1
|
Edres HA, Elmassry IH, Lebda MA, Othman SI, El-Karim DRSG, Rudayni HA, Ebied SKM, Allam AA, Hashem AE. Berberine and Cyperus rotundus extract nanoformulations protect the rats against Staphylococcus-induced mastitis via antioxidant and anti-inflammatory activities: role of MAPK signaling. Cell Biochem Biophys 2025; 83:2167-2183. [PMID: 39707026 DOI: 10.1007/s12013-024-01628-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/14/2024] [Indexed: 12/23/2024]
Abstract
Berberine (BER) and Cyperus rotundus rhizomes extract (CRE) are phytochemicals characterized by broad-spectrum pharmacological activity that could tackle the side effects of conventional mastitis therapies, however, they undergo a modest bioavailability. In the current study, nanoformulations of BER and CRE chitosan hydrogel (BER/CH-NPs, CRE/CH-NPs) were investigated for their antibacterial, antioxidant, anti-inflammatory and anti-apoptotic effects against S. aureus-induced mastitis in a rat model. The experiment was conducted on 80 early lactating female albino rats allocated into 6 groups; control, mastitis, BER/CH-NPs (1 and 0.5 mg), CRE/CH-NPs (0.5 and 0.25 mg), BER/CH-NPs + CRE/CH-NPs (0.5 + 0.25 and 0.25 + 0.125 mg). The nanoparticles were given by oral gavage once every other day from day 2 to day 12 after parturition. On the 13th day, intra-mammary inoculation with 100 µl of S. aureus suspension containing 2.1 × 108 CFU/ml in all groups except the control group. The results expressed the effect of BER/CH-NPs and CRE/CH-NPs on mammary gland tissue including significantly diminished viable bacterial load as well as attenuated the levels of MPO, MDA, caspase-3 with elevating Nrf2 level, and modulating glutathione redox. Also, the nanoformulations resulted in attenuation of the mRNA expression of TLR2, NOD2, Keap-1 and MAPK signaling pathway additional to the immune reactivity of NF-κB P65 and p-ERK as well as the preservation of the regular alveolar architecture. The supplementation of the berberine and Cyperus rotundus extract nanoformulations could be a prospective protective approach against Staphylococcal mastitis via their antibacterial, antioxidant, antiapoptotic, anti-inflammatory and modulation of MAPK signaling pathway.
Collapse
Affiliation(s)
- Hanan A Edres
- Department of Biochemistry, Faculty of Veterinary Medicine, Alexandria University, Alexandria, 21944, Egypt
| | - Ingi H Elmassry
- Department of Biochemistry, Faculty of Veterinary Medicine, Alexandria University, Alexandria, 21944, Egypt
| | - Mohamed A Lebda
- Department of Biochemistry, Faculty of Veterinary Medicine, Alexandria University, Alexandria, 21944, Egypt.
