Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 168
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Physiol Rev ; 101(2): 417-425, 2021 04 01.
Article in English | MEDLINE | ID: mdl-32790578

ABSTRACT

The host immune system shapes the fate of tumor progression. Hence, manipulating patients' immune system to activate host immune responses against cancer pathogenesis is a promising strategy to develop effective therapeutic interventions for metastatic and drug-resistant cancers. Understanding the dynamic mechanisms within the tumor microenvironment (TME) that contribute to heterogeneity and metabolic plasticity is essential to enhance the patients' responsiveness to immune targeted therapies. Riera-Domingo et al. (Riera-Domingo C, Audige A, Granja S, Cheng WC, Ho PC, Baltazar F, Stockmann C, Mazzone, M. Physiol Rev 100: 1-102, 2020) describe the immune landscape within the TME and highlight the significance of metabolic and hypoxic signatures that impact immune function and response to immunotherapy strategies. Current literature in this field confirms that targeting tumor metabolism and the acidic microenvironment commonly associated with tumors may present viable strategies to modulate the host immune system in favor of response to immune targeted therapies. However, development of better tools to understand tumor-immune interactions and identify mechanisms driving nonresponders, more innovative clinical trial design, and new therapies will need to be identified to move the field forward. Personalized immune therapies incorporating metabolic and microbiome-based gene signatures to influence the therapeutic response and novel methods to generate immunologically "hot" tumors are at the forefront of immunotherapy currently. The combination of these approaches with clinically approved immunotherapies will be valuable moving forward.


Subject(s)
Immunotherapy/methods , Neoplasms/therapy , Animals , Antineoplastic Agents/pharmacology , Humans , Immunotherapy/trends , Tumor Microenvironment/immunology
2.
Proc Natl Acad Sci U S A ; 120(49): e2315096120, 2023 Dec 05.
Article in English | MEDLINE | ID: mdl-38011564

ABSTRACT

Hidradenitis suppurativa (HS) is a complex inflammatory skin disease with undefined mechanistic underpinnings. Here, we investigated HS epithelial cells and demonstrated that HS basal progenitors modulate their lineage restriction and give rise to pathogenic keratinocyte clones, resulting in epidermal hyperproliferation and dysregulated inflammation in HS. When comparing to healthy epithelial stem/progenitor cells, in HS, we identified changes in gene signatures that revolve around the mitotic cell cycle, DNA damage response and repair, as well as cell-cell adhesion and chromatin remodeling. By reconstructing cell differentiation trajectory and CellChat modeling, we identified a keratinocyte population specific to HS. This population is marked by S100A7/8/9 and KRT6 family members, triggering IL1, IL10, and complement inflammatory cascades. These signals, along with HS-specific proinflammatory cytokines and chemokines, contribute to the recruitment of certain immune cells during the disease progression. Furthermore, we revealed a previously uncharacterized role of S100A8 in regulating the local chromatin environment of target loci in HS keratinocytes. Through the integration of genomic and epigenomic datasets, we identified genome-wide chromatin rewiring alongside the switch of transcription factors (TFs), which mediated HS transcriptional profiles. Importantly, we identified numerous clinically relevant inflammatory enhancers and their coordinated TFs in HS basal CD49fhigh cells. The disruption of the S100A enhancer using the CRISPR/Cas9-mediated approach or the pharmacological inhibition of the interferon regulatory transcription factor 3 (IRF3) efficiently reduced the production of HS-associated inflammatory regulators. Our study not only uncovers the plasticity of epidermal progenitor cells in HS but also elucidates the epigenetic mechanisms underlying HS pathogenesis.


