Next Article in Journal
Carotenoids in Drug Discovery and Medicine: Pathways and Molecular Targets Implicated in Human Diseases
Next Article in Special Issue
Oleuropein-Rich Gellan Gum/Alginate Films as Innovative Treatments against Photo-Induced Skin Aging
Previous Article in Journal
Investigating Localized Electrochemical of Ferrocenyl-Imidazolium in Ionic Liquid Using Scanning Electrochemical Microscopy Configuration
Previous Article in Special Issue
Effects of UV and UV-vis Irradiation on the Production of Microalgae and Macroalgae: New Alternatives to Produce Photobioprotectors and Biomedical Compounds
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Photolyase Production and Current Applications: A Review

1
Tecnologico de Monterrey, School of Engineering and Sciences, Monterrey 64849, Mexico
2
Tecnologico de Monterrey, Institute of Advanced Materials for Sustainable Manufacturing, Monterrey 64849, Mexico
3
Department of Basic Sciences, Universidad de Monterrey, Av. Ignacio Morones Prieto 4500 Pte, San Pedro Garza Garcia 66238, Mexico
4
Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
5
Division of Gastroenterology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Molecules 2022, 27(18), 5998; https://doi.org/10.3390/molecules27185998
Submission received: 1 July 2022 / Revised: 20 August 2022 / Accepted: 24 August 2022 / Published: 15 September 2022
(This article belongs to the Special Issue New Trends in Skin Care: Topical Delivery of Cosmeceutical Molecules)

Abstract

:
The photolyase family consists of flavoproteins with enzyme activity able to repair ultraviolet light radiation damage by photoreactivation. DNA damage by the formation of a cyclobutane pyrimidine dimer (CPD) and a pyrimidine-pyrimidone (6-4) photoproduct can lead to multiple affections such as cellular apoptosis and mutagenesis that can evolve into skin cancer. The development of integrated applications to prevent the negative effects of prolonged sunlight exposure, usually during outdoor activities, is imperative. This study presents the functions, characteristics, and types of photolyases, their therapeutic and cosmetic applications, and additionally explores some photolyase-producing microorganisms and drug delivery systems.

1. Introduction

In the late 1940s, Albert Kelner reported the phenomenon of enzymatic photoreactivation for the first time, studying in situ the repair of major DNA lesions produced by ultraviolet (UV) radiation by a light-induced enzymatic cleavage of a thymine dimer to yield two thymine monomers. This enzyme is called photolyase, and it detects and binds to dimers contained in single- and double-stranded DNA [1]. Photolyase is also active against cytosine dimers and cytosine–thymine dimers, which are also formed by UV irradiation, but with much less frequently.
Photolyases are evolutionary ancient flavoproteins found in archaea, bacteria, and eukarya domains [2,3]. In vertebrates, photolyases are found in fish, amphibians, birds, and reptiles. Interestingly, mammals have lost this mechanism of protection throughout evolution. Hence, most mammals, including humans, can only repair these DNA lesions through a process called nucleotide excision repair. UV light is a known source of damage to our DNA that can be repaired by this mechanism. However, the deficient repair of UV-induced DNA damage which, for example, may occur after excessive unprotected sunbathing, can cause damage to DNA by inducing the formation of a cyclobutane pyrimidine dimer (CPD) and a pyrimidine-pyrimidone (6-4) photoproduct (6-4PP). Both CPD and 6-4PP can lead to cellular apoptosis and mutagenesis, which can eventually lead to skin cancer.
According to the World Cancer Research Fund International [4], between 2 and 3 million non-melanoma skin cancers and 132,000 melanoma skin cancers are estimated to occur globally each year. Importantly, the main factors that promote the development of melanoma are exposure to the sun and a history of sunburn. Commonly, this chronic, long-term sun exposure causes lesions such as premature aging and actinic keratosis, both associated with skin cancer [5].
The increasing incidence of skin cancer and the consequent burden to the healthcare system poses great problems, since the primary topical product defense has been broad-spectrum (UVA/UVB) sunscreen application. Nonetheless, it has been shown that some sunscreen products contain chemicals that can enter the bloodstream [6], and some products were found to contain cancer-causing substances. In addition to these deleterious effects on health, the environmental impacts of sunscreen ingredients include increasing concerns regarding coral reefs. Therefore, the use of repair DNA enzyme-based products represents a feasible alternative to prevent and treat DNA damage and consequent skin lesions.
This review focuses on the characterization of photolyases and therapeutic and cosmetic applications available.

2. Photolyase

Photolyase is a protein that has various functions, among which is the repair of DNA damaged from exposure to UV rays from the sun [7].
Additionally, the presence of photolyases has been observed in fish, amphibians, birds, and a few marsupials [8]; nevertheless, in higher plants and animals, the ability to repair DNA was lost during evolution (Figure 1). Therefore, their function is limited to regulating growth and acting as blue-light photoreceptors; these enzymes are known as cryptochromes [9].
The first precedent of enzymes with photolyase-like activity was discovered in 1993 in a plant of the Brassicaceae family native to Europe, Arabidopsis thaliana [10]. In other plants such as white mustard (Sinapis alba), the same photolyase cofactors are present; however, these plants lack DNA repair activity [11]. However, the photoreactivation process and DNA repair with photolyase are available and have been demonstrated in Streptomyces griseus [12] and bacteriophages [13].
In cyanobacteria, the existence of photolyase is reported in Anacystis nidulans [14]. The activity of photolyase enzymes has been reported mainly in environments with high exposure to UV rays, such as the case of twelve species of diatoms from Antarctica that demonstrated a DNA repair response to UV radiation damage [15].
In eukaryotic organisms, the Antarctic alga Chlamydomonas sp. ICE-L, which is developed in high-irradiation environments, can use mechanisms that reduce UV radiation damage [16].
Figure 2 shows the evolutionary relationship of several members of the photolyase superfamily. From the members analyzed, the phylogenetic tree highlights two main groups: one dominated by photolyases from microalgae and plants, and the other with more diversified evolutionary relationships. Interestingly, some microalgae species express photolyase at a relatively near distance to human photolyases.

2.1. Type of Enzyme

Photolyases are monomeric proteins with a molecular mass from 50 to 61 kDa. They are made up of 450–550 amino acids and two unsorted covalently bound chromophores as cofactors. One of the cofactors is always flavin adenine dinucleotide FAD, and the second is methenyltetrahydrofolate (MTHF) or 8-hydroxy-7, 8-didemethyl-5-deazariboflavin (8-HDF) [20]. Light is an indispensable resource in the photoreactivation process for the conversion of enzyme–substrate complexes into additional DNA repair products [21]. The surface of photolyases is characterized by a positive charge near the substrate binding which promotes the interaction with DNA [22]. The structure of photolyase consists of two domains: a C-terminal α-helical catalytic domain that contains the flavin cofactor and an N-terminal α/β domain [23].
The superfamily of chromophores/photolyases (CRY/PHR) consists of subfamilies (Figure 3) [24,25], of which there are three types of photolyases that have been identified to repair a specific type of dimer: (1) CPD photolyase, responsible for repairing CPD, (6-4); (2) photolyase, which repairs (6-4) pyrimidine pyrimidone; and (3) cryptochrome-DASH, which causes a variety of physiological changes to DNA [2,26].

2.2. Photolyase and Microorganisms

Photolyase biosynthesis begins with the transfer of an electron from the anionic chromophore FADH, promoted by light. In a catalytically active form, this binds to damaged DNA in a high-affinity, light-independent step [27,28].
To study numerous characteristics of the photolyases obtained from various organisms such as structural, physical, and mechanical properties, researchers have employed genetic manipulation approaches to promote the overexpression of DNA repair enzymes, since biosynthesis naturally provides low concentrations. Subsequently, extraction and purification processes are performed to obtain better quality enzymes (Table 1) [29]. After these steps, generally, between 15 and 25 mg of photolyase with a purity greater than 98% is obtained. The quality of the enzyme obtained is reflected in the color of the extract: a dark blue color indicates a good quality product. Therefore, it is important to measure and quantify the absorbance spectrum after enzyme purification [29].

2.3. DNA Damage by UV Irradiation

The organisms and cells that inhabit the earth naturally are continuously exposed to genotoxic agents present in the environment. Sunlight, as a source of UV rays, is one of the main genotoxic agents; nonetheless, it is essential for the development of life, for example, in the process of photosynthesis [40].
The damage caused by exposure to UV radiation (Figure 4) can trigger various skin reactions, mainly (as mentioned before) by affecting pyrimidine dimers; erythema, immunosuppression, and melanogenesis are just some of the disorders that can occur. Hence, it has been proven that overexposure to sunlight almost irreversibly damages skin cells [41].

