Volume 21, number 2
 Views: (Visited 192 times, 1 visits today)  

Nerurkar R, Khan S. A, Mehrotra N. Impact of Gut Microbiome Dynamics on Epidermal Health. Biotech Res Asia 2024;21(2).
Manuscript received on : 05-02-2024
Manuscript accepted on : 15-05-2024
Published online on:  10-06-2024

Plagiarism Check: Yes

Reviewed by: Dr. Suresh C

Second Review by: Dr. Kulvinder Kochar Kaur

Final Approval by: Dr. Eugene A. Silow

How to Cite    |   Publication History    |   PlumX Article Matrix

Impact of Gut Microbiome Dynamics on Epidermal Health  

Rutuja Nerurkar, Sara A. Khan* and Nupur Mehrotra

Department of Biochemistry, SVKM’s Mithibai College of Arts, Chauhan Institute of Science and Amrutben Jivanlal College of Commerce and Economics (Autonomous) Affiliated to University of Mumbai India.

Corresponding Author E-mail: sara.khan@mithibai.ac.in

ABSTRACT: The trillions of bacteria that reside on and inside the body constitute the human microbiome, which is essential to maintaining host health. The gut microbiome is one of these microbial communities that has attracted a lot of attention due to its effects on the skin and other physiological systems. An extensive synopsis of the complex connection between gut microbiota and epidermal health is given in this review. There are ways in which the gut microbiota influences the health of the epidermis. It has been demonstrated that gut microbe-produced metabolites, such as vitamins, secondary metabolites, and short-chain fatty acids, affect skin barrier function and immune response modulation. Furthermore, the complex interaction of immunological signalling pathways, which govern communication between the gut and skin, regulates the gut-skin axis. Rosacea, psoriasis, eczema and other dermatological disorders are related to dysbiosis of the gut microbiome. Gaining knowledge of mechanisms underlying this relationship could be extremely beneficial for the creation of novel treatment approaches targeted at enhancing health and treating or preventing a range of dermatological disorders. In order to maintain optimal epidermal health, future research endeavours should concentrate on clarifying the precise microbial taxa and metabolites that are essential to this complex interplay in gut-skin axis.

KEYWORDS: Dysbiosis; Epidermal Health; Gut-Skin Axis; Gut Microbiome; Probiotics

Copy the following to cite this article:

Nerurkar R, Khan S. A, Mehrotra N. Impact of Gut Microbiome Dynamics on Epidermal Health. Biotech Res Asia 2024;21(2).

Copy the following to cite this URL:

Nerurkar R, Khan S. A, Mehrotra N. Impact of Gut Microbiome Dynamics on Epidermal Health. Biotech Res Asia 2024;21(2). Available from: https://bit.ly/4aUvQpd

Introduction

The 10-100 trillion symbiotic microbial cells that each individual carries, predominantly bacteria in the stomach, make up the human microbiota1. Numerous illnesses, such as inflammatory bowel disease, multiple sclerosis, diabetes (types 1 and 2), allergies, and asthma, have been linked to dysbiosis in the microbiome2, 3, 4. They enhance the immune system, provide pathogen defence, support metabolic activities, and through these vital tasks directly or indirectly affect most of our physiological systems5. The microbes in the gut which harbours around 1000 species of bacteria and around 5 million genes carry out many of the tasks necessary for human survival as well as physiology6. It is becoming widely recognized that the gut microbiome and human health are corelated. The healthy gut microbiota has specialized roles in the host’s food metabolism, the metabolism of xenobiotics and drugs, the preservation of the immunomodulation and the structural integrity of the gut as mucosal barrier and pathogen defence7. The human body’s largest organ is the skin, which behave like a protective barrier against wounds and microbial attack8. The host’s health and longevity are directly related to the billions of microbial communities that make up the gut, which is known as a virtual organ. The homeostasis of both gut and skin tissues are both influenced positively and negatively by the gut microbiota9, 10. Skin symptoms of Gastro-Intestinal (GI) illnesses are frequently present., The gut microbiota seems to have an important role in the pathogenesis of many inflammatory conditions11, 12. The microbiome functions to conserve homeostasis by interacting bidirectionally with tissues and organs, making it a crucial immune system regulator. As a result, imbalance in the skin and gut microbiota is linked to a diversified immune response, which promotes the evolution of skin illnesses like atopic dermatitis, psoriasis, acne vulgaris, dandruff, and melanomas13. The diversified gut microbiome may influence the cutaneous flora, according to mounting evidence, although the precise mechanism by which this happens is unclear14. Studies have connected gut microbiome imbalance to inflammatory skin illnesses, making it possible to modulate gut microbiota to treat skin conditions. Natural products are becoming more popular as synthetic medicine substitutes today. In this regard, oral probiotics may be a straightforward, risk-free, and affordable therapeutic management strategy for skin irritation15. Consequently, the gut microbiota is a possible target for controlling the host’s immunological reactions. Numerous skin conditions are characterized by persistent inflammation. The health of the skin may be impacted by imbalances in the gut microbiota that result in a condition of systemic inflammation. The generation of inflammatory chemicals, which can affect the skin’s inflammatory response, is controlled by the gut microbiota. It is possible that an unhealthy gut microbiota will make skin inflammation worse. The gut microbiome aids in the maturation of immune cells and the gut is home to a sizeable percentage of the immune system cells. When the gut microbiota is in equilibrium, the immune system is better trained to distinguish between benign and hazardous substances. The capacity of immune system to stop inappropriate inflammatory reactions that can cause skin diseases is influenced by this equilibrium10. The epidermal barrier of the skin guards against infections. It is interesting to note that the gut microbiota has an effect on the expression of proteins necessary for skin barrier maintenance. While imbalances may jeopardize the integrity of the epidermal barrier, a balanced gut microbiota can support a healthy epidermal barrier. Numerous researches have looked into the relationship between skin allostasis and homeostasis with gastrointestinal health in recent years, and corroborating evidence suggests that the skin and gut have a strong bidirectional association10, 16. This is demonstrated by the existence of translocation of bacterial DNA, which has recently been linked to the gut microbiome composition and suggests that recent active plaque psoriasis breakouts may be related to circulating bacterial DNA in blood from the intestinal lumen17. The alterations in gut microbial metabolism and immunological responses are brought on by the imbalance of the diversity of the gut microbiome and its composition, which also results in intestinal microecological disturbance. These changes are crucial for maintaining human health, since they are strongly related to physiological and pathological processes. The structural diversity of gut microbiota resists pathogen invasion and lessens the feeding conflict between symbiotic and possibly hazardous microorganisms. The adult innate and adaptive immune systems are stimulated by the gut microbiota, which is important in the metabolism of bile acid, vitamins, amino acids, and short-chain fatty acids.18. Modifications in the microbiome are associated with the onset of cancer and may have an effect the immune system and treatment response. Recent research has demonstrated that the gut microbiota, in particular, can modify the response to melanoma immunotherapy19. However, the significance of the skin microbiota in the microenvironment of epidermal tumours has not been thoroughly researched, and the interrelation between the gut microbiome and skin microbiome in the advancement of melanoma has not been looked into19. The gut-brain-skin axis, which was initially proposed by Arck et al.20, postulated that a number of gastrointestinal and skin disorders are caused by excessively shared signals and cellular protagonists, complex gut and skin innervation, and the predominance of neurogenic inflammation. The human microbiome project (HMP), which was started in 2007, aims to characterize the resident bacteria of the oral cavity, skin, nose, vagina, and stool by examining the diverse microenvironments of the human body. The HMP identified the “normal” human microbiome in the end, with the help of samples taken from distinct anatomical environments at various times21. Previous restrictions have been overcome by more recent methods including metagenomic sequencing22 and 16S ribosomal RNA (16S rRNA) amplification21. This review explores the intricate relationship between gut microbiota and epidermal health, elucidate their interconnections and their consequences for overall well-being.

Figure 1: Plot of the global prevalence of psoriasis, atopic dermatitis, seborrheic dermatitis, acne vulgaris, skin cancer, vitiligo, urticaria and rosacea23, 24, 25, 26, 27, 28, 29, 30

Click here to view Figure

Gut microbiome

The bacteria, viruses, and eukaryotic organisms living in and on our body make up the microbiome of humans. These bacteria have a great capacity to affect both the health and illness of our physiological system. They play a role in the metabolic processes, provide defence against pathogens, prepare the immune system, and through these fundamental processes affect the majority of our physiologic processes directly or indirectly5. It is becoming widely understood that the gut microbiota and human health are related. It is now well understood that a healthy gut microbiota has a significant impact in the host’s overall wellbeing. Bacteroidetes and Firmicutes make up the majority of the normal human gut microbiota7. The Human Microbiome Project and the Metagenome of the Human Intestinal tract (MetaHIT) investigations predict that the human microbiome may contain about ten million genes that are non-redundant, if seen from the perspective of all bacterial genes31. The phyla Actinobacteria and Verrucomicrobia are then listed after this. Even while this basic profile does not change, the distribution of the gut microbiota shows temporal and spatial variations at the genus level and above. As one travels from the oesophagus and stomach to the rectum, there will be a discernible change in the variety and quantity of bacteria with values ranging from 101 per gram of contents in the former to 1012 per gram of contents in the latter and distal gut7. The microbiota of the gastrointestinal system is divided into two groups: transverse and longitudinal, and its composition is a reflection of the physiological characteristics of a particular area32. Chemical, nutritional, and immunological gradients in and along the gut are believed to have an influence on the density and constitution of the microbiota. High quantities of acids, oxygen, and antimicrobials with a quick transit time are normally seen in the small intestine33. Numerous environmental factors, such as geographic location, medical procedures, smoking, depression, and type of housing, whether it is urban or rural have been linked to forming the microbiota34. The microbial communities that live in different parts of the human gut have an impact on a variety of health-related factors. In a healthy state, they support the host’s metabolism and immune system by providing nutrients and energy to the host through the large intestine’s fermentation of dietary components that are indigestible.

