Content uploaded by Marija Glavas Dodov
Author content
All content in this area was uploaded by Marija Glavas Dodov on Feb 07, 2023
Content may be subject to copyright.
__________________
* magl@ff.ukim.edu.mk
Macedonian pharmaceutical bulletin, 68 (1) 9 - 26 (2022)
ISSN: 1857-8969
UDC: 665.584 615.262
DOI: 10.33320/maced.pharm.bull.2022.68.01.002
Review paper
Probiotics in dermatological and cosmetic products –
application and efficiency
Edita Hyseni and Marija Glavas Dodov*
Center of Pharmaceutical nanotechnology, Faculty of Pharmacy,
Ss Cyril and Methodius University in Skopje, Majka Tereza 47, 1000 Skopje, N. Macedonia
Received: September 2022; Accepted: November 2022; Published: 25th January 2023
Abstract
The term “probiotics” has first been used in 1907 by Elie Metchnikoff. Since then, probiotics have been part of research not only in
regards of digestive health, but also inflammatory diseases. Lately, there has been an increased interest of probiotic’s effects in skincare.
The management of atopic dermatitis, acne, psoriasis, photo aging, skin cancer, intimate care, oral care, wound healing is getting harder
each passing day, due to increased antibiotic resistance and other side effects of conventional therapy. Therefore, new ingredients have
been investigated and probiotics have been proved to be effective in treating various skin conditions.
This review aims to evaluate the scientific evidence on topical and oral probiotics, and to evaluate the efficacy of cosmetic and
dermatological products containing probiotics. Many studies have shown that skin and gut microbiome alterations have an important role
in skin health. Although this is a new topic in dermatology and cosmetology, there have been some promising results in lots of research
studies that the use of probiotics in cosmetic products may help improve the patient’s outcome. While oral probiotics have been shown to
promote gut health, which influences the host immune system and helps treat different skin diseases, the mechanism of action of topical
probiotics is not yet fully understood. Although the number of commercial probiotic cosmetic products released in the market is increasing
and most of the studies have not shown any serious side effect of probiotics, further studies, in larger and heterogeneous groups are needed.
Keywords: probiotics, acne, atopic dermatitis, photoaging, psoriasis, immune system, wound healing, oral care, intimate care, skin
microbiome
Introduction
The integumentary system is formed by the skin and
its derivative structures. The skin is composed of three
layers: the epidermis, the dermis, and subcutaneous tissue
(Kanitakis, 2002). The outermost level, the epidermis,
consists of a specific constellation of cells known as
keratinocytes, which function to synthesize keratin, a long,
threadlike protein with a protective role. The middle layer,
the dermis, is fundamentally made up of the fibrillar
structural protein known as collagen. The dermis lies on the
subcutaneous tissue, or panniculus, which contains small
lobes of fat cells known as lipocytes. The thickness of these
layers varies considerably, depending on the geographic
location on the anatomy of the body. The eyelid, for
example, has the thinnest layer of the epidermis, measuring
less than 0.1 mm, whereas the palms and soles of the feet
have the thickest epidermal layer, measuring
approximately 1.5 mm. The dermis is thickest on the back,
where it is 30-40 times as thick as the overlying epidermis
(James et al., 2006).
Skin is the largest organ of the human body whose
primary functions consist of thermoregulation, immune
response, a physical barrier that protects from exogenous
factors. It is colonized by a diverse and large number of
E. Hyseni, M. Glavas Dodov
10
Maced. pharm. bull., 68 (1) 9 – 26 (2022)
microorganisms. Some of them are commensal and some
are transient. Most of them are beneficial to the host in
normal conditions, but some of them may be pathogenic.
So far, only about 200 microorganisms has been
characterized as pathogenic. The rest are either commensal
or facultative pathogenic. Due to differences in the
environment in different anatomic sites, the abundance of
bacteria is also different. Moist areas harbor mostly
Staphylococcus and Corynebacteria species. Sebaceous
sites host lipophilic species such as Propionibacterium. Dry
areas host predominantly Staphylococcus,
Propionibacterium, Micrococcus, Corynebacterium,
Enhydrobacter and Streptococcus species.
Understanding the skin’s microbiome is crucial to
gain insight about the benefits and/or skin diseases caused
by its alterations. Culture based methods have been widely
used in the identification of skin bacteria. However, some
bacteria require fastidious growth conditions and are
difficult to isolate. The development of molecular
techniques to identify and quantify microbial organisms has
enhanced our understanding of the skin bacterial diversity.
Genomic approaches to characterize skin bacteria rely on
sequence analysis of the 16S ribosomal RNA genes, which
is present in all bacteria and archaea, but not in eukaryotes.
The 16S rRNA gene contains variable regions, enabling
taxonomic classification and conserved regions, serving as
binding sites for PCR primers. Moreover, an organism does
not need to be cultured to determine its type by 16S rRNA
sequencing (Dethlefsen et al., 2007; Turnbaugh et al.,
2007). The disadvantage of this method is that it cannot
distinguish between methicillin-resistant and methicillin-
sensitive Staphylococcus spp. isolates.
Beside metagenomic analysis, metabolomic
identification, through mass spectrometry of metabolites,
lipids and peptides; bioinformatic software for analysis of
phylogenetics, metaproteomics and metabolomics; and
artificial intelligence-based data management and
prediction of microbial interactions (i.e. artificially created
microbial ecosystems [ACMEs]) are now being used in
combination for the quantification and profiling of
microorganisms, and their genetic material, environmental
conditions, and interrelations.
Skin microbiome
The microbiota refers to any microorganism present
in and on the body, such as gut, nose, oral mucosa,
pulmonary mucosa, scalp and the skin. The skin
microbiome is the genome of the microorganisms present
on the skin to which microorganisms maintain a complex
relationship.
The early age development of skin microbiome plays
a pivotal role in the establishment of the cutaneous
homeostasis and skin immune functions. Because of the
environmental changes, from mostly sterile environment in
the womb to a gaseous one with constant microbial
interaction, the first days after birth mark significant
changes in skin barrier composition and function. These
changes include reduction in transepidermal water loss,
skin pH and sebaceous activity and increase in water
content. The early colonization is dominated by
Staphylococci, but it decreases by the end of the first year
of life, contributing to an evenness of population. The
predominant species of infant skin are Firmicutes, followed
in decreasing order by Actinobacteria, Proteobacteria and
Bacteriodetes.
The skin region specific bacteria predominance
depends on a lot of factors such as: moisture levels, sebum
content, temperature, pH and UV exposure. It is also
influenced by the place of residence, either urban or rural.
Alterations in skin microbiome caused by intrinsic
and extrinsic factors is called dysbiosis. Dysbiosis does not
only occur between bacteria, but also between bacteria and
other skin commensals, such as bacteria and fungi. A
disequilibrium between bacteria and fungi strains on the
scalp, has been observed in subjects prone to dandruff.
Host skin cells continuously sample microorganisms
that reside in the epidermis and dermis through pattern
recognition receptors (PRRs). Part of the activated immune
system and how changes are regulated, distinguish a
commensal organism from a potential pathogen. Functions
of the skin microbiota in health and disease.
Probiotics in skin homeostasis
The term probiotic has been defined as “living
microorganisms which, when consumed in adequate
amounts, confer a health effect on the host” (World Health
Organization and Food and Agriculture Organization of the
United Nations, 2001). Lactobacillus and Bifidobacterium
have emerged as two of the most commonly used probiotics
(Ouwehand et al., 2002), although newer strains such as
Bacillus coagulans are being investigated with positive
results (Benson et al., 2012). Most commonly formulated
as fermentation products, probiotics counter pathogenic
bacteria, support barrier function, and contribute to the
regulation of the innate and adaptive immune responses
(Hacini-Rachinel et al., 2009). Probiotics can now be found
in household items ranging from yogurt to children’s
popsicles to facial cream. In Table 1, the role of probiotics
in dermatology as well as in cosmetology is presented.
Table 1. Potential probiotics role in dermatology and
cosmetology
Probiotics role in dermatology and cosmetic products
Acne treatment
Di Marzio et al. (2008)
Photo aging
Satoh et al. (2015)
Atopic dermatitis
Huang et al. (2017)
Immune regulation
Savan et al. (2006)
Wound healing
Peral et al. (2009)
Probiotics in dermatological and cosmetic products – application and efficiency
11
Макед. фарм. билт., 68 (1) 9 – 26 (2022)
Another example of potential use of probiotics for skin
health is Lactobacillus pes-odoris, which specifically
inhibits the odor-producing bacteria of the feet and
Lactobacillus ala-odoris, which prevents the formation of
odor in armpits. Both Lactobacilli cultures can improve the
effectiveness of deodorants, foot sprays, or lotions.
Additionally, probiotics can result in significant
benefits in protecting and restoring the skin barrier. In
patients with acne and rosacea, living microorganisms on
the skin are recognized as foreign by the body’s immune
system. The immune system springs into action to counter
this potential threat resulting in the inflammation, redness,
or bumps common in these skin conditions. Probiotics
applied topically sit on the skin’s surface and prevent the
skin cells from seeing the bad bacteria and parasites that
can cause this immune system response. This is known as
“bacterial interference,” as probiotics could protect the skin
and interfere with the ability of the bacteria and/or parasites
to provoke an immune reaction (Bowe, 2013; Sánchez-
Pellicer et al., 2022).
When certain types of probiotics are placed in contact
with skin cells, they calm the parts of the cells that may
want to react to the presence of pathogenic bacteria that
they see as a threat. These healthy signals produced by the
probiotics stop the skin cells from sending “attack”
messages to the immune system that result in flares of acne
or rosacea (Yan and Polk, 2011).
Mechanism of action of probiotics
The mechanisms whereby probiotics may play a role
in skin physiology are not fully elucidated. However, it is
proposed that, as shown for other commensal bacteria,
probiotics could be directly sampled in the lumen by
mucosal dendritic cells, which express tight junction
proteins and penetrate the gut epithelial monolayer (Uhlig
and Powrie, 2003). It is postulated that upon interaction of
the probiotic bacteria (or their components) with the
intestinal epithelium and/or direct interaction with
dendritic cells, other immune cells, such as Band T
lymphocytes may be activated (primed) and immune
mediators, including cytokines, may subsequently be
released. These cytokines, bacterial fractions, and primed
immune cells may be transported via the blood to other
organs, including the skin, where they could modulate the
immune status.
In addition, the improvement of reactive skin after
probiotic supplementation could also result from a direct
activity of the ingredient on neurosensitive mechanisms.
On the one hand, immune-regulating properties of
probiotics at the skin level could modulate the
inflammatory reactions generated by the release of
neuromediators involved in skin neurosensitivity
(Nickoloff and Naidu, 1994; Ohta et al., 2000; Thurin and
Baumann, 2003). On the other hand, the capacity of certain
probiotics to modulate the production of regulating
cytokines and growth factors (as Transforming growth
factor beta) may play a role in the proliferation and
differentiation of skin keratinocytes, which are important
for skin barrier repair (Von der Wied et al., 2001). Such
possible effects on the process of generating the stratum
corneum allow the quality of the cutaneous barrier function
and skin dryness to be improved (Thurin and Baumann,
2003).
Probiotics in dermatotherapy and cosmetology
Acne treatment
Acne is a chronic inflammatory disease that is caused
by hyperproliferation of Propionibacterium acnes in hair
follicles, hyperkeratinisation and sebum overproduction.
Beside androgen levels and pathogenic influence, the
alterations in the skin microbiome play an important role
in the presence and severity of acne. It is one of the 10 most
prevalent disorders in the world and has severe adverse
effects on the quality of the life in patients.
The co-morbidity of chronic skin conditions and
mental health disorders has long been recognized, and in
recent years’ specialty psychodermatology and
neurodermatology groups have emerged. Acne vulgaris is
a common dermatological disorder frequently associated
with depression, anxiety and other psychological sequelae.
The mental health impairment scores among acne patients
are higher vs. a number of other chronic, non-psychiatric
medical conditions, including epilepsy and diabetes. Along
with the psychological fallout, there have also been
indications that acne patients are at a higher risk for
gastrointestinal distress. For example, one study involving
over 13,000 adolescents showed that those with acne were
more likely to experience gastrointestinal symptoms such
as constipation, halitosis, and gastric reflux. In particular,
abdominal bloating was 37% more likely to be associated
with acne and other seborrheic diseases.
Stokes and Pillsbury made numerous references to the
use of L. acidophilus and L. acidophilus-fermented milk
products as a treatment modality in the context of the brain-
gut-skin inflammatory process. Indeed, other physicians
writing in the 1930s made reference to the popularity of L.
acidophilus cultures among the general public as an
internal means to treat acne. However, despite the apparent
appeal of what would later be described as probiotics, there
was little research to determine efficacy. The first formal
clinical case report series on the potential value of
Lactobacillus probiotics was published in 1961. A
physician from Union Memorial Hospital in Baltimore,
Robert H. Siver, followed 300 patients who were
administered a commercially available probiotic (Laxtinex
tablets providing a mixture of L. acidophilus and L.
bulgaricus). He used a protocol of probiotic
supplementation for 8 days followed by two-week wash
out then re-introduction for an additional eight days. The
rationale for such a dosing regimen is unclear. In any case,
he reported that 80 percent of those with acne had some
degree of clinical improvement, and that the intervention
E. Hyseni, M. Glavas Dodov
12
Maced. pharm. bull., 68 (1) 9 – 26 (2022)
was most valuable in cases of inflammatory acne. Without
a placebo control, Dr Siver concluded merely that ‘interactions of skin manifestations of acne vulgaris and of
metabolic processes of the intestinal tract are suggestive’.
