Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Jan 26.
Article in English | MEDLINE | ID: mdl-38187708

ABSTRACT

The goal of therapeutic cancer vaccines and immune checkpoint therapy (ICT) is to eliminate cancer by expanding and/or sustaining T cells with anti-tumor capabilities. However, whether cancer vaccines and ICT enhance anti-tumor immunity by distinct or overlapping mechanisms remains unclear. Here, we compared effective therapeutic tumor-specific mutant neoantigen (NeoAg) cancer vaccines with anti-CTLA-4 and/or anti-PD-1 ICT in preclinical models. Both NeoAg vaccines and ICT induce expansion of intratumoral NeoAg-specific CD8 T cells, though the degree of expansion and acquisition of effector activity was much more substantial following NeoAg vaccination. Further, we found that NeoAg vaccines are particularly adept at inducing proliferating and stem-like NeoAg-specific CD8 T cells. Single cell T cell receptor (TCR) sequencing revealed that TCR clonotype expansion and diversity of NeoAg-specific CD8 T cells relates to their phenotype and functional state associated with specific immunotherapies employed. Effective NeoAg vaccines and ICT required both CD8 and CD4 T cells. While NeoAg vaccines and anti-PD-1 affected the CD4 T cell compartment, it was to less of an extent than observed with anti-CTLA-4, which notably induced ICOS+Bhlhe40+ Th1-like CD4 T cells and, when combined with anti-PD-1, a small subset of Th2-like CD4 T cells. Although effective NeoAg vaccines or ICT expanded intratumoral M1-like iNOS+ macrophages, NeoAg vaccines expanded rather than suppressed (as observed with ICT) M2-like CX3CR1+CD206+ macrophages, associated with the vaccine adjuvant. Further, combining NeoAg vaccination with ICT induced superior efficacy compared to either therapy in isolation, highlighting the utility of combining these modalities to eliminate cancer.

2.
Cells ; 12(3)2023 01 19.
Article in English | MEDLINE | ID: mdl-36766706

ABSTRACT

Immunotherapy has brought new hope for cancer patients in recent times. However, despite the promising success of immunotherapy, there is still a need to address major challenges including heterogeneity in response among patients, the reoccurrence of the disease, and iRAEs (immune-related adverse effects). The first critical step towards solving these issues is understanding the epigenomic events that play a significant role in the regulation of specific biomolecules in the context of the immune population present in the tumor immune microenvironment (TIME) during various treatments and responses. A prominent advantage of this step is that it would enable researchers to harness the reversibility of epigenetic modifications for their druggability. Therefore, we reviewed the crucial studies in which varying epigenomic events were captured with immuno-oncology set-ups. Finally, we discuss the therapeutic possibilities of their utilization for the betterment of immunotherapy in terms of diagnosis, progression, and cure for cancer patients.


Subject(s)
Epigenomics , Neoplasms , Humans , Neoplasms/therapy , Neoplasms/drug therapy , Immunotherapy/adverse effects , Medical Oncology , Epigenesis, Genetic , Tumor Microenvironment
3.
Cancer Immunol Res ; 10(5): 597-611, 2022 05 03.
Article in English | MEDLINE | ID: mdl-35181783