Figure 2: Microbiome composition in organs of the gastrointestinal tract

Click here to view Figure

Negative effects, however, may include being causes of infection and inflammation, being connected to gastrointestinal disorders, and perhaps making a difference in the development of diabetes mellitus and obesity35. In some circumstances, changes to the gut microbiome are noticeable years before a disease manifest itself, providing biomarkers for early disease risk detection and chances for preventive therapies. The growing body of information linking microbial causes of disease to specific pathogenic mechanisms, which in turn identifies new microbial targets for drug development, is fundamental to all of these observations. Following attempts to restore a disturbed gut microbiota through dietary intervention, microbial supplementation, or faecal microbiota transplant, positive effects have been seen for several disease indications36.

Gut-Skin axis

The processes through which intestinal microbiota affect skin homeostasis are yet unknown, but they seem to be connected to the modulatory impact of gut commensals on systemic immunity10. According to recent research, the intestinal microbes may have a more direct influence on the physiology, pathology, and immune response of the skin due to the gut microbiota and their metabolites’ skin metastasis10, 37, 38. The mechanisms via which the gut microbiota has an effect on the skin health is unknown38. Both the skin and the gut are active, intricate immunological and neuroendocrine organs that are frequently exposed to the outside world and support a diverse array of microbiomes39, 40. Gut-skin axis microbiota research have an influence to research and to acknowledge the link between the gut microbiome and alterations to the skin microbiome as well as the ensuing epidermal illnesses such as atopic dermatitis, psoriasis, acne vulgaris and other disorders. These studies go beyond the aetiological factors such as environmental, nutritional, or medicinal exposures41. According to the study conducted by Liedtke42, the management of dermatoses with probiotics and prebiotics as well as dietary and lifestyle changes point to the presence of a crucial gut-skin axis. It is essential to balance the gut microbiota in order to develop aesthetically healthy skin. In order to effectively treat inflammatory skin illnesses, treatments that raise or repair the intestine are necessary as adjuvant therapy. These procedures may also increase the efficacy of conventional dermal therapy. 43 The immune system and gut microbiome appear to interact as well as train the regulatory T lymphocytes, which can result in the inflammation in other parts of the body43, 44. Although the significance of the gut microbiome in autoimmune and inflammatory skin disorders is still being investigated, regulatory T cells appear to have a significant role in these conditions45. 46, 47.

Figure 3: Effect of gut microbiome on skin dyshomeostasis

Click here to view Figure

Psoriasis

A chronic inflammatory skin condition called psoriasis is characterized by whitish scale-covered, clearly delineated erythematous plaques48. Psoriasis is a chronic skin disorder that frequently has systemic symptoms. Although psoriasis can appear at any age, it usually first appears between the ages of 15 and 30. The clinical course is uncertain49. A first-degree relative who also has psoriasis affects about one-third of individuals. Research indicates that inheritance is complex49. Psoriasis risk and severity both rise with smoking. Alcohol misuse and obesity are also connected to psoriasis. These correlations may not be causal; psoriasis sufferers may be more prone to these patterns of alcohol misuse and obesity50. The most well-known and approachable human illness that is interceded by T lymphocytes and dendritic cells is psoriasis vulgaris. Interleukin-23 and interleukin-12 are released by inflammatory myeloid dendritic cells, which then stimulate T lymphocytes, Type 1 T helper (cells to produce large amounts of the psoriatic cytokines IL-17, interferons, tumour necrotic factors, and IL-22. These cytokines influence keratinocytes in a way that intensifies psoriatic inflammation51. Many studies revealed that intestinal permeability, the immune system, and metabolism are all significantly influenced by gut bacteria52, 53. The psoriasis comorbidities metabolic syndrome, depression, cardiovascular disease, and inflammatory bowel illnesses have all been linked to the dysbiosis of the gut microbiome, also known as gut dysbiosis54, 55, 56, 57. Psoriasis and changes in gut microbial composition are significantly correlated, but there is significant variation among various researches58. Psoriatic flare-ups and microbiome imbalances, defined by modified diversity and organisation as well as blooms of opportunistic pathogens, are closely related59. Zakostelska et al.60 demonstrated that imiquimod stimulates milder psoriasis-like skin inflammation in mice that were germ-free than in conventional mice by enhancing the Th17 response, indicating that gut dysbiosis acts as a probable pathogenic element for psoriasis. The intestinal microbiota promotes balance between Th17 effector cells and their equivalent regulatory T cells61. In a study conducted by Huang et al.62, 35 psoriasis sufferers and 27 healthy controls had their faeces sequenced by 16S rRNA and then the results were examined using informatics techniques. It was discovered that the psoriasis group’s microbiome was unlike that of the healthy group. The microbiome of people with severe psoriasis is different from those of people with more mild cases and also from healthy controls62. The microbiome has been shown to have a crucial role in the production of immunoglobulin A and the preservation of the equilibrium between effector and regulatory T lymphocytes in the gastrointestinal tract. Furthermore, chronic inflammatory skin diseases like psoriasis are linked to gut bacterial dysbiosis. Consequently, it may be said that the microbiota is a potent therapeutic target for curing this condition63. The composition of the human gut microbiome begins to form shortly after birth and stabilizes at the age of two, however significant changes can also be observed later in life due to food, lifestyle, comorbidities, antibiotic courses, and other factors.64. Gram-negative and Gram-positive anaerobic bacteria, such as those in the Bacteroides, Bifidobacterium, Eubacterium, Fusobacterium, and Ruminococcus genera, make up the great majority of the intestinal microbiome, outnumbering aerobic bacteria by a factor of over a hundred. Bacteroidetes along with Firmicutes are the two phyla that make up the majority of the intestinal microbiome65. In a study performed by Arumugam et al.66 three distinct gut microbiome clusters known as enterotypes were identified according to dominating genera. Bacteroides, Prevotella, and Ruminococcus are the three enterotypes. Enterotype three, which includes the Ruminococcus genus in addition to the Akkermansia genus, is the most prevalent. As studied by Polak et al.64 the most remarkable discoveries regarding changed gut physiology in psoriatic patients appear to be microbiota dysbiosis, depletion in short chain fatty acid synthesis, an increase in generated trimethylamine-N-oxide, and dysregulation of pathways controlling the balance of lymphocyte populations69.

Figure 4: Long-term consumption of a high-saturated-fat or high-fructose diet disrupts the balance of intestinal flora, which causes an increase in permeability and impaired gut barrier function.

Click here to view Figure

The bacterial composition of the stomach may be impacted by the medications used to treat psoriasis. A different form of psoriasis called guttate psoriasis, which is more frequent in children and adolescents than in adults, is traditionally brought on by a streptococcal infection69. Guttate psoriasis exhibits rapid involution and extended remission than other kinds of psoriasis, although little is known about its clinical history67. Based on the finding that patients’ gut microbiota profiles were dysbiotic when compared to healthy controls, abnormal cytokine levels are linked to abnormal cytokine profiles. As therapeutic targets or psoriasis biomarkers, it was anticipated that gut microbiota will be crucial in the clinical diagnosis and treatment of psoriasis70. The last 20 years have seen the discovery of causative immunological course of psoriasis that link up on adaptive immune pathways including interleukin (IL)-17 and IL-2371. Mice fed with Lactobacillus pentosus showed a decrease of psoriasis-related, pro-inflammatory, and Th17-associated cytokines like tumour necrosis factor (TNF)-, IL-17A, and IL-2372. The clinical value of the immune system’s and microbiota’s interaction is crucial. Secukinumab, ixekizumab, and guselkumab, monoclonal antibodies that target IL-17A and selective IL-23, respectively were all very successful in treating psoriasis, but their effects in treating inflammatory bowel disease (IBD) were inconsistent. IBD has become worse as a result of clinical studies for biologics that block IL-17A or its receptor73, 74. In a study done by Huang et al.71 showed that the patients with psoriasis who received therapies with IL-23 and IL-17 inhibitors experienced different changes in the composition of their gut bacteria. According to significant changes within the comparative plethora of bacterial taxa between responders and non-responders, IL-23 and IL-17 inhibitors may work in conjunction with the gut microbiome to lessen cutaneous inflammation. It looks quite complicated how skin psoriasis and gut microbiota are related. Just about all studies of the gut microbiome found consequential changes in psoriatic sufferers. The complete therapeutic potential of the alterations in the gut microbiota in patients affected by psoriasis needs to be further investigated in the future64.

Atopic dermatitis

Atopic dermatitis (AD), also known as atopic eczema, is a habitual relapsing seditious epidermal condition. Prevalence of atopic dermatitis has shot up two to three-fold within bucolic nations, impacting roughly 15 to 20 of children and one to three of grown-ups worldwide75. Although atopic dermatitis shows indications of skin barrier disfigurement and immunological divagation, the fundamental mechanism of action of eczema isn’t recognised properly, and the therapy is frequently veritably delicate. There is substantial data that AD cases have an imbalanced microbial anatomy and decreased microbial diversity in their skin and gut in contrast to controls, which contributes to complaint onset along with atopic march76. As compared to healthy individuals, diversity of the gut microbiome reduced and the relative plethora of the beneficial bacteria such as Bifidobacterium and Lactobacillus consequently decreased, in contrast, the magnitude of Escherichia coli, Clostridium difficile and Staphylococcus aureus was higher in people suffering from AD. Principally, the microbial colonization and adaptations were illustrated preceding any clinical manifestations in early life, stipulating gut microbial imbalance as one of the reasons for AD77. In addition, host-commensal interactions affect how children’ immune systems grow and, as a result, how often illnesses like atopic dermatitis manifest. The cutaneous microbiota influences the onset and progression of skin diseases later in life. As a result, the severity of the condition and a rise in the colonization of harmful bacteria such Staphylococcus aureus are correlated in AD patients with a decrease in microbiome diversity78. More severe clinical signs of AD are caused by skin microbiota, mainly S. aureus and Malassezia blooms in the lesions79. Diet, lifestyle choices, and psychological stress all have an impact on the dynamic and special ecosystem known as intestinal microecology. The alterations in gut bacterial metabolism and immunological reactions are brought on by the dysbiosis of gut microbial diversity and anatomy, which also results in intestinal microecological disturbance. These changes are crucial for maintaining human health since they are strongly related to physiological and pathological processes.  The innate and adaptive immune response mature as a result of the gut microbiome’s role in the metabolism of short-chain fatty acids, amino acids, vitamins, and bile acids80. The “gut-skin” axis has also been suggested as a new goal for the prevention and therapeutic modalities of AD10. In a study performed by Atarashi et al.81 the relationship between the gut microbiota and the T and B lymphocytes might result in systemic effects which are distant from the gut location. Additionally, intestinal dendritic cells’ migration of microbial antigens from the gut to the thymus caused gut microbial colonization to increase growth of microbiota and T lymphocytes in the thymus82. Myeloperoxidase and lipid hydroperoxide levels were considerably higher in a recent study of 56 children with atopic dermatitis. In contrast, a lower blood level of the total antioxidant potential value suggest that AD may be linked to an increase in oxidative stress reactions and a decrease in antioxidant defence75. As a result, changes in gut microbes are intimately related to immunological responses and are critical in the emergence of disorders involving abnormal immune activities80.