2.4. Photolyase Mechanism of Action

There are multiple mechanisms of DNA repair: direct reversal [43], base excision, nucleotide excision [44], mismatch [45], single-strand break, and double-strand break repair [46].
Photolyases are light-driven DNA repair enzymes which function specifically in the reversal of genomic lesions induced by UV radiation [47]. An important DNA repair mechanism for mutagenic and cytotoxic UV-induced photolesions in DNA is photoreactivation, which utilizes enzyme photolyase for reverting modified nitrogenous bases into normal form, employing blue wavelength [48]. DNA repair to minimize mutagenic changes is divided into two main mechanisms: single-strand (ss) and double-strand (ds) DNA damage repair. In the same way, ss and ds DNA repair are divided into direct reversal repair, nucleotide excision repair, base excision repair, and mismatch repair for ss DNA repair and homologous recombination and non-homologous end-joining repairs for ds DNA repairs [7].
The photolyase for the CPD and 6-4PP lesions can be divided into CPD photolyases based on the photoproduct that they recognize, which are subdivided in relation to the amino acid sequence they possess, and 6-4PP photolyases, respectively [49]. Photolyases can possess flavin adenine dinucleotide (FAD) in four different redox states: oxidized (FAD), anionic semiquinone (FAD), neutral semiquinone (FADH), and anionic hydroquinone (FADH) [7]. Different redox states will act differently in the absorption spectrum. While FAD and FAD mainly absorb UV-A, and blue light, FADH absorbs blue, green, and red light, and FADH does not absorb visible light. An absorption spectrum was determined from two organisms by following flavin intermediates during the catalytic process, showing the mode of action on the DNA repair by photolyase based on the absorption properties of FAD [50].
According to Wang et al. [50], DNA repair by photolyase can be divided into three steps (Figure 5): (i) Recognition, which is a light-independent process where CPD or (6-4) photoproduct in the damaged DNA forms a photolyase/DNA complex by flipping into the active site containing the flavin cofactor of the DNA photolyase. (ii) During the catalytic reaction [51] taking place when FAD is at a fully reduced state (FADH), the methenytetrahydrofolate (MTHF), a photolyase chromophore, transfers energy to FADH through the absorption of photons from the blue-light spectrum, changing FADH to an exciting form of FADH. In this step, CPD or the (6-4) photoproduct ring is opened by bond dissociation in the dimer radical anion, and electron transfers from FADH to the lesions occur [7,52]. (iii) Finally, the separation step completes the repair process by the departure of repaired DNA from photolyases.
As previously mentioned, FAD oxidation states influence the light absorption spectra; hence, catalytic reactions for DNA repair from FADH take place under blue-light conditions. Considering the poor permeability of living tissues to blue light, works that aim to remove the barrier of this range of light have been developed using harvesting or antenna chromophores. Antenna chromophores are utilized by some photolyase to gain the photoreception ability of a light range, and the development and application of artificial antenna chromophores demonstrate an increase of up to 1.5-fold in DNA repair activity. This is a promising strategy area for the optimization of photolyase DNA repair [52]. Nevertheless, reports on the application of artificial antenna chromophores remain scarce.

2.5. Immobilization and Biocarriers

Photolyase offers outstanding protection and recovery mechanisms against sunlight-induced DNA damage, but if the enzyme lacks the capacity of reaching the site where it should intervene, or lacks the stability necessary to complete this, its utilization will go to waste.
Currently, immobilization strategies such as the encapsulation of photolyases within liposomes are the most used method to stabilize and deliver the protein; nonetheless, other methods, such as the application of nanomaterials, are being explored [49]. There are an assortment of technologies and systems currently being used as an approach to deliver active ingredients in skincare, such as antioxidants that can be useful and might offer an alternative to liposomes. Some of these technologies include the use of different formulations, such as gels, hydrogels, and emulsions (micro and nano-emulsions), the usage of other vesicular delivery systems instead of liposomes, such as ethosomes, transfersomes, niosomes, and non-vesicular particles such as solid lipid nanoparticles, and nanostructured lipid carriers, polymeric nanoparticles, nanocrystals, and—lastly—carbon, metal, or metal oxide nanoparticles. These immobilization technologies seek to improve long-term stability and sensitivity in order to provide a better catalytic activity; however, it is necessary to explore their efficiency according to their final applications [54,55,56,57,58,59,60,61].

3. Photolyase Applications

The sun is the most vital star in our solar system. The radiation from this star promotes photosynthesis in photoautotroph organisms, and in mammalian species, light promotes the synthesis of vitamins necessary for the bone structure, such as the production of vitamin D, among other things [62,63]. However, everything in excess can be harmful, and high exposure to UV rays can be harmful to cells [64,65]. For this reason, some organisms, such as bacteria and algae, which are constantly exposed to UV radiation, have evolved to develop several mechanisms that prevent or even revert damages caused by the exposure of different UV wavelengths. Unfortunately, most organisms, humans included, do not have the mechanisms to mitigate the effects of prolonged exposure to UV rays, with the consequence of causing damage to DNA which ultimately can result in the development of skin diseases, or even skin cancer.
The use of enzymes that promote DNA repair, such as photolyase, represents a key alternative for a wide range of applications in skin care products aimed to prevent or reverse the damage prompted by extensive sunlight exposure. Advances in immobilization within liposomes and nanomaterials have made the application of photolyase more accessible in a wide range of commercial products, due to its higher stability. The activity of the enzyme is maintained even in harsh conditions [49]. Photolyase is typically used in topical creams or sunscreens which are used to prevent UV damage, and in therapy to restore the skin from conditions such as premature photoaging, actinic keratosis, and squamous cell carcinoma.
In the following sections, we will discuss the different applications of photolyases in different fields and cosmetics, and explore some examples of them being used and their effects on different organisms or skin conditions. Table 2 presents some studies related to the use of sunscreens containing photolyase and their effects on the treatment of premature photoaging, actinic keratosis, and some types of skin cancer.

3.1. Current Photolyase Production

As stated previously in the manuscript, there are many organisms capable of producing photolyase to repair DNA damage from UV exposure, and currently, the industry is focusing on the production of this enzyme to take advantage of its properties to improve crops and its potential use in therapeutic products [28].
The current application of the photolyase enzyme is focused on the enhancement of UV resistance not only in microorganisms, but also in other organisms and areas. An example of photolyase application is the use of it to improve agriculture practices, which has been reported to improve plants. For example, in Japan, researchers successfully modified African rice to overexpress the gene. CPD repaired enzyme photolyase, demonstrating an increased UVB resistance compared to the rice plants without these modifications [66]. Similarly, there is open research about improving the UV irradiation resistance of fungal insecticides to improve their pest control [67].

3.2. Sunscreen with Photolyase as an Ingredient

The use of products to protect the skin from the sun is something humankind has undertaken for centuries, from the Egyptians’ use of different plant extracts to the sunscreens we know today, composed of a wide variety of filters offering protection beyond UV radiation [68,69]. In the last few decades, the use of sunscreens has been promoted and advertised to prevent damage to the skin by UV radiation; thus, the sunscreen market is expected to reach USD 24.4 billion by 2029 [68,70].
Despite the proven advantages of regular sunscreen application (from reducing the effects of photoaging to protecting against skin cancer) [71,72], there are some concerns about the systematic and regular use of sunscreens and the effect on vitamin D synthesis, especially in older people; nonetheless, there is plenty of evidence that deny those claims and assure that the usage of sunscreens does not affect levels of vitamin D [73,74,75]. Another concern with sunscreens is the skin absorption of the most used active ingredients, such as avobenzone and oxybenzone, among others, resulting in high plasmatic levels [5,6]. In addition to these health concerns, the damage these organic ingredients cause to the environment has been studied [76,77]. However, these findings prove the importance and the need to further assess the products used in sunscreens, and to find active ingredients that can offer the same benefits for all ages and skin types without a detrimental effect on human and environmental health [78].
The use of the photolyase has been around for the last few decades, with the first patent in 1988 [79], and since then it has been tested and proven that this enzyme can prevent and reverse sunlight-induced skin damage when used as an active ingredient in traditional sunscreen formulations [7,80,81,82].
There are sunscreens containing DNA repair enzymes obtained from microalgae and labeled as “plankton extract” available on the market. Some companies that offer this product are Isdin (Barcelona, Spain) with the product Eryfotona® AK-NMSC, Kwizda Pharma GmbH (Vienna, Austria) with Ateia®, and Pharma Cosmetics (Oradell, NJ, USA), with a variety of products such as Neova Active®, Neova Everyday®, and Neova Silc Sheer® 2.0. Many other companies are also releasing new products containing photolyase [82]. For a more in-depth listing of all the products currently available on the market, we recommend consulting the Supplementary Material of the review by Yarosh et al. [83].