Table 2. Beneficial properties and application of some probiotics in dermatology and cosmetology
Probiotic
Benefit
References
Streptococcus thermophilus
Increases ceramide production
Di Marzio et al. (1999)
Enterococcus faecalis
Reduces inflammatory lesions
Kanget a l. (2009)
Streptococcus salivarius
Immune modulator
Cosseau et al (2008)
Bifidobacterium longum
Skin inflammation
Guéniche et al. (2010)
Lactobacillus paracasei
Skin inflammation
Guéniche et al. (2010)
Lactobacillus johnsonii
UV protection
Bouilly-Gauthier et al. (2010)
Lactobacillus plantarum HY7714
Anti photoaging effect
Lee et al. (2015)
Lactobacillus acidophilus IDCC 3302
tyndallizate
UVB protection
Smith, (1996)
Lactobacillus buchneri
Photoprotection
Kang et al. (2020)
Nitrosomonas eutropha
Anti-wrinkles
Notay et al. (2019)
Lactobacillus fermentum
SCORAD reduction
Huang et al. (2017)
Lactobacillus plantarum CJLP133
Prevention of AD symptoms
Han et al. (2012)
Bifidobacterium lactis UABLA-12
AD improvement
Gerasimov et al. (2010)
Vitreoscilla filiformis lysate
Improves AD symptoms
Gueniche et al. (2008)
Bifidobacterium dentium
Reduces the incidence of AD
Avershina et al. (2017)
Pseudomonas aeruginosa
Wound healing
Valdéz et al. (2005)
Streptococcus thermophilus
Acne treatment
Di Marzio et al. (2008)
Enterococcus faecalis SL-5
Acne treatment
Kang et al. (2009)
Bifidobacterium longum
Acne treatment
Guéniche, A et al. (2010)
Lactobacillus paracasei
Acne treatment
Wang and Wang, (2015)
Lactobacillus johnsonii
Photoaging
Bouilly-Gauthier et al., (2010)
Lactobacillus plantarum HY7714
Photoaging
Lee et al., (2015)
Lactobacillus acidophilus IDCC 3302
tyndallizate
Photoaging
Smith WP, (1996)
Lactobacillus buchneri
Photoaging
Kang YM et al., (2020)
Nitrosomonas eutropha
Photoaging
Notay et al. (2019)
Lactobacillus fermentum
Atopic dermatitis
Huang et al. (2017)
Lactobacillus salivarius
Atopic dermatitis
Huang et al. (2017)
Lactobacillus plantarum CJLP133
Atopic dermatitis
Han et al., (2012)
Lactobacillus rhamnosus
Atopic dermatitis
Kalliomaki et al. (2001)
Lactobacillus paracasei
Atopic dermatitis
Wang and Wang, (2015)
Lactobacillus fermentum
Atopic dermatitis
Wang and Wang, (2015)
Bifidobacteria lactis
Atopic dermatitis
Sistek et al. (2006)
Lactobacillus sakei
Atopic dermatitis
Woo et al. (2010)
Vitreoscilla filiformis lysate
Atopic dermatitis
Gueniche et al., (2008)
Propionibacterium freudenreichii
Atopic dermatitis
Sikorska and Smoragiewicz (2013)
Lactococcus lactis
Atopic dermatitis
Sikorska and Smoragiewicz (2013)
Lactobacillus bulgaricus
Atopic dermatitis
Sikorska and Smoragiewicz (2013)
Bifidobacterium dentium
Atopic dermatitis
Avershina e. et al., (2017)
Bifidobacterium lactis HN019
Atopic dermatitis
Wickens K. et al, 2018
Lactobacillus casei CRL 431
Immune regulation
Galdeano et al. (2006)
Lactobacillus paracasei CNCM I-1518
Immune regulation
Galdeano et al. (2006)
Lactobacillus plantarum
Wound healing
Peral et al. (2009)
Probiotics in dermatological and cosmetic products – application and efficiency
13
Макед. фарм. билт., 68 (1) 9 – 26 (2022)
The effectiveness of conventional acne treatment
including oral and topical antibiotics has been decreasing,
due to the increased antibiotic resistance. Another major
concern caused by antibiotics is the damage to stable skin
microbiome and the resistance of non-target bacteria. In
order to overcome these limits and find new ways to
improve the treatment outcome, it is recommended to avoid
the use of topical and oral antibiotics as monotherapy or
polytherapy and supports a combination of a topical
retinoid and antimicrobial, eg. Benzoyl peroxide, as the
first line therapy.
Recent studies have brought new insights in
understanding the pathophysiology of acne. It has been
found by metagenomic studies that the abundance of P.
acnes between healthy individuals and acne patients is
similar. Instead, the disbalance between phylotypes of P.
acnes appears to have a significant role in the triggering of
acne. The predominant P. acnes strain in acne patients was
found to be the phylotype IA1 (also known as the CC18
clonal complex or A1 SLST-type). Moreover, the virulence
of phylotypes IA1 and IB in acne is higher than in normal
skin. Finally, P. acnes types IA and IB were found to
induce greater levels of production of the human
antimicrobial peptide (AMP), β-defensin 2 (hBD2), from
cultured sebocytes, and displayed higher levels of lipase
activity than a type II isolate.
As for intestinal permeability in acne vulgaris, there
have been hints that the intestinal lining may be
compromised. One older study used a blood serum
complement fixation test and reported that acne patients
were more likely to show enhanced reactivity to bacterial
strains isolated from stool. Approximately 66% of the 57
patients with acne showed positive reactivity to stool
isolated coliforms, this compared to none of the control
patients without active skin disease. Furthermore, a study
involving 40 acne patients showed both the presence of,
and high reactivity to, lipopolysaccharide (LPS)
endotoxins in the blood as measured by the stellate fibrin
crystal test. None of the matched healthy controls reacted
to the E. coli lipopolysaccharide endotoxin (E. coli LPS),
while 65% of the acne patients did have a positive reaction.
The inference of these results is that circulating endotoxins
derived from gut microbes is not an uncommon feature of
acne vulgaris, and one indicating that intestinal
permeability is a potential issue for a sizable group of acne
patients.
The first report that ‘topical bacterio-therapy’ (via
local Lactobacillus bulgaricus application) may be helpful
in acne and seborrhea was published in 1912. However, it
was not until 1999 that proper scientific technique was used
to evaluate some of the potential skin-specific benefits of
lactic acid bacterial application. Specifically, researchers
showed that the lactic acid bacteria Streptococcus
thermophilus, a species found in most yogurts, can increase
ceramide production when applied to the skin for 7 days as
a cream. This work, which has since been replicated, is of
relevance to acne, particularly when considering that some
of the ceramide sphingolipids, most notably
phytosphingosine (PS), provide both antimicrobial activity
against Propionibacterium acnes (P. acnes) and direct
anti-inflammatory activity. Sphingolipids have been noted
to be low in acne, and the seasonal loss of ceramides may
be a driving force behind much higher dermatological
office visits for acne during winter months. Indeed, topical
application of 0.2% PS reduced papules and pustules by
89% in a recent 2-month pilot study. Additional studies
hinting at the value of topical probiotics in acne include
recent reports that strains of Bifidobacterium longum and
Lactobacillus paracasei can attenuate skin inflammation
mediated by substance P. This is of relevance because
substance P may be a primary mediator of stress-induced
amplification of inflammation and sebum production in
acne. Two separate reports have also indicated that various
probiotic lactic acid bacteria can provide in vitro
antimicrobial activity against P. acnes. The latter study
also used a clinical arm, the results showing that topical
application of an Enterococcus faecalis probiotic lotion for
8 weeks reduced inflammatory lesions by over 50% vs.
placebo. Certain substances secreted by bacterial strains,
such as antimicrobial peptides, have been shown to inhibit
growth of P. acnes. Streptococcus salivarius, a prominent
member of the oral microbiota of healthy humans, has been
shown to secrete a bacteriocin-like inhibitory substance
(BLIS-like substance) capable of inhibiting P. acnes. In
addition to the antimicrobial activity, S. salivarius bacterial
cells themselves inhibit a number of inflammatory
pathways, thus acting as immune modulators. Finally, the
application of select bacteria to the skin may provide a
protective shield, similar to a physical barrier. This so-
called bacterial interference, through competitive
inhibition of binding sites, is thought to prevent
colonization by other, potentially pathogenic, bacterial
strains.
In healthy skin, P. acnes plays a beneficial role in the
cutaneous microbiota of the pilosebaceous unit. It limits
the growth of S. aureus, such as community-acquired
methicillin-resistant S. aureus, as well as that of S.
pyogenes in maintaining an acidic pH of the pilosebaceous
follicle by hydrolysing sebum triglycerides and by
secreting propionic acid.
However, during puberty, over-colonisation of the
pilosebaceous unit by P. acnes may lead to a loss of
diversification and dysbiosis, potentially causing acne. A
recent clinical study using the single-locus sequence typing
method investigated P. acnes subgroups on the face and
back in patients with severe acne and in healthy subjects.
In almost 75% of the acne patients, P. acnes phylotypes
were identical on the face and back, whereas this was only
the case in about 45% of the healthy subjects. In the healthy
group, phylotypes IA1 (39%) and II (43%) were the main
phylotypes, whereas in the acne group IA1 (84%),
especially on the back (96%), was the main phylotype. This
may confirm the hypothesis that acne severity may be
associated with a loss of diversity of P. acnes phylotypes,
E. Hyseni, M. Glavas Dodov
14
Maced. pharm. bull., 68 (1) 9 – 26 (2022)
following a selection of phylotype IA1/clonal complex
(CC) 18 present in all acne patients. Therefore, different
inflammatory profiles, depending on the phylotype (i.e.,
phylotype IA1, which has been mainly observed on the face
and back of acne patients and cluster of P. acnes activating
the innate immunity via the expression of protease-
activated receptors (PARs), tumor necrosis factor-α, and
the production of interferon-γ and interleukins [IL-8], have
been observed. Moreover, P. acnes activates the release of
lipases, matrix metalloproteinases, and hyaluronidases,
leading to hyperkeratinisation of the pilosebaceous unit and
finally to comedones, papules, and pustules.
The application of live bacteria on skin poses several
challenges. Postbiotic is a relatively new term used to
describe microbial metabolites. These include short-chain
fatty acids, extracellular metabolites, functional proteins,
cell lysates and other products derived from a probiotic that
can influence the microbiome composition. Postbiotics
have a long shelf life, safety and possess multiple health
benefits. They have been evaluated for anti-inflammatory,
immunomodulatory, anti-obesogenic, antihypertensive,
hypocholesterolemic, antiproliferative and antioxidant
benefits.
Various research groups have reported the presence of
bacteriocins in the Bacillus genus. LactoSporin is an
extracellular metabolite purified from Bacillus coagulans
MTCC 5856 fermented broth with an INCI name Bacillus
ferment filtrate extract. The potential mechanism of
LactoSporin as an antimicrobial agent is by pH drop,
microbial biofilms inhibition, and draining the ions from
the targeted cells.
A study suggests that LactoSporin is a postbiotic that
is thermostable and stable at an acidic pH. LactoSporin 2%
w/w cream was a safe antiacne formulation with efficacy
comparable to the standard treatment of benzoyl peroxide
2.5% gel. The onset of efficacy was very early for
LactoSporin as early as three days especially for closed
comedones, providing a quicker benefit than benzoyl
peroxide. The major finding of this study shows that
LactoSporin reduces the sebaceous secretion by its 5-alpha
reductase inhibitory and antimicrobial properties, which are
better than benzoyl peroxide and can be a potential
ingredient for other Seborrheic conditions as well.
Considering the clinical efficacy, continuous treatment and
patient satisfaction, LactoSporin is highly suitable for
treating subjects with mild-to-moderate acne vulgaris.
Further studies on a larger sample in multiple ethnic
populations may substantiate our findings.
Di Marzio et al. (2008) conducted an in vitro study
during which he added the bacterium Streptococcus
thermophilus to human keratinocyte cell cultures and found
an increase in the production of ceramides. He believed this
was due to S. thermophilus’ possession of
sphingomyelinase, an enzyme that hydrolyzes
sphingomyelin into ceramides. Many bacteria have been
reported to produce extracellular sphingomyelinase
including the genera Bacillus, Listeria, Staphylococcus,
Mycobacterium, Chlamydia, Pseudomonas, Leptospira,
and some species of Helicobacter. Although this enzyme
primarily functions as a virulence factor for the bacteria, its
ability to increase ceramide production may provide a
beneit in treating skin diseases. In the next phase of the
study, Di Marzio tested this theory in vivo on 17 healthy
subjects with normal skin. The subjects were instructed to
apply 0.5 g of a topical probiotic formulation consisting of
Streptococcus thermophilus twice a day to the volar surface
of one of their forearms. They applied the vehicle alone to
the contralateral forearm for comparison. An additional
four subjects were treated with sphingomyelinase purified
from Bacillus cereus to ensure that the results produced
were specific to the sphingomyelinase and not another
component within the bacterium. After seven consecutive
days of application, the probiotic formulation containing S.
thermophilus caused an increase in the production of
ceramides in the stratum corneum, which was comparable
to the results seen using the sphingomyelinase extracted
from B. cereus. These results demonstrated that the
sphingomyelinase produced by S. thermophilus may
improve skin barrier function.