Seborrheic dermatitis

A chronic relapsing erythematous scaly skin condition called seborrheic dermatitis affects 1 to 3% of the population in United States. It has two incidence peaks: the first occurs through the first three months of life, and the second starts throughout puberty and peaks between the ages of 40 and 6084. Malassezia yeasts, hormones (androgens), sebum levels, and immunological response are all recognized to have important contribution in the development of seborrheic dermatitis, despite the fact that the actual cause is still unknown. Seborrheic dermatitis may be made worse by additional elements such as medicines, cold weather and stress85. Seborrheic dermatitis and dandruff are frequently linked by a fungal component. The most prevalent yeast species in the skin’s mycobiome, Malassezia spp. are lipophilic and dominating fungi that colonize the scalp of humans86. It is believed that Malassezia spp. increase oleic acid synthesis, disrupting the stratum corneum cells and causing an inflammatory reaction on the scalp13. When compared to a placebo treatment, a clinical trial on the consumption of probiotics (Lactobacillus paracasei strain) revealed significant reductions in the intensity and signs of moderate to severe dandruff. It is yet unclear, nevertheless, how the composition of the gut microbiota affects seborrheic dermatitis and dandruff87. It is debatable if gut dysbiosis and dandruff or seborrheic dermatitis are related. Patients with seborrheic dermatitis have intestinal mucosal abnormalities that have been identified88.

Acne vulgaris

Inflammation, increased sebum production, hyperkeratinisation, and primarily Propionibacterium acnes (formerly known as Cutibacterium acnes) proliferation are the main causes of acne, which can leave behind crippling psychological scars89. As a follicular illness, acne is characterized by the impingement and distention of the follicles by a keratinous plug, which results in the initial comedown. Keratinocyte hyperproliferation and improper differentiation result in keratinous plugs. The production of antimicrobial peptides and cytokines by activated keratinocytes can have both a direct antimicrobial effect and result in the recruitment and regulation of immune cells90. There is currently little consensus on the precise processes through which the gut microbiota affects the start as well as progression of acne91. Endotoxemia and increased intestinal permeability have been linked to acne in studies conducted many years ago 92. Due to increased serum reactivity to faecal coliforms in 66% of patients compared to 0% of controls in a complement fixation test, intestinal permeability may be higher in acne patients92. Gut flora and its metabolites can invade the bloodstream, collect in the epidermis, and affect skin homeostasis when intestinal barrier is compromised40. The link between gastrointestinal problems and acne may have its roots in the brain. The aggravation of acne brought on by stress lends credence to this hypothesis. Stress affects the normal gut microbiota, most significantly Lactobacillus and Bifidobacterium species, according to experimental animal and human investigations93. There is evidence that the Western diet and its accompanying intestinal flora cause inflammatory acne. For example, diets high in fat decrease the quantity of gut microbiota and raise the quantities of lipopolysaccharides which affect colonic epithelial integrity and barrier function, which reduces the thickness of the mucus layer and increases release of pro-inflammatory cytokines, all of which lead to systemic inflammation94. Additionally, Yan et al.95 discovered that acne sufferers’ levels of Lactobacillus, Bifidobacterium, Butyricicoccus, Coprobacillus, and Allobaculum were lower than those of controls offers fresh insight into the relationship between acne and changes in gut flora. Common probiotic species like Lactobacillus and Bifidobacterium regulate the intestinal microbiota by digesting unabsorbed oligosaccharides in the upper intestine96. Additionally, Bifidobacterium and Lactobacillus promote the development of regulatory dendritic cells and CD4+Foxp3+ T cells (regulatory T cells), which inhibits the response of T helper cells and B cells as well as the generation of cytokines97. In an earlier investigation, blood serum complement fixation testing was utilized to determine whether patients had increased reactivity to bacterial strains isolated from stool93.  Acne sufferers were more prone to have this enhanced reactivity. Comparatively to none of the control patients with no active skin disease, almost 66% of the 57 acne patients displayed positive reaction to stool-isolated coliforms93. It is important to conduct more research to understand the enteral microbiome of acne sufferers and the alterations in gut microbiota that occur after using oral antibiotics and isotretinoin96.

Skin Cancer

The most prevalent group of malignant neoplasms in the white population is skin cancer. Melanoma and non-melanoma skin cancer incidence rates are rising globally99. In general, Caucasians have a lifetime risk of acquiring melanoma of 2.4%, Blacks have a risk of 0.1%, and Hispanics have a risk of 0.5% 100. Age raises the likelihood of developing melanoma. At the time of diagnosis, the usual age is around 60. Males are around 1.5 times more likely than females to develop melanoma. It has been demonstrated that the incidence rate does not differ considerably until the age of 40, but after the age of 75, the incidence is nearly three times higher in men than in women101, 102. The methods by which the gut microbiome may raise or lower the risk of particular malignancies have been relatively thoroughly investigated, and they may also apply to the skin microbiome. Furthermore, through encouraging systemic inflammation, the gut flora may directly affect the risk of cancer in skin and other organs103. Alterations in the gut flora have been linked to carcinogenesis, immune evasion, and chronic inflammation, with particular bacteria being responsible for the emergence of particular tumours. Recently, it has come to light that the gut microbiota may be a novel element in the progression and management involved in malignant melanoma104. Indicators that the gut microbiome may be associated with the aetiology of melanoma were discovered by Vitali et al.105. In another study conducted by Luo106 it was discovered that Lactobacillus reuteri FLRE5K1 could increase the formation of anti-oncogenic cytokines in mice, stop the migration of the melanoma cell line B16-F10, and delay start of skin cancer thus prolonging survival. According to research by Li et al.107 introduction of 11 bacterial strains that are abundant in ubiquitin ligase RNF5 negative mice results in the establishment of anti-tumour immunity and limits the formation of melanoma in germ-free mice. Meanwhile, Pereira et al.108 demonstrated that the microbiota and IL-6 of obese mice promote the progression of melanoma, and faecal transfer experiments from leptin-deficient (obesity) mice revealed tumour evolution in lean mice. These two experiments show an alternative melanoma treatment approach to the conventional methods now in use104.

Vitiligo

An autoimmune condition called vitiligo is characterized by white patches of skin caused by the loss of melanocytes109. 0.5 to 1% of the global population is affected by this autoimmune skin disorder110. A loss of melanocytes that occurs over time in the skin and occasionally in the hair follicles is what defines vitiligo. The loss of melanocytic cells is now well established to be caused by CD8+ T lymphocytes drawn to the epidermis. In patients with vitiligo, Genome-Wide Association Studies found more than 50 loci are susceptible for the condition related to melanogenesis and the immune system111. Numerous inflammatory and autoimmune illnesses have been linked to gut and, to a lesser extent, skin dysbiosis. Surprisingly, there are still few microbiome investigations of vitiligo109. It is recognized that the primary factor in the degeneration of melanocytes is autoreactive CD8+ T lymphocytes. Additionally, a number of parameters, such as the heightened inflammatory environment brought on by excessive proinflammatory substances, affect how CD8 is activated. One such cytokine is IL-1. It has been shown that CD4+ T cells, such as Th1 and Th17 cells, aid CD8+ T cells’ abnormal response in vitiligo112, 113. There may be a relation between vitiligo and intestinal microbiome, even though the relationship of the intestinal microbiome and the autoimmune state in vitiligo has never been studied. According to Hadi et al.114, vitiligo patients frequently also have inflammatory bowel disease (IBD), another autoimmune condition linked to an abnormal gut microbiome115, 116. In an experiment conducted by Dellacecca et al.117 link between gut dysbiosis and ampicillin-induced depigmentation has been suggested by the correlation between ampicillin treatment and faster depigmentation, decreased bacteria in faecal pellets, and altered T cell distribution in tissues and blood. According to a recent study by Ganju et al.118 the skin lesions associated with vitiligo had a specific skin microbiome distribution. However, it is believed that the skin microbiome is very changeable and influenced by a variety of factors, including skin locations and various microenvironments. In contrast, the anatomy of the gut microbiome has a tendency to remain consistent since early childhood, despite the possibility of rapid changes in illness state, particularly for autoimmune disorders111. Probiotics-based therapy and preservation of the skin’s microbiota may result in novel immune therapies for vitiligo119.

