scholarly journals Suppressive Effect of Two Cucurbitane-Type Triterpenoids from Momordica charantia on Cutibacterium acnes-Induced Inflammatory Responses in Human THP-1 Monocytic Cell and Mouse Models

Molecules ◽  
2021 ◽  
Vol 26 (3) ◽  
pp. 579
Author(s):  
Lu-Te Chuang ◽  
Wen-Cheng Huang ◽  
Yu-Chen Hou ◽  
Jong-Ho Chyuan ◽  
Hsiang Chang ◽  
...  

Cutibacterium acnes (formerly Propionibacterium acnes) is one of the major bacterial species responsible for acne vulgaris. Numerous bioactive compounds from Momordica charantia Linn. var. abbreviata Ser. have been isolated and examined for many years. In this study, we evaluated the suppressive effect of two cucurbitane-type triterpenoids, 5β,19-epoxycucurbita-6,23-dien-3β,19,25-triol (Kuguacin R; KR) and 3β,7β,25-trihydroxycucurbita-5,23-dien-19-al (TCD) on live C. acnes-stimulated in vitro and in vivo inflammatory responses. Using human THP-1 monocytes, KR or TCD suppressed C. acnes-induced production of interleukin (IL)-1β, IL-6 and IL-8 at least above 56% or 45%, as well as gene expression of these three pro-inflammatory cytokines. However, a significantly strong inhibitory effect on production and expression of tumor necrosis factor (TNF)-α was not observed. Both cucurbitanes inhibited C. acnes-induced activation of the myeloid differentiation primary response 88 (MyD88) (up to 62%) and mitogen-activated protein kinases (MAPK) (at least 36%). Furthermore, TCD suppressed the expression of pro-caspase-1 and cleaved caspase-1 (p10). In a separate study, KR or TCD decreased C. acnes-stimulated mouse ear edema by ear thickness (20% or 14%), and reduced IL-1β-expressing leukocytes and neutrophils in mouse ears. We demonstrated that KR and TCD are potential anti-inflammatory agents for modulating C. acnes-induced inflammation in vitro and in vivo.

2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Shinta Marito ◽  
Sunita Keshari ◽  
Supitchaya Traisaeng ◽  
Do Thi Tra My ◽  
Arun Balasubramaniam ◽  
...  

AbstractStaphylococcus 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.


2021 ◽  
Author(s):  
Shinta Marito ◽  
Sunita Keshari ◽  
Supitchaya Traisaeng ◽  
Do Thi Tra My ◽  
Arun Balasubramaniam ◽  
...  

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 minutes 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.


2021 ◽  
Vol 9 (7) ◽  
pp. 1486
Author(s):  
Marcela Espinoza-Monje ◽  
Jorge Campos ◽  
Eduardo Alvarez Villamil ◽  
Alonso Jerez ◽  
Stefania Dentice Maidana ◽  
...  

Previously, we isolated lactic acid bacteria from the slime of the garden snail Helix aspersa Müller and selected Weissella viridescens UCO-SMC3 because of its ability to inhibit in vitro the growth of the skin-associated pathogen Cutibacterium acnes. The present study aimed to characterize the antimicrobial and immunomodulatory properties of W. viridescens UCO-SMC3 and to demonstrate its beneficial effect in the treatment of acne vulgaris. Our in vitro studies showed that the UCO-SMC3 strain resists adverse gastrointestinal conditions, inhibits the growth of clinical isolates of C. acnes, and reduces the adhesion of the pathogen to keratinocytes. Furthermore, in vivo studies in a mice model of C. acnes infection demonstrated that W. viridescens UCO-SMC3 beneficially modulates the immune response against the skin pathogen. Both the oral and topical administration of the UCO-SCM3 strain was capable of reducing the replication of C. acnes in skin lesions and beneficially modulating the inflammatory response. Of note, orally administered W. viridescens UCO-SMC3 induced more remarkable changes in the immune response to C. acnes than the topical treatment. However, the topical administration of W. viridescens UCO-SMC3 was more efficient than the oral treatment to reduce pathogen bacterial loads in the skin, and effects probably related to its ability to inhibit and antagonize the adhesion of C. acnes. Furthermore, a pilot study in acne volunteers demonstrated the capacity of a facial cream containing the UCO-SMC3 strain to reduce acne lesions. The results presented here encourage further mechanistic and clinical investigations to characterize W. viridescens UCO-SMC3 as a probiotic for acne vulgaris treatment.