Ceramides not only have a role in water permeability,
but they also play a part in the antimicrobial and anti-
inflammatory properties of the skin. The exact
antimicrobial mechanism of ceramides has not been
confirmed; however, there are many theories: reduction of
bacteria adherence to epithelial cells, inhibition of bacterial
protein kinases, and/or damage to the cell wall of the
bacteria. Aware of their antimicrobial properties, Pavicic et
al. performed a study in 2007 to evaluate the role of
ceramides in patients with acne. The study consisted of
both an in vitro and in vivo phase. In vitro, he found that
phytosphingosine (PS), one of the four types of sphingoid
bases that make up ceramides, inhibited growth of
Propionibacterium acnes, an important contributor to acne
formation. From these findings, he performed a two-part in
vivo pilot study testing a 0.2% PS formulation on subjects
with acne. In the first part, 30 subjects with acne applied a
topical medication containing PS with benzoyl peroxide
(PS-BPO) to half of their face versus benzoyl peroxide
(BPO) alone to the contralateral side of their face two times
per day. After 2 months, comedones were reduced by 72%
and inflammatory papules and pustules by 88% in the PS-
BPO group versus 22 and 32%, respectively, in BPO only
group. In another arm of the trial, 10 subjects applied PS
alone to half of their face and a placebo cream alone to the
other side of their face twice a day. After 2 months, the
placebo increased comedones by 43% compared to only
6% in the PS group. More significant results were seen in
inflammatory acne numbers with an 89% reduction
observed in the PS group compared to no change in the
placebo group.
A similar study in 2009 by Kang et al. demonstrated
the effects of the bacterium, Enterococcus faecalis SL-5 (a
very common inhabitant of the human gastrointestinal
tract) and its effect on P. acnes. He conducted in vitro and
Probiotics in dermatological and cosmetic products – application and efficiency
15
Макед. фарм. билт., 68 (1) 9 – 26 (2022)
in vivo studies. In the in vitro aspect of the study, E. faecalis
proved to be bacteriocidal to P. acnes due to a bacteriocidin
named ESL5. In the clinical trial, 70 subjects with mild-to-
moderate acne were enrolled in an 8-week double-blind,
randomized, placebo-controlled phase III study. Subjects
were randomized into the probiotic or placebo group.
Those in the experimental group applied a lotion
containing ESL5 to the areas of the face involved with acne
twice per day, and the control group applied a placebo
lotion twice daily. After 8 weeks of application, a decrease
in the number of comedones was seen in the probiotic
group compared to the placebo group; however, these
results were not statistically significant. In the
inflammatory lesion counts, a statistically significant
reduction of greater than 50% was observed in the E.
faecalis group compared to placebo.
Photoaging
Skin aging has intrinsic and extrinsic components.
Intrinsic aging is related to genetic factors and is a set of
physiologic processes related to the passage of time that
includes thinning of epidermal and dermal skin layers and
increasing dryness. The extrinsic skin-aging process is
characterized by coarse wrinkles, loss of elasticity,
epidermal thickening, dryness, laxity, rough appearance,
and pigmentation disorder.
Photoaging refers to premature aging of the skin due to
repeated light exposure. Its clinical manifestations,
histopathology, and biochemical changes are different
from the natural aging of skin. Photoaging reduces the
amount of mature type I collagen and elastic fibers in the
dermis of the skin. The clinical characteristics of
photoaging occur mainly on exposed skin, such as the face,
neck, and forearm, where rough skin, loss of elasticity,
deepening and thickening of wrinkles, a leather-like
appearance, pigmentation, and dilated capillaries can occur
(Qin et al., 2018). Studies have shown that approximately
65% of patients with melanoma and 90% of patients with
non-melanoma skin cancers, including BCC and SCC, are
associated with skin photoaging (Damiani and Ullrich,
2016). Many external factors are attributed to photoaging
of the skin, such as ultraviolet (UV), infrared, chemical
smog, dust, and smog, among which UV radiation is the
most significant (Markiewicz and Idowu, 2018).
The occurrence and development of skin photoaging
mediated by UV radiation involves multiple pathways,
including apoptosis, proliferation, autophagy, DNA repair,
checkpoint signal transduction, cell transduction, and
inflammation. UV radiation is generally categorized
according to wavelength into long-wave UVA (315–400
nm), medium-wave UVB (280–315 nm), and short-wave
UVC (200–280 nm). Although UVB radiation (280–315
nm) accounts for only 1–2% of the UV rays of the Sun, it
is considered to be the main environmental carcinogen that
causes skin cancer and is related to the occurrence and the
development of tumors (Panich et al., 2016; Gherardini et
al., 2019). Experimental models are the most widely used
photoaging models. This type of experimental model often
uses UVB because the skin tissue changes caused by UVB
are very similar to photoaging of human skin (Kim et al.,
2019b).
It has been reported that probiotic bacteria may be
highly effective in protecting the skin from photoaging, as
oral supplementation prior to UVB exposure was
demonstrated to prevent TEWL, increase epidermal
thickness, and alleviate damage to tight junction structures
and the basement membrane in a mouse model (Bouilly-
Gauthier et al., 2010; Satoh et al., 2015). The probiotic
gram-positive bacterium Lactobacillus acidophilus
inhabits the intestines and serves an important role in the
maintenance of gut health; however, it is not known
whether it can protect against photoaging induced by UV
radiation.
Recent clinical trials have shown protective effects of
dietary supplements containing Lactobacillus johnsonii
alone or combined with carotenoids against early UV-
induced skin via regulation of immune cells and
inflammatory cytokines (Bouilly-Gauthier et al., 2010).
Recent experiments in hairless mice have suggested
that in addition to regulating immune responses in the skin,
orally administered probiotics may exert anti-aging effects
by suppressing wrinkle formation and increasing skin
elasticity. Furthermore, our recent experiments in hairless
mice have shown that oral administration of L. plantarum
HY7714 (HY7714) exerts anti-photoaging effects through
reduction of wrinkle formation and suppression of
epidermal thickening and that skin hydration increases in
association with increasing ceramide level via regulation
of serine palmitoyltransferase and ceramidase expression
in the mice skin (Lee et al., 2015).
A study demonstrated the protective effect of L.
acidophilus IDCC 3302 tyndallizate on HaCaT
keratinocyte damage induced by UVB exposure.
Tyndallizate was analyzed for its nutrients including
carbohydrate, crude protein, crude fat, moisture, and ash
(Smith WP, 1996). For small molecule analysis, lactic acid
was detected as a major chemical component and it is
reported that topical lactic acid has cosmetic benefit
(LeBlanc et al., 2017). In previous studies beneficial
effects of short-chain fatty acids and vitamins produced by
probiotics have been reported (Preidis et al., 2011). In this
study Im et al. (2019), tyndallizate was hypothesized to be
helpful in skin rejuvenation. The detailed mechanisms
regarding anti-wrinkle activity require further study.
Sharma et al. (2016) explain that probiotics can restore
acidic skin pH, alleviate oxidative stress, attenuate
photoaging, improve skin barrier function, and enhance
hair quality.
Plant extracts fermented with Lactobacillus buchneri
(PELB), found in Kimchi, were evaluated regarding their
anti-photoaging effects. The authors investigated the anti-
photoaging effect of PELB in ultraviolet light (UVB)-
induced photoaging in in vitro models. They pre-treated
normal human dermal fibroblasts and epidermal
E. Hyseni, M. Glavas Dodov
16
Maced. pharm. bull., 68 (1) 9 – 26 (2022)
keratinocytes with PELB and that had been exposed to
UVB. They found that PELB decreased elastase activity
and increased type I collagen expression in the UVB-
induced photoaging in vitro model, reduced collagenase
activity, and promoted the expression of moisture factor
and antioxidant enzymes. The study results show that the
PELB could be useful in the cosmetic industry due to its
protective effects against UVB-induced photoaging (Kang
et al., 2020).
Notay et al. (2019) investigated the use of topical
Nitrosomonas eutropha for cosmetic improvement of facial
wrinkles. They found a significant improvement in wrinkle
depth severity, hyperpigmentation of the forehead and
glabella in the group receiving high topical concentration
of the probiotic formula.
Atopic dermatitis
Atopic dermatitis is a chronic inflammatory disease
that affects 1 in 10 people in their lifetime. The etiology of
atopic dermatitis is very complex. It can be caused by
microbiome alterations, genetic factors, environmental
factors, immune dysregulation and is characterized by
scaly, pruritic, erythematous lesions.
AD is known to initiate with Th2, Th22 and Th17 cell
activation (acute phase), before chronicity, defined by the
onset of a Th1 cell response alongside the continued
activation of Th2 and Th22 cells. This mechanism could be
explained by the presence of a positive feedback loop made
by TSLP (thymic stromal lymphopoietin), IL-4 and IL-13.
TSLP, produced by keratinocytes, drives Th2 polarization
and activates dendritic cells, while IL-4 and IL-13 act on
keratinocytes to further increase TSLP level. Thymus and
activation-regulated chemokine (TARC) levels also
increase in the stratum corneum of skin lesions of AD
patients, which is correlated with disease severity,
especially with erythema, oedema/papules, and
oozing/crusts. These two cytokines have been used as
indicators of skins inflammation in AD lesions.
The conventional treatment strategy for AD includes
hydration with emollients, avoidance of individual trigger
factors, anti-inflammatory therapy with topical
corticosteroids or calcineurin inhibitors and treatment of
secondary infections. For severe AD treatment, a recent
antibody has been approved, Dupilumab, which is an
injectable human lgG4 monoclonal antibody that inhibits
IL-4 and IL-13 cytokine responses, and has provided
remarkable results. However, its use is still debated
regarding its suitability as a routine treatment. Although
anti-inflammatory drugs are considered as the first
pharmacological treatment, in recent years, concerns have
arisen due to the high incidence of side effects. In addition,
corticosteroids are not feasible for delicate parts of the
body, such as the eyelids, and are discouraged in childhood.
Increasing evidence shows that patients affected by AD
have a modified composition of the gut microbiome and
lack microbial diversity compared to healthy controls. This
bacterial dysbiosis may be considered a possible target for
the treatment and prevention of AD. Therefore, the
supplementation of the diet with probiotics, prebiotics and
postbiotics may have a role in the management of AD
because of their capacity of modulation of the microbiota.
Recent studies have shown that gut microbiomes have
been implied in the development and shaping of the host
immune system. Gut dysbiosis-alterations of microbial
diversity and abundance due to lifestyle and diet
correspond to the impaired inflammatory response to a
particular antigen. This phenomenon has been
demonstrated in patients with AD (Lee JH et al., 2016). In
general, it is known for its short-chain fatty acid- (SCFA-)
producing capability. Bifidobacterium, Propionibacterium,
Coprococcus, Blautia, and Eubacterium are significantly
reduced in patients with AD (Ellis et al., 2019; Zheng et al.,
2016). Since SCFA can induce regulatory T (Treg) cell
expansion, reduction of SCFA may result in a shift of Treg-
)17 balance towards )17, T helper cell known for its role in
orchestrating proinflammatory immune response.
Repopulating gut microbiota has been suggested to be an
alternative mechanism to treat various immunologic
diseases, including AD (Forbes et al., 2016). However, the
approach is still controversial (Williams et al., 2008).
An analysis of the gut microbiota of patients with AD
has shown an intraspecies compositional change in
Faecalibacterium prausnitziithat reduces the number of
high butyrate and propionate producers. Butyrate and
propionate are microbial-produced short-chain fatty acids
with an anti-inflammatory role. Moreover, butyrate has
been shown to be a key player in maintaining gut barrier
integrity. Therefore, reduced levels in the microbiota of
both butyrate and propionate producers may result in a pro-
inflammatory state in the gut and a loss of barrier integrity.
All these data indicate the potential role of probiotics as
microbiota recovery players, and consequently as potential
nutritional supplements in AD treatment.
Huang et al. (2017) included 13 randomized
controlled studies (1,070 total subjects) in a recent meta-
analysis. The data suggested an overall benefit of probiotics
supplementation in children with AD, indicated by a
reduction of the SCORAD values of −4.50 (95%
confidence interval=−7.45 to −1.54, p value < 0.001) in
children aged 1–18 years. The beneficial effect was
especially evident for Lactobacillus fermentum (mean
difference=−11.42, 95% confidence interval=−13.81 to
−9.04), Lactobacillus salivarius (mean difference=−7.21,
95% confidence interval=−9.63 to −4.78) and a mixture of
different strains (mean difference=−3.52, 95% confidence
interval=−5.61 to −1.44).
A randomized, double-blind, placebo-controlled
study investigated the effects of the use of the L. plantarum
CJLP133 strain in the prevention of AD symptoms. The
study was performed for a time period of 12 weeks among
children who were one and 12 years old. It was found that
there was an improvement in AD scores (SCORAD), with
a concomitant decrease in IFN-γ, eosinophil, and
Interleukin-4 counts (Han et al., 2012).
Probiotics in dermatological and cosmetic products – application and efficiency
17
Макед. фарм. билт., 68 (1) 9 – 26 (2022)
The effect of Lactobacillus rhamnosus in the
management of AD has been assessed by Kalliomaki et al.
(2001) in a double-blind, placebo-controlled, randomized
trial. In this trial 159 pregnant women with a family history
of allergic disorders such as asthma, allergic rhinitis or
eczema received placebo or Lactobacillus rhamnosus for 2
to 4-weeks before delivery and for a 6 month period after.