Urticaria

A skin condition known as urticaria is distinguished by wheals (hives), angioedema, or both. The most common type of urticaria, the chronic spontaneous urticaria (CSU) is indicated by repetitive itchy wheals and/or angioedema that last for greater than six weeks without any known eliciting factors. It is defined as when a person has transient wheals that last longer than 6 weeks and occur almost every day121. Around 1% of the general population worldwide experiences CU at some or the other point in their lives, and in recent years, there has been a surge in prevalence 122. The activation and degranulation of epidermal mast cells, thereafter followed by the release of histamine along with other mediators that cause sensory nerve activation, vasodilatation, plasma extravasation, and cellular recruitment, resulting in urticaria, a frequent and varied inflammatory skin condition123, 124, 125. It has been proposed that the pathophysiology of CSU may be influenced by pro-inflammatory responses brought on by changes in the gut microbiome and mediated by an imbalance of the cytokines Th1/Th2/Th17126. Although the gut microbiota of CSU patients has been examined in the past, only a small number of research have so far found a substantial distinction between the makeup of the gut flora in CSU participants and healthy controls127, 128, 129. In an experiment performed by Wang et al.130, profiling of intestinal microbiome of CSU patients was performed using 16S rRNA gene sequencing. And metabolomics analysis was utilised to analyse metabolites of the patients. The major findings showed a significant increase in Lactobacillus, Turicibacter, and Lachnobacterium, whereas Phascolarctobacterium was decreased. The perturbed gut flora may further contribute to the progression of CSU via G protein coupled receptors. Thus, insinuating that CSU is potentially associated with the gut microbiome being disturbed. The findings of another study show that the Enterobacteriaceae family was found in the faecal samples of all patients and healthy controls, and that the quantities of A. muciniphila, C. leptum, and F. prausnitzii in healthy controls’ stool samples were considerably higher than in patients with CSU131. There were some changes in the gut microbiota’s phylum, order, family, genus, and species abundance between urticaria patients and healthy people. Patients with urticaria had a predominance of Firmicutes, Bacteroidetes, Proteobacteria, Verrucomicrobia, and Actinobacteria in their gut microbiomes132, 133, 134. Genus Coprococcus 3 and Genus Defluviitaleaceae UCG011135 were discovered to have an increased risk against urticaria. A crucial genus of gut bacteria, Coprococcus is a member of the Phylum Firmicutes. According to earlier studies, Coprococcus was important for immunological responses128 and was connected to the severity of atopic disease136. Additionally, Yun-Zhou et al.135 discovered a connection between Coprococcus and urticaria and it was discovered that Defluviitaleaceae UCG011 was positively related with urticaria for the first time. Defluviitaleaceae and butyric acid levels have been discovered to have a positive association with Defluviitaleaceae 138 and a negative association with butyric acid levels139 but the precise effect is yet unknown.

Rosacea

A widespread, chronic skin condition affecting the face known as rosacea is categorized by erythema, papules, pustules, telangiectasias, flushing, phymatous differences, and ocular clinical manifestations. Avoiding triggers, taking care of your skin, and getting treatments that target specific traits are all part of management140. Rosacea is a long-lasting inflammatory condition with a complex pathophysiology that includes genetic and environmental factors, as well as dysfunctionality of innate as well as adaptive immune system, neurovascular reactions, microbiome colonization or infection, leading to recurring inflammation. Inflammatory bowel illness, celiac disease, irritable bowel syndrome, gastroesophageal reflux disease, Helicobacter pylori (HP) infection, as well as small intestine bacterial overgrowth (SIBO) are just a few of the gastrointestinal conditions that have been linked to rosacea141. In comparison to healthy controls, rosacea patients had lower levels of the following genus-level bacteria: Peptococcaceae family, Methanobrevibacter, Slackia, Coprobacillus, Citrobacter, Desulfovibrio, Lactobacillus, Hemophilus, Roseburia, and Clostridium142, 143. One can speculate that the gut microbiota may be involved in the etiology of the disease given that rosacea has been connected to small intestine bacterial overgrowth and inflammatory bowel disease144. It has long been assumed that Helicobacter pylori has a role in the pathophysiology of the disease145. Pernicious chemicals may enter the bloodstream and harm peripheral sites if the gut mucosa is compromised, whether as a result of autoimmune illness or changes in the microbiome146. Improvements in inflammatory bowel disease and the clinical manifestations of rosacea with oral metronidazole treatment support the theory that resident gut flora may act as the underlying stimulant to an inflated immune response147.  Several factors, such as the mode of birth, food, age, stress, and antibiotics, may affect the components of the gut microbiota and therefore, may cause rosacea148. Certain foods and beverages may behave as “triggers” for rosacea exacerbations. These may be split into heat-related, alcohol-related, capsaicin-related, and cinnamaldehyde-related149. Hot drinks in particular, such as hot coffee (33%) and hot tea (30%), served as a trigger. Another common trigger was alcohol, which included both wine (52%) and hard liquor (42%). Certain spices and peppers contain the compound capsaicin. Spices (identified as a trigger by 75% of respondents), spicy sauce (54%), cayenne pepper (47%), and red pepper (37%), were also often mentioned. Finally, foods that don’t seem to have anything to do with each other, like tomatoes, citrus, cinnamon, and chocolate, contain cinnamaldehyde150. Another matter of concern is to the connection between rosacea and inflammatory bowel disease (IBD). In contrast to matched controls, a Taiwanese countrywide cohort experiment of more than 89,000 rosacea patients discovered an independent link with IBD incidence151. But the mechanisms underlying the contribution of gut bacteria to the pathophysiology of rosacea remain unclear. Dietary alteration may have a role in the management of rosacea149.

Probiotics as a therapeutic agent

Probiotics are well recognized to be beneficial in certain illnesses, and numerous clinical surveys have shown that they can have peculiar influence on the epidermis, directly or indirectly, that can be significant from a variety of angles. Probiotic bacteriotherapy has the potential to be very effective in treating and preventing skin disorders such as atopic dermatitis, acne, and allergic inflammation; it can also be used as a cosmetic product and to treat skin hypersensitivity, UV-induced skin damage, and injury protection152. Studies show that probiotics can enhance the functioning of the epithelial barrier of the intestine and have antibacterial, competitive exclusion, and immunomodulatory properties. Probiotics’ benefits for human health are becoming more well documented in science, and the information that is now available supports the benefits of probiotics against a number of illnesses153. In order to determine the probiotic strains’ ability to stick to human skin and their antibacterial effectiveness against particular infections, Lopes et al.154 conducted a study which showed the ability of probiotic strains’ cell-free culture supernatants to stop or eliminate specific pathogens’ ability to build established biofilms was also assessed by these investigators, who also looked for possible quorum-sensing antagonists. The scientists discovered that certain probiotic strains’ CFCS had antimicrobial action against Propionibacterium acnes, Escherichia coli, and Pseudomonas aeruginosa, but none of them could stop Staphylococcus aureus from growing154. Five paediatric and 10 adult participants participated in a trial examining Roseomonas mucosa as a potential therapy for AD. With no negative side effects or consequences, treatment with Roseomonas mucosa was linked to considerable reductions in the severity of the disease, the need for topical steroids, and the S. aureus load. The study’s preliminary findings encourage further assessment of Roseomonas mucosa therapy through a placebo-controlled experiment155. By decreasing mast cells and raising T-reg cytokines, topical use of Vitreoscilla filiformis and Lactococcus has been proven to be effective in the treatment of seborrheic dermatitis and atopic eczema156. It may also be hypothesized that topical probiotics, like gut microbiome, may similarly lower the incidence of skin cancer by enhancing immune surveillance and decreasing chronic inflammation. Additionally, a topical probiotic may modify the tumour microenvironment by changing immune responses, which may have therapeutic effects, if it is injected into or administered to a cutaneous tumour157. Probiotics have been shown in numerous studies to be effective in treating long-term inflammatory diseases like rosacea while having few negative effects150, 151. Most studies have found that the initial beneficial effect of probiotics cannot be sustained upon discontinuation and that probiotics do not necessarily allow for recolonization of beneficial gut microorganisms159. The severity of dermatologic illnesses increases and treatment effectiveness is decreased as patients become resistant to antimicrobial medicines160.

Conclusion

In conclusion, the complex relationship between the gut microbiota and skin health are an emerging field of study that has the potential to completely transform how we think about skincare and general well-being. The data highlights the significant function of the gut microbiota in regulating the immune system, nutritional absorption, and inflammatory responses—all of which have an immediate effect on the epidermal health. Imbalance in the stomach can cause a wide range of dermatological problems, from acne to eczema and beyond, and it is now abundantly obvious that they play a crucial part in preserving good skin. The creation of bioactive substances, immunological regulation, and inflammatory modulation are just a few of the many ways that the gut microbiome affects skin health. This discovery not only opens the door to novel therapeutic methods but also offers crucial insights into the underlying pathophysiology of skin problems. Although no therapeutic modalities have yet been discovered to provide persistent relief to these dermatological conditions, probiotics, prebiotics, and dietary modifications are becoming more and more effective weapons in the fight against these illnesses, giving patients additional options for controlling and even averting skin-related problems. But it’s crucial to recognize that there is still a lot to learn and that the field of gut-skin health is still in its infancy. Subsequent investigations will surely deepen our comprehension of these complex relationships, possibly opening the door to innovative therapies and interventions for people with a variety of skin conditions. The more we learn about this interesting interplay between the gut and the skin, the more obvious it is that maintaining a diverse and well-balanced gut microbiota is essential to good health in general and is also a key component of a holistic skincare regimen. Once thought of as a remote environment, the stomach is now recognized as a vital component in the pursuit of healthy skin. Accepting this paradigm change gives people with skin problems hope for a better future by providing them with a more efficient and all-encompassing route to recovery and wellbeing.

Acknowledgment

This author is thankful to the Department of Biochemistry and SVKM’s Mithibai College of Arts, Chauhan Institute of Science & Amrutben Jivanlal College of Commerce and Economics (Autonomous) for their constant support and providing all the resources.

Conflict of Interest

There is no conflict of interest.

Funding sources

Financial support to the Department of Biochemistry from its management Shri Vile Parle Kelavani Mandal is gratefully acknowledged.