3.3. Photoaging

Normal signs of aging are the appearance of fine lines, pigmentation, and wrinkles in the skin, all of which appear over time and are generally attributed to getting older; however, there is evidence that multiple factors contribute to accelerating the aging process, including lifestyle factors such as smoking, sleep, diet, the chronic use of drugs, environmental factors such as contact with polluted air, exposure to visible and infrared light [84], and UV radiation, with the last one accounting for up to 90% of visible changes on the skin [85,86].
Photoaging is a term that has been used since 1986 [87] to reference the effect on the skin produced by chronic exposure to UV radiation that causes damage to DNA [82,84,88,89] and leads to premature aging. All of these signs overlap with natural signs of aging, such as wrinkles, thin and dry skin due to a loss of underlying fat, more fragile skin, and pigmentation; however, the effects of photoaging go beyond these appearances with, depending on the skin type, the formation of fine to coarse wrinkles, a leather-like appearance to the skin, and hyperpigmentation or dyschromia. All of these signs are presented even when normal signs of aging have not appeared [85,90,91,92].
Given that photoaging only occurs in areas of the skin that have been exposed to the sun for a prolonged time, it is recommended to avoid sun exposure and apply sunscreens that provide protection against UVA and UVB radiation and can even in some cases reverse damage [71,85,93]. Listed below are some studies related to the use of photolyase in clinical trials intended to assess the efficacy of this enzyme to mitigate the effects of photoaging.
Corinne Granger et al. conducted a study in which they tested, over 28 days, a tinted sunscreen containing encapsulated photolyase on 30 women of ages ranging from 45 to 65 with slight-to-moderate photoaging signs [94]. The patients were evaluated on day zero and on the last day of use and signs of photoaging were analyzed and evaluated. The results showed an improvement between 6 and 12% for each factor analyzed in the treated women compared to the control.
Another study was conducted to prove the efficacy of using photolyase from the microalgae Anacystis nidulans to prevent damage to DNA produced by UV radiation on the skin. This was performed by comparing the use of a regular sunscreen with a sun protection factor (SPF) 50, and the same sunscreen supplemented with photolyase [95].
The results from this study revealed that, for the 10 participants (5 males and 5 females, with ages ranging from 26 to 36 years old and Fitzpatrick skin type II), the formation of CPDs and apoptosis of the skin cells was reduced by 93%, and 82%, respectively, with the use of the sunscreen containing photolyase compared with the control that received only radiation.
Similarly, Emanuele et al. [96] compared a novel topical product containing a traditional sunscreen with SPF 50, a mixture of DNA repair enzymes encapsulated in liposomes, one of them being photolyase, and an antioxidant complex versus other topical products with similar characteristics. The study demonstrated the novel topical products as the most effective in reducing the three parameters analyzed: the formation of CPD, 8-oxo-7,8-dihydro-2′-deoxyguanosine (8OHdG), and protein carbonylation (PC). This was attributed to the synergistic effect of all the components within the product.
Table 2. Studies on the use of photolyase in different skin conditions.
Table 2. Studies on the use of photolyase in different skin conditions.
StudySkin ConditionTherapy EvaluationDurationAssessmentResultsReference
Clinical seriesAKEryfotona3 monthsClinical photographyGreat improvement in AK lesions count.[97]
Retrospective case studyXeroderma PigmentosumEryfotona12 monthsHistological recordsReduction of 65% for AK, 56% for BCC, and 100% for SCC lesions.[98]
Longitudinal, observational clinicalAKEryfotona3 monthsClinical, dermoscopy, and confocal microscopy analysisGrade I AK clinical and dermoscopy improvement.
Reduction in desquamation.
Improvement in the epidermal architectural pattern.
Grade II AK, no improvements.
[99]
Pilot studyAKEryfotona1 monthClinical, dermoscopy, and reflectance confocal microscopy assessmentsErythema and scaling improvement.[100]
ClinicalAKEryfotona9 monthsTelethermografyHyperthermic halos area reduced from 3.46 to 0.64 cm2.[101]
Randomized, assessor-blinded parallel-groupAKEryfotona9 monthsLesion countSignificant reduction in new AK lesions.
No additional photodynamic therapy required.
[102]
Prospective observational studyAKEryfotona
Cryotherapy
6 monthsEpidemiologic, clinical, and therapeutic variablesNo adverse cutaneous effects and
84% improvement in AK lesion count.
[103]
Prospective, single-arm, case-seriesAKEryfotona3 monthsClinical photographyPartial response in 100% of patients.
50% reduction in lesion count.
[104]
Randomized, double-blind parallel-group
Pilot study
AKEryfotona6 monthsClinical, dermoscopy, and reflectance confocal Microscopy evaluationSignificant reduction in mean AK lesion number up to 31%.[105]
--PhotoagingTinted facial sunscreen with high sun protection, peptide complex, and encapsulated photolyase1 monthPeriocular wrinkles, skin firmness and elasticity, UV spots, and patient subjective questionnaireWrinkle count −6.9%.
Wrinkle volume −10.4%.
UV spots area −9%.
Firmness +8.2.
Elasticity +11.3%.
[94]
--UV exposureSunscreen amended with photolyase4 daysSkin biopsies after experimental irradiations93% prevention of CPD formation.
82% apoptosis prevention.
[95]
Head-to-head comparison studiesUV exposureTriple-protection factor broad-spectrum sunscreen (TPF50)--Skin biopsies after experimental irradiationsReduction in CPD and protein carboxylation.[96]
Randomized, double-blind, factorial clinical trialAKSunscreen amended with photolyase2 monthsClinical and demographic variablesNo significant differences with common sunscreen.[106]
--AKEryfotona1 monthHistopathological and molecular assessmentImprovement in the field of cancerization.
Restoration of normal phenotype through CPI-17 up-regulation.
[107]
Randomized, clinical studyAKSunscreen amended with photolyase6 monthsFluorescence diagnostics using methylaminolevulinate
Skin biopsies
Superior to sunscreen in reduction in field cancerization and UVR-associated molecular signatures.[108]
AK: actinic keratosis. SCC: squamous cell carcinoma. BCC: basal cell carcinoma. CPD: cyclobutane pyrimidine dimer.

3.4. Actinic Keratosis

Actinic keratosis (AK) is a skin disease that is characterized by squamous lesions that histologically show keratinocyte neoplasms occurring on skin that has had long-term exposure to UV radiation [109]. AK is typically presented in people with light skin belonging to the Fitzpatrick skin types of I to III in areas of the skin on which they experience solar exposure regularly, such as the head (especially in areas with hair loss), ears, neck, forearm, and the dorsum of the hand [80,110]. The lesions of AK can progress to keratinocyte carcinoma, the most common type of skin cancer in the United States [111]. Therefore, AK must be prevented or treated early to avoid further disease progression. The method to prevent skin damage is to avoid sun exposure, but when the exposure is inevitable it is recommended to use protective clothes and at least 2 mg per cm2 of sunscreen in the exposed areas [112]. However, these measures are not enough once the skin damage is present and the use of active molecules capable of stopping and even reversing this damage is essential [113].
Some studies have been published with diverse findings. For example, the efficacy of photolyase in sunscreen and a combination of topical antioxidants in the treatment of patients with AK were assessed in Brazil [106]. A total of 80 patient forearms were tested using either regular sunscreen or sunscreen containing photolyase during the day and the night. They either applied topical antioxidant or placebo cream to one forearm for 8 weeks. The researchers found that all groups tested showed a significant improvement at the end of the study; however, there were little to no significant differences between the groups using regular sunscreen and those with additional photolyase. The authors attributed this to the short time period of the treatment.
Puig-Butillé et al. [107] evaluated the use of a film-forming medical device containing photolyase in liposomes on a small group of patients from a wide range of ages, composed mainly of males that presented multiple AK and two patients with xeroderma pigmentosum. The study was conducted for 4 weeks, and at the end of the study, all groups showed an improvement in their condition. Notably, some showed total clearance on the assessed area from lesions caused by UV radiations. With the same medical device with photolyase, Eibenschutz et al. [102] analyzed the effect of the product with the enzyme compared to a regular sunscreen on 30 patients that underwent photodynamic therapy (PDT) with a total of 225 AK lesions for 9 months. At the end of the treatment, it was shown that the group treated with the film-forming medical device did better than the group treated with regular sunscreen. PDT was not needed, nor was any other medical procedure, and no new AK lesions were observed.
A research study in 2015 compared the effects of sunscreen containing photolyase and traditional sunscreens in 28 patients during 6 months of treatment [108]. The findings showed that both treatments reduced hyperkeratosis; however, for the field cancerization and the levels of CPDs, the results showed a better performance in the group that used the sunscreen containing photolyase than the group that used the traditional sunscreens.