The percentage of children aged 2 years old with atopic
eczema was 23% in the probiotic group and 46% on
placebo (p= 0.008). A subgroup analysis conducted on 57
breastfed infants confirmed the benefit of a defensive effect
against eczema from supplementation with Lactobacillus
rhamnosus (Rautava, 2002). In recent trials by Wickens et
al., supplementation with Lactobacillus rhamnosus HN001
or Bifidobacterium lactis HN019 to mothers and infants
showed a significant protective effect only in the groups
receiving the supplementation with Lactobacillus
rhamnosus HN001 by 2 years of age (Wickens et al., 2008)
and 4 years of age (Wickens et al., 2012). The reported data
suggest that many probiotics may share common properties
but their effect may be strain-specific (Allen et al., 2003).
For instance, Gruber et al. (2007) found that
Lactobacillus rhamnosus strain GG (LGG) exerted no
therapeutic effects in infants with mild-to-moderate AD.
Another randomized, double-blind, placebo controlled
study investigated the use of L. paracasei (LP), L.
fermentum (LF), and LP+LF together in children, and it
was observed that the SCORAD scores were lower in the
group that received probiotics than those of the placebo
group 4 months after discontinuing the probiotic treatment
(Wang and Wang, 2015).
Sistek et al. (2006) conducted a 12-week trial in the
UK and found that a combination of LR and Bifidobacteria
lactis (BL) improved AD symptoms in food-sensitive
children.
Woo et al. (2010) compared children who received L.
sakei supplementation to those who received a placebo in
a double-blind, placebo-controlled trial. It was found that
the supplementation of L. sakei was associated with
substantial clinical improvement with concomitant
decrease in chemokine levels.
Gerasimov et al. (2010) reported that Lactobacillus
acidophilus DDS-1 and Bifidobacterium lactis UABLA-12
afforded significant clinical improvements in children with
moderate-to-severe AD.
A prospective, double-blind, placebo-controlled study
tested a lotion containing 5% of nonpathogenic Vitreoscilla
filiformis lysate. Seventy-five volunteers with AD applied
the lotion or placebo twice a day for 30 days. Then, the
severity of the disease (Scoring Atopic Dermatitis –
SCORAD), the transepidermal water loss, the microbiome,
and the patient’s report on itchiness and slvitreoscieep loss
were evaluated. The lysate has significantly improved AD
in all evaluated items, reducing skin colonization by S.
aureus. Authors concluded that the results are not only
because of S. aureus bacterial load reduction but also
immunomodulatory effect on the skin. Later, the lysate’s
immunomodulatory action was proved through analysis of
dendritic cells differentiation and effector functions of
dendritic and T-helper cells in vitro and in vivo. The topical
treatment with the bacteria significantly reduced
inflammation in mice, and the combination of allergen and
lysate showed lower induced dermatitis, indicating active
immunomodulation. It was observed that the innate
sensibility of nonpathogenic bacteria for Toll-Like
Receptor 2 (TLR2) induces dendritic tolerogenic cells and
Tr1 regulatory cells, suppressing T-effector cells and
cutaneous inflammation (Gueniche et al., 2008).
A revision performed by Sikorska and Smoragiewicz
(2013) found a series of evidence that various strains of
Lactobacillus and Bifidobacterium isolated from a variety
of sources inhibit in vitro growth of S. aureus. The most
active strains were Lactobacillus reuteri, Lactobacillus
rhamnosus GG, Propionibacterium freudenreichii, P.
acnes, L. paracasei, L. acidophilus, L. casei, Lactobacillus
plantarum, Lactobacillus bulgaricus, Lactobacillus
fermentum, and Lactococcus lactis. This revision also
included evidence that probiotics may also eliminate or
reduce colonization of methicillin-resistant S. aureus.
According to authors, their effects are mediated by both
cellular competitive exclusion and acid or Bacteriocin-
Like Inhibitors’ secretion.
Administration of the strain Bifidobacterium dentium
in mothers from 36-week gestation to 3 months post-
partum reduced the incidence of AD in offspring by 40%.
The group responding to the probiotic intervention had a
gut microbiota similar to the non-atopic children
(Avershina et al., 2017).
In a randomized double blind control trial using
Lactobacillus rhamnosus HN001 or Bifidobacterium lactis
HN019 taken (dose 6X109 colony-forming units (CFU)
daily) from 35 weeks of gestation to 6 months post-partum
in mothers while breastfeeding and from birth to age 2
years in infants, the authors showed that HN001
significantly protected against the development of eczema
at 2, 4, 6, and 11 years and atopic sensitization at 6 years,
while the strain HN019 had no significant effect on these
outcomes (Wickens et al, 2018).
In a randomized double blind control trial from 36
weeks of gestation until 3 months’ post-partum, 415
pregnant women were randomized to receive either
probiotics (LGG, L acidophilus La-5, and B animalis subsp
lactis Bb-12 dose: 5 × 1010 CFU daily) or placebo. At 6
years, there was a trend toward a lower cumulative
incidence of AD in the probiotic group, but the prevalence
of asthma and atopic sensitization was not significantly
affected by the probiotic regime (Simpson MR et al.,
2015). In a RDBCT, a total of 290 participants were
randomized to receive a daily mixture of LGG and B
animalis subsp lactis BB-12 each at a dose of 109 CFU, or
placebo. During the follow-up (mean age: 16.1 months), a
significantly lower incidence of eczema was observed in
the probiotic group (Schmidt et al., 2019).
Probiotic’s mechanism of action in atopic dermatitis
relies on their immunomodulatory effect, normalizing
microbial composition and metabolic effect. The
E. Hyseni, M. Glavas Dodov
18
Maced. pharm. bull., 68 (1) 9 – 26 (2022)
immunomodulatory effect is achieved by their ability to
inhibit the T-helper cell type 2 (Th2) mediated response and
improve the Th1/Th2 ratio; inhibiting Th2 cell response,
cytokines such as IL-4, IL-5, IL-6 and IL-13 are no longer
released, (Nwanodi, 2018) phagocytosis is stimulated,
serum IgA is increased. Probiotics can reduce inflammation
by reducing proinflammatory cytokines, IL4, IL6, tumor
necrosis factor-α (TNF-α), INF-γ and high sensitivity C
reactive protein (hsCRP) and by increasing expression of
IL-10 and Treg-related cytokines at mesenteric lymph
nodes. A new mechanism proposed to demonstrate the
effectiveness of probiotics is the inhibition of the mature
dendritic cell differentiation and transformation of naive T
cells into Th2. Immunomodulation decreases the
susceptibility to inflammatory and allergic factors
modulating the intestine skin axis. Probiotics also modulate
brain function including stress response on the intestine-
brain axis in rats and human subjects (Messaoudi M. et al.,
2010). Normalization of microbial composition provides
protection against pathogens at the mucosal surface. It has
been shown that the levels of Bifidobacterium longum in
the feces of healthy children were significantly higher than
in children with allergy, suggestigLDEng the role of this
strain in preventing the occurrence of bronchial asthma and
allergic dermatitis (Akay, 2014).
The metabolic effect via probiotic consumption, can
be associated with the reduction of blood glucose,
insulinemia and insulin resistance.
Psoriasis
Psoriasis is a systemic inflammatory disease
characterized by scaly lesioned plaques with defined
borders. These lesions are mainly located on the scalp and
large areas of the extremities but can occur at any site of the
body. The prevalence of psoriasis is around 1–3%, with
differences between countries, corresponding the highest
prevalence to Western countries.
The etiopathogenesis of psoriasis is not fully known,
although most authors postulate that it would be a skin
disorder of genetic origin, finally triggered by external
factors that would cause changes at the immune level. The
disease is associated with inflammation in other systems
and organs, as evidenced by the fact of finding a correlation
with inflammatory bowel disease, where between 7 and
11% of diagnosed patients also suffer from psoriasis. Other
components as triggers of psoriasis are age, the
comorbidity, environmental and external factors.
Although data on probiotic supplementation in
psoriasis treatment are limited, promising outcomes have
been documented with oral probiotics interventions, and
more studies are needed to explore the role of topical
probiotics in psoriasis therapy.
Immune regulation
One of the most important properties required for a
potential probiotic strain is the capacity of sticking to the
epithelial cells. In this regard, Galdeano et al. (2006)
demonstrated using electronic microscopy that 2 probiotic
microorganisms, L. casei CRL 431 and L. paracasei CNCM
I-1518, adhere to the intestinal epithelial cells (IECs)
through the Toll-like receptors (TLRs) and mediate
immune stimulation. Following this interaction, an increase
in the cytokines production such as IL-6 and macrophage
chemoattractant protein 1 from the IECs occurred, without
altering the intestinal barrier; only a slight increase in the
mononuclear cell infiltration of small intestine was
observed. The authors also demonstrated that only
fragments of the probiotic bacteria, and not the whole
bacteria, were internalized inside the IECs. As a
consequence, the IECs initiate a complex network of
signals that stimulate the immune cells associated with the
lamina propria and activate mainly the innate response and
the cytokines released by T cells.
The immunomodulatory effect of probiotics is
attributed to the release of cytokines, including interleukins
(ILs), tumor necrosis factors (TNFs), interferons (IFNs),
transforming growth factor (TGF), and chemokines from
immune cells (lymphocytes, granulocytes, macrophages,
mast cells, epithelial cells, and dendritic cells (DCs))
(Savan et al., 2006) which further regulate the innate and
adaptive immune system (Follign’e et al., 2010). In
addition, two surface phagocytosis receptors (FcRIII and
tool-like receptor (TLR)) are also upregulated by NO
(Delcenserie et al., 2008; Schwandner et al., 1999).
Probiotics have been reported to interact with enterocytes
and dendritic, T1, T2, and Treg cells in the intestine and to
modulate the adaptive immunity into pro- and/or anti-
inflammatory action. Studies with BALB/c (20–30 g)
inbred mice and Fisher-344 inbred rats demonstrated that
Lactobacillus paracasei subsp. Paracasei DC412 strain
and L. acidophilus NCFB 1748 induced early innate
immune responses and specific immune markers through
phagocytosis, polymorphonuclear (PMN) cell recruitment,
and TNFalpha (TNF-α) production (Kourelis A. et al.,
2010). In another experimental animal model involving
BALB/c mice, oral administration of L. casei favored rapid
activation of immune cells and produced a higher number
of specific markers such as CD-206 and TLR2 cells
(Galdeano et al., 2006), while TLRs improve the
immunological defense mechanism in terms of pro- and
anti-inflammatory cytokine production upon the detection
of foreign objects (Anderson, 2000).
One of the ways probiotics promote human health is
by inhibiting the growth of pathogenic bacteria through the
synthesis of low molecular weight compounds such as
organic acid and large molecular weight antimicrobial
compounds termed bacteriocins. Organic acids are acetic
and lactic acids. These compounds have been proven to
exhibit strong inhibitory effects against pathogenic gram-
negative bacteria such as H. pylori. Some bacteriocins
produced by probiotics are lactacin B from L. acidophilus,
bifidocin B produced by Bifidobacterium bifidum NCFB,
plantaricin from L. plantarum, and nisin from Lactococcus
lactis.
Probiotics modulate the composition of gut microbial
Probiotics in dermatological and cosmetic products – application and efficiency
19
Макед. фарм. билт., 68 (1) 9 – 26 (2022)
species by maintaining the balance and suppressing the
growth of potential pathogenic bacteria in the gut. It has
been reported that L. acidophilus or L. casei increased LAB
with a concomitant decrease of fecal coliforms and
anaerobes. In addition, a study by Li et al. (2016) found
that probiotics caused shifts in the gut microbiota
composition toward specific beneficial bacteria, for
example, Prevotella and Oscillibacter. These bacteria are
known to produce anti-inflammatory metabolites, which
subsequently decreased the Th17 polarization and favored
the differentiation of anti-inflammatory Treg/Type 1
regulatory T (Tr1) cells in the gut.
Wound healing
Skin microorganisms contribute to the host immunity
via different pathways. S. epidermidis has been shown to
boost the host immunity by inducing AMPc such as β
defensins 2 and 3 to S. aureus, by activating mast cell-
mediated antiviral immunity, by suppressing uncontrolled
inflammatory reactions during wound healing, inducing
skin’s AMP production and by stimulating cutaneous T-
cell maturation.
The use of Lactobacillus plantarum as a potential
therapeutic agent for the local treatment of Pseudomonas
aeruginosa burn infections have shown positive results.
The same bacteria has been investigated for the treatment
of non-diabetic and diabetic ulcers. Peral et al. (2009)
found that it reduced bacterial load, neutrophils, and
apoptotic and necrotic cells, modified interleukin-8
production, and induced wound healing.
Regulatory aspects (labeling, marketing)
The U.S. Food and Drugs Administration (FDA)
categorizes probiotics into different categories, such as
dietary supplements, foods, food additives, cosmetics, or
drugs, based on each individual product. However, there is
no regulation for topical probiotics, and currently there are
no topical probiotic products approved by the FDA, which
shows a need to improve the regulations for these products.
There are scores of topical cosmetic probiotic skin care
products marketed and sold in the U.S., and the number
continues to grow. While there is no restriction on
including probiotics in cosmetic products, and FDA pre-
market approval is not required, companies must still
ensure their products are safe before they are marketed to
consumers. In addition to testing for safety, products that
include live microorganisms should be tested to ensure
stability for the anticipated life of the product under normal
use indications. FDA continues to evaluate scientific data
on the safety of probiotics, but, as of the date of
publication, has not issued any guidance documents
addressing the use of such ingredients in cosmetics.
As with any other cosmetic product, companies that
market topical probiotic skin care products are proscribed
from claiming the product affects the structure or function
of the body; such claims effectively convert a product from
a legally marketed cosmetic into an unapproved drug. For
example, claims that a probiotic skin care product can
reverse the signs of aging, stimulate the growth of collagen
and elastin, or treat acne are structure/function claims that
cannot be used to legally market cosmetics in the U.S.