References

  1. Ursell LK, Metcalf JL, Parfrey LW, Knight R. Defining the human microbiome. Nutr Rev. 2012;70 Suppl 1(Suppl 1):S38-S44. doi:10.1111/j.1753-4887.2012.00493.x
  2. Petersen, C., Round, J.L. Defining dysbiosis and its influence on host immunity and disease. Cell Microbiol., 2014; 16:1024–33.
  3. Trompette A, Gollwitzer ES, Yadava K, Sichelstiel AK, Sprenger N, Ngom-Bru C, Blanchard C, Junt T, Nicod LP, Harris NL, Marsland BJ. Gut microbiota metabolism of dietary fiber influences allergic airway disease and hematopoiesis. Nat Med. 2014 Feb;20(2):159-66. doi: 10.1038/nm.3444. Epub 2014 Jan 5. PMID: 24390308.
  4. Backhed, F., Fraser, CM, Ringel Y, Sanders, M.E., Sartor, R.B., Sherman, P.M., et al., Defining a healthy human gut microbiome: current concepts, future directions, and clinical applications. Cell Host Microbe., 2012; 12:611–22.
  5. Shreiner, A.B., Kao, J.Y., and Young, V.B. Curr Opin Gastroenterol. 2015; Jan, 31(1): 69–75.
  6. D’Argenio, V., Salvatore, F., The role of the gut microbe in the healthy adult status, Clinica Chimica Acta, 2015; Vol 451, Part A, Pages 97-102, ISSN 0009-8981.
  7. Jandhyala, S.M., Taludka, R., Subramanyam, Vuyyuru, H., Sasikala, M., Reddy, D., World J Gastroenterol. 2015; 8787–8803.
  8. Hsu, D.K., Fung, M.A., Chen, H.L. Role of skin and gut microbiota in the pathogenesis of psoriasis, an inflammatory skin disease. Med Microecol. 2020; 4:100016.
  9. Ellis, S.R., Nguyen, M., Vaughn, A.R., Notay, M., Burney, W.A., Sandhu, S., Sivama, R.K. The skin and gut microbiome and its role in common dermatologic conditions. Microorganisms. 2019; ,7(11):550.
  10. O’Neill, C.A., Monteleone, G., McLaughlin, J.T., Paus, R. The gut-skin axis in health and disease: A paradigm with therapeutic implications. Bioessays, 2016; 38, 1167–1176.
  11. Shah, K. R., Boland, C. R., Patel, M., Thrash, B., and Menter, A. Cutaneous manifestations of gastrointestinal disease: part I. JAAD 2013; 68, 189.e1-189.e21.
  12. Thrash, B., Patel, M., Shah, K. R., Boland, C. R., and Menter, A. Cutaneous manifestations of gastrointestinal disease: part II. JAAD 2013; 68, 211.e1-211.e33.
  13. De Pessemier, B., Grine, L., Debaere, M., Maes, A., Paetzold, B., Callewaert, C. Gut–Skin Axis: Current Knowledge of the Interrelationship between Microbial Dysbiosis and Skin Conditions. Microorganisms., 2021; 9(2):353.
  14. Sinha, S., Lin, G., Ferenczi, K., The skin microbiome and the gut-skin axis, Clinics in Dermatology, 2021; Volume 39, Issue 5, Pages 829-839, ISSN 0738-081X,
  15. Szántó, M., Dózsa, A., Antal, D., Szabó, K., Kemény, L., Bai, P. 2019;
  16. Levkovich, T., Poutahidis, T., Smillie, C., Varian, B.J., Ibrahim, Y.M., Lakritz, J.R., Alm, E.J., Erdman, S.E. Probiotic bacteria induce a ‘glow of health’. PLoS ONE, 2013; 8, e53867.
  17. Ramírez-Boscá, A., Navarro-López, V., Martínez-Andrés, A., Such, J., Francés, R., de la Parte, J.H., Asín-Llorca, M. Identification of bacterial DNA in the peripheral blood of patients with active psoriasis. JAMA Dermatol.2015; 151, 670–671.
  18. O’Hara, A.M., Shanahan, F. The Gut Flora as a Forgotten Organ. EMBO Rep 2006; 7(7):688–93.
  19. Mekadim, C., Skalnikova, H.K., Cizkova, J. et al. Dysbiosis of skin microbiome and gut microbiome in melanoma progression. BMC Microbiol 2022; 22, 63.
  20. Arck, P., Handjiski, B., Hagen, E., et al., Is there a ‘gut-brain-skin axis’? Exp Dermatol, 2010; 19:401-5.
  21. Huttenhower, C., Gevers, D., Knight, R., Abubucker, S., Badger, J.H., Chinwalla, A.T., et al. Structure, function and diversity of the healthy human microbiome. Nature. 2012; 486:207–214.
  22. Qin, J., Li, R., Raes, J., Arumugam, M., Burgdorf, K.S., Manichanh, C., et al. A human gut microbial gene catalogue established by metagenomic sequencing. Nature. 2010; 464:59–65.
  23. Damiani G, Bragazzi NL, Karimkhani Aksut C, et al. The Global, Regional, and National Burden of Psoriasis: Results and Insights From the Global Burden of Disease 2019 Study. Front Med (Lausanne). 2021;8:743180. Published 2021 Dec 16. doi:10.3389/fmed.2021.743180
  24. Tian J, Zhang D, Yang Y, Huang Y, Wang L, Yao X, Lu Q. Global epidemiology of atopic dermatitis: a comprehensive systematic analysis and modelling study. Br J Dermatol. 2023 Dec 20;190(1):55-61. doi: 10.1093/bjd/ljad339. PMID: 37705227.
  25. Xue Y, Bao W, Zhou J, et al. Global Burden, Incidence and Disability-Adjusted Life-Years for Dermatitis: A Systematic Analysis Combined With Socioeconomic Development Status, 1990-2019. Front Cell Infect Microbiol. 2022;12:861053. Published 2022 Apr 12. doi:10.3389/fcimb.2022.861053
  26. Tan, J.K.L. & Bhate, Ketaki. (2015). A global perspective on the epidemiology of acne. British Journal of Dermatology. 172. 10.1111/bjd.13462.
  27. Urban K, Mehrmal S, Uppal P, Giesey RL, Delost GR. The global burden of skin cancer: A longitudinal analysis from the Global Burden of Disease Study, 1990-2017. JAAD Int. 2021;2:98-108. Published 2021 Jan 4. doi:10.1016/j.jdin.2020.10.013
  28. Bibeau K, Pandya AG, Ezzedine K, et al. Vitiligo prevalence and quality of life among adults in Europe, Japan and the USA. J Eur Acad Dermatol Venereol. 2022;36(10):1831-1844. doi:10.1111/jdv.18257
  29. Peck G, Hashim MJ, Shaughnessy C, Muddasani S, Elsayed NA, Fleischer AB Jr. Global Epidemiology of Urticaria: Increasing Burden among Children, Females and Low-income Regions. Acta Derm Venereol. 2021 Apr 22;101(4):adv00433. doi: 10.2340/00015555-3796. PMID: 33847367; PMCID: PMC9364256.
  30. Gether L, Overgaard LK, Egeberg A, Thyssen JP. Incidence and prevalence of rosacea: a systematic review and meta-analysis. Br J Dermatol. 2018 Aug;179(2):282-289. doi: 10.1111/bjd.16481. Epub 2018 May 31. PMID: 29478264.
  31. Le Chatelier, E., Nielsen, T., Qin, J., Prifti, E., Hildebrand, F., Falony, G., Almeida, M., Arumugam, M., Batto, J.M., Kennedy, S., et al. Richness of human gut microbiome correlates with metabolic markers. Nature., 2013; 500:541–546.
  32. Macpherson, A.J. and McCoy, K.D. Stratification and compartmentalisation of immunoglobulin responses to commensal intestinal microbes. Semin. Immunol. 2013; 25, 358–363
  33. Donaldson, G.P., Lee, S.M. and Mazmanian, S.K. Gut biogeography of the bacterial microbiota. Nat. Rev. Microbiol. 2015; 14, 20–32
  34. Thursby, E., Juge, N., Introduction to the human gut microbiota. Biochem J 1 474 2017; (11): 1823–1836.
  35. Flint, H., Scott, K., Louis, P. et al. The role of the gut microbiota in nutrition and health. Nat Rev Gastroenterol Hepatol 9, 2012; 577–589.
  36. Durack, J., Lynch, S.V., J Exp Med 216 2019; (1): 20–40.
  37. Samuelson, D. R., Welsh, D. A., and Shellito, J. E. Regulation of lung immunity and host defense by the intestinal microbiota. Front. Microbiol. 2015; 6:1085.
  38. Mahmud, M.R., Akter, S., Tamanna, S.K., Mazumder, L., Esti, I.Z., Banerjee, S., Hasan, M.R., Acharjee, M., Hossain, M.S. & Pirttilä, A.M. 2022; Article: 2096995
  39. Coates, M., Lee, M.J., Norton, D., MacLeod, A.S., Norton, D., MacLeod, A.S., MacLeod, A.S. The skin and intestinal microbiota and their specific innate immune systems. Front Immunol. 2019; 10:10.
  40. Salem, I., Ramser, A., Isham, N., Ghannoum, M.A. The gut microbiome as a major regulator of the gut-skin axis. Front Microbiol. 2018; 9:1459.
  41. Alghamdi, H.A., Behieldin, A., Edris, S. Gut microbiome skin axis in the development of atopic dermatitis. JPMA. The Journal of the Pakistan Medical Association. 2021; 71(4):1221-1227 PMID: 34125775.
  42. Liedtke, F. S. Gut-Skin axis and healthy skin: a systematic review. International Journal of Nutrology, 2023; 16(2).
  43. Russler-Germain, E.V., Rengarajan, S., Hsieh, C.S. Antigen-specific regulatory T-cell responses to intestinal microbiota. Mucosal Immunol. 2017; 10(6):1375-1386