Subject(s)
Hidradenitis Suppurativa , Humans , Hidradenitis Suppurativa/genetics , Skin/metabolism , Epigenomics , Epigenesis, Genetic , Stem Cells/metabolism , Chromatin/metabolism
3.
Semin Cell Dev Biol ; 128: 120-129, 2022 08.
Article in English | MEDLINE | ID: mdl-35131152

ABSTRACT

Hidradenitis suppurativa (HS) is characterized by deep-seated, highly inflamed, and painful lumps/abscesses, fistulae, and sinus tracts that grow extensively deep in the dermis and are highly immunogenic in nature. In about one-third of the HS patients there is strong evidence for the role of γ-secretase mutations along with dysregulated Notch signaling. However, the contribution of dysregulated Notch signaling in HS pathogenesis in relation to hair follicle alterations and hyper-activation of the immune system remains undefined. A genome-wide association study (GWAS), proteomic data and functional investigations of identified sequence variants in HS pathology are not fully revealing. The disease initiation or progression may involve bacterial infection besides intrinsic functional defects in keratinocytes, which may be key to further exacerbate immune cell infiltration and cytokine production in and around the lesional tissue. The absence of a suitable animal model that could fully recapitulate the pathogenesis of HS is a major impediment for proper understanding the underlying mechanisms and development of effective treatments. The presence of extracellular matrix (ECM) degradation products along with dysregulation in keratinocytes and, dermal fibroblasts ultimately affect immune regulation and are various components of HS pathogenesis. Bacterial infection further exacerbates the complexity of the disease progression. While anti-TNFα therapy shows partial efficacy, treatment to cure HS is absent. Multiple clinical trials targeting various cytokines, complement C5a and ECM products are in progress. This review provides state-of-the-art information on these aspects with a focus on dysregulated keratinocyte and immune cells; and role of ECM, and Keratin functions in this regard.


Subject(s)
Hidradenitis Suppurativa , Animals , Cytoskeletal Proteins/metabolism , Genome-Wide Association Study , Hidradenitis Suppurativa/genetics , Hidradenitis Suppurativa/pathology , Humans , Keratins/genetics , Keratins/metabolism , Proteomics , Signal Transduction/genetics
4.
J Biol Chem ; 299(1): 102746, 2023 01.
Article in English | MEDLINE | ID: mdl-36436565

ABSTRACT

Retinoid X receptors (RXRs) are nuclear transcription factors that partner with other nuclear receptors to regulate numerous physiological processes. Although RXR represents a valid therapeutic target, only a few RXR-specific ligands (rexinoids) have been identified, in part due to the lack of clarity on how rexinoids selectively modulate RXR response. Previously, we showed that rexinoid UAB30 potentiates all-trans-retinoic acid (ATRA) signaling in human keratinocytes, in part by stimulating ATRA biosynthesis. Here, we examined the mechanism of action of next-generation rexinoids UAB110 and UAB111 that are more potent in vitro than UAB30 and the FDA-approved Targretin. Both UAB110 and UAB111 enhanced ATRA signaling in human organotypic epithelium at a 50-fold lower concentration than UAB30. This was consistent with the 2- to 5- fold greater increase in ATRA in organotypic epidermis treated with UAB110/111 versus UAB30. Furthermore, at 0.2 µM, UAB110/111 increased the expression of ATRA genes up to 16-fold stronger than Targretin. The less toxic and more potent UAB110 also induced more changes in differential gene expression than Targretin. Additionally, our hydrogen deuterium exchange mass spectrometry analysis showed that both ligands reduced the dynamics of the ligand-binding pocket but also induced unique dynamic responses that were indicative of higher affinity binding relative to UAB30, especially for Helix 3. UAB110 binding also showed increased dynamics towards the dimer interface through the Helix 8 and Helix 9 regions. These data suggest that UAB110 and UAB111 are potent activators of RXR-RAR signaling pathways but accomplish activation through different molecular responses to ligand binding.