ABSTRACT

Immune checkpoint therapy (ICT) using antibody blockade of programmed cell death protein 1 (PD-1) or cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) can provoke T cell-dependent antitumor activity that generates durable clinical responses in some patients. The epigenetic and transcriptional features that T cells require for efficacious ICT remain to be fully elucidated. Herein, we report that anti-PD-1 and anti-CTLA-4 ICT induce upregulation of the transcription factor BHLHE40 in tumor antigen-specific CD8+ and CD4+ T cells and that T cells require BHLHE40 for effective ICT in mice bearing immune-edited tumors. Single-cell RNA sequencing of intratumoral immune cells in BHLHE40-deficient mice revealed differential ICT-induced immune cell remodeling. The BHLHE40-dependent gene expression changes indicated dysregulated metabolism, NF-κB signaling, and IFNγ response within certain subpopulations of CD4+ and CD8+ T cells. Intratumoral CD4+ and CD8+ T cells from BHLHE40-deficient mice exhibited higher expression of the inhibitory receptor gene Tigit and displayed alterations in expression of genes encoding chemokines/chemokine receptors and granzyme family members. Mice lacking BHLHE40 had reduced ICT-driven IFNγ production by CD4+ and CD8+ T cells and defects in ICT-induced remodeling of macrophages from a CX3CR1+CD206+ subpopulation to an iNOS+ subpopulation that is typically observed during effective ICT. Although both anti-PD-1 and anti-CTLA-4 ICT in BHLHE40-deficient mice led to the same outcome-tumor outgrowth-several BHLHE40-dependent alterations were specific to the ICT that was used. Our results reveal a crucial role for BHLHE40 in effective ICT and suggest that BHLHE40 may be a predictive or prognostic biomarker for ICT efficacy and a potential therapeutic target.


Subject(s)
Neoplasms , Tumor Microenvironment , Animals , Basic Helix-Loop-Helix Transcription Factors , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Cell Line, Tumor , Granzymes , Homeodomain Proteins , Humans , Interferon-gamma , Mice , Neoplasms/drug therapy
4.
Sci Rep ; 11(1): 11980, 2021 06 07.
Article in English | MEDLINE | ID: mdl-34099789

ABSTRACT

Ultraviolet irradiation induces melanin accumulation, which can be reduced by the use of chemical whitening products. However, the associated safety concerns of such products have prompted the search for natural and harmless alternatives. This study aimed to identify a natural acidic formulation to reduce skin pigmentation. The metabolite propionic acid (CH3CH2COOH, PA) was the most abundant fatty acid in the filtrate from Pluronic F68 (PF68) fermentation of Cutibacterium acnes (C. acnes) and reduced the DOPA-positive melanocytes by significantly inhibiting cellular tyrosinase activity via binding to the free fatty acid receptor 2 (FFAR2). Moreover, 4 mM PA treatment did not alter melanocyte proliferation, indicating that it is an effective solution for hyperpigmentation, causing no cellular damage. The reduced DOPA-positive melanocytes and tyrosinase activity were also observed in mice ear skin tissue injected with a mixture of C. acnes and PF68, supporting that the inhibition of melanogenesis is likely to be mediated through fermentation metabolites from C. acnes fermentation using PF68 as a carbon source. Additionally, PA did not affect the growth of its parent bacteria C. acnes, hence is a potent fermentation metabolite that does not disrupt the balance of the skin microbiome.


Subject(s)
Melanins/chemical synthesis , Propionates/metabolism , Propionibacterium acnes/metabolism , Animals , Cell Proliferation , Ear , Female , Fermentation , Humans , Hyperpigmentation , Melanocytes/cytology , Melanocytes/metabolism , Metabolome , Mice, Inbred ICR , Photochemical Processes , Propionates/chemistry , Receptors, G-Protein-Coupled/radiation effects , Skin , Skin Pigmentation , Ultraviolet Rays
5.
Sci Rep ; 11(1): 12001, 2021 06 07.
Article in English | MEDLINE | ID: mdl-34099817

ABSTRACT

Staphylococcus epidermidis (S. epidermidis) ATCC 12228 was incubated with 2% polyethylene glycol (PEG)-8 Laurate to yield electricity which was measured by a voltage difference between electrodes. Production of electron was validated by a Ferrozine assay. The anti-Cutibacterium acnes (C. acnes) activity of electrogenic S. epidermidis was assessed in vitro and in vivo. The voltage change (~ 4.4 mV) reached a peak 60 min after pipetting S. epidermidis plus 2% PEG-8 Laurate onto anodes. The electricity produced by S. epidermidis caused significant growth attenuation and cell lysis of C. acnes. Intradermal injection of C. acnes and S. epidermidis plus PEG-8 Laurate into the mouse ear considerably suppressed the growth of C. acnes. This suppressive effect was noticeably reversed when cyclophilin A of S. epidermidis was inhibited, indicating the essential role of cyclophilin A in electricity production of S. epidermidis against C. acnes. In summary, we demonstrate for the first time that skin S. epidermidis, in the presence of PEG-8 Laurate, can mediate cyclophilin A to elicit an electrical current that has anti-C. acnes effects. Electricity generated by S. epidermidis may confer immediate innate immunity in acne lesions to rein in the overgrowth of C. acnes at the onset of acne vulgaris.