3.5. Skin Cancer

Skin cancer remains a major global public health threat [114]. As the human body naturally grows, cells are divided when needed and die when they lose their normal function, or due to natural cell-aging. Cancer starts as a result of an interference in the cycle of cell growth division and death. This condition is characterized by an overproduction of cell division and the permanency of abnormal cells, instead of their death [115].
According to the American Cancer Society, there are five types of skin cancer: basal and squamous cell skin cancer, melanoma skin cancer, Merkel cell skin cancer, lymphoma of the skin, and Kaposi sarcoma. The basal and squamous cell skin cancers are mostly found on the body areas commonly exposed to the sun without protection, such as the head, neck, and arms. These two types are the most common, and they start in the epidermis [115]. In early-stage cases, a skin excision is the treatment for squamous cell carcinoma (SCC) [116].
The precursor cell of SCC is AK, and for BCC it is hypothesized that its occurrence is related to interfollicular epidermal basal keratinocytes with retained basal morphology from the follicular outer root sheath or sebaceous gland-derived keratinocytes [117]. Malignant melanoma is a serious form of skin cancer that begins in cells known as melanocytes. While it is less common than SCC and BCC, melanoma is the most severe type of skin cancer due to its capacity to spread if it is not treated at an early stage [118,119]. Merker cell, lymphoma, and Kaposi cancers are less common types of skin cancer [115].
Field cancerization refers to the replacement of the normal cell population by a cancer-primed cell population that may show no morphological change [101]. Some studies have focused on this topic and the role of photolyase as a potential treatment. A study with a topic product categorized as a medical device containing photolyase showed positive results for treating cancerization areas with long-term use versus the use of commercially available sunscreen, not only in terms of Baseline Severity Index (BSI) and total Clinical Score (TCS), but also by reducing the occurrence of new AK lesions [105].
In 2016, Naverrete-Dechent et al. [104] showed that subjects with skin field cancerization showed a partial positive response to the treatment with a photolyase-added sunscreen and at least a 50% reduction in their lesion number. These findings are consistent with the work of Laino et al. [101], where 30 individuals with AK were treated with a photolyase-added medical device, which improved their lesions.

4. Perspectives

Photolyase’s potential for DNA repair has been widely studied in vitro and in vivo. Especially, numerous studies have investigated the impact of the use of products containing DNA repair enzymes in patients. Still, we found that one of the major limitations of these studies is the duration of the treatment, making it difficult to determine the effect of photolyase; thus, several details remain as open questions. In this sense, it would be advisable for researchers to consider longer-term treatments in order to achieve solid information regarding the effect of photolyase in cosmetic use.
On the other hand, the mechanism of photolyase DNA repair has been widely studied, which makes it plausible to begin the evaluation of possible modifications to increase the activation of photolyase, even under unfavorable conditions. The use of artificial antenna chromophores is an area of opportunity that has already shown results, but the literature remains scarce. Additionally, microalgae biomass is a potential source of photolyase enzymes. This represents an alternative source in the application of technologies where microalgae can be implemented for bioremediation processes such as CO2 sequestration and water treatment, and the produced microalgae biomass could further be used to extract photolyase. These biomass production processes also represent a low-cost approach for generating the enzyme, as a substrate is not needed during the production of microalgal biomass.

5. Conclusions

The exploration of photolyase production improvement can be implemented to take advantage of several microorganisms. Biotechnologies for the development of new products that mitigate the DNA damage produced by UV radiation have been explored. Technological advances have allowed the extraction and use of photolyases in different products. Furthermore, new efforts for immobilization and delivery systems are needed to develop new topical formulations to battle UV-damaged DNA affections.

Author Contributions

Conceptualization, M.M.-R., A.L.D.-Z. and D.R.-G.; methodology, H.R.-P., E.R.M.-S., K.R.V.-Z. and M.E.L.-A.; validation, H.R.-P., K.G.C.-A. and M.E.L.-A.; formal analysis, H.R.-P.; investigation, A.L.D.-Z., D.R.-G. and K.G.C.-A.; resources, M.M.-R.; data curation, E.R.M.-S. and K.R.V.-Z.; writing—original draft preparation, M.M.-R., A.L.D.-Z., D.R.-G., S.A. and K.G.C.-A.; writing—review and editing, M.M.-R., S.A., A.L.D.-Z., D.R.-G., K.G.C.-A. and A.G.-M.; visualization, M.E.L.-A., E.R.M.-S. and D.R.-G.; supervision, M.M.-R., J.E.S.-H. and A.G.-M.; project administration, R.P.-S., and H.M.N.I.; funding acquisition, M.M.-R., R.P.-S. and H.M.N.I. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by NET ZERO RESEARCH FUND (2021 call) from The Bank of Nova Scotia. The funding number is not applicable.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