  44. Sun, M., He, C., Cong, Y., Liu, Z. Regulatory immune cells in regulation of intestinal inflammatory response to microbiota. Mucosal Immunol. 2015; 8(5):969-978.
  45. Haeberle, S., Wei, X., Bieber, K., et al. (2018) Regulatory T-cell deficiency leads to pathogenic bullous pemphigoid antigen 230 autoantibody and autoimmune bullous disease. J Allergy Clin Immunol. 2018
  46. Melnik, B.C., John, S.M., Chen, W., Plewig, G. T helper 17 cell/regulatory T-cell imbalance in hidradenitis suppurativa/acne inversa: the link to hair follicle dissection, obesity, smoking and autoimmune comorbidities. Br J Dermatol. 2018
  47. Owczarczyk-Saczonek, A., Czerwinska, J., Placek, W. The role of regulatory T cells and anti-inflammatory cytokines in psoriasis. Acta Dermatovenerol Alp Pannonica Adriat. 2018; 27(1):17-23.
  48. Boehncke, W.H., Schon, M.P. Psoriasis. Lancet386, 2015; 983–994.
  49. Kamiya, K., Kishimoto, M., Sugai, J., Komine, M., Ohtsuki, M., Risk Factors for the Development of Psoriasis. International Journal of Molecular Sciences., 2019; 20(18):4347.
  50. Weigle, N., McBane, S., American Family Physician, Volume 87, Number 9 2013
  51. Lowes, M.A., Suárez-Fariñas, M., and Krueger, J.G. Annual Review of Immunology 32:1, 2014; 227-255
  52. Spencer, S.P., Fragiadakis, G.K., Sonnenburg, J.L. Pursuing Human-Relevant Gut Microbiota-Immune Interactions. Immunity, 51, 2019; 225–239.
  53. Chelakkot, C., Ghim, J., Ryu, S.H. Mechanisms regulating intestinal barrier integrity and its pathological implications. Exp. Mol. Med., 50, 2018; 103
  54. Stevens, B.R., Roesch, L., Thiago, P., Russell, J.T., Pepine, C.J., Holbert, R.C., Raizada, M.K., Triplett, E.W. Depression phenotype identified by using single nucleotide exact amplicon sequence variants of the human gut microbiome. Mol. Psychiatry. 2020
  55. Vallianou, N., Stratigou, T., Christodoulatos, G.S., Dalamaga, M. Understanding the Role of the Gut Microbiome and Microbial Metabolites in Obesity and Obesity-Associated Metabolic Disorders: Current Evidence and Perspectives. Curr. Obes. Rep. 8, 2019; 317–332.
  56. Kitai, T., Tang, W.H.W. Gut microbiota in cardiovascular disease and heart failure. Clin. Sci., 132, 2018; 85–91.
  57. Nishida, A., Inoue, R., Inatomi, O., Bamba, S., Naito, Y., Andoh, A. Gut microbiota in the pathogenesis of inflammatory bowel disease.  J. Gastroenterol.11, 2018; 1–10.
  58. Sikora, M., Stec, A., Chrabaszcz, M., Knot, A., Waskiel-Burnat, A., Rakowska, A., Olszewska, M., Rudnicka, L. Gut Microbiome in Psoriasis: An Updated Review. Pathogens., 2020; 9(6):463.
  59. Chen, L., Li, J., Zhu, W., Kuang, Y., Liu, T., Zhang, W., Chem, X., Peng, C. Front. Microbiol., Sec. Microbial Immunology Volume 11. 2020
  60. Zakostelska, Z., Malkova, J., Klimesova, K., Rossmann, P., Hornova, M., Novosadova, I., et al. Intestinal microbiota promotes psoriasis-like skin inflammation by enhancing Th17 response. 2016; PLoS One 11: e0159539.
  61. Van Beelen, A. J., Teunissen, M. B., Kapsenberg, M. L., and de Jong, E. C. Interleukin-17 in inflammatory skin disorders. Curr. Opin. Allergy Clin. Immunol. 7, 2007; 374–381.
  62. Huang, L., Gao, R., Yu, N. et al. Dysbiosis of gut microbiota was closely associated with psoriasis. Sci. China Life Sci. 62, 2019; 807–815.
  63. Alesa, D.I., Alshamrani, H.M., Alzahrani, Y.A., Alamssi, D.N., Alzahrani, N.S., Almohammadi, M.E. The role of gut microbiome in the pathogenesis of psoriasis and the therapeutic effects of probiotics. J Family Med Prim Care. 2019; 5,8(11):3496-3503.
  64. Polak, K., Bergler-Czop, B., Szczepanek, M., Wojciechowska, K., Frątczak, A., Kiss, N. Psoriasis and Gut Microbiome—Current State of Art. International Journal of Molecular Sciences. 2021; 22(9):4529.
  65. Malinowska, M., Tokarz-Deptuła, B., Deptuła, W. The human microbiome. Post. Mikrobiol., 2017; 56, 33–42.
  66. Arumugam, M., Raes, J., Pelletier, E., Le Paslier, D., Yamada, T., Mende, D., Fernandes, G., Tap, J., Bruls, T., Batto, J., et al. Enterotypes of the human gut microbiome. Nature., 2011; 473, 174–180.
  67. Hyun-Chang, K.O., Seung-Wook, J.W.A., Margaret, S., Moon-Bum, K., Kyung-Sool, Moon-Bum Kim, M.D., Department of Dermatology, School of Medicine, Pusan National University, 2010; 1-10 Ami-dong, Seo-gu, Busan 602-739, Korea
  68. Ji Y, Yin Y, Sun L, Zhang W. The Molecular and Mechanistic Insights Based on Gut–Liver Axis: Nutritional Target for Non-Alcoholic Fatty Liver Disease (NAFLD) Improvement. International Journal of Molecular Sciences. 2020; 21(9):3066. https://doi.org/10.3390/ijms21093066
  69. Saleh, D., Tanner, L. Suttate Psoriasis. In: StatPearls. StatPearls Publishing, Treasure Island (FL). 2022; PMID: 29494104.
  70. Zhang, X., Shi, L., Sun, T., Guo K. & Geng, S., BMCMicrobiology volume 21, Article number 78. Urticaria patientis j. Microbiol. Biotechnol. 2021; 31, 747-755
  71. Huang Y.H., Chang, L.C., Chang, Y.C., Chung, W.H., Yang, S.F., Su, S.C. Compositional Alteration of Gut Microbiota in Psoriasis Treated with IL-23 and IL-17 Inhibitors. International Journal of Molecular Sciences., 2023; 24(5):4568.
  72. Nermes, M., Kantele, J.M., Atosuo, T.J., Salminen, S., Isolauri, E. Interaction of orally administered Lactobacillus rhamnosus GG with skin and gut microbiota and humoral immunity in infants with atopic dermatitis. Clin. Exp. Allergy, 2011; 41, 370–377.
  73. Targan, S.R., Feagan, B., Vermeire, S., Panaccione, R., Melmed, G.Y., Landers, C., Li, D., Russell, C., Newmark, R., Zhang, N., et al. A randomized, double-blind, placebo-controlled Phase 2 study of brodalumab in patients with moderate-to-severe Crohn’s disease. Am. J. Gastroenterol. 2016; 111, 1599–1607.
  74. Hueber, W., Sands, B.E., Lewitzky, S., Vandemeulebroecke, M., Reinisch, W., Higgins, P.D., Wehkamp, J., Feagan, B.G., Yao, M.D., Karczewski, M., et al. Secukinumab, a human anti-IL-17A monoclonal antibody, for moderate to severe Crohn’s disease: unexpected results of a randomised, double-blind placebo-controlled trial. Gut, 2012; 61, 1693–1700.
  75. Avena-Woods C. Overview of atopic dermatitis. The American Journal of Managed Care. 2017; 23(8 Suppl): S115-S123. PMID: 28978208.
  76. Kim, J.E., Kim, H.S. Microbiome of the Skin and Gut in Atopic Dermatitis (AD): Understanding the Pathophysiology and Finding Novel Management Strategies. Journal of Clinical Medicine., 2019; 8(4):444.
  77. Zachariassen, L.F., Krych, L., Engkilde, K., Nielsen, D.S., Kot, W., Hansen, C.H., et al. Sensitivity to Oxazolone Induced Dermatitis is Transferable with Gut Microbiota in Mice. Sci Rep. 2017; 7:44385.
  78. Paller, A.S., Kong, H.H., Seed, P., Naik, S., Scharschmidt, T.C., Gallo, R.L., Luger, T., Irvine, A.D., The microbiome in patients with atopic dermatitis, Journal of Allergy and Clinical Immunology, Volume 143, Issue 1, 2019; Pages 26-35, ISSN 0091-6749.
  79. Reginald, K., Westritschnig, K., Linhart, B., Focke-Tejkl, M., Jahn-Schmid, B., Eckl-Dorna, J., et al. Staphylococcus Aureus Fibronectin-Binding Protein Specifically Binds Ige from Patients with Atopic Dermatitis and Requires Antigen Presentation for Cellular Immune Responses. J Allergy Clin Immunol. 2011; 128(1):82–91. e8.
  80. Fang, Z., Li, L., Zhang, H., Zhao, J., Lu, W. and Chen, W. Gut Microbiota, Probiotics, and Their Interactions in Prevention and Treatment of Atopic Dermatitis: A Review. Front. Immunol. 2021; 12:720393.
  81. Atarashi, K., Tanoue, T., Oshima, K., Suda, W., Nagano, Y., Nishikawa, H., et al. Treg Induction by A Rationally Selected Mixture of Clostridia Strains from the Human Microbiota. Nature 2013; 500(7461):232–6.
  82. Zegarra-Ruiz, D.F., Kim, D.V., Norwood, K., Kim, M., Wu, W.H., Saldana-Morales FB, et al. Thymic Development of Gut-Microbiota-Specific T Cells. Nature 2021; 594(863):413–17.