| | - Sarah I Othman
- Department of Biology, college of Science, Princess Nourah bint Abdulrahman University, P.O. BOX 84428, Riyadh, 11671, Saudi Arabia
| | - Dina R S Gad El-Karim
- Department of Pathology and Clinical Pathology, Faculty of Veterinary Medicine, Alexandria University, Alexandria, 21944, Egypt
| | - Hassan A Rudayni
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University, Riyadh, 11623, Saudi Arabia
| | - Sawsan Kh M Ebied
- Bacteriology Unit, Animal Health Research Institute, Alexandria Province, Alexandria, 21944, Egypt
| | - Ahmed A Allam
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University, Riyadh, 11623, Saudi Arabia
- Department of Zoology, Faculty of Science, Beni-suef University, Beni-suef, 65211, Egypt
| | - Aml E Hashem
- Department of Biochemistry, Faculty of Veterinary Medicine, Alexandria University, Alexandria, 21944, Egypt
| |
Collapse
|
2
|
Yuan B, Yu J, Dong J, Mao Z, Wang X. Bacteria in hypertrophic scars promote scar formation through HSBP1-mediated autophagy. Wound Repair Regen 2025; 33:e13253. [PMID: 39823159 PMCID: PMC11740274 DOI: 10.1111/wrr.13253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 12/11/2024] [Accepted: 01/02/2025] [Indexed: 01/19/2025]
Abstract
Bacterial colonisation in hypertrophic scars (HSs) has been reported, yet the precise mechanism of their contribution to scar formation remains elusive. To address this, we examined HS and normal skin (NS) tissues through Gram staining and immunofluorescence. We co-cultured fibroblasts with heat-inactivated Staphylococcus aureus (S. aureus) and evaluated their levels of apoptosis and proliferation by flow cytometry and Cell Counting Kit-8 assay, respectively. Additionally, we performed proteomic analysis and western blotting to identify upregulated proteins. To assess autophagy levels, we examined light chain 3 (LC3) expression through western blotting and immunofluorescence, and transmission electron microscopy (TEM) was performed to detect autophagy-associated vesicles. Our results demonstrated a notable increase in bacterial load, primarily S. aureus, in HS tissues. Furthermore, S. aureus promoted fibroblast proliferation and enhanced the expression of profibrotic markers such as transforming growth factor β1 (TGF-β1), vascular endothelial growth factor (VEGF), collagen I, collagen III and α smooth muscle actin (α-SMA). Proteomic analysis highlighted heat shock factor-binding protein 1 (HSBP1) as a key upregulated protein mediating the profibrotic effects induced by S. aureus. Knockdown of HSBP1 reversed these effects. Intriguingly, HSBP1 also upregulated LC3 and Beclin-1 expression and increased the number of autophagosomes in fibroblasts. Finally, when fibroblasts stimulated by S. aureus were treated with HSBP1 siRNA, autophagy levels decreased significantly. Collectively, our findings suggest that S. aureus, via HSBP1, stimulates fibroblast proliferation and promotes their transition into myofibroblasts, triggering autophagy and fibrosis. These results underscore the potential of HSBP1 as a therapeutic target for the management of HSs.
Collapse
Affiliation(s)
- Bo Yuan
- Department of Burn, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Jiarong Yu
- Department of Burn, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Jiaoyun Dong
- Department of Burn, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Zhigang Mao
- Department of Plastic Surgery, Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xiqiao Wang
- Department of Burn, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
3
|
Arsaute S, Reinoso EB, Cecchini ME, Moliva MV, Montironi ID, Cariddi LN. Minthostachys verticillata essential oil modulates cytokine synthesis and Staphylococcus aureus internalization in MAC-T cells at least through TLR4/MyD88/NFkB pathway. Vet Res Commun 2024; 48:3727-3742. [PMID: 39249174 DOI: 10.1007/s11259-024-10526-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 08/30/2024] [Indexed: 09/10/2024]
Abstract
The aim of this study was to evaluate the activation pathway(s) triggered by Minthostachys verticillata essential oil (EO) in bovine mammary epithelial cells (MAC-T) challenged with a strain of bovine Staphylococcus aureus. MAC-T cells were stimulated with EO, S. aureus or pre-treated with EO and then challenged with S. aureus. Cytokine's release was measured by ELISA. The mRNA for TLR2, TLR4, NOD2, MyD88 and NFκB was quantified by RT-qPCR. S. aureus adherence and internalization was also evaluated. MAC-T cells stimulated with S. aureus synthesized high levels of IL-1ß and IL-6 were kept up to 48 h, while IL-4 levels were not altered. Cells pre-treated with EO for 2 and 6 h and then challenged with S. aureus showed a significant increase of IL-1β and IL-6. However, in these cells, a decrease in IL-1ß and IL-6 levels and an increase of IL-4 values was observed from 24 h. No significant increase in the expression levels of TLR2 or NOD2 was detected in all stimulated cells. However, the expression of TLR4, MyD88 and NFκB was increased in cells stimulated with S. aureus at 2 and 6 h as well as in cells pre-treated with EO between 2 and 6 h and then challenged with S. aureus. The NFκB expression levels was similar to control at 24 h in all stimulated cells, although pro-inflammatory cytokine levels and TLR4 and MyD88 expression levels remained high in cells stimulated with S. aureus. This results suggested the activation of other pathways independent of MyD88 by the pathogen that involucrated the activation of others transcription factors. Pre-treatment with EO during 2, 6 and 24 h did not affect S. aureus adherence but decreased its internalization. In conclusion, pre-treatment with EO increased the IL-1β and IL-6 synthesis during the first hours post-challenged with S. aureus up-regulating TLR4/MyD88/NFκB pathway. Furthermore, EO increased the IL-4 levels from 6 to 24 h down-regulating the NFκB and possibly other transcription factors activated by the pathogen, which decreased its internalization into MAC-T cells.
