1
|
Louisa M, Wanafri E, Arozal W, Sandhiutami NMD, Basalamah AM. Nanocurcumin preserves kidney function and haematology parameters in DMBA-induced ovarian cancer treated with cisplatin via its antioxidative and anti-inflammatory effect in rats. Pharm Biol 2023; 61:298-305. [PMID: 36708211 PMCID: PMC9888479 DOI: 10.1080/13880209.2023.2166965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 11/25/2022] [Accepted: 01/05/2023] [Indexed: 06/18/2023]
Abstract
CONTEXT Cisplatin, as a first-line treatment for ovarian cancer, is associated with debilitating adverse effects, including nephrotoxic and haematotoxic effects. OBJECTIVE This study determines whether nanocurcumin, combined with cisplatin, would give additional benefit to kidney function and haematological parameters in rats with ovarian cancer. MATERIALS AND METHODS Twenty-five Wistar rats were divided into five untreated rats and 20-dimethylbenz(a)anthracene (DMBA)-induced ovarian cancer rats. The 20 ovarian cancer rats were divided into four treatment groups: vehicle, cisplatin, cisplatin-curcumin, and cisplatin-nanocurcumin. Cisplatin was given at the dose of 4 mg/kg BW once weekly, while curcumin or nanocurcumin was administered at 100 mg/kg BW daily for four weeks. At the end of treatment, we analysed kidney function, haematological parameters, and inflammatory and oxidative stress markers from plasma. RESULTS Nanocurcumin alleviates the increase in kidney function markers and abnormalities in haematological indices in rats treated with cisplatin. Compared to cisplatin-treated rats, plasma urea levels decreased from 66.4 to 47.7 mg/dL, creatinine levels lowered from 0.87 to 0.82 mg/dL, and neutrophil gelatinase-associated lipocalin (NGAL) levels declined from 8.51 to 3.59 mIU/mg protein. Furthermore, the therapy increased glutathione activities (from 2.02 to 3.23 U/µL), reduced lipid peroxidation (from 0.54 to 0.45 nmol/mL), and decreased plasma TNF-α (from 270.6 to 217.8 pg/mL). CONCLUSIONS Cisplatin with nanocurcumin in an ovarian cancer rat model may provide additional benefits as a preventive agent against renal impairment and cisplatin-induced haematological toxicity. However, further research is required to prove that using nanocurcumin for a more extended time would not affect its anticancer properties.
Collapse
Affiliation(s)
- Melva Louisa
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Erico Wanafri
- Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Wawaimuli Arozal
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | | | | |
Collapse
|
2
|
Li C, Gong L, Jiang Y, Huo X, Huang L, Lei H, Gu Y, Wang D, Guo D, Deng Y. Sanguisorba officinalis ethyl acetate extract attenuates ulcerative colitis through inhibiting PI3K-AKT/NF-κB/ STAT3 pathway uncovered by single-cell RNA sequencing. Phytomedicine 2023; 120:155052. [PMID: 37717310 DOI: 10.1016/j.phymed.2023.155052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 07/17/2023] [Accepted: 08/25/2023] [Indexed: 09/19/2023]
Abstract
BACKGROUND Ulcerative colitis (UC) accounts for the untreatable illness nowadays. Bloody stools are the primary symptom of UC, and the first-line drugs used to treat UC are associated with several drawbacks and negative side effects. S. officinalis has long been used as a medicine to treat intestinal infections and bloody stools. However, what the precise molecular mechanism, the exact etiology, and the material basis of the disease remain unclear. PURPOSE This work aimed to comprehensively explore pharmacological effects as well as molecular mechanisms underlying the active fraction of S. officinalis, and to produce a comprehensive and brand-new guideline map of its chemical base and mechanism of action. METHODS First, different polarity S. officinalis extracts were orally administered to the DSS-induced UC model mice for the sake of investigating its active constituents. Using the UPLC-orbitrap high-resolution mass spectrometry (UPLC-Q-Orbitrap-HRMS) technique, the most active S. officinalis (S. officinalis ethyl acetate fraction, SOEA) extract was characterized. Subsequently, the effectiveness of its active fraction on UC was evaluated through phenotypic observation (such as weight loss, colon length, and stool characteristics), and histological examination of pathological injuries, mRNA and protein expression. Cell profile, cell-cell interactions and molecular mechanisms of SOEA in different cell types of the colon tissue from UC mice were described using single-cell RNA sequencing (scRNA-seq). As a final step, the molecular mechanisms were validated by appropriate molecular biological methods. RESULTS For the first time, this study revealed the significant efficacy of SOEA in the treatment of UC. SOEA reduced DAI and body weight loss, recovered the colon length, and mitigated colonic pathological injuries along with mucosal barrier by promoting goblet cell proliferation. Following treatment with SOEA, inflammatory factors showed decreased mRNA and protein expression. SOEA restored the dynamic equilibrium of cell profile and cell-cell interactions in colon tissue. All of these results were attributed to the ability of SOEA to inhibit the PI3K-AKT/NF-κB/STATAT pathway. CONCLUSIONS By integrating the chemical information of SOEA derived from UPLC-Q-Orbitrap-HRMS with single-cell transcriptomic data extracted from scRNA-seq, this study demonstrates that SOEA exerts the therapeutic effect through suppressing PI3K-AKT/NF-B/STAT3 pathway to improve clinical symptoms, inflammatory response, mucosal barrier, and intercellular interactions in UC, and effectively eliminates the interference of cellular heterogeneity.