This work was also partially supported by Consejo Nacional de Ciencia y Tecnología (CONACYT) under Sistema Nacional de Investigadores (SNI) program awarded to Manuel Martínez Ruiz (CVU: 418151), Juan Eduardo Sosa-Hernández (CVU: 375202), Roberto Parra-Saldívar (CVU: 35753) and Hafiz M.N. Iqbal (CVU: 735340). Figures were created with BioRender.com.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Kelner, A. Photoreactivation of ultraviolet-irradiated Escherichia coli, with special reference to the dose-reduction principle and to ultraviolet-induced mutation. J. Bacteriol. 1949, 58, 511–522. [Google Scholar] [CrossRef] [PubMed]
  2. Selby, C.P.; Sancar, A. A cryptochrome/photolyase class of enzymes with single-stranded DNA-specific photolyase activity. Proc. Natl. Acad. Sci. USA 2006, 103, 17696–17700. [Google Scholar] [CrossRef] [PubMed]
  3. Lucas-Lledó, J.I.; Lynch, M. Evolution of mutation rates: Phylogenomic analysis of the photolyase/cryptochrome family. Mol. Biol. Evol. 2009, 26, 1143–1153. [Google Scholar] [CrossRef]
  4. World Cancer Research Fund International. Skin Cancer Statistics. Available online: https://www.wcrf.org/cancer-trends/skin-cancer-statistics/ (accessed on 16 August 2022).
  5. Matta, M.K.; Zusterzeel, R.; Pilli, N.R.; Patel, V.; Volpe, D.A.; Florian, J.; Oh, L.; Bashaw, E.; Zineh, I.; Sanabria, C.; et al. Effect of Sunscreen Application Under Maximal Use Conditions on Plasma Concentration of Sunscreen Active Ingredients. JAMA 2019, 321, 2082. [Google Scholar] [CrossRef]
  6. Matta, M.K.; Florian, J.; Zusterzeel, R.; Pilli, N.R.; Patel, V.; Volpe, D.A.; Yang, Y.; Oh, L.; Bashaw, E.; Zineh, I.; et al. Effect of Sunscreen Application on Plasma Concentration of Sunscreen Active Ingredients. JAMA 2020, 323, 256. [Google Scholar] [CrossRef] [PubMed]
  7. Kavakli, I.H.; Ozturk, N.; Gul, S. DNA repair by photolyases. Adv. Protein Chem. Struct. Biol. 2019, 115, 1–19. [Google Scholar]
  8. Menck, C.F.M. Shining a light on photolyases. Nat. Genet. 2002, 32, 338–339. [Google Scholar] [CrossRef]
  9. Chaves, I.; Pokorny, R.; Byrdin, M.; Hoang, N.; Ritz, T.; Brettel, K.; Essen, L.O.; Van Der Horst, G.T.J.; Batschauer, A.; Ahmad, M. The cryptochromes: Blue light photoreceptors in plants and animals. Annu. Rev. Plant Biol. 2011, 62, 335–364. [Google Scholar] [CrossRef]
  10. Ahmad, M.; Cashmore, A.R. HY4 gene of A. thaliana encodes a protein with characteristics of a blue-light photoreceptor. Nature 1993, 366, 162–166. [Google Scholar] [CrossRef]
  11. Malhotra, K.; Kim, S.T.; Batschauer, A.; Dawut, L.; Sancar, A. Putative blue-light photoreceptors from Arabidopsis thaliana and Sinapis alba with a high degree of sequence homology to DNA photolyase contain the two photolyase cofactors but lack DNA repair activity. Biochemistry 1995, 34, 6892–6899. [Google Scholar] [CrossRef] [PubMed]
  12. Kelner, A. Effect of Visible Light on the Recovery of Streptomyces Griseus Conidia from Ultra-violet Irradiation Injury. Proc. Natl. Acad. Sci. USA 1949, 35, 73–79. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Dulbecco, R. Reactivation of ultra-violet-inactivated bacteriophage by visible light. Nature 1949, 163, 949–950. [Google Scholar] [CrossRef] [PubMed]
  14. Aubert, C.; Mathis, P.; Eker, A.P.M.; Brettel, K. Intraprotein electron transfer between tyrosine and tryptophan in DNA photolyase from Anacystis nidulans. Proc. Natl. Acad. Sci. USA 1999, 96, 5423–5427. [Google Scholar] [CrossRef]
  15. Karentz, D.; Cleaver, J.E.; Mitchell, D.L. Cell survival characteristics and molecular responses of Antarctic phytoplankton to ultraviolet-B radiation. J. Phycol. 1991, 27, 326–341. [Google Scholar] [CrossRef]
  16. Li, C.; Ma, L.; Mou, S.; Wang, Y.; Zheng, Z.; Liu, F.; Qi, X.; An, M.; Chen, H.; Miao, J. Cyclobutane pyrimidine dimers photolyase from extremophilic microalga: Remarkable UVB resistance and efficient DNA damage repair. Mutat. Res. 2015, 773, 37–42. [Google Scholar] [CrossRef]
  17. Saitou, N.; Nei, M. The neighbor-joining method: A new method for reconstructing phylogenetic trees. Mol. Biol. Evol. 1987, 4, 406–425. [Google Scholar] [CrossRef]
  18. Zuckerkandl, E.; Pauling, L. Evolutionary Divergence and Convergence in Proteins. In Evolving Genes and Proteins; Elsevier: London, UK, 1965; pp. 97–166. [Google Scholar]
  19. Tamura, K.; Stecher, G.; Kumar, S. MEGA11: Molecular Evolutionary Genetics Analysis Version 11. Mol. Biol. Evol. 2021, 38, 3022–3027. [Google Scholar] [CrossRef]
  20. Sancar, A. Structure and Function of DNA Photolyase and Cryptochrome Blue-Light Photoreceptors. Chem. Rev. 2003, 103, 2203–2238. [Google Scholar] [CrossRef]
  21. Sancar, G.B. DNA photolyases: Physical properties, action mechanism, and roles in dark repair. Mutat. Res. 1990, 236, 147–160. [Google Scholar] [CrossRef]
  22. Müller, M.; Carell, T. Structural biology of DNA photolyases and cryptochromes. Curr. Opin. Struct. Biol. 2009, 19, 277–285. [Google Scholar] [CrossRef]
  23. Zhang, M.; Wang, L.; Zhong, D. Photolyase: Dynamics and Mechanisms of Repair of Sun-Induced DNA Damage. Photochem. Photobiol. 2017, 93, 78–92. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Mei, Q.; Dvornyk, V. Evolutionary History of the Photolyase/Cryptochrome Superfamily in Eukaryotes. PLoS ONE 2015, 10, e0135940. [Google Scholar] [CrossRef] [PubMed]
  25. Öztürk, N.; Song, S.H.; Özgür, S.; Selby, C.P.; Morrison, L.; Partch, C.; Zhong, D.; Sancar, A. Structure and function of animal cryptochromes. Cold Spring Harb. Symp. Quant. Biol. 2007, 72, 119–131. [Google Scholar] [CrossRef] [PubMed]
  26. Essen, L.O.; Klar, T. Light-driven DNA repair by photolyases. Cell. Mol. Life Sci. 2006, 63, 1266–1277. [Google Scholar] [CrossRef]
  27. Graf, D.; Wesslowski, J.; Ma, H.; Scheerer, P.; Krauß, N.; Oberpichler, I.; Zhang, F.; Lamparter, T. Key Amino Acids in the Bacterial (6-4) Photolyase PhrB from Agrobacterium fabrum. PLoS ONE 2015, 10, e0140955. [Google Scholar] [CrossRef]
  28. He, Y.; Qu, C.; Zhang, L.; Miao, J. DNA photolyase from Antarctic marine bacterium Rhodococcus sp. NJ-530 can repair DNA damage caused by ultraviolet. 3 Biotech 2021, 11, 102. [Google Scholar] [CrossRef]
  29. An, M.; Zheng, Z.; Qu, C.; Wang, X.; Chen, H.; Shi, C.; Miao, J. The first (6-4) photolyase with DNA damage repair activity from the Antarctic microalga Chlamydomonas sp. ICE-L. Mutat. Res.-Fundam. Mol. Mech. Mutagen. 2018, 809, 13–19. [Google Scholar] [CrossRef]
  30. Marizcurrena, J.J.; Martínez-López, W.; Ma, H.; Lamparter, T.; Castro-Sowinski, S. A highly efficient and cost-effective recombinant production of a bacterial photolyase from the Antarctic isolate Hymenobacter sp. UV11. Extremophiles 2019, 23, 49–57. [Google Scholar] [CrossRef]
  31. Kiontke, S.; Geisselbrecht, Y.; Pokorny, R.; Carell, T.; Batschauer, A.; Essen, L.O. Crystal structures of an archaeal class II DNA photolyase and its complex with UV-damaged duplex DNA. EMBO J. 2011, 30, 4437–4449. [Google Scholar] [CrossRef]
  32. An, M.; Qu, C.; Miao, J.; Sha, Z. A Class II CPD Photolyase and a 6-4 Photolyase with Photorepair Activity from the Antarctic Moss Pohlia nutans M211. Photochem. Photobiol. 2021, 97, 1527–1533. [Google Scholar] [CrossRef]
  33. An, M.; Qu, C.; Miao, J.; Sha, Z. Two class II CPD photolyases, PiPhr1 and PiPhr2, with CPD repair activity from the Antarctic diatom Phaeodactylum tricornutum ICE-H. 3 Biotech 2021, 11, 377. [Google Scholar] [CrossRef] [PubMed]
  34. Öztürk, N.; Kao, Y.T.; Selby, C.P.; Kavakli, I.H.; Partch, C.L.; Zhong, D.; Sancar, A. Purification and Characterization of a Type III Photolyase from Caulobacter crescentus. Biochemistry 2008, 47, 10255. [Google Scholar] [CrossRef] [PubMed]
  35. Navarro, E.; Niemann, N.; Kock, D.; Dadaeva, T.; Gutiérrez, G.; Engelsdorf, T.; Kiontke, S.; Corrochano, L.M.; Batschauer, A.; Garre, V. The DASH-type Cryptochrome from the Fungus Mucor circinelloides Is a Canonical CPD-Photolyase. Curr. Biol. 2020, 30, 4483–4490.e4. [Google Scholar] [CrossRef] [PubMed]
  36. Tagua, V.G.; Pausch, M.; Eckel, M.; Gutiérrez, G.; Miralles-Durán, A.; Sanz, C.; Eslava, A.P.; Pokorny, R.; Corrochano, L.M.; Batschauer, A. Fungal cryptochrome with DNA repair activity reveals an early stage in cryptochrome evolution. Proc. Natl. Acad. Sci. USA 2015, 112, 15130–15135. [Google Scholar] [CrossRef] [PubMed]