Collapse
Affiliation(s)
- Sofía Arsaute
- Facultad de Ciencias Exactas Físico-Químicas y Naturales, Departamento de Microbiología e Inmunología, Universidad Nacional de Río Cuarto, Río Cuarto, Córdoba, 5800, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Biotecnología Ambiental y Salud (INBIAS), Ruta 36 Km 601, Río Cuarto, Córdoba, 5800, Argentina
| | - Elina Beatriz Reinoso
- Facultad de Ciencias Exactas Físico-Químicas y Naturales, Departamento de Microbiología e Inmunología, Universidad Nacional de Río Cuarto, Río Cuarto, Córdoba, 5800, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Biotecnología Ambiental y Salud (INBIAS), Ruta 36 Km 601, Río Cuarto, Córdoba, 5800, Argentina
| | - María Eugenia Cecchini
- Facultad de Ciencias Exactas Físico-Químicas y Naturales, Departamento de Microbiología e Inmunología, Universidad Nacional de Río Cuarto, Río Cuarto, Córdoba, 5800, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Biotecnología Ambiental y Salud (INBIAS), Ruta 36 Km 601, Río Cuarto, Córdoba, 5800, Argentina
| | - Melina Vanesa Moliva
- Facultad de Ciencias Exactas Físico-Químicas y Naturales, Departamento de Microbiología e Inmunología, Universidad Nacional de Río Cuarto, Río Cuarto, Córdoba, 5800, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Biotecnología Ambiental y Salud (INBIAS), Ruta 36 Km 601, Río Cuarto, Córdoba, 5800, Argentina
| | - Ivana Dalila Montironi
- Facultad de Ciencias Exactas Físico-Químicas y Naturales, Departamento de Microbiología e Inmunología, Universidad Nacional de Río Cuarto, Río Cuarto, Córdoba, 5800, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Biotecnología Ambiental y Salud (INBIAS), Ruta 36 Km 601, Río Cuarto, Córdoba, 5800, Argentina
| | - Laura Noelia Cariddi
- Facultad de Ciencias Exactas Físico-Químicas y Naturales, Departamento de Microbiología e Inmunología, Universidad Nacional de Río Cuarto, Río Cuarto, Córdoba, 5800, Argentina.
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Biotecnología Ambiental y Salud (INBIAS), Ruta 36 Km 601, Río Cuarto, Córdoba, 5800, Argentina.