Collapse
Affiliation(s)
- Congcong Li
- State Key Laboratory of Southwestern Chinese Medicine Resource, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Leiqiang Gong
- State Key Laboratory of Southwestern Chinese Medicine Resource, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yu Jiang
- Department of Nursing, Sichuan Nursing Vocational College, Deyang 618000, China
| | - Xueyan Huo
- State Key Laboratory of Southwestern Chinese Medicine Resource, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Lijun Huang
- State Key Laboratory of Southwestern Chinese Medicine Resource, School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Haoran Lei
- State Key Laboratory of Southwestern Chinese Medicine Resource, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yucheng Gu
- Syngenta Limited, Jealott's Hill International Research Centre, Berkshire RG42 6EY, UK
| | - Dong Wang
- State Key Laboratory of Southwestern Chinese Medicine Resource, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Dale Guo
- State Key Laboratory of Southwestern Chinese Medicine Resource, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Yun Deng
- State Key Laboratory of Southwestern Chinese Medicine Resource, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
3
|
Doğan GT, Kepekçi RA, Bostancıeri N, Tarakçıoğlu M. Protective effect of Arum maculatum against dextran sulfate sodium induced colitis in rats. Biotech Histochem 2023; 98:456-465. [PMID: 37394993 DOI: 10.1080/10520295.2023.2225226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/04/2023] Open
Abstract
Ulcerative colitis (UC) is an inflammatory disease of the large intestine that is characterized by diarrhea, bloody stools, abdominal pain and mucosal ulceration. UC is treated with nonsteroidal anti-inflammatory drugs, corticosteroids or immunosuppressants, but long-term use of these drugs can cause adverse effects. Arum maculatum is used as a traditional treatment for digestive system disorders, but its use for treatment of UC has not been investigated rigorously. We investigated the possible protective effect of a methanol extract of A. maculatum against dextran sulfate sodium (DSS) induced experimental UC in rats. Total phenolic and flavonoid contents of the extract were 32.919 ± 1.125 mg gallic acid equivalent (GAE)/g and 52.045 ± 7.902 µg rutin equivalent (RE)/mg, respectively. The half-maximal inhibitory concentration (IC50) for the extract was 105.76 µg/ml according to the 1,1-diphenyl-2-picrylhydrazyl (DPPH) radical scavenging activity assay. Effects of A. maculatum extract on UC induced by DSS were assessed both macroscopically and histologically. We also investigated effects of A. maculatum extract on malondialdehyde (MDA) levels and the oxidative stress index (OSI) in normal rats and rats with UC. We found that treatment with A. maculatum extract protected the colon against DSS induced UC in a dose-dependent manner.
Collapse
Affiliation(s)
- Gülsüm Toparlı Doğan
- Biochemistry Science and Technology Department, Gaziantep University, Gaziantep, Turkey
| | | | - Nuray Bostancıeri
- Histology and Embryology Department, Gaziantep University, Gaziantep, Turkey
| | - Mehmet Tarakçıoğlu
- Department of Biochemistry and Clinical Biochemistry, Gaziantep University, Gaziantep, Turkey
| |
Collapse
|
4
|
Dil E, Topcu A, Mercantepe T, Tumkaya L, Akyildiz K, Saral S, Yilmaz A. Agomelatine on cisplatin-induced nephrotoxicity via oxidative stress and apoptosis. Naunyn Schmiedebergs Arch Pharmacol 2023; 396:2753-2764. [PMID: 37480488 DOI: 10.1007/s00210-023-02632-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 07/14/2023] [Indexed: 07/24/2023]
Abstract
Drug-induced nephrotoxicity is the greatest deterrent to the use of cisplatin, which is a frequently used chemotherapeutic with proven effectiveness in cancer therapy. Agomelatine, which is used in the treatment of sleep disorders and depression, has gained attention in recent years with its antioxidative and anti-inflammatory effects. In this study, the effects of the synthetic melatonin agonist agomelatine on nephrotoxicity were investigated in a rat model of cisplatin-induced nephrotoxicity using biochemical, histological, and immunohistochemical methods. Thirty-two male rats were divided into 4 groups: 1. control group, 2. agomelatine group, 3. cisplatin group, 4. cisplatin + agomelatine group. In the cisplatin group, there were widespread atypical glomerular structures and vacuolization in tubular epithelial cells, necrotic tubules, deterioration of brush border structure in proximal tubules, and fibrotic areas characterized by diffuse polymorphonuclear leukocyte (PNL) and extensive collagen deposition in the interstitial spaces. However, in the cisplatin + agomelatine group, we observed a reduction in glomeruli of atypical structure and necrotic tubules, in PNL infiltration in interstitial spaces, and fibrotic areas compared to the cisplatin group. The cisplatin + agomelatine group showed lower malondialdehyde (MDA) serum creatinine, serum urea levels, and higher glutathione (GSH) levels compared to the cisplatin group. Immunohistochemical analyses revealed that the elevated NF-kβ/p65, 8-OHdG, and cleaved caspase-3 positivity in the cisplatin group had significantly decreased in the cisplatin + agomelatine group. In conclusion, agomelatine showed a nephroprotective effect against cisplatin-induced nephrotoxicity.