Nutrients ◽  
2020 ◽  
Vol 12 (9) ◽  
pp. 2620
Author(s):  
Mi-Jin Yim ◽  
Jeong Min Lee ◽  
Hyun-Soo Kim ◽  
Grace Choi ◽  
Young-Mog Kim ◽  
...  

Acne vulgaris is a chronic inflammatory condition of skin sebaceous follicles. To explore its effects on acne vulgaris, we investigated the antibacterial and anti-inflammatory activities of Sargassum miyabei Yendo (a brown alga) ethanolic extract (SMYEE) on Cutibacterium acnes (C. acnes)-stimulated inflammatory responses, both in vivo and in vitro. To induce inflammation in vivo, C. acnes was intradermally injected into the dorsal skin of mice, to which SMYEE was applied. The antimicrobial activity of SMYEE was evaluated by the determination of minimum inhibitory concentrations (MICs). To explore in vitro anti-inflammatory effects, HaCaT cells were stimulated with C. acnes after treatment with SMYEE. The levels of IL-8 and the underlying molecular effects in C. acnes-stimulated HaCaT cells were assessed by enzyme-linked immunosorbent assay, Western blotting, and an electrophoretic mobility shift assay. Mouse skin lesions improved after treatment with SMYEE (50 μg/mouse). Neutrophil infiltration was significantly reduced in SMYEE-treated compared to SMYEE-untreated skin lesions. SMYEE reversed the C. acnes-induced increase in IL-8 levels in HaCaT cells and suppressed dHL-60 cell migration. SMYEE also inhibited C. acnes-induced phosphorylation of the extracellular signal-regulated kinase and inhibited activator protein-1 signaling. SMYEE may be a useful treatment for C. acnes-induced acne vulgaris.


Author(s):  
Vicky Bronnec ◽  
Hinnerk Eilers ◽  
Anika C. Jahns ◽  
Hélène Omer ◽  
Oleg A. Alexeyev

Acne vulgaris is the most common dermatological disorder worldwide affecting more than 80% of adolescents and young adults with a global prevalence of 231 million cases in 2019. The involvement of the skin microbiome disbalance in the pathophysiology of acne is recognized, especially regarding the relative abundance and diversity of Propionibacterium acnes a well-known dominant human skin commensal. Biofilms, where bacteria are embedded into a protective polymeric extracellular matrix, are the most prevalent life style for microorganisms. P. acnes and its biofilm-forming ability is believed to be a contributing factor in the development of acne vulgaris, the persistence of the opportunistic pathogen and antibiotic therapy failures. Degradation of the extracellular matrix is one of the strategies used by bacteria to disperse the biofilm of competitors. In this study, we report the identification of an endogenous extracellular nuclease, BmdE, secreted by Propionibacterium granulosum able to degrade P. acnes biofilm both in vivo and in vitro. This, to our knowledge, may represent a novel competitive mechanism between two closely related species in the skin. Antibiotics targeting P. acnes have been the mainstay in acne treatment. Extensive and long-term use of antibiotics has led to the selection and spread of resistant bacteria. The extracellular DNase BmdE may represent a new bio-therapeutical strategy to combat P. acnes biofilm in acne vulgaris.


Molecules ◽  
2020 ◽  
Vol 25 (18) ◽  
pp. 4277
Author(s):  
Lu-Te Chuang ◽  
Ya-Hsin Shih ◽  
Wen-Cheng Huang ◽  
Lie-Chwen Lin ◽  
Chin Hsu ◽  
...  