| |
Collapse
|
4
|
Khan MZ, Li L, Wang T, Liu X, Chen W, Ma Q, Zahoor M, Wang C. Bioactive Compounds and Probiotics Mitigate Mastitis by Targeting NF-κB Signaling Pathway. Biomolecules 2024; 14:1011. [PMID: 39199398 PMCID: PMC11352841 DOI: 10.3390/biom14081011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 08/10/2024] [Accepted: 08/12/2024] [Indexed: 09/01/2024] Open
Abstract
Mastitis is a significant inflammatory condition of the mammary gland in dairy cows. It is caused by bacterial infections and leads to substantial economic losses worldwide. The disease can be either clinical or sub-clinical and presents challenges such as reduced milk yield, increased treatment costs, and the need to cull affected cows. The pathogenic mechanisms of mastitis involve the activation of Toll-like receptors (TLRs), specifically TLR2 and TLR4. These receptors play crucial roles in recognizing pathogen-associated molecular patterns (PAMPs) and initiating immune responses through the NF-κB signaling pathway. Recent in vitro studies have emphasized the importance of the TLR2/TLR4/NF-κB signaling pathway in the development of mastitis, suggesting its potential as a therapeutic target. This review summarizes recent research on the role of the TLR2/TLR4/NF-κB signaling pathway in mastitis. It focuses on how the activation of TLRs leads to the production of proinflammatory cytokines, which, in turn, exacerbate the inflammatory response by activating the NF-κB signaling pathway in mammary gland tissues. Additionally, the review discusses various bioactive compounds and probiotics that have been identified as potential therapeutic agents for preventing and treating mastitis by targeting TLR2/TLR4/NF-κB signaling pathway. Overall, this review highlights the significance of targeting the TLR2/TLR4/NF-κB signaling pathway to develop effective therapeutic strategies against mastitis, which can enhance dairy cow health and reduce economic losses in the dairy industry.
Collapse
Affiliation(s)
- Muhammad Zahoor Khan
- Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, Liaocheng University, Liaocheng 522000, China
| | - Liangliang Li
- Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, Liaocheng University, Liaocheng 522000, China
| | - Tongtong Wang
- Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, Liaocheng University, Liaocheng 522000, China
| | - Xiaotong Liu
- Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, Liaocheng University, Liaocheng 522000, China
| | - Wenting Chen
- Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, Liaocheng University, Liaocheng 522000, China
| | - Qingshan Ma
- Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, Liaocheng University, Liaocheng 522000, China
| | - Muhammad Zahoor
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Sognsvannsveien, 90372 Oslo, Norway
| | - Changfa Wang
- Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, Liaocheng University, Liaocheng 522000, China
| |
Collapse
|
5
|
Yang Y, Yuan T, Wu R, Geng Z, Lian S, Wang J. The effect of bta-miR-1296 on the proliferation and extracellular matrix synthesis of bovine mammary fibroblasts. In Vitro Cell Dev Biol Anim 2024; 60:183-194. [PMID: 38409638 DOI: 10.1007/s11626-024-00851-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Accepted: 12/11/2023] [Indexed: 02/28/2024]
Abstract
Mammary fibrosis in dairy cows is a chronic condition caused by mastitis, and can lead to serious culling of dairy cows resulting in huge economic losses in the dairy industry. MicroRNAs (miRNAs) exert an important role in regulating mammary gland health in dairy cows. This study investigated whether exosomal miRNAs in mammary epithelial cells can regulate the proliferation of bovine mammary fibroblasts (BMFBs) in mastitis. Liposome transfection technology was used to construct a cellular model of the overexpression and inhibition of miRNAs. The STarMir software, dual luciferase reporter gene test, real-time quantitative PCR (qRT-PCR), a Cell Counting Kit-8 (CCK-8), and a Western Blot and plate clone formation test were used to investigate the mechanism by which bta-miR-1296 regulates the proliferation of BMFBs. Target gene prediction results revealed that glutamate-ammonia ligase was a direct target gene by which bta-miR-1296 regulates cell proliferation. It was found that bta-miR-1296 significantly inhibited the proliferation of BMFBs. After BMFBs were transfected with a bta-miR-1296 mimic, mRNA expression in the extracellular matrix (ECM), α-smooth muscle actin (α-SMA), collagen type I alpha 1 chain (COL1α1) and collagen type III alpha 1 chain (COL3α1), and various cell growth factors (basic fibroblast growth factor (bFGF), vascular endothelial growth factor (VEGF), platelet-derived growth factor-BB (PDGF-BB), and transforming growth factor-β1 (TGF-β1)) were down-regulated, and the expressions of α-SMA, COL1α1, COL3α1, phospho-extracellular regulated protein kinases, phospho-protein kinase B, TGF-β1, and phospho-Smad family member3 proteins were inhibited. In conclusion, bta-miR-1296 can inhibit the proliferation of BMFBs and the synthesis of ECM in BMFBs, thus affecting the occurrence and development of mammary fibrosis in dairy cows and laying the foundation for further studies to clarify the regulatory mechanism of mammary fibrosis.