Subject(s)
Acne Vulgaris/therapy , Antibiosis/genetics , Bacterial Proteins/genetics , Cyclophilin A/genetics , Propionibacteriaceae/pathogenicity , Staphylococcus epidermidis/drug effects , Acne Vulgaris/microbiology , Animals , Bacterial Proteins/metabolism , Coculture Techniques , Culture Media/chemistry , Culture Media/pharmacology , Cyclophilin A/metabolism , Disease Models, Animal , Ear/microbiology , Electricity , Electrodes , Female , Gene Expression , Laurates/pharmacology , Mice , Mice, Inbred ICR , Polyethylene Glycols/pharmacology , Propionibacteriaceae/growth & development , Skin/microbiology , Staphylococcus epidermidis/physiology , Surface-Active Agents/pharmacology
6.
Int J Mol Sci ; 22(3)2021 Jan 31.
Article in English | MEDLINE | ID: mdl-33572500

ABSTRACT

Collagen type I is a key structural component of dermis tissue and is produced by fibroblasts and the extracellular matrix. The skin aging process, which is caused by intrinsic or extrinsic factors, such as natural aging or free radical exposure, greatly reduces collagen expression, thereby leading to obstructed skin elasticity. We investigated the effective fermentation of Cetearyl isononanoate (CIN), a polyethylene glycol (PEG) analog, as a carbon source with the skin probiotic bacterium Staphylococcus epidermidis (S.epidermidis) or butyrate, as their fermentation metabolites could noticeably restore collagen expression through phosphorylated extracellular signal regulated kinase (p-ERK) activation in mouse fibroblast cells and skin. Both the in vitro and in vivo knockdown of short-chain fatty acid (SCFA) or free fatty acid receptor 2 (FFaR2) considerably blocked the probiotic effect of S. epidermidis on p-ERK-induced collagen type I induction. These results demonstrate that butyric acid (BA) in the metabolites of fermenting skin probiotic bacteria mediates FFaR2 to induce the synthesis of collagen through p-ERK activation. We hereby imply that metabolites from the probiotic S. epidermidis fermentation of CIN as a potential carbon source could restore impaired collagen in the dermal extracellular matrix (ECM), providing integrity and elasticity to skin.


Subject(s)
Butyric Acid/metabolism , Collagen Type I/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Probiotics , Receptors, G-Protein-Coupled/metabolism , Skin Aging/drug effects , Staphylococcus epidermidis/physiology , Animals , Collagen Type I/drug effects , Extracellular Matrix/metabolism , Extracellular Signal-Regulated MAP Kinases/genetics , Fatty Acids, Volatile/metabolism , Female , Fermentation , Fibroblasts , MAP Kinase Signaling System/drug effects , Mice , Phosphorylation , Receptors, G-Protein-Coupled/genetics , Skin/metabolism , Skin/microbiology
7.
Sci Rep ; 10(1): 21585, 2020 12 09.
Article in English | MEDLINE | ID: mdl-33299009

ABSTRACT

Repurposing existing compounds for new indications may facilitate the discovery of skin prebiotics which have not been well defined. Four compounds that have been registered by the International Nomenclature of Cosmetic Ingredients (INCI) were included to study their abilities to induce the fermentation of Staphylococcus epidermidis (S. epidermidis), a bacterial species abundant in the human skin. Liquid coco-caprylate/caprate (LCC), originally used as an emollient, effectively initiated the fermentation of S. epidermidis ATCC 12228, produced short-chain fatty acids (SCFAs), and provoked robust electricity. Application of LCC plus electrogenic S. epidermidis ATCC 12228 on mouse skin significantly reduced ultraviolet B (UV-B)-induced injuries which were evaluated by the formation of 4-hydroxynonenal (4-HNE), cyclobutane pyrimidine dimers (CPD), and skin lesions. A S. epidermidis S2 isolate with low expressions of genes encoding pyruvate dehydrogenase (pdh), and phosphate acetyltransferase (pta) was found to be poorly electrogenic. The protective action of electrogenic S. epidermidis against UV-B-induced skin injuries was considerably suppressed when mouse skin was applied with LCC in combination with a poorly electrogenic S. epidermidis S2 isolate. Exploring new indication of LCC for promoting S. epidermidis against UV-B provided an example of repurposing INCI-registered compounds as skin prebiotics.