Collapse
Affiliation(s)
- Eyup Dil
- Department of Urology Faculty of Medicine, Recep Tayyip Erdogan University, 2 Nolu Sehitler Street, Rize, 53010, Turkey.
| | - Atilla Topcu
- Department Pharmacology, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, 53010, Turkey
| | - Tolga Mercantepe
- Histology and Embryology, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, 53010, Turkey
| | - Levent Tumkaya
- Histology and Embryology, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, 53010, Turkey
| | - Kerimali Akyildiz
- Biochemistry, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, 53010, Turkey
| | - Sinan Saral
- Faculty of Medicine, Recep Tayyip Erdogan University, Rize, 53010, Turkey
| | - Adnan Yilmaz
- Biochemistry, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, 53010, Turkey
| |
Collapse
|
5
|
Vlk AM, Prantner D, Shirey KA, Perkins DJ, Buzza MS, Thumbigere-Math V, Keegan AD, Vogel SN. M2a macrophages facilitate resolution of chemically-induced colitis in TLR4-SNP mice. mBio 2023:e0120823. [PMID: 37768050 DOI: 10.1128/mbio.01208-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 07/18/2023] [Indexed: 09/29/2023] Open
Abstract
Toll-like receptor 4 (TLR4) is an innate immune receptor responsive to lipopolysaccharide (LPS). Single nucleotide polymorphisms (SNPs) in human TLR4 that encode an A896G transition at SNP rs4986790 (D299G) and a C1196T transition at SNP rs4986791 (T399I) render individuals hyporesponsive to LPS. In humans, these SNPs are also associated with increased susceptibility to inflammatory bowel diseases (IBDs). Using knock-in mice engineered to express the murine homologs of these human TLR4 mutations ("TLR4-SNP" mice), we have shown that TLR4-SNP mice develop significantly more severe colitis induced by dextran sodium sulfate (DSS) than wild-type (WT) mice, similar to IBD in humans expressing these SNPs. Previous studies have provided indirect evidence for "tissue repair" M2 macrophages (Mφ) in the resolution of colitis. Signaling through the IL-4/IL-13 receptor, IL-4Rα, and the transcription factor, peroxisome proliferator-activated receptor (PPARγ), have been shown to be required for induction of M2a Mφ, and our data provide direct evidence for the involvement of both in the repair of DSS-induced colonic damage. In response to DSS, colons of TLR4-SNP mice produced reduced levels of M2a Mφ marker mRNA and protein, including PPARγ, and therapeutic administration of the PPARγ agonist ligand, rosiglitazone, resolved colitis in TLR4-SNP mice, and increased expression of the M2a protein, Ym1. Together, these data indicate that the failure of TLR4-SNP mice to resolve DSS-induced colitis may be secondary to their failure to induce "tissue repair" M2a Mφ. IMPORTANCE Inflammatory bowel disease (IBD), including Crohn's disease and ulcerative colitis, impacts millions of individuals worldwide and severely impairs the quality of life for patients. Dysregulation of innate immune signaling pathways reduces barrier function and exacerbates disease progression. Macrophage (Mφ) signaling pathways are potential targets for IBD therapies. While multiple treatments are available for IBD, (i) not all patients respond, (ii) responses may diminish over time, and (iii) treatments often have undesirable side effects. Genetic studies have shown that the inheritance of two co-segregating SNPs expressed in the innate immune receptor, TLR4, is associated with human IBD. Mice expressing homologous SNPs ("TLR4-SNP" mice) exhibited more severe colitis than WT mice in a DSS-induced colonic inflammation/repair model. We identified a critical role for M2a "tissue repair" Mφ in the resolution of colitis. Our findings provide insight into potential development of novel therapies targeting Mφ signaling pathways that aim to alleviate the debilitating symptoms experienced by individuals with IBD.
Collapse
Affiliation(s)
- Alexandra M Vlk
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Daniel Prantner
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Kari Ann Shirey
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Darren J Perkins
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- University of Maryland Marlene & Stewart Greenebaum Comprehensive Cancer Center, Baltimore, Maryland, USA
| | - Marguerite S Buzza
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Vivek Thumbigere-Math
- Division of Periodontics, University of Maryland School of Dentistry, Baltimore, Maryland, USA
| | - Achsah D Keegan
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Stefanie N Vogel
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- University of Maryland Marlene & Stewart Greenebaum Comprehensive Cancer Center, Baltimore, Maryland, USA
| |
Collapse
|
6
|
Lian X, Wang Y, Wang S, Peng X, Wang Y, Huang Y, Chen W. Does inflammatory bowel disease promote kidney diseases: a mendelian randomization study with populations of European ancestry. BMC Med Genomics 2023; 16:225. [PMID: 37752523 PMCID: PMC10521387 DOI: 10.1186/s12920-023-01644-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 08/23/2023] [Indexed: 09/28/2023] Open
Abstract
BACKGROUND This study aimed to investigate a causal relationship between IBD and multiple kidney diseases using two-sample Mendelian randomization (MR) analyses. METHODS We selected a group of single nucleotide polymorphisms (SNPs) specific to IBD as instrumental variables from a published genome-wide association study (GWAS) with 86,640 individuals of European ancestry. Summary statistics for multiple kidney diseases were obtained from the publicly available GWAS. Genetic data from one GWAS involving 210 extensive T-cell traits was used to estimate the mediating effect on specific kidney disease. Inverse-variance weighted method were used to evaluate the MR estimates for primary analysis. RESULTS Genetic predisposition to IBD was associated with higher risk of IgA nephropathy (IgAN) (OR, 1.78; 95% CI, 1.45-2.19), but not membranous nephropathy, diabetic nephropathy, glomerulonephritis, nephrotic syndrome, chronic kidney disease, and urolithiasis. CD4 expression on CD4 + T cell had a significant genetic association with the risk of IgAN (OR, 2.72; 95% CI, 1.10-6.72). Additionally, consistent results were also observed when IBD was subclassified as ulcerative colitis (OR, 1.38; 95% CI, 1.10-1.71) and Crohn's disease (OR, 1.37; 95% CI, 1.12-1.68). MR-PRESSO and the MR-Egger intercept did not identify pleiotropic SNPs. CONCLUSIONS This study provides genetic evidence supporting a positive casual association between IBD, including its subclassification as ulcerative colitis and Crohn's disease, and the risk of IgAN. However, no casual association was found between IBD and other types of kidney diseases. Further exploration of IBD interventions as potential preventive measures for IgAN is warranted.