Collapse
Affiliation(s)
- Yuejie Yang
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural Affairs, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Heilongjiang Province, Daqing, 163319, People's Republic of China
| | - Tao Yuan
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural Affairs, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Heilongjiang Province, Daqing, 163319, People's Republic of China
| | - Rui Wu
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural Affairs, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Heilongjiang Province, Daqing, 163319, People's Republic of China
| | - Zijian Geng
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural Affairs, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Heilongjiang Province, Daqing, 163319, People's Republic of China
| | - Shuai Lian
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural Affairs, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Heilongjiang Province, Daqing, 163319, People's Republic of China.
| | - Jianfa Wang
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural Affairs, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Heilongjiang Province, Daqing, 163319, People's Republic of China.
- Veterinary Medicine Faculty, Heilongjiang Bayi Agricultural University, No. 2 Xinyang Road, Daqing, People's Republic of China.
| |
Collapse
|
6
|
Yao L, Liu Q, Lei Z, Sun T. Development and challenges of antimicrobial peptide delivery strategies in bacterial therapy: A review. Int J Biol Macromol 2023; 253:126819. [PMID: 37709236 DOI: 10.1016/j.ijbiomac.2023.126819] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 09/06/2023] [Accepted: 09/07/2023] [Indexed: 09/16/2023]
Abstract
The escalating global prevalence of antimicrobial resistance poses a critical threat, prompting concerns about its impact on public health. This predicament is exacerbated by the acute shortage of novel antimicrobial agents, a scarcity attributed to the rapid surge in bacterial resistance. This review delves into the realm of antimicrobial peptides, a diverse class of compounds ubiquitously present in plants and animals across various natural organisms. Renowned for their intrinsic antibacterial activity, these peptides provide a promising avenue to tackle the intricate challenge of bacterial resistance. However, the clinical utility of peptide-based drugs is hindered by limited bioavailability and susceptibility to rapid degradation, constraining efforts to enhance the efficacy of bacterial infection treatments. The emergence of nanocarriers marks a transformative approach poised to revolutionize peptide delivery strategies. This review elucidates a promising framework involving nanocarriers within the realm of antimicrobial peptides. This paradigm enables meticulous and controlled peptide release at infection sites by detecting dynamic shifts in microenvironmental factors, including pH, ROS, GSH, and reactive enzymes. Furthermore, a glimpse into the future reveals the potential of targeted delivery mechanisms, harnessing inflammatory responses and intricate signaling pathways, including adenosine triphosphate, macrophage receptors, and pathogenic nucleic acid entities. This approach holds promise in fortifying immunity, thereby amplifying the potency of peptide-based treatments. In summary, this review spotlights peptide nanosystems as prospective solutions for combating bacterial infections. By bridging antimicrobial peptides with advanced nanomedicine, a new therapeutic era emerges, poised to confront the formidable challenge of antimicrobial resistance head-on.
Collapse
Affiliation(s)
- Longfukang Yao
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China; Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China
| | - Qianying Liu
- School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhixin Lei
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China; Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China.
| | - Taolei Sun
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China; Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China.