Cutibacterium acnes (formerly Propionibacterium acnes) is a key pathogen involved in the development and progression of acne inflammation. The numerous bioactive properties of wild bitter melon (WBM) leaf extract and their medicinal applications have been recognized for many years. In this study, we examined the suppressive effect of a methanolic extract (ME) of WBM leaf and fractionated components thereof on live C. acnes-induced in vitro and in vivo inflammation. Following methanol extraction of WBM leaves, we confirmed anti-inflammatory properties of ME in C. acnes-treated human THP-1 monocyte and mouse ear edema models. Using a bioassay-monitored isolation approach and a combination of liquid–liquid extraction and column chromatography, the ME was then separated into n-hexane, ethyl acetate, n-butanol and water-soluble fractions. The hexane fraction exerted the most potent anti-inflammatory effect, suppressing C. acnes-induced interleukin-8 (IL-8) production by 36%. The ethanol-soluble fraction (ESF), which was separated from the n-hexane fraction, significantly inhibited C. acnes-induced activation of mitogen-activated protein kinase (MAPK)-mediated cellular IL-8 production. Similarly, the ESF protected against C. acnes-stimulated mouse ear swelling, as measured by ear thickness (20%) and biopsy weight (23%). Twenty-four compounds in the ESF were identified using gas chromatograph–mass spectrum (GC/MS) analysis. Using co-cultures of C. acnes and THP-1 cells, β-ionone, a compound of the ESF, reduced the production of IL-1β and IL-8 up to 40% and 18%, respectively. β-ionone also reduced epidermal microabscess, neutrophilic infiltration and IL-1β expression in mouse ear. We also found evidence of the presence of anti-inflammatory substances in an unfractionated phenolic extract of WBM leaf, and demonstrated that the ESF is a potential anti-inflammatory agent for modulating in vitro and in vivo C. acnes-induced inflammatory responses.


Pathogens ◽  
2021 ◽  
Vol 10 (2) ◽  
pp. 120
Author(s):  
Jialu Ma ◽  
Shasha Zhao ◽  
Xiao Gao ◽  
Rui Wang ◽  
Juan Liu ◽  
...  

Mycobacterium tuberculosis (MTB) infection is characterized by granulomatous lung lesions and systemic inflammatory responses during active disease. Inflammasome activation is involved in regulation of inflammation. Inflammasomes are multiprotein complexes serving a platform for activation of caspase-1, which cleaves the proinflammatory cytokines such as interleukin-1β (IL-1β) and IL-18 into their active forms. These cytokines play an essential role in MTB control. MTB infection triggers activation of the nucleotide-binding domain, leucine-rich-repeat containing family, pyrin domain-containing 3 (NLRP3) and absent in melanoma 2 (AIM2) inflammasomes in vitro, but only AIM2 and apoptosis-associated speck-like protein containing a caspase-activation recruitment domain (ASC), rather than NLRP3 or caspase-1, favor host survival and restriction of mycobacterial replication in vivo. Interferons (IFNs) inhibits MTB-induced inflammasome activation and IL-1 signaling. In this review, we focus on activation and regulation of the NLRP3 and AIM2 inflammasomes after exposure to MTB, as well as the effect of inflammasome activation on host defense against the infection.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 1256-1256 ◽  
Author(s):  
Angelica A. Silveira ◽  
Clare Cunningham ◽  
Emma Corr ◽  
Wilson Alves Ferreira ◽  
Fernando F. Costa ◽  
...  