Collapse
Affiliation(s)
- Xingji Lian
- Department of Nephrology, The First Affiliated Hospital, NHC Key Laboratory of Nephrology (Sun Yat-sen University), Guangdong Provincial Key Laboratory of Nephrology, Sun Yat-sen University, Guangzhou, 510080 China
- Department of Geriatrics, Guangzhou First People’s Hospital, School of Medicine, National Key Clinic Specialty, South China University of Technology, Guangzhou, 510180 China
| | - Yiqin Wang
- Department of Nephrology, The First Affiliated Hospital, NHC Key Laboratory of Nephrology (Sun Yat-sen University), Guangdong Provincial Key Laboratory of Nephrology, Sun Yat-sen University, Guangzhou, 510080 China
| | - Shuyi Wang
- Department of Rheumatology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080 China
| | - Xiaohui Peng
- Department of Geriatrics, Guangzhou First People’s Hospital, School of Medicine, National Key Clinic Specialty, South China University of Technology, Guangzhou, 510180 China
| | - Yanhui Wang
- Department of Geriatrics, Guangzhou First People’s Hospital, School of Medicine, National Key Clinic Specialty, South China University of Technology, Guangzhou, 510180 China
| | - Yuyu Huang
- Department of Geriatrics, Guangzhou First People’s Hospital, School of Medicine, National Key Clinic Specialty, South China University of Technology, Guangzhou, 510180 China
| | - Wei Chen
- Department of Nephrology, The First Affiliated Hospital, NHC Key Laboratory of Nephrology (Sun Yat-sen University), Guangdong Provincial Key Laboratory of Nephrology, Sun Yat-sen University, Guangzhou, 510080 China
| |
Collapse
|
7
|
Zanardi KR, Grancieri M, Silva CW, Trivillin LO, Viana ML, Costa AGV, Costa NMB. Functional effects of yacon ( Smallanthus sonchifolius) and kefir on systemic inflammation, antioxidant activity, and intestinal microbiome in rats with induced colorectal cancer. Food Funct 2023. [PMID: 37740322 DOI: 10.1039/d3fo02599c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/24/2023]
Abstract
Colorectal cancer (CRC) is one of the most common cancers with high morbidity and mortality. The modulation of intestinal health through the administration of pro- and prebiotics may be a viable alternative to reduce the risk of CRC. This study aimed to evaluate the functional effects of yacon and kefir, isolated or associated, in rats with colorectal cancer. Adult Wistar rats were divided into five groups (n = 8): HC (healthy control AIN-93M diet), CC (CCR + AIN-93M diet), Y (CCR + AIN-93 M + yacon diet), K (CCR + AIN-93-M + kefir diet) and YK (CCR + AIN-93 M + yacon + kefir diet). Colorectal carcinogenesis was induced in groups CC, Y, K, and YK with 1,2-dimethylhydrazine (55 mg kg-1, subcutaneously) for 5 weeks. From the 6th week onwards, the experimental groups were fed the respective diets. In the 15th week, urine was collected for analysis of intestinal permeability and then the animals were euthanized. Yacon increased acetate levels, reduced pH and carcinogenic neoplastic lesions, and increased the abundance of bacteria related to the fermentation of non-digestible carbohydrates, such as the genera Dorea, Collinsela, and Bifidobacteria. On the other hand, kefir increased macroscopic neoplastic lesions and increased the abundance of Firmicutes and Clostridium. The association of yacon + kefir increased the number of carcinogenic lesions, despite a reduction in pH and beneficial bacteria prevalence. Thus, it is concluded that yacon, unlikely kefir, is a promising alternative to mitigate the manifestations of induced carcinogenesis in rats.
Collapse
Affiliation(s)
- Keila Rodrigues Zanardi
- Postgraduate Program in Food Science and Technology, Centre of Agricultural and Engineering Sciences, Federal University of Espirito Santo (Universidade Federal do Espírito Santo - UFES), Alto Universitário, Guararema, 29500-000, Alegre, ES, Brazil.
| | - Mariana Grancieri
- Postgraduate Program in Food Science and Technology, Centre of Agricultural and Engineering Sciences, Federal University of Espirito Santo (Universidade Federal do Espírito Santo - UFES), Alto Universitário, Guararema, 29500-000, Alegre, ES, Brazil.