  37. Schleicher, E.; Heßling, B.; Illarionova, V.; Bacher, A.; Weber, S.; Richter, G.; Gerwert, K. Light-induced reactions of Escherichia coli DNA photolyase monitored by Fourier transform infrared spectroscopy. FEBS J. 2005, 272, 1855–1866. [Google Scholar] [CrossRef]
  38. Brash, D.E.; Franklin, W.A.; Sancar, G.B.; Haseltine, W.A. Escherichia coli DNA photolyase reverses cyclobutane pyrimidine dimers but not pyrimidine-pyrimidone (6-4) photoproducts. J. Biol. Chem. 1985, 260, 11438–11441. [Google Scholar] [CrossRef]
  39. Sancar, G.B.; Sancar, A. Purification and characterization of DNA photolyases. Methods Enzymol. 2006, 408, 121–156. [Google Scholar] [CrossRef]
  40. Mullenders, L.H.F. Solar UV damage to cellular DNA: From mechanisms to biological effects. Photochem. Photobiol. Sci. 2018, 17, 1842–1852. [Google Scholar] [CrossRef]
  41. Marrot, L.; Meunier, J.R. Skin DNA photodamage and its biological consequences. J. Am. Acad. Dermatol. 2008, 58, S139–S148. [Google Scholar] [CrossRef]
  42. Zhang, M.; Wang, L.; Zhong, D. Photolyase: Dynamics and electron-transfer mechanisms of DNA repair. Arch. Biochem. Biophys. 2017, 632, 158–174. [Google Scholar] [CrossRef]
  43. Ahmad, A.; Nay, S.L.; O’Connor, T.R. Direct Reversal Repair in Mammalian Cells. In Advances in DNA Repair; Chen, C., Ed.; InTech: Rijeka, Croatia, 2015. [Google Scholar]
  44. Petruseva, I.O.; Evdokimov, A.N.; Lavrik, O.I. Molecular mechanism of global genome nucleotide excision repair. Acta Nat. 2014, 6, 23–34. [Google Scholar] [CrossRef] [Green Version]
  45. Fukui, K. DNA Mismatch Repair in Eukaryotes and Bacteria. J. Nucleic Acids 2010, 2010, 260512. [Google Scholar] [CrossRef] [PubMed]
  46. Vítor, A.C.; Huertas, P.; Legube, G.; de Almeida, S.F. Studying DNA Double-Strand Break Repair: An Ever-Growing Toolbox. Front. Mol. Biosci. 2020, 7, 24. [Google Scholar] [CrossRef] [PubMed]
  47. Huang, R.; Zhou, P.K. DNA Damage Repair: Historical Perspectives, Mechanistic Pathways and Clinical Translation for Targeted Cancer Therapy; Springer: Greer, SA, USA, 2021; Volume 6, ISBN 4139202100648. [Google Scholar]
  48. Pathak, J.; Rajneesh; Singh, P.R.; Häder, D.P.; Sinha, R.P. UV-induced DNA damage and repair: A cyanobacterial perspective. Plant Gene 2019, 19, 100194. [Google Scholar] [CrossRef]
  49. Ramírez, N.; Serey, M.; Illanes, A.; Piumetti, M.; Ottone, C. Immobilization strategies of photolyases: Challenges and perspectives for DNA repairing application. J. Photochem. Photobiol. B Biol. 2021, 215, 112113. [Google Scholar] [CrossRef] [PubMed]
  50. Wang, J.; Du, X.; Pan, W.; Wang, X.; Wu, W. Photoactivation of the cryptochrome/photolyase superfamily. J. Photochem. Photobiol. C Photochem. Rev. 2015, 22, 84–102. [Google Scholar] [CrossRef]
  51. Brettel, K.; Müller, P.; Yamamoto, J. Kinetics of Electron Returns in Successive Two-Photon DNA Repair by (6-4) Photolyase. ACS Catal. 2022, 12, 3041–3045. [Google Scholar] [CrossRef]
  52. Terai, Y.; Sato, R.; Matsumura, R.; Iwai, S.; Yamamoto, J. Enhanced DNA repair by DNA photolyase bearing an artificial light-harvesting chromophore. Nucleic Acids Res. 2020, 48, 10076–10086. [Google Scholar] [CrossRef]
  53. Kavakli, I.H.; Ozturk, N.; Gul, S. DNA repair by photolyases. In Advances in Protein Chemistry and Structural Biology; Elsevier Inc.: London, UK, 2019; Volume 115, pp. 1–19. [Google Scholar]
  54. Benson, H.A.E.; Grice, J.E.; Mohammed, Y.; Namjoshi, S.; Roberts, M.S. Topical and Transdermal Drug Delivery: From Simple Potions to Smart Technologies. Curr. Drug Deliv. 2019, 16, 444. [Google Scholar] [CrossRef]
  55. Pavlou, P.; Siamidi, A.; Varvaresou, A.; Vlachou, M. Skin Care Formulations and Lipid Carriers as Skin Moisturizing Agents. Cosmetics 2021, 8, 89. [Google Scholar] [CrossRef]
  56. Roberts, M.S.; Cheruvu, H.S.; Mangion, S.E.; Alinaghi, A.; Benson, H.A.E.; Mohammed, Y.; Holmes, A.; van der Hoek, J.; Pastore, M.; Grice, J.E. Topical drug delivery: History, percutaneous absorption, and product development. Adv. Drug Deliv. Rev. 2021, 177, 113929. [Google Scholar] [CrossRef]
  57. Eskens, O.; Amin, S. Challenges and effective routes for formulating and delivery of epidermal growth factors in skin care. Int. J. Cosmet. Sci. 2021, 43, 123–130. [Google Scholar] [CrossRef]
  58. Salvioni, L.; Morelli, L.; Ochoa, E.; Labra, M.; Fiandra, L.; Palugan, L.; Prosperi, D.; Colombo, M. The emerging role of nanotechnology in skincare. Adv. Colloid Interface Sci. 2021, 293, 102437. [Google Scholar] [CrossRef]
  59. Montenegro, L. Nanocarriers for skin delivery of cosmetic antioxidants. J. Pharm. Pharmacogn. Res. 2014, 2, 73–92. [Google Scholar]
  60. Kim, B.; Cho, H.-E.; Moon, S.H.; Ahn, H.-J.; Bae, S.; Cho, H.-D.; An, S. Transdermal delivery systems in cosmetics. Biomed. Dermatol. 2020, 4, 10. [Google Scholar] [CrossRef]
  61. Travis, B.; Darren, S.; Zimei, W. Pharmaceutical Strategies for the Topical Dermal Delivery of Peptides/Proteins for Cosmetic and Therapeutic Applications. Austin J. Pharmacol. Ther. 2014, 2, 1–10. [Google Scholar]
  62. Nair, R.; Maseeh, A. Vitamin D: The sunshine vitamin. J. Pharmacol. Pharmacother. 2012, 3, 118–126. [Google Scholar] [CrossRef]
  63. Wacker, M.; Holick, M.F. Sunlight and Vitamin D: A global perspective for health. Dermatoendocrinology 2013, 5, 51–108. [Google Scholar] [CrossRef]
  64. Hoel, D.G.; Berwick, M.; de Gruijl, F.R.; Holick, M.F. The risks and benefits of sun exposure 2016. Dermatoendocrinology 2016, 8, e1248325. [Google Scholar] [CrossRef]
  65. Mead, M.N. Benefits of Sunlight: A Bright Spot for Human Health. Environ. Health Perspect. 2008, 116, A160–A167. [Google Scholar] [CrossRef]
  66. Mmbando, G.S.; Teranishi, M.; Hidema, J. Transgenic rice Oryza glaberrima with higher CPD photolyase activity alleviates UVB-caused growth inhibition. GM Crop. Food 2021, 12, 435–448. [Google Scholar] [CrossRef]
  67. Tong, S.; Feng, M. Molecular basis and regulatory mechanisms underlying fungal insecticides’ resistance to solar ultraviolet irradiation. Pest Manag. Sci. 2022, 78, 30–42. [Google Scholar] [CrossRef]
  68. Ma, Y.; Yoo, J. History of sunscreen: An updated view. J. Cosmet. Dermatol. 2021, 20, 1044–1049. [Google Scholar] [CrossRef]
  69. Lyons, A.B.; Trullas, C.; Kohli, I.; Hamzavi, I.H.; Lim, H.W. Photoprotection beyond ultraviolet radiation: A review of tinted sunscreens. J. Am. Acad. Dermatol. 2021, 84, 1393–1397. [Google Scholar] [CrossRef]
  70. Guan, L.L.; Lim, H.W.; Mohammad, T.F. Sunscreens and Photoaging: A Review of Current Literature. Am. J. Clin. Dermatol. 2021, 22, 819–828. [Google Scholar] [CrossRef]
  71. Singer, S.; Karrer, S.; Berneburg, M. Modern sun protection. Curr. Opin. Pharmacol. 2019, 46, 24–28. [Google Scholar] [CrossRef]
  72. Seité, S.; Fourtanier, A.M.A. The benefit of daily photoprotection. J. Am. Acad. Dermatol. 2008, 58, S160–S166. [Google Scholar] [CrossRef]
  73. Passeron, T.; Bouillon, R.; Callender, V.; Cestari, T.; Diepgen, T.L.; Green, A.C.; van der Pols, J.C.; Bernard, B.A.; Ly, F.; Bernerd, F.; et al. Sunscreen photoprotection and vitamin D status. Br. J. Dermatol. 2019, 181, 916–931. [Google Scholar] [CrossRef]
  74. Neale, R.E.; Khan, S.R.; Lucas, R.M.; Waterhouse, M.; Whiteman, D.C.; Olsen, C.M. The effect of sunscreen on vitamin D: A review. Br. J. Dermatol. 2019, 181, 907–915. [Google Scholar] [CrossRef]
  75. Marks, R.; Foley, P.A.; Jolley, D.; Knight, K.R.; Harrison, J.; Thompson, S.C. The Effect of Regular Sunscreen Use on Vitamin D Levels in an Australian Population: Results of a Randomized Controlled Trial. Arch. Dermatol. 1995, 131, 415–421. [Google Scholar] [CrossRef]
  76. Siller, A.; Blaszak, S.C.; Lazar, M.; Olasz Harken, E. Update About the Effects of the Sunscreen Ingredients Oxybenzone and Octinoxate on Humans and the Environment. Plast. Surg. Nurs. 2018, 38, 158–161. [Google Scholar] [CrossRef] [PubMed]
  77. Schneider, S.L.; Lim, H.W. Review of environmental effects of oxybenzone and other sunscreen active ingredients. J. Am. Acad. Dermatol. 2019, 80, 266–271. [Google Scholar] [CrossRef]