  83. Simonetti, O., Bacchetti, T., Ferretti, G., Molinelli, E., Rizzetto, G., Bellachioma, L., et al. Oxidative Stress and Alterations of Paraoxonases in Atopic Dermatitis. Antioxidants (Basel) 2021; 10, 697.
  84. Sampaio, A.L.B, Mameri, A., Jeunon, T., Ramos-e-Silva, M., Nunes, A.P., Carneiro, S. Dermatite seborreica. An Bras Dermatol. 2011; 86(6):1061-74.
  85. Mokos, Z.B., Kralj, M., Basta-Juzbašić, A., Jukić, I.L., Acta Dermatovenerol Croat Seborrheic dermatitis, 2012; 20(2):98-104.
  86. Kamamoto, C., Nishikaku, A., Gompertz, O., Melo, A., Hassun, K., Bagatin, E. Cutaneous fungal microbiome: Malassezia yeasts in seborrheic dermatitis scalp in a randomized, comparative and therapeutic trial. Dermato-endocrinology, 2017; 9, e1361573.
  87. Reygagne, P., Bastien, P., Couavoux, M., Philippe, D., Renouf, M., Castiel-Higounenc, I., Gueniche, A. The positive benefit of Lactobacillus paracasei NCC2461 ST11 in healthy volunteers with moderate to severe dandruff. Benef. Microbes, 2017; 8, 671–680.
  88. Odintsova, I., Dyudyun, A. Features of the composition of microorganisms inhabiting the intestinal mucosa in patients with seborrheic dermatitis. Dermatovenerol. Cosmetol. Sexopathol., 2019; 31–34.
  89. Siddiqui, R., Makhlouf, Z. & Khan, N.A. The increasing importance of the gut microbiome in acne vulgaris. Folia Microbiol 2022; 67, 825–835.
  90. Afshar, M., Gallo, R.L. Innate immune defense system of the skin. Vet. Dermatol. 2013; 24, e8–e9
  91. Navarro-López, V., Núñez-Delegido, E., Ruzafa-Costas, B., Sánchez-Pellicer, P., Agüera-Santos, J., Navarro-Moratalla, L. Probiotics in the Therapeutic Arsenal of Dermatologists. Microorganisms, 2021; 9, 1513.
  92. Sánchez-Pellicer, P., Navarro-Moratalla, L., Núñez-Delegido, E., Ruzafa-Costas, B., Agüera-Santos, J., Navarro-López, V. Acne, Microbiome, and Probiotics: The Gut–Skin Axis. Microorganisms. 2022; 10(7):1303.
  93. Bowe, W.P., Logan, A.C. Acne vulgaris, probiotics and the gut-brain-skin axis – Back to the future? Gut Pathog., 2011; 3, 1.
  94. Morales, P., Fujio, S., Navarrete, P., Ugalde, J.A., Magne, F., Carrasco-Pozo, C., Tralma, K., Quezada, M., Hurtado, C., Covarrubias, N., et al. Impact of Dietary Lipids on Colonic Function and Microbiota: An Experimental Approach Involving Orlistat-Induced Fat Malabsorption in Human Volunteers. Clin. Transl. Gastroenterol. 2016; 7, e161.
  95. Yan, H.M., Zhao, H.J., Guo, D.Y., Zhu, P.Q., Zhang, C.L., Jiang, W. Gut microbiota alterations in moderate to severe acne vulgaris patients. J. Dermatol. 2018; 45, 1166–1171
  96. Lee, D.K., Kim, M.J., Ham, J.W., An, H.M., Cha, M.K., Lee, S.W., Park, C.I., Shin, S.H., Lee, K.O., Kim, K.J., et al. In vitro evaluation of antibacterial activities and anti-inflammatory effects of Bifidobacterium spp. addressing acne vulgaris. Arch. Pharmacal. Res., 2012; 35, 1065–1071.
  97. Kwon, H.K., Lee, C.G., So, J.S., Chae, C.S., Hwang, J.S., Sahoo, A., Nam, J.H., Rhee, J.H., Hwang, K.C., Im, S.H. Generation of regulatory dendritic cells and CD4+Foxp3+ T cells by probiotics administration suppresses immune disorders. Proc. Natl. Acad. Sci. USA, 2010; 107, 2159–2164.
  98. Lee, Y.B., Byun, E.J., Kim, H.S. Potential Role of the Microbiome in Acne: A Comprehensive Review. Journal of Clinical Medicine., 2019; 8(7):987.
  99. Leiter, U., Eigentler, T., Garbe, C. Epidemiology of skin cancer. Adv Exp Med Biol., 2014; 810:120–140.
  100. Apalla, Z., Lallas, A., Sotiriou, E., Lazaridou, E., Ioannides, D. Epidemiological trends in skin cancer. Dermatol Pract Concept., 2017; 7(2):1-6.
  101. Chen, S.T., Geller, A.C., Tsao, H. Update on the epidemiology of melanoma. Curr Dermatol Rep., 2013; 2:24–34.
  102. Rastrelli, M., Tropea, S., Rossi, C., Alaibac, M. Melanoma: epidemiology, risk factors, pathogenesis, diagnosis and classification. In Vivo. 2014; 28:1005–1011
  103. Yu, Y., Champer, J., Beynet, D., Kim, J., Friedman, A.J. The role of the cutaneous microbiome in skin cancer: lessons learned from the gut. Journal of Drugs in Dermatology. 2015; JDD.14(5):461-465. PMID: 25942663
  104. Makaranka, S., Scutt, F., Frixou, M., Wensley, K.E., Sharma, R., Greenhowe, J. The gut microbiome and melanoma: A review. Exp. Dermatol., 2022; 31, 1292–1301.
  105. Vitali, F., Colucci, R., Di Paola, M., et al. Early melanoma invasivity correlates with gut fungal and bacterial profiles. Br J Dermatol. 2021; 186: 106-116.
  106. Luo, M., Hu, M.M., Feng, X., XiaoLi, W., Dong, D.G., Wang, W.C. Preventive effect of lactobacillus reuteri on melanoma. Biomed Pharmacother., 2020; 126:109929.
  107. Li, Y., Tinoco, R., Elmén, L., et al. (2019) Gut microbiota dependent anti-tumor immunity restricts melanoma growth in Rnf5 −/− mice. Nat Commun., 2019; 10(1): 1492.
  108. Pereira, F., ACL, M., Silva, M.B., et al. Interleukin-6 and the gut microbiota influence melanoma progression in obese mice. Nutr Cancer. 2021; 73(4): 642-651.
  109. Bzioueche, H., Sjödin, K.S., West, C.E., Khemis, A., Rocchi, S., Passeron, T., Tulic, M.K., Analysis of Matched Skin and Gut Microbiome of Patients with Vitiligo Reveals Deep Skin Dysbiosis: Link with Mitochondrial and Immune Changes, Journal of Investigative Dermatology, Volume 141, Issue 9, 2021; Pages 2280-2290, ISSN 0022-202X.
  110. Ezzedine, K., Eleftheriadou, V., Whitton, M., and van Geel, N. Vitiligo. Lancet 386, 2015; 74–84.
  111. Roberts, G.H.L., Santorico, S.A., Spritz, R.A. The genetic architecture of vitiligo Pigment Cell Melanoma Res, 2020; 33 pp. 8-15
  112. Czarnowicki, T., He, H., Leonard, A., Kim, H. J., Kameyama, N., Pavel, A. B., et al. Blood endotyping distinguishes the profile of vitiligo from that of other inflammatory and autoimmune skin diseases. J. Allergy Clin. Immunol. 2019; 143, 2095–2107.
  113. Kotobuki, Y., Tanemura, A., Yang, L., Itoi, S., Wataya-Kaneda, M., Murota, H., et al. Dysregulation of melanocyte function by Th17-related cytokines: significance of Th17 cell infiltration in autoimmune vitiligo vulgaris. Pigment Cell Melanoma Res. 2012; 25, 219–230.
  114. Hadi, A., Wang, J. F., Uppal, P., Penn, L. A., and Elbuluk, N. Comorbid diseases of vitiligo: a 10-year cross-sectional retrospective study of an urban US population. J. Am. Acad. Dermatol. 2019; 82, 628–633.
  115. Lloyd-Price, J., Arze, C., Ananthakrishnan, A. N., Schirmer, M., Avila-Pacheco, J., Poon, T. W., et al. multi-omics of the gut microbial ecosystem in inflammatory bowel diseases. Nature 2019; 569, 655–662.
  116. Gevers, D., Kugathasan, S., Denson, L. A., Vázquez-Baeza, Y., Van Treuren, W., Ren, B., et al. The treatment-naive microbiome in new-onset Crohn’s disease. Cell Host Microbe 2014; 15, 382–392. DOI: 10.1016/j.chom.2014.02.005
  117. Dellacecca, E. R., Cosgrove, C., Mukhatayev, Z., Akhtar, S., Engelhard, V. H., Rademaker, A. W., et al. Antibiotics drive microbial imbalance and vitiligo development in mice. J. Invest. Dermatol. 2020; 140, 676–687.e676. DOI: 1016/j.jid.2019.08.435
  118. Ganju, P., Nagpal, S., Mohammed, M. H., Nishal Kumar, P., Pandey, R., Natarajan, V. T., et al. Microbial community profiling shows dysbiosis in the lesional skin of Vitiligo subjects. Sci. Rep. 2016; 6:18761. DOI: 10.1038/srep18761
  119. Ni, Qingrong, Ye, Zhubiao,Wang,Yinghan, Chen,Jianru, Zhang,Weigang, Ma,Cuiling, Li,Kai, Liu,Yu, Liu,Ling, Han,Zheyi Gao,Tianwen, Jian,Zhe, Li,Shuli, Li,Chunying. Frontiers in Microbiology. Microbial Dysbiosis in Vitiligo. Gut Microbial Dysbiosis and Plasma Metabolic Profile in Individuals with Vitiligo 2020;
  120. Giri, P.S., Bharti, A., Kemp, E.H., Dwivedi, M.K. Microorganisms in Pathogenesis and Management of Vitiligo. In: Dwivedi, M.K., Amaresan, N., Kemp, E.H., Shoenfeld, Y. (eds) Role of Microorganisms in Pathogenesis and Management of Autoimmune Diseases. Springer, Singapore. 2022;