- Department of Pharmacy and Nutrition, Centre of Exact, Natural and Health Sciences, UFES, Alegre, ES, Brazil
| | - Caroline Woelffel Silva
- Postgraduate Program in Food Science and Technology, Centre of Agricultural and Engineering Sciences, Federal University of Espirito Santo (Universidade Federal do Espírito Santo - UFES), Alto Universitário, Guararema, 29500-000, Alegre, ES, Brazil.
| | - Leonardo Oliveira Trivillin
- Department of Veterinary Medicine, Centre of Agricultural and Engineering Sciences, UFES, Alegre, ES, Brazil
| | - Mirelle Lomar Viana
- Department of Pharmacy and Nutrition, Centre of Exact, Natural and Health Sciences, UFES, Alegre, ES, Brazil
| | - André Gustavo Vasconcelos Costa
- Postgraduate Program in Food Science and Technology, Centre of Agricultural and Engineering Sciences, Federal University of Espirito Santo (Universidade Federal do Espírito Santo - UFES), Alto Universitário, Guararema, 29500-000, Alegre, ES, Brazil.
- Department of Pharmacy and Nutrition, Centre of Exact, Natural and Health Sciences, UFES, Alegre, ES, Brazil
| | - Neuza Maria Brunoro Costa
- Postgraduate Program in Food Science and Technology, Centre of Agricultural and Engineering Sciences, Federal University of Espirito Santo (Universidade Federal do Espírito Santo - UFES), Alto Universitário, Guararema, 29500-000, Alegre, ES, Brazil.
- Department of Pharmacy and Nutrition, Centre of Exact, Natural and Health Sciences, UFES, Alegre, ES, Brazil
| |
Collapse
|
8
|
Takada K, Suzukawa M, Igarashi S, Uehara Y, Watanabe S, Imoto S, Ishii M, Morio Y, Matsui H, Akishita M, Ohta K. Serum IgA augments adhesiveness of cultured lung microvascular endothelial cells and suppresses angiogenesis. Cell Immunol 2023; 393-394:104769. [PMID: 37741001 DOI: 10.1016/j.cellimm.2023.104769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 09/11/2023] [Accepted: 09/15/2023] [Indexed: 09/25/2023]
Abstract
Immunoglobulin A (IgA) is important in local immunity and is also abundant in the blood. This study aimed to evaluate the effects of serum IgA on cultured lung microvascular endothelial cells (HMVEC-Ls), which are involved in the pathogenesis of inflammatory lung diseases. Serum IgA induced adhesion molecules and inflammatory cytokine production from HMVEC-Ls, and enhanced adhesion of peripheral blood mononuclear cells to HMVEC-Ls. In contrast, migration, proliferation, and tube formation of HMVEC-Ls were significantly suppressed by serum IgA. Experiments with siRNAs and western blotting revealed that two known IgA receptors, β1,4-galactosyltransferase 1 (b4GALT1) and asialoglycoprotein receptor 1 (ASGR1), and mitogen-activated protein kinase and nuclear factor-kappa B pathways were partly involved in serum IgA-induced cytokine production by HMVEC-Ls. Collectively, serum IgA enhanced cytokine production and adhesiveness of HMVEC-L, with b4GALT1 and ASGR1 partially being involved, and suppressed angiogenesis. Thus, serum IgA may be targeted to treat inflammatory lung diseases.
Collapse
Affiliation(s)
- Kazufumi Takada
- Clinical Research Center, National Hospital Organization Tokyo National Hospital, 3-1-1 Takeoka, Kiyose-City, Tokyo 204-8585, Japan; Department of Geriatric Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Maho Suzukawa
- Clinical Research Center, National Hospital Organization Tokyo National Hospital, 3-1-1 Takeoka, Kiyose-City, Tokyo 204-8585, Japan.
| | - Sayaka Igarashi
- Clinical Research Center, National Hospital Organization Tokyo National Hospital, 3-1-1 Takeoka, Kiyose-City, Tokyo 204-8585, Japan
| | - Yuuki Uehara
- Clinical Research Center, National Hospital Organization Tokyo National Hospital, 3-1-1 Takeoka, Kiyose-City, Tokyo 204-8585, Japan; Division of Respiratory Medicine and Allergology, Department of Medicine, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi-ku, Tokyo, 173-8605, Japan
| | - Shizuka Watanabe
- Clinical Research Center, National Hospital Organization Tokyo National Hospital, 3-1-1 Takeoka, Kiyose-City, Tokyo 204-8585, Japan; Department of Respiratory Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Sahoko Imoto
- Clinical Research Center, National Hospital Organization Tokyo National Hospital, 3-1-1 Takeoka, Kiyose-City, Tokyo 204-8585, Japan; Division of Respiratory Medicine and Allergology, Department of Medicine, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi-ku, Tokyo, 173-8605, Japan
| | - Masaki Ishii
- Department of Geriatric Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Yoshiteru Morio
- Department of Respiratory Medicine, National Hospital Organization Tokyo National Hospital, 3-1-1 Takeoka, Kiyose-City, Tokyo 204-8585, Japan
| | - Hirotoshi Matsui
- Department of Respiratory Medicine, National Hospital Organization Tokyo National Hospital, 3-1-1 Takeoka, Kiyose-City, Tokyo 204-8585, Japan
| | - Masahiro Akishita
- Department of Geriatric Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Ken Ohta
- Clinical Research Center, National Hospital Organization Tokyo National Hospital, 3-1-1 Takeoka, Kiyose-City, Tokyo 204-8585, Japan; Japan Anti-Tuberculosis Association, JATA Fukujuji Hospital, 3-1-24 Matsuyama, Kiyose-City, Tokyo 204-8522, Japan.
| |
Collapse
|
9
|
Zogopoulos VL, Malatras A, Michalopoulos I. Special Issue on Differential Gene Expression and Coexpression. Biology (Basel) 2023; 12:1226. [PMID: 37759625 PMCID: PMC10525233 DOI: 10.3390/biology12091226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023]
Abstract
The most common approach in transcriptomics (RNA-seq and microarrays) is differential gene expression analysis (DGEA) [...].