| |
Collapse
|
7
|
Wei M, Tang W, Lv D, Liu M, Wang G, Liu Q, Qin L, Huang B, Zhang D. Long-chain noncoding RNA sequencing analysis reveals the molecular profiles of chemically induced mammary epithelial cells. Front Genet 2023; 14:1189487. [PMID: 37745843 PMCID: PMC10514351 DOI: 10.3389/fgene.2023.1189487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 08/21/2023] [Indexed: 09/26/2023] Open
Abstract
Long noncoding RNAs (lncRNAs) were important regulators affecting the cellular reprogramming process. Previous studies from our group have demonstrated that small molecule compounds can induce goat ear fibroblasts to reprogram into mammary epithelial cells with lactation function. In this study, we used lncRNA-Sequencing (lncRNA-seq) to analyze the lncRNA expression profile of cells before and after reprogramming (CK vs. 5i8 d). The results showed that a total of 3,970 candidate differential lncRNAs were detected, 1,170 annotated and 2,800 new lncRNAs. Compared to 0 d cells, 738 lncRNAs were significantly upregulated and 550 were significantly downregulated in 8 d cells. Heat maps of lncrnas and target genes with significant differences showed that the fate of cell lineages changed. Functional enrichment analysis revealed that these differently expressed (DE) lncRNAs target genes were mainly involved in signaling pathways related to reprogramming and mammary gland development, such as the Wnt signaling pathway, PI3K-Akt signaling pathway, arginine and proline metabolism, ECM-receptor interaction, and MAPK signaling pathway. The accuracy of sequencing was verified by real-time fluorescence quantification (RT-qPCR) of lncRNAs and key candidate genes, and it was also demonstrated that the phenotype and genes of the cells were changed. Therefore, this study offers a foundation for explaining the molecular mechanisms of lncRNAs in chemically induced mammary epithelial cells.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Ben Huang
- Guangxi Key Laboratory of Eye Health, Guangxi Academy of Medical Sciences, People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Dandan Zhang
- Guangxi Key Laboratory of Eye Health, Guangxi Academy of Medical Sciences, People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| |
Collapse
|
8
|
Yu J, Mao Z, Zhou Z, Yuan B, Wang X. Microbiome dysbiosis occurred in hypertrophic scars is dominated by S. aureus colonization. Front Immunol 2023; 14:1227024. [PMID: 37701435 PMCID: PMC10494536 DOI: 10.3389/fimmu.2023.1227024] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 08/01/2023] [Indexed: 09/14/2023] Open
Abstract
Background The mechanisms of hypertrophic scar formation and its tissue inflammation remain unknown. Methods We collected 33 hypertrophic scar (HS) and 36 normal skin (NS) tissues, and detected the tissue inflammation and bacteria using HE staining, Gram staining, and transmission electronic microscopy (TEM), in situ hybridization and immunohistochemistry for MCP-1, TNF-α, IL-6 and IL-8. In addition, the samples were assayed by 16S rRNA sequencing to investigate the microbiota diversity in HS, and the correlation between the microbiota and the indices of Vancouver Scar Scale(VSS)score. Results HE staining showed that a dramatically increased number of inflammatory cells accumulated in HS compared with NS, and an enhanced number of bacteria colonies was found in HS by Gram staining, even individual bacteria could be clearly observed by TEM. In situ hybridization demonstrated that the bacteria and inflammation cells co-localized in the HS tissues, and immunohistochemistry indicated the expression of MCP-1, TNF-α, IL-6, and IL-8 were significantly upregulated in HS than that in NS. In addition, there was a significantly different microbiota composition between HS and NS. At the phylum level, Firmicutes was significantly higher in HS than NS. At the genus level, S. aureus was the dominant species, which was significantly higher in HS than NS, and was strongly correlated with VSS indices. Conclusion Microbiome dysbiosis, dominated by S. aureus, occurred in HS formation, which is correlated with chronic inflammation and scar formation, targeting the microbiome dysbiosis is perhaps a supplementary way for future scar management.