Subject(s)
Tetrahydronaphthalenes , Tretinoin , Humans , Retinoid X Receptors/metabolism , Bexarotene , Ligands , Tetrahydronaphthalenes/pharmacology , Tretinoin/pharmacology , Tretinoin/metabolism , Epidermis/metabolism
5.
Int J Mol Sci ; 23(3)2022 Feb 05.
Article in English | MEDLINE | ID: mdl-35163747

ABSTRACT

Type I interferons (IFNs) are important enhancers of immune responses which are downregulated in human cancers, including skin cancer. Solar ultraviolet (UV) B radiation is a proven environmental carcinogen, and its exposure contributes to the high prevalence of skin cancer. The carcinogenic effects of UV light can be attributed to the formation of cyclobutane pyrimidine dimers (CPD) and errors in the repair and replication of DNA. Treatment with a single dose of UVB (100 mJ/cm2) upregulated IFNα and IFNß in the skin of C57BL/6 mice. IFNα and IFNß were predominantly produced by CD11b+ cells. In mice lacking the type I IFN receptor 1 (IFNAR1), the repair of CPD following cutaneous exposure to a single dose of UVB (100 mJ/cm2) was decreased. UVB induced the expression of the DNA repair gene xeroderma pigmentosum A (XPA) in wild-type (WT) mice. In contrast, such treatment in IFNAR1 (IFNAR1-/-) mice downregulated XPA. A local UVB regimen consisting of UVB radiation (150 mJ/cm2) for 4 days followed by sensitization with hapten 2,4, dinitrofluorobenzene (DNFB) resulted in significant suppression of immune responses in both WT and IFNAR1-/- mice. However, there were significantly higher CD4+CD25+Foxp3+ regulatory T-cells in the draining lymph nodes of IFNAR1-/- mice in comparison to WT mice. Overall, our studies reveal a previously unknown action of type I IFNs in the repair of photodamage and the prevention of UVB-induced immune suppression.


Subject(s)
Interferon Type I , Skin Neoplasms , Xeroderma Pigmentosum , Animals , DNA Damage , DNA Repair , Interferon Type I/genetics , Interferon Type I/metabolism , Mice , Mice, Inbred C57BL , Pyrimidine Dimers/metabolism , Skin/metabolism , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Ultraviolet Rays/adverse effects , Xeroderma Pigmentosum/metabolism
6.
Mol Carcinog ; 60(1): 60-70, 2021 01.
Article in English | MEDLINE | ID: mdl-33283918

ABSTRACT

Ultraviolet (UV) B irradiation of the skin induces acute inflammation, as characterized by erythema, edema, and immunosuppression, and is subsequently linked to the progression of skin cancer. Toll-like receptor 4 (TLR4), a component of innate immunity, has been shown to play an important role in cancer. To elucidate the role of TLR4 in UVB-induced tumor development, TLR4-proficient (C3H/HeN) and TLR4-deficient (C3H/HeJ) mice were exposed to multiple doses of UVB radiation (200 mJ/cm2 ) for 40 weeks. Photocarcinogenesis was retarded in terms of tumor incidence, and tumor latency, in mice deficient in TLR4 compared with TLR4-proficient mice, whereas significantly greater numbers of tumors occurred in TLR4-proficient mice. There was significant upregulation of inflammatory markers like COX-2, PGE2 , S100A8, and S100A9 in the skin of TLR4-proficient mice than the skin of TLR4-deficient mice. Furthermore, we found that TLR4-proficient mice had a significantly higher number of Gr1+CD11b+ myeloid cells CD4+CD25+ regulatory T-cells than TLR4-deficient mice. Furthermore, the levels of interferon (IFN)-γ cytokine was increased and the levels of interleukin (IL)-4, IL-10, and IL-17 cytokines were decreased in serum, skin, and tumor lysates of TLR4-deficient mice in comparison with samples from TLR4-proficient mice. Together, our data indicate that TLR4-mediated inflammation may cause suppression of antitumor responses and trigger the development of UVB-induced skin cancers. Thus, strategies to inhibit TLR4-mediated immune suppression may allow us to develop preventive and therapeutic approaches for the management of UVB-induced cutaneous tumors.