Collapse
Affiliation(s)
- Vasileios L. Zogopoulos
- Centre of Systems Biology, Biomedical Research Foundation, Academy of Athens, 11527 Athens, Greece;
- Section of Cell Biology and Biophysics, Department of Biology, National and Kapodistrian University of Athens, 15701 Athens, Greece
| | - Apostolos Malatras
- Biobank.cy Centre of Excellence in Biobanking and Biomedical Research, University of Cyprus, 2029 Nicosia, Cyprus
| | - Ioannis Michalopoulos
- Centre of Systems Biology, Biomedical Research Foundation, Academy of Athens, 11527 Athens, Greece;
| |
Collapse
|
10
|
Han X, Luo R, Qi S, Wang Y, Dai L, Nie W, Lin M, He H, Ye N, Fu C, You Y, Fu S, Gao F. "Dual sensitive supramolecular curcumin nanoparticles" in "advanced yeast particles" mediate macrophage reprogramming, ROS scavenging and inflammation resolution for ulcerative colitis treatment. J Nanobiotechnology 2023; 21:321. [PMID: 37679849 PMCID: PMC10483867 DOI: 10.1186/s12951-023-01976-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 06/30/2023] [Indexed: 09/09/2023] Open
Abstract
Ulcerative colitis (UC) faces some barriers in oral therapy, such as how to safely deliver drugs to the colon and accumulate in the colon lesions. Hence, we report an advanced yeast particles system loaded with supramolecular nanoparticles with ROS scavenger (curcumin) to treat UC by reducing oxidative stress state and inflammatory response and accelerating the reprogramming of macrophages. In this study, the dual-sensitive materials are bonded on β-cyclodextrin (β-CD), the D-mannose (Man) is modified to adamantane (ADA), and then loaded with curcumin (CUR), to form a functional supramolecular nano-delivery system (Man-CUR NPs) through the host-guest interaction. To improve gastrointestinal stability and colonic accumulation of Man-CUR NPs, yeast cell wall microparticles (YPs) encapsulated Man-CUR NPs to form Man-CUR NYPs via electrostatic adsorption and vacuum extrusion technologies. As expected, the YPs showed the strong stability in complex gastrointestinal environment. In addition, the Man modified supramolecular nanoparticles demonstrated excellent targeting ability to macrophages in the in vitro cellular uptake study and the pH/ROS sensitive effect of Man-CUR NPs was confirmed by the pH/ROS-dual stimulation evaluation. They also enhanced lipopolysaccharide (LPS)-induced inflammatory model in macrophages through downregulation of pro-inflammatory factors, upregulation of anti-inflammatory factors, M2 macrophage polarization, and scavenging the excess ROS. Notably, in DSS-induced mice colitis model, Man-CUR NYPs can reduce the inflammatory responses by modulating TLR4/NF-κB signaling pathways, alleviate oxidative stress by Nrf2/HO-1 signaling pathway, promote macrophages reprogramming and improve the favorable recovery of the damaged colonic tissue. Taken together, this study not only provides strategy for "supramolecular curcumin nanoparticles with pH/ROS sensitive and multistage therapeutic effects" in "advanced yeast particles", but also provided strong theoretical support multi-effect therapy for UC.
Collapse
Affiliation(s)
- Xiaoqin Han
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, 611130, Chengdu, China
| | - Ruifeng Luo
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, 611130, Chengdu, China
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, 999078, Taipa, Macau, China
| | - Shanshan Qi
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, 611130, Chengdu, China
| | - Yanli Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, 611130, Chengdu, China
| | - Linxin Dai
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, 611130, Chengdu, China
| | - Wenbiao Nie
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, 611130, Chengdu, China
| | - Meisi Lin
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, 611130, Chengdu, China.
| | - Haoqi He
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, 611130, Chengdu, China
| | - Naijing Ye
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, 610072, Chengdu, China
| | - Chaomei Fu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, 611130, Chengdu, China
| | - Yu You
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, 611130, Chengdu, China.
| | - Shu Fu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, 611130, Chengdu, China.
| | - Fei Gao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, 611130, Chengdu, China.