  78. Rigel, D.S.; Lim, H.W.; Draelos, Z.D.; Weber, T.M.; Taylor, S.C. Photoprotection for all: Current gaps and opportunities. J. Am. Acad. Dermatol. 2022, 86, S18–S26. [Google Scholar] [CrossRef] [PubMed]
  79. Mezei, M.; Gulasekharam, V.; Straubinger, R.; Hong, K.; Friend, D. Purification and administration of dna repair enzymes. Int. J. Pharm. 1988, 173, 3415–3422. [Google Scholar]
  80. Puig, S.; Granger, C.; Garre, A.; Trullàs, C.; Sanmartin, O.; Argenziano, G. Review of Clinical Evidence over 10 Years on Prevention and Treatment of a Film-Forming Medical Device Containing Photolyase in the Management of Field Cancerization in Actinic Keratosis. Dermatol. Ther. 2019, 9, 259–270. [Google Scholar] [CrossRef]
  81. Garinis, G.A.; Jans, J.; van der Horst, G.T.J. Photolyases: Capturing the light to battle skin cancer. Futur. Oncol. 2006, 2, 191–199. [Google Scholar] [CrossRef]
  82. Luze, H.; Nischwitz, S.P.; Zalaudek, I.; Müllegger, R.; Kamolz, L.P. DNA repair enzymes in sunscreens and their impact on photoageing—A systematic review. Photodermatol. Photoimmunol. Photomed. 2020, 36, 424–432. [Google Scholar] [CrossRef] [PubMed]
  83. Yarosh, D.B.; Rosenthal, A.; Moy, R. Six critical questions for DNA repair enzymes in skincare products: A review in dialog. Clin. Cosmet. Investig. Dermatol. 2019, 12, 617–624. [Google Scholar] [CrossRef]
  84. Addor, F.A.S. Beyond photoaging: Additional factors involved in the process of skin aging. Clin. Cosmet. Investig. Dermatol. 2018, 11, 437–443. [Google Scholar] [CrossRef]
  85. Huang, A.H.; Chien, A.L. Photoaging: A Review of Current Literature. Curr. Dermatol. Rep. 2020, 9, 22–29. [Google Scholar] [CrossRef]
  86. The Skin Cancer Foundation. Photoaging: What You Need to Know about the Other Kind of Aging. Available online: https://www.skincancer.org/blog/photoaging-what-you-need-to-know/ (accessed on 25 January 2022).
  87. Kligman, L.H. Photoaging. Manifestations, prevention, and treatment. Dermatol. Clin. 1986, 4, 517–528. [Google Scholar] [CrossRef]
  88. Gilchrest, B.A. Photoaging. J. Invest. Dermatol. 2013, 133, E2–E6. [Google Scholar] [CrossRef] [PubMed]
  89. Megna, M.; Lembo, S.; Balato, N.; Monfrecola, G. “Active” photoprotection: Sunscreens with DNA repair enzymes. Ital. J. Dermatol. Venereol. 2017, 152, 302–307. [Google Scholar] [CrossRef]
  90. Berneburg, M.; Plettenberg, H.; Krutmann, J. Photoaging of human skin. Photodermatol. Photoimmunol. Photomed. 2000, 16, 239–244. [Google Scholar] [CrossRef]
  91. Han, A.; Chien, A.L.; Kang, S. Photoaging. Dermatol. Clin. 2014, 32, 291–299. [Google Scholar] [CrossRef] [PubMed]
  92. Krutmann, J.; Schalka, S.; Watson, R.E.B.; Wei, L.; Morita, A. Daily photoprotection to prevent photoaging. Photodermatol. Photoimmunol. Photomed. 2021, 37, 482–489. [Google Scholar] [CrossRef]
  93. Fisher, G.J.; Kang, S.; Varani, J.; Bata-Csorgo, Z.; Wan, Y.; Datta, S.; Voorhees, J.J. Mechanisms of photoaging and chronological skin aging. Arch. Dermatol. 2002, 138, 1462–1470. [Google Scholar] [CrossRef]
  94. Granger, C.; Trullàs, C.; Bauza, G.; Garre, A. 17895 Anti-photoaging effect of a novel tinted facial sunscreen with high sun protection, peptide complex, and encapsulated photolyase after 1 month of use. J. Am. Acad. Dermatol. 2020, 83, AB202. [Google Scholar] [CrossRef]
  95. Emanuele, E. Reduced ultraviolet-induced DNA damage and apoptosis in human skin with topical application of a photolyase-containing DNA repair enzyme cream: Clues to skin cancer prevention. Mol. Med. Rep. 2011, 5, 570–574. [Google Scholar] [CrossRef]
  96. Emanuele, E.; Spencer, J.M.; Braun, M. An experimental double-blind irradiation study of a novel topical product (TPF 50) compared to other topical products with DNA repair enzymes, antioxidants, and growth factors with sunscreens: Implications for preventing skin aging and cancer. J. Drugs Dermatol. 2014, 13, 309–314. [Google Scholar]
  97. Puviani, M.; Barcella, A.; Milani, M. Efficacy of a photolyase-based device in the treatment of cancerization field in patients with actinic keratosis and non-melanoma skin cancer. G. Ital. Dermatol. Venereol. 2013, 148, 693–698. [Google Scholar] [PubMed]
  98. Giustini, S.; Miraglia, E.; Berardesca, E.; Milani, M.; Calvieri, S. Preventive long-term effects of a topical film-forming medical device with ultra-high uv protection filters and dna repair enzyme in xeroderma pigmentosum: A retrospective study of eight cases. Case Rep. Dermatol. 2014, 6, 222–226. [Google Scholar] [CrossRef] [PubMed]
  99. Rstom, S.A.; Abdalla, B.M.Z.; Rezze, G.G.; Paschoal, F.M. Evaluation of the effects of a cream containing liposomeencapsulated photolyase and SPF 100 sunscreen on facial actinic keratosis: Clinical, dermoscopic, and confocal microscopy based analysis. Surg. Cosmet. Dermatol. 2014, 6, 226–231. [Google Scholar]
  100. Malvehy, J.S.P. Field Cancerisation Improvement with Topical Application of a Film-Forming Medical Device Containing Photolyase and UV Filters in Patients with Actinic Keratosis, a Pilot Study. J. Clin. Exp. Dermatol. Res. 2014, 5, 220. [Google Scholar] [CrossRef]
  101. Laino, L.; Elia, F.; Desiderio, F.; Scarabello, A.; Sperduti, I.; Cota, C.; DiCarlo, A. The efficacy of a photolyase-based device on the cancerization field: A clinical and thermographic study. J. Exp. Clin. Cancer Res. 2015, 34, 84. [Google Scholar] [CrossRef]
  102. Eibenschutz, L.; Silipo, V.; De Simone, P.; Buccini, P.L.; Ferrari, A.; Carbone, A.; Catricalà, C. A 9-month, randomized, assessor-blinded, parallel-group study to evaluate clinical effects of film-forming medical devices containing photolyase and sun filters in the treatment of field cancerization compared with sunscreen in patients after successful p. Br. J. Dermatol. 2016, 175, 1391–1393. [Google Scholar] [CrossRef]
  103. Vañó-Galván, S.; Jiménez, N.; Grillo, E.; Ballester, A. Estudio observacional sobre la efectividad y tolerabilidad de un producto tópico con fotoliasa y filtros solares en pacientes con queratosis actínicas tratados con crioterapia en condiciones de uso habitual. Piel 2016, 31, 532–536. [Google Scholar] [CrossRef]
  104. Navarrete-Dechent, C.; Molgó, M. The use of a sunscreen containing DNA-photolyase in the treatment of patients with field cancerization and multiple actinic keratoses: A case-series. Dermatol. Online J. 2016, 23, 4–7. [Google Scholar] [CrossRef]
  105. Moscarella, E.; Argenziano, G.; Longo, C.; Aladren, S. Management of cancerization field with a medical device containing photolyase: A randomized, double-blind, parallel-group pilot study. J. Eur. Acad. Dermatol. Venereol. 2017, 31, e401–e403. [Google Scholar] [CrossRef]
  106. Alvares, B.A.; Miola, A.C.; Schimitt, J.V.; Miot, H.A.; Abbade, L.P.F. Efficacy of sunscreen with photolyase or regular sunscreen associated with topical antioxidants in treating advanced photodamage and cutaneous field cancerization: A randomized clinical trial. An. Bras. Dermatol. 2022, 97, 157–165. [Google Scholar] [CrossRef]
  107. Puig-Butillé, J.A.; Malvehy, J.; Potrony, M.; Trullas, C.; Garcia-García, F.; Dopazo, J.; Puig, S. Role of CPI-17 in restoring skin homoeostasis in cutaneous field of cancerization: Effects of topical application of a film-forming medical device containing photolyase and UV filters. Exp. Dermatol. 2013, 22, 494–496. [Google Scholar] [CrossRef] [PubMed]
  108. Carducci, M.; Pavone, P.S.; De Marco, G.; Lovati, S.; Altabas, V.; Altabas, K.; Emanuele, E.; De Marco, G.; Lovati, S.; Altabas, V.; et al. Comparative Effects of Sunscreens Alone vs. Sunscreens Plus DNA Repair Enzymes in Patients With Actinic Keratosis: Clinical and Molecular Findings from a 6-Month, Randomized, Clinical Study. J. Drugs Dermatol. 2015, 14, 986–990. [Google Scholar] [PubMed]
  109. Eisen, D.B.; Asgari, M.M.; Bennett, D.D.; Connolly, S.M.; Dellavalle, R.P.; Freeman, E.E.; Goldenberg, G.; Leffell, D.J.; Peschin, S.; Sligh, J.E.; et al. Guidelines of care for the management of actinic keratosis. J. Am. Acad. Dermatol. 2021, 85, e209–e233. [Google Scholar] [CrossRef]