Subject(s)
Prebiotics/administration & dosage , Probiotics , Skin/drug effects , Staphylococcus epidermidis/drug effects , Aldehydes/metabolism , Animals , Drug Repositioning , Mice , Pyrimidine Dimers/metabolism , Skin/metabolism , Skin/microbiology , Skin/radiation effects , Ultraviolet Rays
8.
Int J Mol Sci ; 21(14)2020 Jul 19.
Article in English | MEDLINE | ID: mdl-32707723

ABSTRACT

The probiotic activity of skin Staphylococcus epidermidis (S. epidermidis) bacteria can elicit diverse biological functions via the fermentation of various carbon sources. Here, we found that polyethylene glycol (PEG)-8 Laurate, a carbon-rich molecule, can selectively induce the fermentation of S. epidermidis, not Cutibacterium acnes (C. acnes), a bacterium associated with acne vulgaris. The PEG-8 Laurate fermentation of S. epidermidis remarkably diminished the growth of C. acnes and the C. acnes-induced production of pro-inflammatory macrophage-inflammatory protein 2 (MIP-2) cytokines in mice. Fermentation media enhanced the anti-C. acnes activity of a low dose (0.1%) clindamycin, a prescription antibiotic commonly used to treat acne vulgaris, in terms of the suppression of C. acnes colonization and MIP-2 production. Furthermore, PEG-8 Laurate fermentation of S. epidermidis boosted the activity of 0.1% clindamycin to reduce the sizes of C. acnes colonies. Our results demonstrated, for the first time, that the PEG-8 Laurate fermentation of S. epidermidis displayed the adjuvant effect on promoting the efficacy of low-dose clindamycin against C. acnes. Targeting C. acnes by lowering the required doses of antibiotics may avoid the risk of creating drug-resistant C. acnes and maintain the bacterial homeostasis in the skin microbiome, leading to a novel modality for the antibiotic treatment of acne vulgaris.


Subject(s)
Clindamycin/administration & dosage , Laurates/metabolism , Polyethylene Glycols/metabolism , Propionibacteriaceae/drug effects , Staphylococcus epidermidis/metabolism , Acne Vulgaris/drug therapy , Acne Vulgaris/microbiology , Animals , Anti-Bacterial Agents/administration & dosage , Fermentation , Gram-Positive Bacterial Infections/drug therapy , Gram-Positive Bacterial Infections/microbiology , Humans , In Vitro Techniques , Mice , Mice, Inbred ICR , Probiotics/metabolism , Propionibacteriaceae/growth & development , Propionibacterium acnes/drug effects , Propionibacterium acnes/growth & development , Skin/drug effects , Skin/metabolism , Skin/microbiology
9.
J Clin Med ; 9(2)2020 Jan 22.
Article in English | MEDLINE | ID: mdl-31979095