| |
Collapse
|
11
|
Shi F, Yin W, Adu-Frimpong M, Li X, Xia X, Sun W, Ji H, Toreniyazov E, Qilong W, Cao X, Yu J, Xu X. In-vitro and in-vivo evaluation and anti-colitis activity of esculetin-loaded nanostructured lipid carrier decorated with DSPE-MPEG2000. J Microencapsul 2023; 40:442-455. [PMID: 37191893 DOI: 10.1080/02652048.2023.2215345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 05/12/2023] [Indexed: 05/17/2023]
Abstract
OBJECTIVE Encapsulation of esculetin into DSPE-MPEG2000 carrier was performed to improve its water solubility and oral bioavailability, as well as enhance its anti-inflammatory effect on a mouse model of ulcerative colitis that was induced with dextran sulphate sodium (DSS). METHODS We determined the in-vitro and in-vivo high-performance liquid chromatographic (HPLC) analysis method of esculetin; Esculetin-loaded nanostructure lipid carrier (Esc-NLC) was prepared using a thin-film dispersion method, wherein a particle size analyser was used to measure the particle size (PS) and zeta potential (ZP) of the Esc-NLC, while a transmission electron microscope (TEM) was employed to observe its morphology. Also, HPLC was used to measure its drug loading (DL), encapsulation efficiency (EE) and the in-vitro release of the preparation, as well as investigate the pharmacokinetic parameters. In addition, its anti-colitis effect was evaluated via histopathological examination of HE-stained sections and detection of the concentrations of tumour necrosis factor-alpha (TNF-α), interleukin (IL)-1 beta (β), and IL-6 in serum with ELISA kits. RESULTS The PS of Esc-NLC was 102.29 ± 0.63 nm with relative standard deviation (RSD) of 1.08% (with poly-dispersity index-PDI of 0.197 ± 0.023), while the ZP was -15.67 ± 1.39 mV with RSD of 1.24%. Solubility of esculetin was improved coupled with prolonged release time. Its pharmacokinetic parameters were compared with that of free esculetin, wherein the maximum concentration of the drug in plasma was increased by 5.5 times. Of note, bioavailability of the drug was increased by 1.7 times, while the half-life was prolonged by 2.4 times. In the anti-colitis efficacy experiment, the mice in Esc and Esc-NLC groups exhibited significantly reduced levels of TNF-α, IL-1β, and IL-6 in their sera comparable to the DSS group. Colon histopathological examination revealed that mice with ulcerative colitis in both Esc and Esc-NLC groups displayed improved inflammation, amid the Esc-NLC groups having the best prophylactic treatment effect. CONCLUSION Esc-NLC could ameliorate DSS-induced ulcerative colitis by improving bioavailability, prolonging drug release time and regulating cytokine release. This observation confirmed the potential of Esc-NLC to reduce inflammation in ulcerative colitis, albeit the need for follow-up research to verify the application of this strategy to clinical treatment of ulcerative colitis.
Collapse
Affiliation(s)
- Feng Shi
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, Jiangsu, CN, P.R. China
- Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, CN, P.R. China
| | - Wenxiong Yin
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, Jiangsu, CN, P.R. China
| | - Michael Adu-Frimpong
- Department of Biochemistry and Forensic Sciences, School Chemical and Biochemical Sciences C. K. Tedam University of Technology and Applied Sciences (CKT-UTAS), Navrongo, GH, 0215-5321, UK
| | - Xiaoxiao Li
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, Jiangsu, CN, P.R. China
| | - Xiaoli Xia
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, Jiangsu, CN, P.R. China
| | - Weigang Sun
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, Jiangsu, CN, P.R. China
| | - Hao Ji
- Jiangsu Tian Sheng Pharmaceutical Co., Ltd, Zhenjiang, CN, P.R. China
| | - Elmurat Toreniyazov
- Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, CN, P.R. China
- Tashkent State Agricultural University (Nukus branch), Nukus, UZ, P.R. China
| | - Wang Qilong
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, Jiangsu, CN, P.R. China
- Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, CN, P.R. China
| | - Xia Cao
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, Jiangsu, CN, P.R. China
- Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, CN, P.R. China
| | - Jiangnan Yu
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, Jiangsu, CN, P.R. China
- Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, CN, P.R. China
| | - Ximing Xu
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, Jiangsu, CN, P.R. China
- Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, CN, P.R. China
| |
Collapse
|
12
|
Kobayashi S, Sullivan C, Bialkowska AB, Saltz JH, Yang VW. Computational immunohistochemical mapping adds immune context to histological phenotypes in mouse models of colitis. Sci Rep 2023; 13:14386. [PMID: 37658187 PMCID: PMC10474139 DOI: 10.1038/s41598-023-41574-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 08/29/2023] [Indexed: 09/03/2023] Open
Abstract
Inflammatory bowel disease (IBD) is characterized by chronic, dysregulated inflammation in the gastrointestinal tract. The heterogeneity of IBD is reflected through two major subtypes, Crohn's Disease (CD) and Ulcerative Colitis (UC). CD and UC differ across symptomatic presentation, histology, immune responses, and treatment. While colitis mouse models have been influential in deciphering IBD pathogenesis, no single model captures the full heterogeneity of clinical disease. The translational capacity of mouse models may be augmented by shifting to multi-mouse model studies that aggregate analysis across various well-controlled phenotypes. Here, we evaluate the value of histology in multi-mouse model characterizations by building upon a previous pipeline that detects histological disease classes in hematoxylin and eosin (H&E)-stained murine colons. Specifically, we map immune marker positivity across serially-sectioned slides to H&E histological classes across the dextran sodium sulfate (DSS) chemical induction model and the intestinal epithelium-specific, inducible Villin-CreERT2;Klf5fl/fl (Klf5ΔIND) genetic model. In this study, we construct the beginning frameworks to define H&E-patch-based immunophenotypes based on IHC-H&E mappings.