Subject(s)
Skin Neoplasms/etiology , Skin Neoplasms/genetics , Toll-Like Receptor 4/genetics , Ultraviolet Rays/adverse effects , Animals , Carcinogenesis/genetics , Carcinogenesis/immunology , Carcinogenesis/radiation effects , Gene Deletion , Immunity , Inflammation/etiology , Inflammation/genetics , Inflammation/immunology , Mice , Skin Neoplasms/immunology , Toll-Like Receptor 4/immunology
7.
J Am Acad Dermatol ; 84(2): 432-470, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32738429

ABSTRACT

Psoriasis is a chronic, inflammatory, multisystem disease that affects up to 3.2% of the United States population. This guideline addresses important clinical questions that arise in psoriasis management and care and provides recommendations based on the available evidence. The treatment of psoriasis with topical agents and with alternative medicine will be reviewed, emphasizing treatment recommendations and the role of dermatologists in monitoring and educating patients regarding benefits as well as risks that may be associated. This guideline will also address the severity assessment methods of psoriasis in adults.


Subject(s)
Complementary Therapies/methods , Dermatologic Agents/administration & dosage , Dermatology/methods , Psoriasis/therapy , Academies and Institutes/standards , Administration, Cutaneous , Combined Modality Therapy/methods , Combined Modality Therapy/standards , Complementary Therapies/standards , Dermatology/standards , Evidence-Based Medicine/methods , Evidence-Based Medicine/standards , Foundations/standards , Humans , Patient Education as Topic/standards , Psoriasis/diagnosis , Severity of Illness Index , Treatment Outcome , United States
8.
Adv Exp Med Biol ; 1290: 89-98, 2021.
Article in English | MEDLINE | ID: mdl-33559857

ABSTRACT

The tumor microenvironment (TME), which assists in the development, progression, and metastasis of malignant cells, is instrumental in virtually every step of tumor development. While a healthy TME can protect against malignancy, in an unhealthy state, it can result in aberrant cellular behavior and augment tumor progression. Cytokines are one component of the TME, therefore, understanding the composition of the cytokine milieu in the tumor microenvironment is critical to understand the biology of malignant transformation. One cytokine, interleukin (IL)-23, has received particular scrutiny in cancer research because of its ability to manipulate host immune responses, its role in modulating the cells in TME, and its capacity to directly affect a variety of premalignant and malignant tumors. IL-23 belongs to the IL-12 cytokine family, which is produced by activated dendritic cells (DC) and macrophages. IL-23 acts by binding to its receptor consisting of two distinct subunits, IL-12Rß1 and IL-23R. This, in turn, leads to janus kinase (JAK) activation and signal transducer and activator of transcription (STAT) 3/4 phosphorylation. There have been contradictory reports of pro- and antitumor effects of IL-23, which likely depend on the genetic background, the type of tumor, the causative agent, and the critical balance of STAT3 signaling in both the tumor itself and the TME. Clinical trials of IL-12/23 inhibitors that are used to treat patients with psoriasis, have been scrutinized for reports of malignancy, the most common being nonmelanoma skin cancers (NMSCs). Continued investigation into the relationship of IL-23 and its downstream pathways holds promise in identifying novel targets for the management of cancer and other diseases.


Subject(s)
Interleukin-23 , Tumor Microenvironment , Humans , Interleukin-12 , Janus Kinases/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction
9.
J Am Acad Dermatol ; 82(1): 161-201, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31703821

ABSTRACT

Psoriasis is a chronic, multisystem, inflammatory disease that affects approximately 1% of children, with onset most common during adolescence. This guideline addresses important clinical questions that arise in psoriasis management and provides evidence-based recommendations. Attention will be given to pediatric patients with psoriasis, recognizing the unique physiology, pharmacokinetics, and patient-parent-provider interactions of patients younger than 18 years old. The topics reviewed here mirror those discussed in the adult guideline sections, excluding those topics that are irrelevant to, or lack sufficient information for, pediatric patients.