ABSTRACT

Pruritus and inflammation associated with accumulation of calcium phosphate (CaP) under the skin are common problems among dialysis patients with chronic kidney disease (CKD). The role of skin commensal microbiota in the CaP-induced uremic pruritus remains uncharacterized. Skin Cutibacterium acnes (C. acnes) can solubilize CaP by the production of short-chain fatty acids (SCFAs), such as butyric acid, through glucose fermentation. Like butyric acid, the N-[2-(2-Butyrylamino-ethoxy)-ethyl]-butyramide (BA-NH-NH-BA), a butyric acid derivative, remarkably induced acetylation of histone H3 lysine 9 (AcH3K9) in keratinocytes. Topical application of fermenting C. acnes, butyric acid or BA-NH-NH-BA onto mouse skin effectively ameliorated CaP-induced skin itching, interleukin (IL)-6 up-regulation in keratinocytes, and extracellular signal-regulated kinase (ERK) 1/2 activation in dorsal root ganglia (DRG). Activation of ERK 1/2 by CaP was markedly reduced in IL-6 knockout mice. Genus Cutibacterium was detected in relatively low abundance in itchy skin of patients with CKD. Our results identify a role for the skin fermenting C. acnes in ameliorating CaP-induced activation of IL-6/p-ERK signaling and resulting skin inflammation. Furthermore, we provide evidence for the potential therapeutic efficacy of BA-NH-NH-BA as a postbiotic for the treatment of uremic pruritus.

10.
Int J Mol Sci ; 20(18)2019 Sep 11.
Article in English | MEDLINE | ID: mdl-31514281

ABSTRACT

The glycerol fermentation of probiotic Staphylococcus epidermidis (S. epidermidis) in the skin microbiome produced butyric acid in vitro at concentrations in the millimolar range. The exposure of dorsal skin of mice to ultraviolet B (UVB) light provoked a significant increased production of pro-inflammatory interleukin (IL)-6 cytokine. Topical application of butyric acid alone or S. epidermidis with glycerol remarkably ameliorated the UVB-induced IL-6 production. In vivo knockdown of short-chain fatty acid receptor 2 (FFAR2) in mouse skin considerably blocked the probiotic effect of S. epidermidis on suppression of UVB-induced IL-6 production. These results demonstrate that butyric acid in the metabolites of fermenting skin probiotic bacteria mediates FFAR2 to modulate the production of pro-inflammatory cytokines induced by UVB.


Subject(s)
Butyric Acid/pharmacology , Interleukin-6/metabolism , Microbiota/drug effects , Probiotics/pharmacology , Receptors, G-Protein-Coupled/metabolism , Skin/microbiology , Staphylococcus epidermidis/chemistry , Ultraviolet Rays , Acetolactate Synthase/metabolism , Animals , Down-Regulation/drug effects , Down-Regulation/radiation effects , Fatty Acids, Volatile/metabolism , Female , Fermentation , Glycerol/pharmacology , Inflammation/pathology , Mice, Inbred ICR , Microbiota/radiation effects , Skin/drug effects , Skin/pathology , Skin/radiation effects
11.
FASEB J ; 33(11): 12036-12046, 2019 11.
Article in English | MEDLINE | ID: mdl-31365830

ABSTRACT

Uremic pruritus with elevated levels of calcium phosphate (CaP) in skin is a common symptom in patients with chronic kidney disease (CKD). In this study, we demonstrate that intradermal injection of CaP into mice triggered scratching by up-regulating the IL-6 in skin and phosphorylation of ERKs in dorsal root ganglion (DRG) in a dose-dependent manner. IL-6 is essential because the CaP-induced up-regulation of phosphorylated (p)-ERK in DRG was considerably reduced in the IL-6 knockout mice. Microarray analysis in conjunction with real-time PCR revealed a higher mRNA expression of Bruton's tyrosine kinase (BTK) gene in DRG after CaP injection. The inhibition of BTK by ibrutinib noticeably diminish the CaP-induced up-regulation of IL-6 and p-ERK in mice. A high amount of IL-6 was detected in itchy skin and blood of patients with CKD. The expressions of p-BTK and p-ERK in DRG primary cells reached maximum levels at 1 and 10 min, respectively, after treatment of recombinant IL-6 and were significantly reduced by treatment of IL-6 along with ibrutinib. The mechanism by which the CaP-induced pruritus mediated by the IL-6/p-BTK/p-ERK signaling was revealed.-Keshari, S., Sipayung, A. D., Hsieh, C.-C., Su, L.-J., Chiang, Y.-R., Chang, H.-C., Yang, W.-C., Chuang, T.-H., Chen, C.-L., Huang, C.-M. IL-6/p-BTK/p-ERK signaling mediates calcium phosphate-induced pruritus.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Ganglia, Spinal/metabolism , Interleukin-6/metabolism , Pruritus/metabolism , Adenine/analogs & derivatives , Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Agammaglobulinaemia Tyrosine Kinase/genetics , Animals , Calcium Phosphates , Extracellular Signal-Regulated MAP Kinases/genetics , Female , Ganglia, Spinal/drug effects , Gene Expression Profiling/methods , Interleukin-6/genetics , Interleukin-6/pharmacology , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation/drug effects , Piperidines , Pruritus/chemically induced , Pruritus/genetics , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Signal Transduction/drug effects , Signal Transduction/genetics , Skin/drug effects , Skin/metabolism , Skin/pathology
12.
Microorganisms ; 7(8)2019 Aug 05.
Article in English | MEDLINE | ID: mdl-31387211