Collapse
Affiliation(s)
- Soma Kobayashi
- Department of Biomedical Informatics, Renaissance School of Medicine at Stony, Brook University, Stony Brook, NY, USA
| | - Christopher Sullivan
- Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| | - Agnieszka B Bialkowska
- Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| | - Joel H Saltz
- Department of Biomedical Informatics, Renaissance School of Medicine at Stony, Brook University, Stony Brook, NY, USA
- Department of Pathology, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| | - Vincent W Yang
- Department of Biomedical Informatics, Renaissance School of Medicine at Stony, Brook University, Stony Brook, NY, USA.
- Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA.
- Department of Physiology and Biophysics, Renaissance School of Medicine at Stony, Brook University, Stony Brook, NY, USA.
| |
Collapse
|
13
|
El-Dawy K, Barakat N, Ali H, Sindi IA, Adly HM, Saleh SA. Dexpanthenol improved stem cells against cisplatin-induced kidney injury by inhibition of TNF-α, TGFβ-1, β-catenin, and fibronectin pathways. Saudi J Biol Sci 2023; 30:103773. [PMID: 37635837 PMCID: PMC10450985 DOI: 10.1016/j.sjbs.2023.103773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 07/30/2023] [Accepted: 08/03/2023] [Indexed: 08/29/2023] Open
Abstract
Introduction Cisplatin interacts with DNA and induces an immunological response and reactive oxygen species, which are nephrotoxic mediators. Stem cells self-renew through symmetric divisions and can develop into other cell types due to their multipotency. Dexpanthenol has been proven to protect against renal injury. Aim This study aims to demonstrate that dexpanthenol could improve the effect of adipose-derived mesenchymal stem cells (ADMSC) against cisplatin-induced acute kidney injury. Methods Sixty male Sprague-Dawley rats were divided into 5 groups (N = 12): control, cisplatin, cisplatin & dexpanthenol, cisplatin & ADMSC, and cisplatin & dexpanthenol & ADMSCs. On the 5th day following cisplatin injection, half the rats in each group were sacrificed, and the other half were sacrificed on the 12th day. Histopathological examination, molecular studies (IL-6, Bcl2, TGFβ-1, Caspase-3, Fibronectin, and β-catenin), antioxidants (superoxide dismutase and catalase), and renal function were all investigated. Results In contrast to cisplatin group, the dexpanthenol and ADMSCs treatments significantly decreased renal function and oxidative stress while significantly enhancing antioxidants. Dexpanthenol improved stem cells by significantly down-regulating caspase-3, IL-6, TGF-β1, Fibronectin, and β-catenin and significantly up-regulating Bcl2 and CD34, which reversed the cisplatin effect. Conclusion Dexpanthenol enhanced ADMSCs' ability to protect against cisplatin-induced AKI by decreasing inflammation, apoptosis, and fibrosis.
Collapse
Affiliation(s)
- Khalifa El-Dawy
- Biochemistry Dept., Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Nashwa Barakat
- Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| | - Hala Ali
- Biochemistry Dept., Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Ikhlas A. Sindi
- Preparatory Year Program, Batterjee Medical College, Jeddah 21442, Saudi Arabia
- Department of Biology, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Heba M. Adly
- Community Medicine and Pilgrims Healthcare Department, Faculty of Medicine, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Saleh A.K. Saleh
- Biochemistry Department, Faculty of Medicine, Umm Al-Qura University, Makkah 21955, Saudi Arabia
- Oncology Diagnostic Unit, Faculty of Medicine, Ain Shams University, Cairo 11435, Egypt
| |
Collapse
|
14
|
E L, Li W, Hu Y, Deng L, Yao J, Zhou X. Methyl cinnamate protects against dextran sulfate sodium-induced colitis in mice by inhibiting the MAPK signaling pathway. Acta Biochim Biophys Sin (Shanghai) 2023. [PMID: 37654075 DOI: 10.3724/abbs.2023124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/02/2023] Open
Abstract
Effective and non-toxic therapeutic agents are lacking for the prevention and treatment of colitis. Previous studies found that methyl cinnamate (MC), extracted from galangal ( Alpinia officinarum Hance), has anti-inflammatory properties. However, whether MC is effective as anti-colitis therapy remains unknown. In this study, we investigate the therapeutic effects of MC on dextran sulfate sodium (DSS)-induced colitis in mice and further explore its potential mechanism of action. MC treatment relieves symptoms associated with DSS-induced colitis, including the recovery of DSS-induced weight loss, decreases the disease activity index score, and increases the colon length without toxic side effects. MC treatment protects the integrity of the intestinal barrier in mice with DSS-induced colitis and inhibits the overexpression of pro-inflammatory cytokines in vivo and in vitro. Moreover, the MAPK signaling pathway is found to be closely related to the treatment with MC of colitis. Western blot analysis show that phosphorylation of the p38 protein in colon tissues treated with MC is markedly reduced and phosphorylation levels of the p38, JNK and ERK proteins are significantly decreased in RAW 264.7 cells treated with MC, indicating that the mechanism of MC in treating DSS-induced colitis could be achieved by inhibiting the MAPK signaling pathway. Furthermore, 16S RNA sequencing analysis show that MC can improve intestinal microbial dysbiosis in mice with DSS-induced colitis. Altogether, these findings suggest that MC may be a novel therapeutic candidate with anti-colitis efficacy. Furthermore, MC treatment relieves the symptoms of colitis by inhibiting the MAPK signaling pathway and improving the intestinal microbiota.
Collapse
|