  110. Salasche, S.J. Epidemiology of actinic keratoses and squamous cell carcinoma. J. Am. Acad. Dermatol. 2000, 42, S4–S7. [Google Scholar] [CrossRef] [PubMed]
  111. Albert, M.R.; Weinstock, M.A. Keratinocyte Carcinoma. CA. Cancer J. Clin. 2003, 53, 292–302. [Google Scholar] [CrossRef]
  112. Krutmann, J.; Berking, C.; Berneburg, M.; Diepgen, T.L.; Dirschka, T.; Szeimies, M. New Strategies in the Prevention of Actinic Keratosis: A Critical Review. Skin Pharmacol. Physiol. 2015, 28, 281–289. [Google Scholar] [CrossRef] [Green Version]
  113. Rosenthal, A.; Stoddard, M.; Chipps, L.; Herrmann, J. Skin cancer prevention: A review of current topical options complementary to sunscreens. J. Eur. Acad. Dermatol. Venereol. 2019, 33, 1261–1267. [Google Scholar] [CrossRef]
  114. Parker, E.R. The influence of climate change on skin cancer incidence—A review of the evidence. Int. J. Women’s Dermatol. 2021, 7, 17–27. [Google Scholar] [CrossRef]
  115. American Cancer Society; The American Cancer Society. Skin Cancer|Skin Cancer Facts|Common Skin Cancer Types. Available online: https://www.cancer.org/cancer/skin-cancer.html (accessed on 27 January 2022).
  116. Chabaane, M.; Ayadi, K.; Rkhami, M.; Drissi, C.; Houimli, S.; Bahri, K.; Zammel, I.; Badri, M. Management of a recurrence of a squamous cell carcinoma of the scalp with extension to the brain: A case report and literature review. Surg. Neurol. Int. 2020, 11, 347. [Google Scholar] [CrossRef]
  117. Cobanoglu, H.B.; Constantinides, M.; Ural, A. Nonmelanoma Skin Cancer of the Head and Neck: Molecular Mechanisms. Facial Plast. Surg. Clin. N. Am. 2012, 20, 437–443. [Google Scholar] [CrossRef]
  118. The American Cancer Society. What Is Melanoma Skin Cancer? Available online: https://www.cancer.org/cancer/melanoma-skin-cancer/about/what-is-melanoma.html (accessed on 27 January 2022).
  119. Agamohammadi, D.; Reihan, M.D.; Mirzaei, F.; Payandeh, Z.; Farzin, H.; Marahem, M. New Therapies for Melanoma Cancer Strategies. Crescent J. Med. Biol. Sci. 2021, 8, 3–9. [Google Scholar]
Figure 1. Presence of photolyase in different organisms. Due to evolution, some species lost the ability to repair DNA. Created with BioRender.com.
Figure 1. Presence of photolyase in different organisms. Due to evolution, some species lost the ability to repair DNA. Created with BioRender.com.
Molecules 27 05998 g001
Figure 2. Phylogenetic tree of photolyase. An evolutionary relationship among several members of the photolyase family is shown. This relationship was inferred using the neighbor-joining method [17] with the Poisson correction method [18] in MEGA11 [19]. Amino acid sequences from different species were obtained from the NCBI protein database (*) and CyanoBase database (#), and ID number is between square brackets. Colors are used as group identifiers: black for animals, green for plants, blue for microalgae (eukaryotic and cyanobacteria), red for fungi, orange for bacteria (except for cyanobacteria), and purple for arquea.
Figure 2. Phylogenetic tree of photolyase. An evolutionary relationship among several members of the photolyase family is shown. This relationship was inferred using the neighbor-joining method [17] with the Poisson correction method [18] in MEGA11 [19]. Amino acid sequences from different species were obtained from the NCBI protein database (*) and CyanoBase database (#), and ID number is between square brackets. Colors are used as group identifiers: black for animals, green for plants, blue for microalgae (eukaryotic and cyanobacteria), red for fungi, orange for bacteria (except for cyanobacteria), and purple for arquea.
Molecules 27 05998 g002
Figure 3. Integration of the CRY/PHR superfamily and the emergence of subfamilies due to evolutionary changes. The main distinctions are the ability to repair specific DNA and loss of photorepair capacity. Created with BioRender.com.
Figure 3. Integration of the CRY/PHR superfamily and the emergence of subfamilies due to evolutionary changes. The main distinctions are the ability to repair specific DNA and loss of photorepair capacity. Created with BioRender.com.
Molecules 27 05998 g003
Figure 4. UV light damage to the skin. UV radiation is composed of UVA and UVB rays, and has the ability to penetrate the skin layers. Various UV rays will have different impacts and effects on the skin; UVA and UVB rays are mainly linked to long-term skin damage and play a key role in some skin cancers [42]. Created with BioRender.com.
Figure 4. UV light damage to the skin. UV radiation is composed of UVA and UVB rays, and has the ability to penetrate the skin layers. Various UV rays will have different impacts and effects on the skin; UVA and UVB rays are mainly linked to long-term skin damage and play a key role in some skin cancers [42]. Created with BioRender.com.
Molecules 27 05998 g004
Figure 5. DNA repair by photolyase. DNA damage can be repaired by the enzyme photolyase in three steps: recognition of the damage, binding of the photolyase and the DNA damage, and lastly the separation of the enzyme resulting in the repaired DNA [47,53,50]. Created with BioRender.com.
Figure 5. DNA repair by photolyase. DNA damage can be repaired by the enzyme photolyase in three steps: recognition of the damage, binding of the photolyase and the DNA damage, and lastly the separation of the enzyme resulting in the repaired DNA [47,53,50]. Created with BioRender.com.
Molecules 27 05998 g005
Table 1. Studies demonstrating the presence of different types of photolyases in various organisms and the extraction and purification methods necessary to prove their DNA repair activity.
Table 1. Studies demonstrating the presence of different types of photolyases in various organisms and the extraction and purification methods necessary to prove their DNA repair activity.
MicroorganismsGenusType PhotolyaseExtraction and PurificationReferences
Agrobacterium fabrumProkaryote(6-4) PhotolyaseHeated and cleared by centrifugation HPLC column from Macherey and Nagel[30]
Rhodococcus sp. NJ-530Marine bacteriumCPD Class IDisrupted with ultrasonication, Ni-NTA resin[31]
Chlamydomonas sp. ICE-LPsychrophilic microalga(6-4) PhotolyaseDisrupted with ultrasonication, Ni-NTA resin[32]
Hymenobacter sp.Antarctic bacteriumCPD Class ILysed with sonication, Ni-NTA resin[33]
Methanosarcina mazei Mm0852ArchaeaCPD Class IICell disruption with lysozyme, EDTA and PMSF with an emulsifier, Ni-NTA resin[34]
Pohlia nutans M211Antarctic MossCPD Class II and (6-4) PhotolyaseUltrasonic cell disruptor, Ni-NTA resin[35]
Phaeodactylum tricornutum ICE-HAntarctic diatomCPD Class IIUltrasonic cell disruptor, Ni-NTA resin[36]
Caulobacter crescentusOligotrophic bacteriumCPD Class IIIHeated and cleared with centrifugation, purified by affinity chromatography on amylose resin[37]
Mucor circinelloidesFungusCRY-DASHDisrupted with a French press, affinity chromatography-HisTrap HP column[38]
Phycomyces blakesleeanus (NRRL1555)FungusCRY-DASHDisrupted with a French press, affinity chromatography-His Trap HP column[39]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ramírez-Gamboa, D.; Díaz-Zamorano, A.L.; Meléndez-Sánchez, E.R.; Reyes-Pardo, H.; Villaseñor-Zepeda, K.R.; López-Arellanes, M.E.; Sosa-Hernández, J.E.; Coronado-Apodaca, K.G.; Gámez-Méndez, A.; Afewerki, S.; et al. Photolyase Production and Current Applications: A Review. Molecules 2022, 27, 5998. https://doi.org/10.3390/molecules27185998

AMA Style

Ramírez-Gamboa D, Díaz-Zamorano AL, Meléndez-Sánchez ER, Reyes-Pardo H, Villaseñor-Zepeda KR, López-Arellanes ME, Sosa-Hernández JE, Coronado-Apodaca KG, Gámez-Méndez A, Afewerki S, et al. Photolyase Production and Current Applications: A Review. Molecules. 2022; 27(18):5998. https://doi.org/10.3390/molecules27185998

Chicago/Turabian Style

Ramírez-Gamboa, Diana, Ana Laura Díaz-Zamorano, Edgar Ricardo Meléndez-Sánchez, Humberto Reyes-Pardo, Karen Rocio Villaseñor-Zepeda, Miguel E. López-Arellanes, Juan Eduardo Sosa-Hernández, Karina G. Coronado-Apodaca, Ana Gámez-Méndez, Samson Afewerki, and et al. 2022. "Photolyase Production and Current Applications: A Review" Molecules 27, no. 18: 5998. https://doi.org/10.3390/molecules27185998

Article Metrics

Back to TopTop