Collapse
Affiliation(s)
- Jiarong Yu
- The Department of Burn, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhigang Mao
- The Department of Plastic Surgery, Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zengding Zhou
- The Department of Burn, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Bo Yuan
- The Department of Burn, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiqiao Wang
- The Department of Burn, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
9
|
N6-Methyladenosine Modification Profile in Bovine Mammary Epithelial Cells Treated with Heat-Inactivated Staphylococcus aureus. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1704172. [PMID: 35251466 PMCID: PMC8890870 DOI: 10.1155/2022/1704172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 01/04/2022] [Accepted: 02/01/2022] [Indexed: 11/18/2022]
Abstract
The symptoms of mastitis caused by Staphylococcus aureus (S. aureus) in dairy cows are not obvious and difficult to identify, resulting in major economic losses. N6-Methyladenosine (m6A) modification has been reported to be closely associated with the occurrence of many diseases. However, only a few reports have described the role of m6A modification in S. aureus-induced mastitis. In this study, after 24 h of treatment with inactivated S. aureus, MAC-T cells (an immortalized bovine mammary epithelial cell line) showed increased expression levels of the inflammatory factors IL-1β, IL-6, TNF-α, and reactive oxygen species. We found that the mRNA levels of METLL3, METLL14, WTAP, and ALKBH5 were also upregulated. Methylated RNA immunoprecipitation sequencing analysis revealed that 133 genes were m6A hypermethylated, and 711 genes were m6A hypomethylated. Biological functional analysis revealed that the differential m6A methylated genes were mainly related to oxidative stress, lipid metabolism, inflammatory response, and so on. In the present study, we also identified 62 genes with significant changes in m6A modification and mRNA expression levels. These findings elucidated the m6A modification spectrum induced by S. aureus in MAC-T cells and provide the basis for subsequent m6A research on mastitis.
Collapse
|
10
|
Ling H, Luo L, Dai X, Chen H. Fallopian tubal infertility: the result of Chlamydia trachomatis-induced fallopian tubal fibrosis. Mol Cell Biochem 2021; 477:205-212. [PMID: 34652537 DOI: 10.1007/s11010-021-04270-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 09/29/2021] [Indexed: 11/26/2022]
Abstract
Chlamydia trachomatis is one of the most common pathogens of sexually transmitted diseases, and its incidence in genital tract infections is now 4.7% in south China. Infertility is the end result of C. trachomatis-induced fallopian tubal fibrosis and is receiving intense attention from scientists worldwide. To reduce the incidence of infertility, it is important to understand the pathology-related changes of the genital tract where C. trachomatis infection is significant, especially the mechanism of fibrosis formation. During fibrosis development, the fallopian tube becomes sticky and occluded, which will eventually lead to tubal infertility. At present, the mechanism of fallopian tubal fibrosis induced by C. trachomatis infection is unclear. Our study attempted to summarize the possible mechanisms of fibrosis caused by C. trachomatis infection in the fallopian tube by reviewing published studies and further providing potential therapeutic targets to reduce the occurrence of infertility. This study also provides ideas for future research. Factors leading to fallopian tube fibrosis include inflammatory factors, miRNA, ECT, cHSP, and host factors. We hypothesized that C. trachomatis mediates the transcription and translation of EMT and ECM via upregulating TGF signaling pathway, which leads to the formation of fallopian tube fibrosis and ultimately to tubal infertility.
Collapse
Affiliation(s)
- Hua Ling
- The First People's Hospital of Chenzhou, The First School of Clinical Medicine, Southern Medical University, Chenzhou, 423000, People's Republic of China
| | - Lipei Luo
- The First People's Hospital of Chenzhou, The First School of Clinical Medicine, Southern Medical University, Chenzhou, 423000, People's Republic of China
| | - Xingui Dai
- The First People's Hospital of Chenzhou, The First School of Clinical Medicine, Southern Medical University, Chenzhou, 423000, People's Republic of China.
- The First People's Hospital of Chenzhou, Chenzhou, 423000, People's Republic of China.
- The First Affiliated Hospital of Xiangnan University, Chenzhou, 423000, People's Republic of China.
| | - Hongliang Chen
- The First People's Hospital of Chenzhou, The First School of Clinical Medicine, Southern Medical University, Chenzhou, 423000, People's Republic of China.
- The First People's Hospital of Chenzhou, Chenzhou, 423000, People's Republic of China.
- The First Affiliated Hospital of Xiangnan University, Chenzhou, 423000, People's Republic of China.
| |
Collapse
|