Subject(s)
Biological Products/therapeutic use , Dermatologic Agents/therapeutic use , Methotrexate/therapeutic use , Photochemotherapy , Psoriasis/drug therapy , Psoriasis/epidemiology , Adolescent , Adrenal Cortex Hormones/therapeutic use , Anthralin/therapeutic use , Calcineurin Inhibitors/therapeutic use , Cardiovascular Diseases/epidemiology , Child , Child, Preschool , Coal Tar/therapeutic use , Comorbidity , Cyclosporine/therapeutic use , Dyslipidemias/epidemiology , Evidence-Based Medicine , Humans , Infant , Infant, Newborn , Inflammatory Bowel Diseases/epidemiology , Insulin Resistance , Mental Health , Metabolic Syndrome/epidemiology , Nicotinic Acids/therapeutic use , Obesity/epidemiology , Psoriasis/psychology , Retinoids/therapeutic use
10.
J Am Acad Dermatol ; 82(6): 1445-1486, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32119894

ABSTRACT

Psoriasis is a chronic inflammatory disease involving multiple organ systems and affecting approximately 2% of the world's population. In this guideline, we focus the discussion on systemic, nonbiologic medications for the treatment of this disease. We provide detailed discussion of efficacy and safety for the most commonly used medications, including methotrexate, cyclosporine, and acitretin, and provide recommendations to assist prescribers in initiating and managing patients on these treatments. Additionally, we discuss newer therapies, including tofacitinib and apremilast, and briefly touch on a number of other medications, including fumaric acid esters (used outside the United States) and therapies that are no longer widely used for the treatment of psoriasis (ie, hydroxyurea, leflunomide, mycophenolate mofetil, thioguanine, and tacrolimus).


Subject(s)
Psoriasis/drug therapy , Acitretin/therapeutic use , Cyclosporine/therapeutic use , Drug Monitoring , Humans , Methotrexate/therapeutic use , Piperidines/therapeutic use , Pyrimidines/therapeutic use , Thalidomide/analogs & derivatives , Thalidomide/therapeutic use
11.
Exp Dermatol ; 28(2): 136-141, 2019 02.
Article in English | MEDLINE | ID: mdl-30506967

ABSTRACT

BACKGROUND: Studies have begun to investigate the complex relationship between host and microorganisms in non-infectious pathologies such as acne, atopic dermatitis and psoriasis. Though the skin is exposed to environmental stressors such as ultraviolet radiation (UVR), no studies exist examining the effects of both UVA and UVB on the skin microbiome. OBJECTIVE: To test the effect of UVA and UVB on human skin microbiome. METHODS: To test whether UV will alter the cutaneous microbiome, participants were exposed to doses of UVA (22-47 J/cm2 ) or UVB (100-350 mJ/cm2 ) and samples were collected. DNA was isolated and sequenced to identify the microbial composition of each sample. RESULTS: There was vast intra- and inter-subject variation at all time points, and phylum and species-level differences were identified. These included an increase in the phylum Cyanobacteria and a decrease in the family Lactobacillaceae and Pseudomonadaceae. The sensitivity of microbes to UVR and their re-colonization potential following exposure differed in UVA vs UVB samples. LIMITATIONS: The sample size was small, and the study was limited to males. CONCLUSION: The results demonstrate that UVR has profound qualitative and quantitative influences on the composition of the skin microbiome, possibly effecting skin pathology in which UVR is a factor.


