu, F. Zhu, pH and Glucose Dual- Therapy, Adv. Funct. Mater. 30 (2020).
Responsive Injectable Hydrogels with Insulin and Fibroblasts as Bioactive [214] H.J. Chen, D.A. Lin, F. Liu, L. Zhou, D. Liu, Z. Lin, C. Yang, Q. Jin, T. Hang, G. He,
Dressings for Diabetic Wound Healing, ACS Appl. Mater. Interfaces 9 (2017) X. Xie, Transdermal Delivery of Living and Biofunctional Probiotics through
37563–37574. Dissolvable Microneedle Patches, ACS Appl Bio Mater 1 (2018) 374–381.
[194] N. Charernsriwilaiwat, T. Rojanarata, T. Ngawhirunpat, P. Opanasopit, [215] R. Jamaledin, C.K.Y. Yiu, E.N. Zare, L.N. Niu, R. Vecchione, G. Chen, Z. Gu, F.R.
Electrospun chitosan/polyvinyl alcohol nanofibre mats for wound healing, Tay, P. Makvandi, Advances in Antimicrobial Microneedle Patches for
Int. Wound J. 11 (2014) 215–222. Combating Infections, Adv. Mater. 32 (2020) e2002129.
[195] S. Alven, B.A. Aderibigbe, Fabrication of Hybrid Nanofibers from Biopolymers [216] M. Mir, A.D. Permana, N. Ahmed, G.M. Khan, A.U. Rehman, R.F. Donnelly,
and Poly (Vinyl Alcohol)/Poly (e-Caprolactone) for Wound Dressing Enhancement in site-specific delivery of carvacrol for potential treatment of
Applications, Polymers (Basel) 13 (2021). infected wounds using infection responsive nanoparticles loaded into
[196] M.E. Cam, S. Yildiz, H. Alenezi, S. Cesur, G.S. Ozcan, G. Erdemir, U. Edirisinghe, dissolving microneedles: A proof of concept study, Eur. J. Pharm. Biopharm.
D. Akakin, D.S. Kuruca, L. Kabasakal, O. Gunduz, M. Edirisinghe, Evaluation of 147 (2020) 57–68.
burst release and sustained release of pioglitazone-loaded fibrous mats on [217] I. Woodhouse, S. Nejati, V. Selvamani, H. Jiang, S. Chittiboyina, J. Grant, Z.
diabetic wound healing: an in vitro and in vivo comparison study, J. R. Soc. Mutlu, J. Waimin, N.S. Abutaleb, M.N. Seleem, R. Rahimi, Flexible Microneedle
Interface 17 (2020) 20190712. Array Patch for Chronic Wound Oxygenation and Biofilm Eradication, ACS
[197] O. Berezina, A. Vasilyeva, N. Sidorova, A. Savushkin, N. Marcova, The effect of Appl Bio Mater 4 (2021) 5405–5415.
polyvinylpyrrolidone nanowires on the metabolic activity of Lactobacillus [218] J. Xu, R. Danehy, H. Cai, Z. Ao, M. Pu, A. Nusawardhana, D. Rowe-Magnus, F.
acidophilus, IOP Conference Series: Materials Science and Engineering, IOP Guo, Microneedle Patch-Mediated Treatment of Bacterial Biofilms, ACS Appl.
Publishing (2019). Mater. Interfaces 11 (2019) 14640–14646.
[198] P. Chandrashekhar, F. Minooei, W. Arreguin, M. Masigol, J.M. Steinbach- [219] X. Zhang, X. Fu, G. Chen, Y. Wang, Y. Zhao, Versatile Ice Microneedles for
Rankins, Perspectives on existing and novel alternative intravaginal probiotic Transdermal Delivery of Diverse Actives, Adv Sci (Weinh) 8 (2021) e2101210.
delivery methods in the context of bacterial vaginosis infection, AAPS J. 23 [220] F. Wang, X. Zhang, G. Chen, Y. Zhao, Living Bacterial Microneedles for Fungal
(2021) 1–16. Infection Treatment, Research (Wash D C) 2020 (2020) 2760594.

16

You might also like