While a number of company’s market probiotic cosmetic
products making such claims, they do so at their peril and
run the risk FDA will categorize the products as
unapproved adulterated drugs that cannot lawfully be
marketed in the U.S. without pre-market approval. To
avoid regulatory challenges and possible product recall,
companies marketing probiotic skin care products should
ensure all product claims are truthful and not misleading
and refrain from making structure/function or disease
claims on the product label or labeling accompanying the
product, as well as on company web sites and social media
accounts.
In the EU, as with all cosmetics, prebiotics, probiotics
and postbiotics must meet key cosmetic principles.
However, there is still no specific regulation or guidelines
regarding these cosmetically active substances in relation
to the microbiome. Namely, although the skin microbiota
cannot be separated from the skin, in cosmetic regulation,
skin microbiome / microbiota not yet defined, it is not even
described. However, the expectation is that scientific
results will be implemented in the regulation for cosmetic
products and the basic ones will be introduced definitions
and security aspects. Most importantly, it should be noted
that these cosmetic substances are not prohibited (for
example, not listed in Annex II of EC12323 / 2009) and
can therefore be used safely. Therefore, they can be
considered for "classic" substances, which clearly indicates
the need to comply with cosmetics definition and key
principles.
Prebiotics and postbiotics can be used and are already
used as cosmetics substances. Probiotics, when used in
cosmetics, have in order to protect the skin from external
influences and to feed healthy bacteria for affect the pH or
hydration of the skin, for example. Consequently, in the
absence of a cosmetic policy on probiotics, living
organisms may for the time being are considered classic
cosmetic ingredients that fall under the general definition
of cosmetic raw materials.
It is worth mentioning that the Voluntary International
Group of Regulatory Authorities for Cosmetics
(International Cooperation on Cosmetics Regulation - from
Brazil, Canada, China, EU, Japan, Republic of Korea and
USA) in 2018 formed a joint working group precisely in
order to examine in detail the probiotic-based products in
the cosmetics sector. Based on the observations, the group
was identified the potential need to develop internationally
accepted terminology guidelines, safety and quality of
these substances in cosmetic products.
In this regard, it is important to emphasize the
cosmetic key principles and their association when it
comes to probiotics as active substances. Namely, if we
E. Hyseni, M. Glavas Dodov
20
Maced. pharm. bull., 68 (1) 9 – 26 (2022)
look at the cosmetic regulatory framework, each country
has its own definition of what 31 is a cosmetic product but
has similarities in criteria such as location of application
and cosmetic function. As such, to be considered a cosmetic
product, the product must contain the following:
Place of application: The product must be used on the
outside of the body, e.g. epidermis, nails, teeth, hair, lips
and external genitalia organs.
Function: The function of the product should be to
clean, beautify, correct body odors or otherwise to keep the
body in good condition. While probiotic-based products
meet the above criteria above, cosmetic key principles
include additional factors such as safety, microbiological
quality and permissible claims, as well as shelf life and
packaging.
Safety: The cosmetic product (and its ingredients)
must be safe for its intended use. Safety assessment is a
mandatory requirement in EU cosmetics legislation (and
widely sought after by companies and regulators in North
America, ASEAN countries and China) as proof that the
product is safe to use. Security is the responsibility of
industry. Probiotics are living organisms that are dynamic
and therefore different from other cosmetic substances.
Hence, in the cosmetics industry there is a need to adjust
and design a safety assessment applicable to cosmetics
based on probiotic. For example, it is essential to prove that
the microorganism used does not produce toxins, to prove
the absence of pathogenicity and to determine antibiotic
resistance and metabolic activity (common criteria for a
similar safety assessment as in the food sector).
Microbiological quality: Cosmetic products are not
expected to be sterile, but they still must meet the
microbiological quality requirements (total number of
aerobic mesophilic microorganisms (bacteria, yeasts and
molds) and the absence of specific microorganisms
(Candida albicans, Staphylococcus aureus, Pseudomonas
aeruginosa, Escherichia coli). It is described in the
instructions of EU Scientific Committee on Consumer
Safety (SCCS): even if it is not legally required, this
microbiological test is necessary to guarantee the quality of
the cosmetic product and the safety of the consumer
(Regulation (EC) No 1223/2009 of the European
Parliament and of the Council of 30 November 2009 on
cosmetic products).
Hence, it remains unclear how well probiotics meet
the requirements for microbiological quality that generally
applies to all cosmetic products. Microorganisms are
generally considered contaminants, with a focus on
pathogens microorganisms, because cosmetics are
formulated to minimize their growth. Therefore, it is
important to educate both cosmetic companies and
consumers/patients about the specifics when it comes to
probiotics as active substances in cosmetics products.
Claims: Cosmetic claims must be confirmed, not false
or unproven. In the context of naming a particular product
as a probiotic, that means that the final manufactured
cosmetic product must contain probiotics and most
importantly, probiotics must be alive throughout shelf life
of the product. However, more and more products on the
market contain claims to contain probiotic ingredients, but,
as already mentioned, research has shown that many of
these products on the market contain primarily postbiotics
and prebiotics, not live bacteria. As a result, inappropriate
use of various undefined terms (probiotic, prebiotic and
postbiotic) creates confusion about these products.
It is therefore clear that it is necessary to develop
internationally accepted definitions of key terms that need
to be used consistently in order to minimize confusion and
to avoid misunderstanding, both with regulators and
consumers. In the food and food supplements industry,
each European country has its own country-specific
labeling rules, but in some European countries
manufacturers cannot even put the word "probiotic" on the
label of the product.
For all the above reasons, the joint working group
“Microbiome and ICCR's Cosmetics” works to educate key
stakeholders (regulatory bodies, cosmetics industry and
consumers) in order to achieve a better understanding and
augmentation.
Conclusion
Topical probiotics are definitely a promising range of
products in treating various skin conditions. It has been
demonstrated that different probiotics strains can help
improve the outcomes of acne, atopic dermatitis, psoriasis,
wound healing, oral and intimate care, but there is yet a lot
to be understood.
The lack of knowledge regarding skin’s microbiome,
the site-specific diversity of microorganisms makes it
difficult for topical probiotics developers to achieve the
desired formulation. The optimal dosage and strain for
various skin conditions has yet to be determined. However,
there is enough topical probiotics evidence to keep
searching new ways or strains to achieve their full potential
worth it.
References
Aguilar-Toalá, J.E., Garcia-Varela, R., Garcia, H.S., Mata-Haro,
V., González-Córdova, A.F., Vallejo-Cordoba, B.,
Hernández-Mendoza, A., 2018. Postbiotics: An evolving
term within the functional foods field. Trends in Food
Science & Technology 75, 105–114. Available at:
https://doi.org/10.1016/j.tifs.2018.03.009
Akay, H.K., Bahar Tokman, H., Hatipoglu, N., Hatipoglu, H.,
Siraneci, R., Demirci, M., Borsa, B.A., Yuksel, P.,
Karakullukcu, A., Kangaba, A.A., Sirekbasan, S., Aka, S.,
Mamal Torun, M., Kocazeybek, B.S., 2014. The
relationship between bifidobacteria and allergic asthma
and/or allergic dermatitis: A prospective study of 0–3
years-old children in Turkey. Anaerobe 28, 98–103.
Available at:
https://doi.org/10.1016/j.anaerobe.2014.05.006
Probiotics in dermatological and cosmetic products – application and efficiency
21
Макед. фарм. билт., 68 (1) 9 – 26 (2022)
Al-Ghazzewi, F.H., Tester, R.F.,2010. Effect of Konjac
glucomannan hydrolysates and probiotics on the growth of
the skin bacterium propionibacterium acnes in vitro.
International Journal of Cosmetic Science 32(2), 139–142.
Available at: 10.1111/j.1468-2494.2009.00555.x
Anderson, K.V., 2000. Toll signaling pathways in the innate
immune respon se. Current Opinion in Immunology 12(1),
13–19. Available at: 10.1016/s0952-7915(99)00045-x
Avershina, E., Cabrera Rubio, R., Lundgård, K., Perez Martinez,
G., Collado, M. C., Storrø, O., Øien, T., Dotterud, C.K.,
Johnsen, R., Rudi, K., 2017. Effect of probiotics in
prevention of atopic dermatitis is dependent on the intrinsic
microbiota at early infancy. Journal of Allergy and Clinical
Immunology, 139(4). Available at:
https://doi.org/10.1016/j.jaci.2016.09.056
Bek-Thomsen, M., Lomholt, H.B., Kilian, M., 2008. Acne is not
associated with yet-uncultured bacteria. Journal of Clinical
Microbiology 46(10), 3355–3360. Available at:
https://doi.org/10.1128/JCM.00799-08
Bibel, D.J., Aly, R., Shinefield, H.R., 1992. Antimicrobial
activity of sphingosines. Journal of Investigative
Dermatology 98(3), 269–273. Available
at: 10.2340/0001555573407411
https://www.medicaljournals.se
Boehncke, W.-H., Schön, M.P., 2015. Psoriasis. The Lancet
386(9997), 983–994. Available at: 10.1016/S0140-
6736(14)61909-7
Bouilly-Gauthier, D., Jeannes, C., Maubert, Y., Duteil, L.,
Queille-Roussel, C., Piccardi, N., Montastier, C., Manissier,
P., Piérard, G., Ortonne, J.P., 2010. Clinical evidence of
benefits of a dietary supplement containing probiotic and
carotenoids on ultraviolet-induced skin damage. British
Journal of Dermatology 163(3), 536–543. Available at:
https://doi.org/10.1111/j.1365-2133.2010.09888.x
Bowe W.P. 2013. Probiotics in acne and rosacea. Cutis, 92(1), 6–
7. Available at: https://pubmed.ncbi.nlm.nih.gov
Bowe, W.P., Logan, A.C., 2011. Acne vulgaris, probiotics and the
gut-brain-skin axis - back to the future?. Gut Pathog. 3(1), 1.
Available at: 10.1186/1757-4749-3-1
Bowe, W.P., Filip, J.C., DiRienzo, J.M., Volgina, A., Margolis,
D.J., 2006. Inhibition of propionibacterium acnes by
bacteriocin-like inhibitory substances (BLIS) produced by
Streptococcus salivarius. J. Drugs Dermatol. 9, 868-70.
Available at: Inhibition of propionibacterium acnes by
bacteriocin-like inhibitory substances (BLIS) produced by
Streptococcus salivarius - PubMed (nih.gov)
Brook, I., 1999. Bacterial interference. Critical Reviews in
Microbiology 25(3), 155–172. Available at:
https://doi.org/10.1080/10408419991299211
Callewaert, C., Ravard Helffer, K., Lebaron, P., 2020. Skin
microbiome and its interplay with the environment.
American Journal of Clinical Dermatology 21(S1), 4–
11. Available at: 10.1007/s40257-020-00551-x
Capone, K.A., Dowd, S.E., Stamatas, G.N., Nikolovski, J., 2011.
Diversity of the human skin microbiome early in life. Journal
of Investigative Dermatology 131(10), 2026–2032.
Available at: 10.1038/jid.2011.168
Chaikham, P., Apichartsrangkoon, A., Jirarattanarangsri, W., Van
de Wiele, T., 2012. Influence of encapsulated probiotics
combined with pressurized longan juice on colon microflora
and their metabolic activities on the exposure to simulated
dynamic gastrointestinal tract. Food Research International
49(1), 133–142. Available at: 10.1016/j.foodres.2012.07.033
Cho, S., 2014. The role of functional foods in cutaneous anti-
aging. Journal of Lifestyle Medicine 4(1), 8–16. Available
at: 10.15280/jlm.2014.4.1.8
Clavaud, C., Jourdain, R., Bar-Hen, A., Tichit, M., Bouchier, C.,
Pouradier, F., El Rawadi, C., Guillot, J., Ménard-Szczebara,
F., Breton, L., Latgé, J.-P., Mouyna, I., 2013. Dandruff is
associated with disequilibrium in the proportion of the major
bacterial and fungal populations colonizing the scalp. PLoS
ONE 8(3). Available at:
https://doi.org/10.1371/journal.pone.0058203
Cosseau, C., Devine, D.A., Dullaghan, E., Gardy, J.L.,
Chikatamarla, A., Gellatly, S., Yu, L.L., Pistolic, J., Falsafi,
R., Tagg, J., Hancock, R. E., 2008. The commensal
streptococcus salivarius K12 downregulates the innate
immune responses of human epithelial cells and promotes
host-microbe homeostasis. Infection and Immunity 76(9),
4163–4175. Available at: 10.1128/IAI.00188-08
Da Cunha, N.B., Cobacho, N.B., Viana, J.F.C., Lima, L.A.,
Sampaio, K.B.O., Dohms, S.S.M., Ferreira, A.C.R., de la
Fuente-Núñez, C., Costa, F.F., Franco, O.L., Dias, S.C.,
2017. The next generation of antimicrobial peptides (AMPS)
as molecular therapeutic tools for the treatment of diseases
with social and economic impacts. Drug Discovery Today
22(2), 234–248. Available at:
https://doi.org/10.1016/j.drudis.2016.10.017
Dagnelie, M., Corvec, S., Saint-Jean, M., Bourdès, V., Nguyen,
J., Khammari, A., Dréno, B., 2018. Decrease in diversity of
Propionibacterium acnes phylotypes in patients with severe
acne on the back. Acta Dermato Venereologica 98(2), 262–
267. Available at: 10.2340/00015555-2847
Del Rosso, J.Q., 2019. Monoclonal antibody therapies for Atopic
Dermatitis: Where Are We Now in the Spectrum of Disease
Management?. J. Clin. Aesthet. Dermatol. 12(2), 39-41.