ABSTRACT

Staphylococcus epidermidis (S. epidermidis) is a common bacterial colonizer on the surface of human skin. Lactate is a natural constituent of skin. Here, we reveal that S. epidermidis used sodium l-lactate as a carbon source to undergo fermentation and yield malodors detected by gas colorimetric tubes. Several furan compounds such as furfural originating from the fermentation metabolites play a role in the negative feedback regulation of the fermentation process. The 5-methyl furfural (5MF), a furfural analog, was selected as an inhibitor of sodium l-lactate fermentation of S. epidermidis via inhibition of acetolactate synthase (ALS). S. epidermidis treated with 5MF lost its ability to produce malodors, demonstrating the feasibility of using 5MF as an ingredient in deodorants targeting malodor-causing bacteria in the skin microbiome.

13.
Expert Rev Vaccines ; 18(5): 433-437, 2019 05.
Article in English | MEDLINE | ID: mdl-30920859

ABSTRACT

INTRODUCTION: Acne vulgaris afflicts many people, and despite the multitude of the anti-acne products on the market, there is still no effective treatment that can prevent and cure this disease. The severity of acne vulgaris is highly associated with the inflammatory response to Propionibacterium acnes (P. acnes) now referred to as Cutibacterium acnes (C. acnes), an opportunistic skin bacterium in the human skin microbiome. Areas covered: We here provide the prospects of creating acne vaccines targeting secreted virulence factors of C. acnes including secretory Christie-Atkins-Munch-Peterson (CAMP) factor. Neutralization of secreted virulence factors by either active or passive vaccination may have a lower risk of disturbing the microbial ecosystem in the human skin microbiome. Expert opinion: Major steps could be taken to start a public vaccination program at an early age to prevent the future occurrence of acne vulgaris. Future therapeutic monoclonal antibodies can be designed to specifically neutralize virulence factors of C. acnes including CAMP factors without disrupting the optimal balance of C. acnes in the human skin microbiome and lowering the risk of creating drug-resistant C. acnes. Targeting secreted virulence factors without disturbing the commensal relationship of host can be a novel gateway towards the therapeutic treatment of acne vulgaris.


Subject(s)
Acne Vulgaris/prevention & control , Acne Vulgaris/therapy , Antibodies, Bacterial/immunology , Antigens, Bacterial/immunology , Bacterial Vaccines/immunology , Propionibacterium acnes/immunology , Virulence Factors/immunology , Acne Vulgaris/immunology , Antibodies, Bacterial/administration & dosage , Antibodies, Neutralizing/administration & dosage , Antibodies, Neutralizing/immunology , Bacterial Vaccines/administration & dosage , Humans , Treatment Outcome , Virulence Factors/antagonists & inhibitors
14.
Int J Mol Sci ; 20(1)2018 Dec 20.
Article in English | MEDLINE | ID: mdl-30577530