Subject(s)
Microbiota/radiation effects , Skin/microbiology , Skin/radiation effects , Ultraviolet Rays , Acne Vulgaris/microbiology , Adult , DNA/radiation effects , Dermatitis, Atopic/microbiology , Humans , Inflammation/microbiology , Male , Psoriasis/microbiology , Young Adult
13.
J Am Acad Dermatol ; 81(3): 775-804, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31351884

ABSTRACT

Psoriasis is a chronic inflammatory disease involving multiple organ systems and affecting approximately 3.2% of the world's population. In this section of the guidelines of care for psoriasis, we will focus the discussion on ultraviolet (UV) light-based therapies, which include narrowband and broadband UVB, UVA in conjunction with photosensitizing agents, targeted UVB treatments such as with an excimer laser, and several other modalities and variations of these core phototherapies, including newer applications of pulsed dye lasers, intense pulse light, and light-emitting electrodes. We will provide an in-depth, evidence-based discussion of efficacy and safety for each treatment modality and provide recommendations and guidance for the use of these therapies alone or in conjunction with other topical and/or systemic psoriasis treatments.


Subject(s)
Dermatology/standards , Phototherapy/standards , Practice Guidelines as Topic , Psoriasis/therapy , Academies and Institutes/standards , Foundations/standards , Humans , Meta-Analysis as Topic , Phototherapy/instrumentation , Phototherapy/methods , Systematic Reviews as Topic , Treatment Outcome , United States
14.
J Am Acad Dermatol ; 80(4): 1073-1113, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30772097
15.
J Am Acad Dermatol ; 80(4): 1029-1072, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30772098

ABSTRACT

Psoriasis is a chronic, inflammatory multisystem disease that affects up to 3.2% of the US population. This guideline addresses important clinical questions that arise in psoriasis management and care, providing recommendations based on the available evidence. The treatment of psoriasis with biologic agents will be reviewed, emphasizing treatment recommendations and the role of the dermatologist in monitoring and educating patients regarding benefits as well as associated risks.


Subject(s)
Biological Products/therapeutic use , Dermatologic Agents/therapeutic use , Psoriasis/drug therapy , Adalimumab/therapeutic use , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Biosimilar Pharmaceuticals/therapeutic use , Certolizumab Pegol/therapeutic use , Drug Therapy, Combination , Etanercept/therapeutic use , Evidence-Based Medicine , Humans , Infliximab/therapeutic use , Piperidines/therapeutic use , Pyrimidines/therapeutic use , Pyrroles/therapeutic use , Ustekinumab/therapeutic use
16.
J Immunol ; 198(2): 950-961, 2017 01 15.
Article in English | MEDLINE | ID: mdl-28003381

ABSTRACT

In animal models, IL-12 and IL-23 participate in the development of malignant neoplasms of keratinocytes. However, the role of these cytokines in pigmented lesion development and their progression to melanoma has received little attention. IL-12p35, IL-23p19, and IL-12/IL-23p40 knockout mice on a C3H/HeN background, subjected to a melanomagenesis protocol, demonstrated profound differences in susceptibility to nevus initiation, transformation, tumorigenicity, and metastatic potential. IL-23 was found to be essential for melanocyte homeostasis, whereas IL-12 supported nevus development. A direct action of IL-23 on primary melanocytes, shown to be IL-23R+, demonstrated that DNA repair of damaged melanocytes requires IL-23. Furthermore, IL-23 modulated the cutaneous microenvironment by limiting regulatory T cells and IFN-γ and inhibiting IL-10 production. Neutralizing Ab to IFN-γ, but not IL-17, inhibited nevus development (p < 0.01).


Subject(s)
DNA Repair/immunology , Interleukin-23/immunology , Melanoma, Experimental/immunology , Skin Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Disease Models, Animal , Flow Cytometry , Interleukin-12 , Melanocytes/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Polymerase Chain Reaction , T-Lymphocyte Subsets/immunology
17.
Photodermatol Photoimmunol Photomed ; 34(1): 50-59, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29044724