Available at: MONOCLONAL ANTIBODY THERAPIES
for Atopic Dermatitis: Where Are We Now in the Spectrum
of Disease Management? - PMC (nih.gov)
Delcenserie, V., Martel, D., Lamoureux, M., Amiot, J., Boutin,
Y., Roy, D., 2008. Immunomodulatory effects of probiotics
in the intestinal tract. Curr. Issues Mol. Biol. 10(1-2), 37-54.
Available at:10.21775/cimb.010.037 www.mdpi.com
Dessinioti, C., Antoniou, C., Katsambas, A., 2014. Acneiform
eruptions. Clinics in Dermatology 32(1), 24–34. Available
at: https://doi.org/10.1016/j.clindermatol.2013.05.023
Di Marzio, L., Centi, C., Cinque, B., Masci, S., Giuliani, M.,
Arcieri, A., Zicari, L., De Simone, C., Cifone, M.G., 2003.
Effect of the lactic acid bacterium streptococcus
thermophilus on stratum corneum ceramide levels and signs
and symptoms of atopic dermatitis patients. Experimental
Dermatology 12(5), 615–620. Available at: 10.1034/j.1600-
0625.2003.00051.x
Di Marzio, L., Cinque, B., Cupelli, F., De Simone, C., Cifone,
M.G., Giuliani, M., 2008. Increase of skin-ceramide levels
in aged subjects following a short-term topical application of
bacterial sphingomyelinase from Streptococcus
Thermophilus. International Journal of Immunopathology
and Pharmacology 21(1), 137–143. Available at:
10.1177/039463200802100115
Di Marzio, L., Cinque, B., De Simone, C., Cifone, M.G., 1999.
Effect of the lactic acid bacterium streptococcus
thermophilus on ceramide levels in human keratinocytes in
vitro and stratum corneum in vivo. Journal of Investigative
Dermatology 113(1), 98–106. Available at: 10.1046/j.1523-
1747.1999.00633.x
E. Hyseni, M. Glavas Dodov
22
Maced. pharm. bull., 68 (1) 9 – 26 (2022)
Dopytalska, K., Sobolewski, P., Błaszczak, A., Szymańska, E., &
Walecka, I., 2018. Psoriasis in special localizations.
Reumatologia/Rheumatology 56(6), 392–398. Available
at: 10.5114/reum.2018.80718
Dréno, B., 2017. What is new in the pathophysiology of acne, an
Overview. Journal of the European Academy of
Dermatology and Venereology 31, 8–12. Available at:
https://doi.org/10.1111/jdv.14374
Dréno, B., Araviiskaia, E., Berardesca, E., Gontijo, G., Sanchez
Viera, M., Xiang, L.F., Martin, R., Bieber, T., 2016.
Microbiome in healthy skin, update for dermatologists.
Journal of the European Academy of Dermatology and
Venereology 30(12), 2038–20. Available
at: 10.1111/jdv.13965
Dréno, B., Dagnelie, M.A., Khammari, A., Corvec, S., 2020. The
skin microbiome: A new actor in inflammatory acne.
American Journal of Clinical Dermatology 21(S1), 18–24.
Available at: 10.1007/s40257-020-00531-1
Ellis, S.R., Nguyen, M., Vaughn, A.R., Notay, M., Burney, W.A.,
Sandhu, S., Sivamani, R.K., 2019. The skin and gut
microbiome and its role in common dermatologic
conditions. Microorganisms 7(11), 550. Available
at: 10.3390/microorganisms7110550
Ereaux, L.P., 1938. Facts, fads and fancies in the treatment of
acne vulgaris. Can. Med. Assoc. J. 39(3), 257-261.
Available at:
https://www.ncbi.nlm.nih.gov/pmc/articles/PMC538466/p
df/canmedaj00196-0051.pdf.
Flores-Díaz, M., Monturiol-Gross, L., Naylor, C., Alape-Girón,
A., Flieger, A., 2016. Bacterial sphingomyelinases and
phospholipases as virulence factors. Microbiology and
Molecular Biology Reviews 80(3), 597–628. Available
at: 10.1128/MMBR.00082-15
Foligné, B., Dewulf, J., Breton, J., Claisse, O., Lonvaud-Funel,
A., Pot, B., 2010. Probiotic properties of non-conventional
lactic acid bacteria: Immunomodulation by Oenococcus
Oeni. International Journal of Food Microbiology 140(2-3),
136–145. Available at:
https://doi.org/10.1016/j.ijfoodmicro.2010.04.007
https://pubmed.ncbi.nlm.nih.gov
Forbes, J.D., Van Domselaar, G., Bernstein, C.N., 2016. The gut
microbiota in immune-mediated inflammatory diseases.
Frontiers in Microbiology 7. Available at:
10.14309/ajg.0000000000000305
https://pubmed.ncbi.nlm.nih.gov
Frazier, W., Bhardwaj, N., 2020. Atopic Dermatitis: Diagnosis
and Treatment. Am. Fam. Physician 101(10), 590-598.
PMID: 32412211. Available at
https://pubmed.ncbi.nlm.nih.gov
Gabriela, P., Carolina, M.G., Ivanna, N.N., 2016. Malnutrition:
Role of the Diet on the Microbiota OPEN and In the
Functioning of the Gut Immune System. SM J. Public
Health Epidemiol. 2(1), 1023. Available at:
https://ri.conicet.gov.ar/bitstream/handle/11336/46821/CO
NICET_Digital_Nro.f50745c0-f846-4fb7-b769-
8c391b627049_A.pdf?sequence=2&isAllowed=y.
Galdeano, C.M., Perdign, G., 2006. The probiotic bacterium
lactobacillus casei induces activation of the gut mucosal
immune system through innate immunity. Clinical and
Vaccine Immunology 13(2), 219–226. Available at:
10.1128/CVI.13.2.219-226.2006
Galdeano, C.M., Perdigon, G., 2004. Role of viability of
probiotic strains in their persistence in the gut and in
mucosal immune stimulation. Journal of Applied
Microbiology 97(4), 673–681. Available at:
10.1111/j.1365-2672.2004.02353.x
Gill, S.R., Pop, M., DeBoy, R.T., Eckburg, P.B., Turnbaugh, P.J.,
Samuel, B.S., Gordon, J.I., Relman, D.A., Fraser-Liggett,
C.M., Nelson, K.E., 2006. Metagenomic analysis of the
human distal gut microbiome. Science 312(5778), 1355–
1359. Available at: 10.1126/science.1124234
Gittler, J.K., Shemer, A., Suárez-Fariñas, M., Fuentes-Duculan,
J., Gulewicz, K.J., Wang, C.Q. F., Mitsui, H., Cardinale, I.,
de Guzman Strong, C., Krueger, J.G., Guttman-Yassky, E.,
2012. Progressive activation of th2/TH22 cytokines and
selective epidermal proteins characterizes acute and
chronic atopic dermatitis. Journal of Allergy and Clinical
Immunology 130(6), 1344–1354. Available at:
10.1016/j.jaci.2012.07.012
Goodarzi, A., Mozafarpoor, S., Bodaghabadi, M., Mohamadi,
M., 2020. The potential of probiotics for treating acne
vulgaris: A review of literature on acne and microbiota.
Dermatologic Therapy 33(3). Available at:
10.1111/dth.13279
Grice, E.A., Segre, J.A., 2011. The skin microbiome. Nature
Reviews Microbiology 9(4), 244–253. Available at:
https://doi.org/10.1038/nrmicro2537
Guéniche, A., Bastien, P., Ovigne, J.M., Kermici, M., Courchay,
G., Chevalier, V., Lionel Breton, L., Castiel-Higounenc, I.,
2010. Bifidobacterium longum lysate, a new ingredient for
reactive skin. Exp. Dermatol. 19:e1-8. Available at:
10.1111/j.1600-0625.2009.00932.x
Gueniche, A., Benyacoub, J., Philippe, D., Bastien, P., Kusy, N.,
Breton, L., Blum, S., Castiel-Higounenc, I., 2010.
Lactobacillus paracasei CNCM I-2116 (ST11) inhibits
substance P-induced skin inflammation and accelerates
skin barrier function recovery in vitro. Eur. J. Dermatol. 20,
731-737. Available at: 10.1684/ejd.2010.1108.
Gueniche, A., Knaudt, B., Schuck, E., Volz, T., Bastien, P.,
Martin, R., Röcken, M., Breton, L., Biedermann, T., 2008.
Effects of nonpathogenic gram- negative bacterium
Vitreoscilla filiformis lysate on atopic dermatitis: a
prospective, randomized, doubleblind, placebo-controlled
clinical study. Br. J. Dermatol. 159, 1357–1363. Available
at: 10.1111/j.1365-2133.2008.08836.x
Gueniche, A., Senyacoub, J., Slum, S., Sreton, L., Castie, I.,
2009. Probiotics for skin benefits. Nutritional Cosmetics:
Beauty from Within 421-439. Available at:
https://doi.org/10.1016/B978-0-8155-2029-0.50029-6
Ha, H.K., Jeon, N.-E., Kim, J.W., Han, K.-S., Yun, S. S., Lee,
M.-R., Lee, W.J., 2016. Physicochemical characterization
and potential prebiotic effect of whey protein isolate/inulin
nano complex. Korean Journal for Food Science of Animal
Resources 36(2), 267–274. Available at:
10.5851/kosfa.2021.e45
Hancox, J.G., Sheridan, S.C., Feldman, S.R., Fleischer, A.B.,
2004. Seasonal variation of Dermatologic Disease in the
USA: A study of office visits from 1990 to 1998.
International Journal of Dermatology 43(1), 6–11.
Available at: 10.1111/j.1365-4632.2004.01828.x
https://onlinelibrary.wiley.com/
Holmes. A.D., Waite, K.A., Chen, M.C., Palaniswamy, K.,
Wiser, T.H., Draelos, Z.D., Rafal, E.S., Werschler, W.P.,
Harvey, A.E., 2015. Dermatological adverse events
associated with topical brimonidine gel 0.33% in subjects
with erythema of rosacea: a retrospective review of clinical
studies. J. Clin. Aesthet. Dermatol. 8, 29–35. Available at:
https://pubmed.ncbi.nlm.nih.gov/26345379/.
Probiotics in dermatological and cosmetic products – application and efficiency
23
Макед. фарм. билт., 68 (1) 9 – 26 (2022)
Huang, R., Ning, H., Shen, M., Li, J., Zhang, J., Chen, X., 2017.
Probiotics for the treatment of atopic dermatitis in children:
A systematic review and meta-analysis of randomized
controlled trials. Frontiers in Cellular and Infection
Microbiology 7. Available at:
https://doi.org/10.3389/fcimb.2017.00392
Im, A., Lee, B., Kang, D., Chae, S., 2019. Protective effects of
tyndallized Lactobacillus acidophilus IDCC 3302 against
UVB-induced photodamage to epidermal keratinocytes
cells". International Journal of Molecular Medicine 43(6),
2499-2506. Available at: org/10.3892/ijmm.2019.4161
Jang, S.O., Kim, H.J., Kim, Y.J., Kang, M.J., Kwon, J.W., Seo,
J.H., Kim, H.Y., Kim, B.J., Yu, J., Hong, S.J., 2012. Asthma
prevention by lactobacillus rhamnosus in a mouse model is
associated with CD4+CD25+Foxp3+T cells. Allergy,
Asthma & Immunology Research 4(3), 150. Available at:
10.4168/aair.2012.4.3.150
Jasson, F., Nagy, I., Knol, A.C., Zuliani, T., Khammari, A.,
Dréno, B., 2013. Different strains of Propionibacterium
acnes modulate differently the cutaneous innate immunity.
Exp. Dermatol. 22(9), 587–92. Available at:
10.1111/exd.12206 https://onlinelibrary.wiley.com/
Jeziorkowska, R., Sysa-Jędrzejowska, A., Samochocki, Z., 2015.
Topical steroid therapy in atopic dermatitis in theory and
practice. Postepy Dermatol. Alergol 32(3), 162-166.
Available at: https://doi.org/10.5114/pdia.2014.40962
Juhlin, L., Bernard, B.A., Dalbiez, C., 1985. Stellate fibrin-
fibronectin microclot formation from keratinocytes and
fibroblasts stimulated by plasma from patients with psoriasis.
British Journal of Dermatology 112(3), 271–276. Available
at: 10.1155/2017/7215072
Kang, B.S., Seo, J.G., Lee, G.S., Kim, J.H., Kim, S.Y., Han,
Y.W., Kang, H., Kim, H.O., Rhee, J.H., Chung, M.J., Park,
Y.M., 2009. Antimicrobial activity of enterocins from
Enterococcus faecalis SL-5 against Propionibacterium
acnes, the causative agent in acne vulgaris, and its
therapeutic effect. The Journal of Microbiology 47(1), 101–
109. Available at: 10.1007/s12275-008-0179-y
Kang, Y.M., Hong, C.H., Kang, S.H., Seo, D.S., Kim, S.O., Lee,
H.Y., Sim, H.J., An, H.J., 2020. Anti-photoaging effect of
plant extract fermented with lactobacillus buchneri on CCD-
986SK fieforbesbroblasts and HaCaT keratinocytes. Journal
of Functional Biomaterials 11(1), 3. Available at:
https://doi.org/10.3390/jfb11010003
Kaur, K., Rath, G., 2019. Formulation and evaluation of UV
protective synbiotic skin care topical formulation. Journal of
Cosmetic and Laser Therapy 21(6), 332–342. Available
at: 10.1080/14764172.2019.1658878
Kim, H.M., Lee, D.E., Park, S.D., Kim, Y.T., Kim, Y. J., Jeong,
J.W., Jang, S. S., Ahn, Y.T., Sim, J.H., Huh, C.S., Chung,
D.K., Lee, J.H., 2014. Oral Administration of Lactobacillus
plantarum HY7714 protects hairless mouse against
ultraviolet B-induced photoaging. Journal of Microbiology
and Biotechnology 24(11), 1583–1591. Available at:
10.4014/jmb.1406.06038
Kim, H.J., Kim, H.Y., Lee, S.Y., Seo, J.H., Lee, E., Hong, S.J.,
2013. Clinical efficacy and mechanism of probiotics in
allergic diseases. Korean Journal of Pediatrics 56(9), 369.