Abstract Intravascular hemolysis results in the release of damaging hemoglobin and free heme into the circulation. A role for heme as a danger associated molecular pattern (DAMP), with a function in sterile inflammatory responses, is becoming increasingly recognized. Whilst heme has known effects on leukocytes, activating their migration, adhesion molecule expression and cytokine expression, more recent data demonstrate that this molecule can induce NLRP3 inflammasome formation in murine bone marrow macrophages, with consequent interleukin (IL)-1β processing and neutrophil recruitment (Dutra et al., Proc. Natl Acad Sci. 111: E4110, 2014). We aimed to investigate whether heme can also induce inflammasome activation in primary human macrophages (hMACs) and to further characterize the pathways by which heme-induced inflammatory responses may be amplified under sterile conditions. CD14+ cells were separated from human peripheral blood (using anti-CD14 magnetic beads) and differentiated into hMACs under M-CSF media supplementation and in the presence of 10% fetal bovine serum. In vitro results are expressed as means ± SEM for triplicate cultures and are representative of three independent experiments. Priming of hMACs with lipopolysaccharide (LPS; 100 ng/mL; 3h) alone induced low level secretion of IL-1β (14.11±9.2 pg/106 cells, as measured by ELISA), while heme (50 µM), in the absence of pre-stimulation with LPS, was unable to induce significant IL-1β secretion within 3h (2.46±1.4 pg/106 cells). In contrast, co-incubation of hMACs with both LPS and heme for 3h significantly enhanced hMAC IL-1β release (490.3±36.3 pg/106 cells; P<0.05 compared to LPS alone). The inflamassome pathway inhibitors, MCC950 (5 µM; a specific inhibitor of NLRP3) and YVAD (40 µM; a caspase-1 inhibitor) significantly inhibited IL-1β secretion in LPS-primed hMACs stimulated with heme (reduced to 35.12±3.9; 184±30.4 pg/106 cells, respectively; 3h; P<0.05 compared to LPS/heme). Co-incubation of the LPS-primed cells with varying concentrations of heme, under the conditions employed, did not induce TNF-α production (data not shown), consistent with the hypothesis that IL-1β processing in heme-induced LPS-primed hMAC was mediated by inflammasome formation. Interestingly, qPCR showed that incubation of hMACs (1x106 cells/mL) with heme (50 µM) for 24h stimulated an approximately 10-fold increase (P<0.01) in the expression of the gene encoding, S100A8, another DAMP known to act as a TLR-4 agonist and to contribute to ischemia/reperfusion injury. Priming of hMACs with 1 µg/ml recombinant S100A8 for 3h and subsequent activation with heme (50 or 100 µM, 14h) significantly augmented the release of IL-1β (42.1±0.4 and 89.4±32.4 pg/106 cells for 50 and 100 µM heme, respectively; P<0.05), compared with S100A8 alone (20.6±3.5 pg/106 cells), without any modulation in TNF-α secretion (P>0.05). Using a model of acute intravascular hemolysis, we confirmed an association between heme release and S100A8 secretion, in vivo. Plasma heme levels increased significantly from 26.3±5 µM (i.v. saline control; N=4) to 87±18 µM in C57BL/6 mice at 1h after receiving i.v. water (150 µl; N=4, P=0.04). A concomitant increase in plasma S100A8 levels was also observed within 1h of the hemolytic stimulus (986±102 pg/mL, compared to 694.2±102 pg/ml in control mice; N=4, P=0.05), which was maintained for 3h (P<0.05). Thus, we present data to demonstrate that heme can induce IL-1β processing in LPS-primed human macrophages under in vitro conditions, probably via formation of the NLRP3/caspase-1 inflammasome machinery. In the absence of LPS, heme-stimulated hMACs can express the S100A8 DAMP; furthermore, a hemolytic stimulus induced mouse S100A8 production in vivo. As such, S100A8 may amplify heme-dependent inflammasome formation in an autocrine fashion, even under sterile conditions. Data provide new insights into the mechanisms by which heme may induce and potentiate inflammatory responses in hemolytic diseases, such as sickle cell disease, and suggest S100A8, together with heme, as potential therapeutic targets for reducing inflammation in these diseases. Disclosures Ferreira: Bayer AG: Research Funding. Almeida:Jassen & Cilag: Other: Currently employed with. Conran:Bayer AG: Research Funding.