ABSTRACT

Skin cancer is the most common type of cancer with increasing incidence rate and public health burden. Solar ultraviolet (UV) radiation causes an array of damaging cellular and molecular events that eventually lead to the development of skin cancer. Despite increased awareness about sun protection, the exposure rate remains high with less than 15% of men and 30% of women using sunscreen on a regular basis. Therefore, there is an imperative need for the development of novel preventive approaches. Skin cancer chemoprevention using phytochemicals either as dietary supplements or by topical applications has gained considerable attention due to their low toxicity, availability, and anticarcinogenic properties. Tea, the second most commonly consumed beverage in the world, is a rich source of promising phytochemicals known as polyphenols. In this review, we discuss the findings of various in vitro, in vivo and human studies signifying the chemopreventive effects of tea polyphenols against UVB-induced skin cancer. This is accomplished by exploring the role of tea polyphenols in DNA repair, inflammation, oxidative stress, signaling pathways, and epigenetics. Finally, this review discusses a variety of innovative delivery methods that enhance the photochemopreventive effects of tea polyphenols against skin cancer.


Subject(s)
DNA Repair/drug effects , Polyphenols/pharmacology , Skin Neoplasms/prevention & control , Tea , Apoptosis/drug effects , Epigenesis, Genetic/drug effects , Humans , Inflammation/prevention & control , Oxidative Stress/drug effects , Polyphenols/therapeutic use , Signal Transduction/drug effects , Skin Neoplasms/etiology , Ultraviolet Rays/adverse effects
18.
Lab Invest ; 97(6): 706-724, 2017 06.
Article in English | MEDLINE | ID: mdl-28218743

ABSTRACT

Ultraviolet B (UVB), in addition to having carcinogenic activity, is required for the production of vitamin D3 (D3) in the skin which supplies >90% of the body's requirement. Vitamin D is activated through hydroxylation by 25-hydroxylases (CYP2R1 or CYP27A1) and 1α-hydroxylase (CYP27B1) to produce 1,25(OH)2D3, or through the action of CYP11A1 to produce mono-di- and trihydroxy-D3 products that can be further modified by CYP27B1, CYP27A1, and CYP24A1. The active forms of D3, in addition to regulating calcium metabolism, exert pleiotropic activities, which include anticarcinogenic and anti-melanoma effects in experimental models, with photoprotection against UVB-induced damage. These diverse effects are mediated through an interaction with the vitamin D receptor (VDR) and/or as most recently demonstrated through action on retinoic acid orphan receptors (ROR)α and RORγ. With respect to melanoma, low levels of 25(OH)D are associated with thicker tumors and reduced patient survival. Furthermore, single-nucleotide polymorphisms of VDR and the vitamin D-binding protein (VDP) genes affect melanomagenesis or disease outcome. Clinicopathological analyses have shown positive correlation between low or undetectable expression of VDR and/or CYP27B1 in melanoma with tumor progression and shorter overall (OS) and disease-free survival (DFS) times. Paradoxically, this correlation was reversed for CYP24A1 (inactivating 24-hydroxylase), indicating that this enzyme, while inactivating 1,25(OH)2D3, can activate other forms of D3 that are products of the non-canonical pathway initiated by CYP11A1. An inverse correlation has been found between the levels of RORα and RORγ expression and melanoma progression and disease outcome. Therefore, we propose that defects in vitamin D signaling including D3 activation/inactivation, and the expression and activity of the corresponding receptors, affect melanoma progression and the outcome of the disease. The existence of multiple bioactive forms of D3 and alternative receptors affecting the behavior of melanoma should be taken into consideration when applying vitamin D management for melanoma therapy.