Available at: http://dx.doi.org/10.3345/kjp.2013.56.9.369
Kim, J., Kim, H., 2019. Microbiome of the skin and gut in atopic
dermatitis (AD): Understanding the pathophysiology and
finding novel management strategies. Journal of Clinical
Medicine 8(4), 444. Available at: 10.3390/jcm8040444
Kober, M.M., Bowe, W.P., 2015. The effect of probiotics on
immune regulation, acne, and photoaging. Int. J. Womens
Dermatol. 1(2), 85-89. Available at:
10.1016/j.ijwd.2015.02.001
Kolarsick, P.A.J., Kolarsick, M.A., Goodwin, C., 2011. Anatomy
and Physiology of the Skin, Journal of the Dermatology
Nurses' Association 3(4), 203-213. Available at:
10.1097/JDN.0b013e3182274a98
Kong, H.H., Segre, J.A., 2012. Skin microbiome: Looking back
to move forward. Journal of Investigative Dermatology
132(3), 933–939; Available at: 10.1038/jid.2011.417
Kourelis, A., Zinonos, I., Kakagianni, M., Christidou, A.,
Christoglou, N., Yiannaki, E., Testa, T., Kotzamanidis, C.,
Litopoulou-Tzanetaki, E., Tzanetakis, N., Yiangou, M.,
2010. Validation of the dorsal air pouch model to predict and
examine immunostimulatory responses in the gut. Journal of
Applied Microbiology 108(1), 274–284. Available at:
10.1111/j.1365-2672.2009.04421.x
Lai, Y., Cogen, A.L., Radek, K.A., Park, H.J., MacLeod, D.T.,
Leichtle, A., Ryan, A.F., Di Nardo, A., Gallo, R.L., 2010.
Activation of TLR2 by a small molecule produced by
Staphylococcus epidermidis increases antimicrobial defense
against bacterial skin infections. Journal of Investigative
Dermatology 130(9), 2211–2221. Available at:
10.1038/jid.2010.123
LeBlanc, J.G., Chain, F., Martín, R., Bermúdez-Humarán, L.G.,
Courau, S., Langella, P., 2017. Beneficial effects on host
energy metabolism of short-chain fatty acids and vitamins
produced by commensal and probiotic bacteria. Microbial
Cell Factories 16(1). Available at: 10.1186/s12934-017-
0691-z
Lee, D.E., Huh, C.S., Ra, J., Choi, I.D., Jeong, J.W., Kim, S.H.,
Ryu, J.H., Seo, Y.K., Koh, J.S., Lee, J.H., Sim, J.H., Ahn,
Y.T., 2015. Clinical Evidence of Effects of Lactobacillus
plantarum HY7714 on Skin Aging: A Randomized, Double
Blind, Placebo-Controlled Study. J. Microbiol. Biotechnol.
25(12), 2160-8. Available at: 10.4014/jmb.1509.09021
Lee, J.H., Son, S.W., Cho, S.H., 2016. A comprehensive review
of the treatment of atopic eczema. Allergy, Asthma &
Immunology Research 8(3), 181. Available at:
10.4168/aair.2016.8.3.181
Lee, W.J., Jung, H.D., Lee, H.J., Kim, B.S., Lee, S.J., Kim, D.W.,
2008. Influence of substance-P on cultured sebocytes.
Archives of Dermatological Research 300(6), 311–316.
Available at: https://doi.org/10.1007/s00403-008-0854-1
Li, J., Sung, C.Y., Lee, N., Ni, Y., Pihlajamäki, J., Panagiotou,
G., El-Nezami, H., 2016. Probiotics modulated gut
microbiota suppresses hepatocellular carcinoma growth in
mice. Proceedings of the National Academy of Sciences
113(9). Available at:
https://doi.org/10.1073/pnas.1518189113
Majeed, M., Majeed, S., Nagabhushanam, K., Lawrence, L.,
Arumugam, S., Mundkur, L., 2020. Skin Protective Activity
of LactoSporin-the Extracellular Metabolite from Bacillus
Coagulans MTCC 5856. Cosmetics 7, 76. Available at
https://doi.org/10.3390/cosmetics7040076
Majeed, M., Majeed, S., Nagabhushanam, K., Mundkur, L.,
Rajalakshmi, H.R., Shah, K., Beede, K., 2020. Novel topical
application of a postbiotic, LactoSporin®, in mild to
moderate acne: A randomized, comparative clinical study to
evaluate its efficacy, tolerability and safety. Cosmetics 7(3),
70. Available at: 10.3390/cosmetics7030070
Mandal, S., Hati, S., 2016. Microencapsulation of bacterial cells
E. Hyseni, M. Glavas Dodov
24
Maced. pharm. bull., 68 (1) 9 – 26 (2022)
by emulsion technique for probiotic application. Cell
Microencapsulation, 273–279. Available at:
10.1111/jdi.13372
Matsubara, V.H., Bandara, H.M., Ishikawa, K.H., Mayer, M.P.,
Samaranayake, L.P., 2016. The role of probiotic bacteria in
managing periodontal disease: A systematic review. Expert
Review of Anti-Infective Therapy 14(7), 643–655.
Available at: 10.1080/14787210.2016.1194198
Messaoudi, M., Lalonde, R., Violle, N., Javelot, H., Desor, D.,
Nejdi, A., Bisson, J.-F., Rougeot, C., Pichelin, M.,
Cazaubiel, M., Cazaubiel, J.M., 2010. Assessment of
psychotropic-like properties of a probiotic formulation
(lactobacillus helveticusr0052 Andbifidobacterium
LONGUMR0175) in rats and human subjects. British
Journal of Nutrition 105(5), 755–764. Available at:
10.1017/S0007114510004319.
Morita, E., Takahashi, H., Niihara, H., Dekio, I., Sumikawa, Y.,
Murakami, Y., Matsunaka, H., 2010. Stratum corneum
TARC level is a new indicator of lesional skin inflammation
in atopic dermatitis. Allergy 65, 1166–1172. Available at:
https://doi.org/10.1111/j.1398-9995.2010.02361.x
Naik, S., Bouladoux, N., Wilhelm, C., Molloy, M.J., Salcedo, R.,
Kastenmuller, W., Deming, C., Quinones, M., Koo, L.,
Conlan, S., Spencer, S., Hall, J.A., Dzutsev, A., Kong, H.,
Campbell, D.J., Trinchieri, G., Segre, J.A., Belkaid, Y.,
2012. Compartmentalized control of skin immunity by
resident commensals. Science 337(6098), 1115–1119.
Available at:10.1126/science.1225152
Nakase, K., Nakaminami, H., Takenaka, Y., Hayashi, N.,
Kawashima, M., Noguchi, N., 2014. Relationship between
the severity of acne vulgaris and antimicrobial resistance of
bacteria isolated from acne lesions in a hospital in Japan.
Journal of Medical Microbiology 63(5), 721–728.
Available at: 10.1099/jmm.0.067611-0
Nielsen, D.S., Cho, G.-S., Hanak, A., Huch, M., Franz, C.M.A.P.,
Arneborg, N., 2010. The effect of bacteriocin-producing
lactobacillus plantarum strains on the intracellular ph of
sessile and planktonic listeria monocytogenes single cells.
International Journal of Food Microbiology 141. Available
at: 10.1016/j.ijfoodmicro.2010.03.040
Notay, M., Saric‐Bosanac, S., Vaughn, A.R., Dhaliwal, S.,
Trivedi, M., Reiter, P.N., Rybak, I., Li, C.C., Weiss, L.B.,
Ambrogio, L., Burney, W., Sivamani, R. K., 2019. The use
of topical nitrosomonas eutropha for cosmetic improvement
of facial wrinkles. Journal of Cosmetic Dermatology 19(3),
689–693. Available at: 10.1111/jocd.13060
Nwanodi, O., 2018. Skin Protective Nutraceuticals: The Current
Evidence in Brief. Healthcare (Basel) 6(2), 40. Available at:
10.3390/healthcare6020040
Parisi, R., Symmons, D.P.M., Griffiths, C.E.M., Ashcroft, D.M.,
2013. Global epidemiology of psoriasis: A systematic
review of incidence and prevalence. Journal of
Investigative Dermatology 133(2), 377–385. Available
at: 10.1038/jid.2012.339
Pascal, M., Perez-Gordo, M., Caballero, T., Escribese, M.M.,
Lopez Longo, M.N., Luengo, O., Manso, L., Matheu, V.,
Seoane, E., Zamorano, M., Labrador, M., Mayorga, C.,
2018. Microbiome and Allergic Diseases. Front Immunol.
9, 1584. Available at: 10.3389/fimmu.2018.01584
Patel, R.M., Denning, P.W., 2013. Therapeutic use of prebiotics,
probiotics, and postbiotics to prevent necrotizing
enterocolitis. Clinics in Perinatology 40(1), 11–25.
Available at: 10.1016/j.clp.2012.12.002
Paul, W.E., Zhu, J., 2010. How are th2-type immune responses
initiated and amplified? Nature Reviews Immunology
10(4), 225–235. Available at: 10.1038/nri2735
Pavicic, T., Wollenweber, U., Farwick, M., Korting, H.C., 2009.
Anti-microbial and -inflammatory activity and efficacy of
phytosphingosine: An in vitro and in vivo study addressing
acne vulgaris. International Journal of Cosmetic Science
29(3), 181–190. Available at:
https://doi.org/10.1111/j.1467-2494.2007.00378.x
Peral, M.C., Huaman Martinez, M.A., Valdez, J.C., 2009.
Bacteriotherapy with lactobacillus Plantarumin Burns.
International Wound Journal 6(1), 73–81. Available at:
https://doi.org/10.1111/j.1742-481X.2008.00577.x
Peral, M.C., Rachid, M.M., Gobbato, N.M., Martinez, M.A.H.,
Valdez, J.C., 2010. Interleukin-8 production by
polymorphonuclear leukocytes from patients with chronic
infected leg ulcers treated with lactobacillus plantarum.
Clinical Microbiology and Infection 16(3), 281–286.
Available at: 10.1111/j.1469-0691.2009.02793.x
Peyri, J., 1912. Topical bacteriotherapy of the skin. J. Cutaneous
Dis. 30, 688-689. Available at: 10.1186/1757-4749-3-1
Preidis, G.A., Hill, C., Guerrant, R.L., Ramakrishna, B.S.,
Tannock, G.W., Versalovic, J., 2011. Probiotics, enteric
and diarrheal diseases, and global health. Gastroenterology
140(1), 8-14 Available at:
https://doi.org/10.1053/j.gastro.2010.11.010
Ra, J., Lee, D.E., Kim, S.H., Jeong, J.W., Ku, H. K., Kim, T.Y.,
Choi, I.D., Jeung, W., Sim, J.H., Ahn, Y.T., 2014. Effect of
oral administration of lactobacillus plantarum HY7714 on
epidermal hydration in ultraviolet B-irradiated hairless
mice. J. Microbiol. Biotechnol. 24(12), 1736-1743.
Available at: https://doi.org/10.4014/jmb.1408.08023
Rather, I.A., Bajpai, V.K., Kumar, S., Lim, J., Paek, W.K., Park,
Y.H., 2016. Probiotics and atopic dermatitis: An overview.
Frontiers in Microbiology 7. Available at:
10.3389/fmicb.2016.00507
Retzler, J., Smith, A., Reaney, M., Rout, R., Hudson, R., 2019.
Process utilities for topical treatment in atopic dermatitis.
Qual. Life Res. 28(9), 2373-2381. Available
at: 10.1007/s11136-019-02174-0
Ruan, Y., Sun, J., He, J., Chen, F., Chen, R., Chen, H., 2015.
Effect of probiotics on glycemic control: A systematic
review and meta-analysis of randomized, controlled trials.
PLOS ONE 10(7). Available at:
10.1371/journal.pone.0132121
Rusu, E., Enache, G., Cursaru, R., Alexescu, A., Radu, R., Onila,
O., Cavallioti, T., Rusu, F., Posea, M., Jinga, M., Radulian,
G., 2019. Prebiotics and probiotics in atopic dermatitis.
Experimental and therapeutic medicine 18(2), 926–931.
Available at: 10.3892/etm.2019.7678
Salem, I., Ramser, A., Isham, N., Ghannoum, M.A., 2018. The
gut microbiome as a major regulator of the gut-skin axis.