Nanomaterials ◽  
2020 ◽  
Vol 10 (6) ◽  
pp. 1095
Author(s):  
Kuang-Hung Hsiao ◽  
Chun-Ming Huang ◽  
Yu-Hsiang Lee

Acne vulgaris is one of the most prevalent dermatological diseases among adolescents and is often associated with overgrowth of Cutibacterium acnes (C. acnes) in the pilosebaceous units. In this study, we aimed to develop novel rifampicin (RIF) and indocyanine green (ICG) co-loaded perfluorocarbon nanodroplets named RIPNDs which can simultaneously provide photo-, chemo-, and probiotic-antimicrobility, and explore their efficacy in treatment of C. acnes in vitro and in vivo. The RIPNDs were first characterized as being spherical in shape, with a size of 238.6 ± 7.51 nm and surface charge of −22.3 ± 3.5 mV. Then, the optimal dosages of Staphylococcus epidermidis–produced fermentation product medium (FPM) and RIPND were determined as 25% (v/v) and [RIF]/[ICG] = 3.8/20 μM, respectively, based on the analyses of inhibition zone and cytotoxicity in vitro. Through the in vivo study using C. acnes–inoculated mice, our data showed that the group treated with FPM followed by RIPNDs + near infrared (NIR) irradiation obtained the least granulocytes/macrophage-inflammatory protein 2 expression level in the epidermis, and showed a significantly lower microbial colony population compared to the groups treated with equal amount of RIF, FPM, RIPNDs, and/or combination of the above ± NIR. These results indicated that the RIPND-mediated photo–chemo–probiotic therapeutics was indeed able to rapidly suppress inflammatory response of the skin and provide a robust antibacterial effect against C. acnes with limited use of antibiotics. Taken altogether, we anticipate that the RIPND is highly potential for use in the clinical treatment of acne vulgaris.


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Tae-Hyun Kim ◽  
Kyungwon Yang ◽  
Minsuk Kim ◽  
Hee-Sun Kim ◽  
Jihee Lee Kang

AbstractApoptosis inhibitor of macrophage (AIM) modulates the signaling in inflammatory responses, including infection, cancer, or other immune diseases. Recent studies suggest that like interleukin-10 (IL-10), AIM is involved in alternatively activated (M2) macrophage polarization. We aimed to understand whether and how AIM is involved in IL-10-induced inhibition of inflammasome activation and resolution of inflammation. First, we demonstrated that IL-10 induced increases in mRNA and protein expression of AIM in murine bone marrow-derived macrophages (BMDM). In addition, genetic and pharmacologic inhibition of STAT3 (signal transducer and activator of transcription 3) reduced IL-10-induced AIM expression. We also found that IL-10-induced STAT3 activity enhanced the AIM promoter activity by directly binding the promoter of the AIM gene. Additionally, reduction of LPS/adenosine triphosphate (ATP)-induced IL-1β production and caspase-1 activation by IL-10 was reversed in BMDM from AIM−/− mice. Treatment of BMDM from both wild type (WT) and IL-10−/− mice with recombinant AIM showed the inhibitory effects on IL-1β and IL-18 production and caspase-1 activation. Endogenous and exogenous AIM inhibited apoptosis-associated speck-like protein containing a caspase activation and recruitment domain (ASC) speck formation. In LPS-induced acute peritonitis, inhibition of IL-1β and IL-18 production in peritoneal lavage fluid (PLF) and serum, reduction of caspase-1 activation in peritoneal macrophages, and reduction of numbers of neutrophils and peritoneal macrophages in PLF by administration of IL-10 were not evident in AIM−/− mice. Our in vitro and in vivo data reveal a novel role of AIM in the inhibition of inflammasome-mediated caspase-1 activation and IL-1β and IL-18 production.


Sign in / Sign up

Export Citation Format

Share Document