  121. Schaefer, P. Acute and chronic Urticaria: evaluation and treatment. Am. Fam. Physician 2017; 95, 717–724.
  122. Gonçalo, M.; Gimenéz-Arnau, A.; Al-Ahmad, M.; Ben-Shoshan, M.; Bernstein, J.A.; Ensina, L.F.; Bernstein, J.A.; Bindslev-Jensen, C.; Brzoza, Z.; Buense Bedrikow, R.; et al. The global burden of chronic urticaria for the patient and society. Br. J. Dermatol., 2021; 184, 226–236.
  123. Church, M. K., Kolkhir, P., Metz, M. & Maurer, M. The role and relevance of mast cells in urticaria. Immunol. Rev. 2018; 282, 232–247.
  124. Lee, S. J. et al. Prevalence and risk factors of urticaria with a focus on chronic urticaria in children. Allergy Asthma Immunol. Res. 2017; 9, 212–219.
  125. Zuberbier, T., Balke, M., Worm, M., Edenharter, G. & Maurer, M. Epidemiology of urticaria: a representative cross-sectional population survey.  Exp. Dermatol.2010; 35, 869–873.
  126. Al Bander, Z.; Nitert, M.D.; Mousa, A.; Naderpoor, N. The gut microbiota and inflammation: An overview. Int. J. Environ. Res. Public Health, 2020; 17, 7618
  127. Krišto, M., Lugović-Mihić, L., Muñoz, M., Rupnik, M., Mahnic, A., Ozretić, P., Jaganjac, M., Ćesić, D., Kuna, M. Gut Microbiome Composition in Patients with Chronic Urticaria: A Review of Current Evidence and Data. Life.; 2023; 13(1):152.
  128. Wilmanski, T.; Rappaport, N.; Diener, C.; Gibbons, S.M.; Price, N.D. From taxonomy to metabolic output: What factors define gut microbiome health? Gut Microbes, 2021; 13, 1907270
  129. Godlewska, U.; Brzoza, P.; Kwiecień, K.; Kwitniewski, M.; Cichy, J. Metagenomic studies in inflammatory skin diseases. Curr. Microbiol., 2020; 77, 3201–3212.
  130. Wang, F.Y., Chi, C.C. Rosacea, Germs, and Bowels: A Review on Gastrointestinal Comorbidities and Gut–Skin Axis of Rosacea. Adv Ther 2021; 38, 1415–1424.
  131. Nabizadeh, E., Jazani, N.H., Bagheri, M., Shahabi, S., Association of altered gut microbiota composition with chronic urticaria, Annals of Allergy, Asthma & Immunology, 2017; Volume 119, Issue 1, Pages 48-53, ISSN 1081-1206,
  132. Lu, T., Chen, Y., Guo, Y., Sun, J., Shen, W., Yuan, M., Zhang, S., He, P., Jiao, X., “Altered Gut Microbiota Diversity and Composition in Chronic Urticaria”, Disease Markers, 2019; vol. 2019, Article ID 6417471, 11 pages.
  133. Wang, Xin and Yi, Wanyu and He, Liting and Luo, Shuaihantian and Wang, Jiaqi and Jiang, Li and Long, Hai and Zhao, Ming and Lu, Qianjin (2021), Abnormalities in Gut Microbiota and Metabolism in Patients With Chronic Spontaneous Urticaria vol 12, 2021; https://www.frontiersin.org/articles/ 10.3389/fimmu.2021.691304
  134. Zhang, X., Shi, L., Sun, T., Guo K. & Geng, S., BMC Microbiology volume 21, Article number 78. Urticaria patientis j. Microbiol. Biotechnol. 2021; 31, 747-755
  135. Yun-Zhou S., Qing-Feng T., Hai-Yan Q., Ying L., Hui Z. Causal relationship between gut microbiota and urticaria: a bidirectional two-sample mendelian randomization study. Frontiers in Microbiology. 2023; (Vol 14).
  136. Xu, X., Zhang, W., Guo, M., Xiao, C., Fu, Z., Yu, S., et al. Integrated analysis of gut microbiome and host immune responses in COVID-19. Front. Med. 2022; 16, 263–275.
  137. Nylund, L., Nermes, M., Isolauri, E., Salminen, S., de Vos, W. M., and Satokari, R. Severity of atopic disease inversely correlates with intestinal microbiota diversity and butyrate-producing bacteria. Allergy 2015; 70, 241–244.
  138. Zha, Z., Lv, Y., Tang, H., Li, T., Miao, Y., Cheng, J., et al. An orally administered butyrate-releasing xylan derivative reduces inflammation in dextran sulphate sodium-induced murine colitis. Int. J. Biol. Macromol. 2020; 156, 1217–1233.
  139. Bordoni, L., Gabbianelli, R., Fedeli, D., Fiorini, D., Bergheim, I., Jin, C. J., et al. Positive effect of an electrolyzed reduced water on gut permeability, fecal microbiota and liver in an animal model of Parkinson’s disease. 2019; PLoS One 14: e0223238.
  140. Caren, G., Solomon, M.D., M.P.H., Editor November 2, N Engl J Med 2017; 377:1754-1764
  141. Wang, F.Y., Chi, C.C. Rosacea, Germs, and Bowels: A Review on Gastrointestinal Comorbidities and Gut–Skin Axis of Rosacea. Adv Ther 2021; 38, 1415–1424.
  142. Chen, L., Li, J., Zhu, W., Kuang, Y., Liu, T., Zhang, W., Chem, X., Peng, C. (2020) Front. Microbiol., Sec. Microbial Immunology Volume 11 – 2020;
  143. Nam, J.H.H.; Yun, Y.Y.; Kim, H.S.S.; Kim, H.N.N.; Jung, H.J.J.; Chang, Y.Y.; Ryu, S.; Shin, H.; Kim, H.L.; Kim, W.S. Rosacea and its association with enteral microbiota in Korean females. Exp. Dermatol., 2018; 27, 37–42.
  144. Egeberg, A.; Weinstock, L.B.; Thyssen, E.P.; Gislason, G.H.; Thyssen, J.P. Rosacea and gastrointestinal disorders: A population-based cohort study. Br. J. Dermatol., 2017; 176, 100–106
  145. Two, A.M., Wu, W., Gallo, R.L., Hata, T.R. Rosacea: part I. Introduction, categorization, histology, pathogenesis, and risk factors. J Am Acad Dermatol.; 2015; 72(5):749–58
  146. Searle, T., Ali, F.R., Carolides, S., Al-Niaimi, F. Rosacea and the gastrointestinal system. Australas J Dermatol. 2020;
  147. Vera N, Patel NU. Seminario-Vidal L. Rosacea comorbidities. Dermatol Clin.; 2018; 36(2):115–22
  148. Hasan, N., Yang, H. Factors affecting the composition of the gut microbiota, and its modulation. PeerJ.; 2019; 7:e7502.
  149. Weiss E, Katta R. (2017) Diet and rosacea: the role of dietary change in the management of rosacea. Dermatol Pract Concept.; 2017; 7(4):31-37.
  150. Scheman, A., Rakowski, E.M., Chou, V., Chhatriwala, A., Ross, J., Jacob, S.E. Balsam of Peru: past and future. Dermatitis; 2013; 24(4):153–160.
  151. Wu C.Y., Chang Y.T., Juan C.K., et al. Risk of inflammatory bowel disease in patients with rosacea: Results from a nationwide cohort study in Taiwan. J Am Acad Dermatol; 2017; (155):1–7
  152. Rahmati Roudsari, R. Karimi, S. Sohrabvandi & A. M. Mortazavian Health Effects of Probiotics on the Skin, Critical Reviews in Food Science and Nutrition, 2015; 55:9, 1219-1240.
  153. El Hage R, Hernandez-Sanabria E, Van de Wiele T. Emerging trends in “smart probiotics”: functional consideration for the development of novel health and industrial applications. Front Microbiol 2017; 8:1889
  154. Lopes EG, Moreira DA, Gullón P, Gullón B, Cardelle-Cobas A, Tavaria FK. Topical application of probiotics in skin: adhesion, antimicrobial and antibiofilm in vitro assays. J Appl Microbiol. 2017;122(2):450–61.
  155. Myles IA, Earland NJ, Anderson ED, et al. First-in-human topical microbiome transplantation with Roseomonas mucosa for atopic dermatitis. JCI Insight. 2018;3(9):e120608.
  156. Yu, Y., Champer, J., Beynet, D., Kim, J., Friedman, A.J. The role of the cutaneous microbiome in skin cancer: lessons learned from the gut. Journal of Drugs in Dermatology: JDD. 2015; 14(5):461-465. PMID: 25942663
  157. Sherwani, M.A.; Tufail, S.; Muzaffar, A.F.; Yusuf, N. The skin microbiome and immune system: Potential target for chemoprevention? Photodermatol. Photoimmunol. Photomed. 2018; 34, 25–34
  158. Porubsky CF B, Alexandria B, Comeau V. The Role of Probiotics in Acne and Rosacea, Probiotics. Current Knowledge and Future Prospects, Shymaa Enany, IntechOpen. 2018;
  159. Kober MM, Bowe WP. The effect of probiotics on immune regulation, acne, and photoaging. Int J Womens Dermatol. 2015;1(2):85-89. doi:10.1016/j.ijwd.2015.02.001
  160. Shah, R. A., Hsu, J. I., Patel, R. R., Mui, U. N., & Tyring, S. K. (2022). Antibiotic resistance in dermatology: The scope of the problem and strategies to address it. Journal of the American Academy of Dermatology, 86(6), 1337–1345.
(Visited 192 times, 1 visits today)

Creative Commons License
This work is licensed under a Creative Commons Attribution 4.0 International License.