Frontiers in Microbiology 9. Available
at: 10.3389/fmicb.2018.01459
Sánchez-Pellicer, P., Navarro-Moratalla, L., Núñez-Delegido, E.,
Ruzafa-Costas, B., Agüera-Santos, J., Navarro-López, V.,
2022. Acne, microbiome, and probiotics: The gut–skin
axis. Microorganisms 10(7), 1303.
https://doi.org/10.3390/microorganisms10071303
Sanford, J.A., Gallo, R.L., 2013. Functions of the skin microbiota
in health and disease. Seminars in Immunology 25(5), 370–
377. Available at: 10.1016/j.smim.2013.09.005
Sano, Y., Masuda, K., Tamagawa-Mineoka, R., Matsunaka, H.,
Probiotics in dermatological and cosmetic products – application and efficiency
25
Макед. фарм. билт., 68 (1) 9 – 26 (2022)
Murakami, Y., Yamashita, R., Morita, E., Katoh, N., 2013.
Thymic stromal lymphopoietin expression is increased in the
horny layer of patients with atopic dermatitis. Clinical and
Experimental Immunology 171(3), 330–337. Available at:
https://doi.org/10.1111/cei.12021
Sari, T.P., Mann, B., Kumar, R., Singh, R.R.B., Sharma, R.,
Bhardwaj, M., Athira, S., 2015. Preparation and
characterization of nanoemulsion encapsulating curcumin.
Food Hydrocolloids 43, 540–546. Available at:
https://doi.org/10.1016/j.foodhyd.2014.07.011
Satoh, T., Murata, M., Iwabuchi, N., Odamaki, T., Wakabayashi,
H., Yamauchi, K., Abe, F., Xiao, J.Z., 2015. Effect of
bifidobacterium breve B-3 on skin photoaging induced by
chronic UV irradiation in mice. Beneficial Microbes 6(4),
497–504. Available at: 10.3920/BM2014.0134
Savan, R., Sakai, M., 2006. Genomics of fish cytokines.
Comparative Biochemistry and Physiology Part D:
Genomics and Proteomics 1(1), 89–101. Available
at: 10.1016/j.cbd.2005.08.005
Schmelz, R.M., 2020. Regulation of probiotic and other live
biologic products. Skin Microbiome Handbook 343–375.
Available at: https://doi.org/10.1002/9781119593058.ch18
Schmidt, R.M., Pilmann Laursen, R., Bruun, S., Larnkjær, A.,
Mølgaard, C., Michaelsen, K.F., Høst, A., 2019. Probiotics
in late infancy reduce the incidence of eczema: A
randomized controlled trial. Pediatric Allergy and
Immunology 30(3), 335–340. Available at:
10.1111/pai.13018
Schwandner, R., Dziarski, R., Wesche, H., Rothe, M.,
Kirschning, C.J., 1999. Peptidoglycan- and lipoteichoic acid-
induced cell activation is mediated by toll-like receptor 2.
Journal of Biological Chemistry 274(25), 17406–17409.
Available at: 10.1074/jbc.274.25.17406
Sharma, D., Kober, M.M., Bowe, W.P., 2016. Anti-aging effects
of probiotics. J. Drugs Dermatol. 15(1), 9–12. Available at:
Anti-Aging Effects of Probiotics - PubMed (nih.gov)
Sikorska, H., Smoragiewicz, W., 2013. Role of probiotics in the
prevention and treatment of meticillin-resistant
Staphylococcus aureus infections. International Journal of
Antimicrobial Agents 42(6), 475–481. Available at:
10.1016/j.ijantimicag.2013.08.003
Simpson, M.R., Dotterud, C.K., Storrø, O., Johnsen, R., Øien, T.,
2015. Perinatal probiotic supplementation in the Prevention
of Allergy Related Disease: 6 year follow up of a randomised
controlled trial. BMC Dermatology 15(1). Available at:
10.1186/s12895-015-0030-1
Smith, W.P., 1996. Epidermal and dermal effects of topical lactic
acid. Journal of the American Academy of Dermatology
35(3), 388–391. Available at: 10.1016/s0190-
9622(96)90602-7
Song, H., Yoo, Y., Hwang, J., Na, Y.C., Kim, H.S., 2016.
Faecalibacterium prausnitzii subspecies–level dysbiosis in
the human gut microbiome underlying atopic dermatitis.
Journal of Allergy and Clinical Immunology 137(3), 852–
860. Available at: 10.1016/j.jaci.2015.08.021
Soumelis, V., Reche, P.A., Kanzler, H., Yuan, W., Edward, G.,
Homey, B., Gilliet, M., Ho, S., Antonenko, S., Lauerma, A.,
Smith, K., Gorman, D., Zurawski, S., Abrams, J., Menon, S.,
McClanahan, T., de Waal-Malefyt, Rd.R., Bazan, F.,
Kastelein, R.A., Liu, Y.J.,2002. Human epithelial cells
trigger dendritic cell mediated allergic inflammation by
producing TSLP. Nat. Immunol. 3(7), 673-680. Available
at: 10.1038/ni805
Strickler, A., Kolmer, J.A., Schamberg, J.F., 1916. Complement
fixation in acne vulgaris. J. Cutaneous Dis. 34, 166-178.
Sugimoto, S., Ishii, Y., Izawa, N., Masuoka, N., Kano, M., Sone,
T., Chiba, K., Miyazaki, K., Ishikawa, F., 2012.
Photoprotective effects of bifidobacterium breve
supplementation against skin damage induced by ultraviolet
irradiation in hairless mice. Photodermatology,
Photoimmunology & Photomedicine 28(6), 312–319.
Available at: 10.1111/phpp.12006
The Human Microbiome Project Consortium, 2012. Structure,
function and diversity of the healthy human microbiome.
Nature 486(7402), 207–214. Available at:
10.1038/nature11234
Udompataikul, M., Sripiroj, P., Palungwachira, P., 2009. An oral
nutraceutical containing antioxidants, minerals and
glycosaminoglycans improves skin roughness and fine
wrinkles. International Journal of Cosmetic Science 31(6),
427–435. Available at: https://doi.org/10.1111/j.1468-
2494.2009.00513.x
Valdéz, J.C., Peral, M.C., Rachid, M., Santana, M., Perdigón, G.,
2005. Interference of lactobacillus plantarum with
pseudomonas aeruginosa in vitro and in infected burns: The
potential use of probiotics in wound treatment. Clinical
Microbiology and Infection 11(6), 472–479. Available
at: 10.1111/j.1469-0691.2005.01142.x
Vierkötter, A., Krutmann, J., 2012. Environmental influences on
skin aging and ethnic-specific manifestations. Dermato-
Endocrinology 4(3), 227–231. Available at:
10.4161/derm.19858
Vinolo, M.A.R., Rodrigues, H.G., Nachbar, R.T., Curi, R., 2011.
Regulation of inflammation by short chain fatty acids.
Nutrients 3(10), 858–876. Available at: 10.3390/nu3100858
Volz, T., Skabytska, Y., Guenova, E., Chen, K.-M., Frick, J.-S.,
Kirschning, C.J., Kaesler, S., Röcken, M., Biedermann, T.,
2014. Nonpathogenic bacteria alleviating atopic dermatitis
inflammation induce il-10-producing dendritic cells and
regulatory tr1 cells. Journal of Investigative Dermatology
134(1), 96–104. Available at: 10.1016/j.jaci.2014.02.017
Wang, L., Clavaud, C., Bar-Hen, A., Cui, M., Gao, J., Liu, Y.,
Liu, C., Shibagaki, N., Guéniche, A., Jourdain, R., Lan, K.,
Zhang, C., Altmeyer, R., Breton, L., 2015. Characterization
of the major bacterial-fungal populations colonizing
dandruff scalps in Shanghai, China, shows Microbial
Disequilibrium. Experimental Dermatology 24(5), 398–400.
Available at: 10.1111/exd.12684
Wang, Y., Kao, M.S., Yu, J., Huang, S., Marito, S., Gallo, R.,
&Huang, C.M., 2016. A precision microbiome approach
using sucrose for selective augmentation of Staphylococcus
epidermidis fermentation against Propionibacterium acnes.
International Journal of Molecular Sciences 7(11), 1870.
Available at: https://doi.org/10.3390/ijms17111870
Weston, S., Halbert, A., Richmond, P., Prescott, S.L., 2005.
Effects of probiotics on atopic dermatitis: a randomised
controlled trial. Archives of Disease in Childhood 90(9),
892–897. Available at:
https://doi.org/10.1136/adc.2004.060673
Wickens, K., Barthow, C., Mitchell, E.A., Kang, J., van Zyl, N.,
Purdie, G., Stanley, T., Fitzharris, P., Murphy, R., Crane, J.,
2018. Effects of lactobacillus rhamnosus HN001 in early life
on the cumulative prevalence of allergic disease to 11 years.
Pediatric Allergy and Immunology 29(8), 808–814.
Available at: 10.1111/pai.12982
E. Hyseni, M. Glavas Dodov
26
Maced. pharm. bull., 68 (1) 9 – 26 (2022)
Williams, H., Stewart, A., von Mutius, E., Cookson, W.,
Anderson, H.R., 2008. Is eczema really on the increase
worldwide? Journal of Allergy and Clinical Immunology
121(4). Available at: 10.1016/j.jaci.2007.11.004
Yamamoto, A., Takenouchi, K., Ito, M., 1995. Impaired water
barrier function in acne vulgaris. Archives of
Dermatological Research 287(2), 214–218. Available
at: 10.1007/BF01262335
Yan, F., Polk, D.B., 2011. Probiotics and immune health. Current
opinion in gastroenterology 27(6), 496–501. Available at:
https://doi.org/10.1097/MOG.0b013e32834baa4d
Yang, A.J., Marito, S., Yang, J.J., Keshari, S., Chew, C.H., Chen,
C.C., & Huang, C.M., 2018. A Microtube array membrane
(MTAM) encapsulated live fermenting staphylococcus
epidermidis as a skin probiotic patch against cutibacterium
acnes. International Journal of Molecular Sciences 20(1),
14. Available at: https://doi.org/10.3390/ijms20010014
Yu, Y., Dunaway, S., Champer, J., Kim, J., Alikhan, A., 2019.
Changing our microbiome: Probiotics in dermatology.
British Journal of Dermatology 182(1), 39-46. Available
at: 10.1111/bjd.18088
Zhang, H., Liao, W., Chao, W., Chen, Q., Zenng H., Wu, C., Wu,
S., Ho, H.I., 2008. Risk factors for sebaceous gland diseases
and their relationship to gastrointestinal dysfunction in Han
adolescents. The Journal of Dermatology 35(9), 555–561.
Available at: 10.1111/j.1346-8138.2008.00523.x
Zheng, H., Liang, H., Wang, Y., Miao, M., Shi, T., Yang, F., Liu,
E., Yuan, W., Ji, Z.S., Li, D.K., 2016. Altered gut
microbiota composition associated with eczema in infants.
PLOS ONE 11(11). Available at:
10.1371/journal.pone.0166026
Zheng, J., Feng, Q., Zheng, S., Xiao, X., 2018. The effects of
probiotics supplementation on Metabolic Health in
pregnant women: An evidence based meta-analysis. PLOS
ONE 13(5). Available at:
https://doi.org/10.1371/journal.pone.0197771
Zouboulis, C.C., Jourdan, E., Picardo, M., 2014. Acne is an
inflammatory disease and alterations of sebum composition
initiate acne lesions. J. Eur. Acad. Dermatol. Venereol.
28(5), 527–32. Available at:
https://doi.org/10.1111/jdv.12298
Резиме
Пробиотици во дерматолошки и козметички
производи - употреба и ефикасност
Едита Хисени и Марија Главаш Додов*
Центар за фармацевтска нанотехнологија, Фармацевтски факултет,
Универзитет „Св. Кирил и Методиј“, Мајка Тереза 47, 1000 Скопје, С. Македонија
Клучни зборови: пробиотици, акни, атопичен дерматитис, фотостареење, псоријаза, имунолошки систем,
заздравување на рани, орална нега, интимна нега, микробиом на кожатa
Терминот „пробиотици“ првпат е употребен во 1907. година од страна на Ели Мечникоф. Оттогаш,
пробиотиците се дел од истражувањата не само за дигестивното здравје, туку и за воспалителните болести. Во
последно време, постои зголемен интерес за ефектите на пробиотиците во негата на кожата. Третирањето на
атопичен дерматитис, акни, псоријаза, фото стареење, рак на кожата, спроведувањето на интимна нега, орална
нега, како и заздравувањето на раните е сѐ потешко, поради зголемената резистентност кон антибиотиците и
другите несакани ефекти од конвенционалната терапија. Согласно претходно наведеното, резултатите од многу
истражувања покажуваат дека пробиотиците се ефикасни во лекувањето на различни кожни заболувања.
Овој преглед има за цел да ги оцени научните докази за локални и орални пробиотици и да ја оцени
ефикасноста на козметичките и дерматолошките производи кои содржат пробиотици. Многу студии покажале дека
промените на микробиомот на кожата и цревата имаат важна улога во здравјето на кожата. Иако ова е нова тема
во дерматологијата и козметологијата, постојат ветувачки резултати добиени од многу истражувачки студии во
однос на подобрување на исходот на пациентот при употребата на пробиотици во козметичките производи. И
покрај тоа што, оралните пробиотици е покажано дека го промовираат здравјето на цревата, што влијае на
имунолошкиот систем на домаќинот и помага во лекувањето на различни кожни болести, механизмот на дејство
на локалните пробиотици сè уште не е целосно разбран. Иако бројот на комерцијални пробиотски козметички
производи достапни на пазарот се зголемува и повеќето од студиите не покажале сериозни несакани ефекти од
пробиотиците, потребно е да се спроведат дополнителни студии, во поголеми и хетерогени групи.