ABSTRACT

Antibiotics without selectivity for acne treatment may destroy the beneficial microbes in the human microbiome that helps to fight Cutibacterium acnes (C. acnes), a bacterium associated with inflammatory acne vulgaris. Probiotic treatment by direct application of live Staphylococcus epidermidis (S. epidermidis) onto the open acne lesions may run the risk of bloodstream infections. Here, we fabricated the polysulfone microtube array membranes (PSF MTAM) to encapsulate probiotic S. epidermidis. We demonstrate that the application of the encapsulation of S. epidermidis in PSF MTAM enhanced the glycerol fermentation activities of S. epidermidis. To mimic the granulomatous type of acne inflammatory acne vulgaris, the ears of mice were injected intradermally with C. acnes to induce the secretion of macrophage inflammatory protein-2 (MIP-2), a murine counterpart of human interleukin (IL)-8. The C. acnes-injected mouse ears were covered with a PST MTAM encapsulated with or without S. epidermidis in the presence of glycerol. The application of S. epidermidis-encapsulated PST MTAM plus glycerol onto the C. acnes-injected mouse ears considerably reduced the growth of C. acnes and the production of MIP-2. Furthermore, no S. epidermidis leaked from PSF MTAM into mouse skin. The S. epidermidis-encapsulated PST MTAM functions as a probiotic acne patch.


Subject(s)
Antibiosis , Probiotics , Propionibacteriaceae/physiology , Skin/microbiology , Staphylococcus epidermidis/physiology , Animals , Chemokine CXCL2/metabolism , Dermatitis/metabolism , Dermatitis/microbiology , Fermentation , Glycerol/metabolism , Humans , Mice
15.
BMC Cancer ; 17(1): 858, 2017 Dec 15.
Article in English | MEDLINE | ID: mdl-29246124

ABSTRACT

BACKGROUND: Well-known anti-malarial drug artemisinin exhibits potent anti-cancerous activities. In-vivo and in-vitro studies showed its anti-tumor and immunomodulatory properties signifying it as a potent drug candidate for study. The studies of mechanisms of cell movement are relevant which can be understood by knowing the involvement of genes in an effect of a drug. Although cytotoxicity and anti-proliferative activity of artemisinin is evident, the genes participating in its anti-migratory and reduced invasive effect are not well studied. The present study reports the alteration in the expression of 84 genes involved in cell motility upon artemisinin treatment in MCF-7 breast cancer cells using pathway focused gene expression PCR array. In addition, the effect of artemisinin on epigenetic modifier HDACs is studied. METHODS: We checked the functional stimulus of artemisinin on cell viability, migration, invasion and apoptosis in breast cancerous cell lines. Using qRT-PCR and western blot, we validated the altered expression of relevant genes associated with proliferation, migration, invasion, apoptosis and mammary gland development. RESULTS: Artemisinin inhibited cell proliferation of estrogen receptor negative breast cancer cells with fewer efficacies in comparison to estrogen receptor positive ones. At the same time, cell viability and proliferation of normal breast epithelial MCF10A cells was un-affected. Artemisinin strongly inhibited cancer cell migration and invasion. Along with orphan nuclear receptors (ERRα, ERRß and ERRγ), artemisinin altered the ERα/ERß/PR/Her expression status of MCF-7 cells. The expression of genes involved in the signaling pathways associated with proliferation, migration, invasion and apoptosis was significantly altered which cooperatively resulted into reduced growth promoting activities of breast cancer cells. Interestingly, artemisinin exhibited inhibitory effect on histone deacetylases (HDACs). CONCLUSIONS: Upregulated expression of tumor suppressor genes along with reduced expression of oncogenes significantly associated with growth stimulating signaling pathways in response to artemisinin treatment suggests its efficacy as an effective drug in breast cancer treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Artemisinins/pharmacology , Breast Neoplasms/drug therapy , Cell Movement/drug effects , Histone Deacetylase Inhibitors/pharmacology , Transcriptome/drug effects , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Artemisinins/therapeutic use , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Survival/drug effects , Epigenesis, Genetic/drug effects , Female , Gene Expression Profiling , Genes, Tumor Suppressor/drug effects , Histone Deacetylase Inhibitors/therapeutic use , Histone Deacetylases/metabolism , Humans , MCF-7 Cells , Neoplasm Invasiveness/prevention & control , Oncogenes/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...