Subject(s)
Melanoma/metabolism , Receptors, Calcitriol , Vitamin D , Animals , Cytochrome P-450 Enzyme System/metabolism , Disease Progression , Epidermis/metabolism , Epidermis/physiology , Humans , Mice , Receptors, Calcitriol/metabolism , Receptors, Calcitriol/physiology , Signal Transduction/physiology , Ultraviolet Rays , Vitamin D/metabolism , Vitamin D/physiology
19.
Mol Carcinog ; 56(12): 2543-2557, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28574612

ABSTRACT

Basal cell carcinoma (BCC) of the skin is driven by aberrant hedgehog signaling. Thus blocking this signaling pathway by small molecules such as vismodegib inhibits tumor growth. Primary cilium in the epidermal cells plays an integral role in the processing of hedgehog signaling-related proteins. Recent genomic studies point to the involvement of additional genetic mutations that might be associated with the development of BCCs, suggesting significance of other signaling pathways, such as WNT, NOTCH, mTOR, and Hippo, aside from hedgehog in the pathogenesis of this human neoplasm. Some of these pathways could be regulated by noncoding microRNA. Altered microRNA expression profile is recognized with the progression of these lesions. Stopping treatment with Smoothened (SMO) inhibitors often leads to tumor reoccurrence in the patients with basal cell nevus syndrome, who develop 10-100 of BCCs. In addition, the initial effectiveness of these SMO inhibitors is impaired due to the onset of mutations in the drug-binding domain of SMO. These data point to a need to develop strategies to overcome tumor recurrence and resistance and to enhance efficacy by developing novel single agent-based or multiple agents-based combinatorial approaches. Immunotherapy and photodynamic therapy could be additional successful approaches particularly if developed in combination with chemotherapy for inoperable and metastatic BCCs.


Subject(s)
Carcinoma, Basal Cell/genetics , Hedgehog Proteins/genetics , Signal Transduction/genetics , Skin Neoplasms/genetics , Carcinoma, Basal Cell/pathology , Carcinoma, Basal Cell/therapy , Cilia/metabolism , Disease Progression , Hedgehog Proteins/metabolism , Humans , Models, Genetic , Mutation , Skin Neoplasms/pathology , Skin Neoplasms/therapy
20.
Am J Pathol ; 186(10): 2637-49, 2016 10.
Article in English | MEDLINE | ID: mdl-27528504

ABSTRACT

Lewisite is a potent arsenic-based chemical warfare agent known to induce painful cutaneous inflammation and blistering. Only a few modestly effective antidotes have so far been described in the literature. However, the discovery of effective antidotes for lewisite was hampered by the paucity of the exact molecular mechanism underlying its cutaneous pathogenesis. We investigated the molecular mechanism underlying lewisite-induced cutaneous blistering and inflammation and describe its novel antidotes. On the basis of our initial screening, we used a highly sensitive murine model that recapitulates the known human pathogenesis of arsenicals-induced cutaneous inflammation and blistering. Topically administered lewisite induced potent acute inflammation and microvesication in the skin of Ptch1(+/-)/SKH-1 mice. Even at a very low dose, lewisite up-regulates unfolded protein response signaling, inflammatory response, and apoptosis. These cutaneous lesions were associated with production of reactive oxygen species and extensive apoptosis of the epidermal keratinocytes. We confirmed that activation of reactive oxygen species-dependent unfolded protein response signaling is the underlying molecular mechanism of skin damage. Similar alterations were noticed in lewisite-treated cultured human skin keratinocytes. We discovered that chemical chaperone 4-phenyl butyric acid and antioxidant N-acetylcysteine, which significantly attenuate lewisite-mediated skin injury, can serve as potent antidotes. These data reveal a novel molecular mechanism underlying the cutaneous pathogenesis of lewisite-induced lesions. We also identified novel potential therapeutic targets for lewisite-mediated cutaneous injury.


Subject(s)
Antidotes/pharmacology , Antioxidants/pharmacology , Blister/drug therapy , Chemical Warfare Agents/adverse effects , Molecular Chaperones/pharmacology , Patched-1 Receptor/genetics , Acetylcysteine/pharmacology , Animals , Arsenicals/adverse effects , Blister/chemically induced , Blister/pathology , Disease Models, Animal , Female , Humans , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/pathology , Keratinocytes/metabolism , Male , Mice , Mice, Hairless , Mice, Inbred C57BL , Patched-1 Receptor/metabolism , Phenylbutyrates/pharmacology , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL