Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 321
Filter
1.
PLoS One ; 19(9): e0307414, 2024.
Article in English | MEDLINE | ID: mdl-39226257

ABSTRACT

Cancer continues to pose a significant global health challenge, with gastrointestinal (GI) cancers among the most prevalent and deadly forms. These cancers often lead to high mortality rates and demand the use of potent cytotoxic chemotherapeutics. For example, 5-fluorouracil (5-FU) forms the backbone of chemotherapy regimens for various GI cancers, including colorectal cancer. While these chemotherapeutics efficiently kill cancer cells, they frequently cause off-target effects such as chemotherapy-induced mucositis (CIM), characterized by debilitating symptoms like pain, nausea, and diarrhoea, necessitating medical intervention. In this study, we elucidated the potential of melatonin and misoprostol to reduce 5-FU-induced small intestinal mucositis. Morphological and cellular changes in the jejunum, along with colonic faecal water content were quantified in rats as markers for CIM. Additionally, the effects of melatonin were investigated in vitro on 5-FU treated murine intestinal organoids. The results showed that melatonin prevented villus atrophy in the rat jejunal mucosa and upheld cell viability in murine intestinal organoids. In contrast, misoprostol alone or in combination with melatonin did not significantly affect CIM caused by 5-FU. These in vivo and in vitro experiments provided promising insights that melatonin may be used as a preventive and/or adjuvant combination therapy to prevent and reduce CIM, holding the potential to enhance cancer treatment outcomes and improve patient quality-of-life.


Subject(s)
Fluorouracil , Intestine, Small , Melatonin , Mucositis , Organoids , Animals , Melatonin/pharmacology , Rats , Organoids/drug effects , Fluorouracil/adverse effects , Fluorouracil/pharmacology , Mice , Intestine, Small/drug effects , Intestine, Small/pathology , Mucositis/chemically induced , Mucositis/pathology , Mucositis/prevention & control , Mucositis/drug therapy , Male , Atrophy/chemically induced , Atrophy/drug therapy , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology
2.
Inflamm Res ; 73(10): 1781-1801, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39180691

ABSTRACT

OBJECTIVE: Intestinal mucositis is one of the common side effects of anti-cancer chemotherapy. However, the molecular mechanisms involved in mucositis development remain incompletely understood. In this study, we investigated the function of receptor-interacting protein kinase 3 (RIP3/RIPK3) in regulating doxorubicin-induced intestinal mucositis and its potential mechanisms. METHODS: Intestinal mucositis animal models were induced in mice for in vivo studies. Rat intestinal cell line IEC-6 was used for in vitro studies. RNA­seq was used to explore the transcriptomic changes in doxorubicin-induced intestinal mucositis. Intact glycopeptide characterization using mass spectrometry was applied to identify α-1,2-fucosylated proteins associated with mucositis. RESULTS: Doxorubicin treatment increased RIP3 expression in the intestine and caused severe intestinal mucositis in the mice, depletion of RIP3 abolished doxorubicin-induced intestinal mucositis. RIP3-mediated doxorubicin-induced mucositis did not depend on mixed lineage kinase domain-like (MLKL) but on α-1,2-fucosyltransferase 2 (FUT2)-catalyzed α-1,2-fucosylation on inflammation-related proteins. Deficiency of MLKL did not affect intestinal mucositis, whereas inhibition of α-1,2-fucosylation by 2-deoxy-D-galactose (2dGal) profoundly attenuated doxorubicin-induced inflammation and mucositis. CONCLUSIONS: RIP3-FUT2 pathway is a central node in doxorubicin-induced intestinal mucositis. Targeting intestinal RIP3 and/or FUT2-mediated α-1,2-fucosylation may provide potential targets for preventing chemotherapy-induced intestinal mucositis.


Subject(s)
Doxorubicin , Fucosyltransferases , Galactoside 2-alpha-L-fucosyltransferase , Mice, Inbred C57BL , Mucositis , Receptor-Interacting Protein Serine-Threonine Kinases , Animals , Doxorubicin/adverse effects , Mucositis/chemically induced , Mucositis/metabolism , Mucositis/pathology , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Fucosyltransferases/genetics , Fucosyltransferases/metabolism , Rats , Cell Line , Intestinal Mucosa/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Male , Mice , Antibiotics, Antineoplastic/toxicity , Antibiotics, Antineoplastic/adverse effects , Mice, Knockout
3.
Int J Biol Macromol ; 276(Pt 1): 133699, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38972652

ABSTRACT

Chemotherapy-induced mucositis (CIM) is the typical side effect of chemotherapy. This study investigates the potential of alginate oligosaccharide (AOS) in ameliorating CIM induced by 5-fluorouracil (5-FU) in a murine model and its underlying mechanisms. AOS effectively mitigated body weight loss and histopathological damage, modulated inflammatory cytokines and attenuated the oxidative stress. AOS restored intestinal barrier integrity through enhancing expression of tight junction proteins via MLCK signaling pathway. AOS alleviated intestinal mucosal damage by inhibiting TLR4/MyD88/NF-κB signaling pathway, downregulating the pro-apoptotic protein Bax and upregulating the anti-apoptotic protein Bcl-2. Moreover, AOS significantly enriched intestinal Akkermansiaceae and increased the production of short-chain fatty acids (SCFAs), most notably butyrate and isovalerate. Pre-treatment with butyrate and isovalerate also alleviated 5-FU-induced CIM. In conclusion, AOS effectively mitigated CIM through strenghthening intestinal barrier, attenuating inflammation, and modulating gut microbiota and intestianl levels of butyrate and isovalerate. These finding indicate that AOS could be potentially utilized as a supplemental strategy for prevention or mitigation of CIM.


Subject(s)
Alginates , Butyrates , Fluorouracil , Intestinal Mucosa , Mucositis , Oligosaccharides , Fluorouracil/adverse effects , Animals , Mucositis/chemically induced , Mucositis/drug therapy , Mucositis/metabolism , Mucositis/pathology , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice , Oligosaccharides/pharmacology , Oligosaccharides/chemistry , Butyrates/pharmacology , Butyrates/metabolism , Alginates/pharmacology , Alginates/chemistry , Gastrointestinal Microbiome/drug effects , Male , Oxidative Stress/drug effects , Signal Transduction/drug effects , Cytokines/metabolism
4.
Int J Mol Sci ; 25(13)2024 Jun 28.
Article in English | MEDLINE | ID: mdl-39000262

ABSTRACT

Radiotherapy in the head-and-neck area is one of the main curative treatment options. However, this comes at the cost of varying levels of normal tissue toxicity, affecting up to 80% of patients. Mucositis can cause pain, weight loss and treatment delays, leading to worse outcomes and a decreased quality of life. Therefore, there is an urgent need for an approach to predicting normal mucosal responses in patients prior to treatment. We here describe an assay to detect irradiation responses in healthy oral mucosa tissue. Mucosa specimens from the oral cavity were obtained after surgical resection, cut into thin slices, irradiated and cultured for three days. Seven samples were irradiated with X-ray, and three additional samples were irradiated with both X-ray and protons. Healthy oral mucosa tissue slices maintained normal morphology and viability for three days. We measured a dose-dependent response to X-ray irradiation and compared X-ray and proton irradiation in the same mucosa sample using standardized automated image analysis. Furthermore, increased levels of inflammation-inducing factors-major drivers of mucositis development-could be detected after irradiation. This model can be utilized for investigating mechanistic aspects of mucositis development and can be developed into an assay to predict radiation-induced toxicity in normal mucosa.


Subject(s)
Mouth Mucosa , Humans , Mouth Mucosa/radiation effects , X-Rays/adverse effects , Radiation Injuries/etiology , Radiation Injuries/pathology , Male , Mucositis/etiology , Mucositis/pathology , Female , Dose-Response Relationship, Radiation , Stomatitis/etiology , Stomatitis/pathology , Adult , Middle Aged
5.
Eur J Pharmacol ; 975: 176669, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38795758

ABSTRACT

Methotrexate (MTX)-induced gastrointestinal mucositis is a common adverse effect characterized by redox imbalance and overproduction of inflammatory mediators that perturb intestinal integrity. Currently, there is no definitive treatment for this condition and its prevention is still far beyond comprehension. Because of its pleiotropic pharmacological actions, we aimed to explore the potential mechanisms through which cilostazol (CILO) can protect against MTX-induced intestinal mucositis. Wistar rats were allocated into 4 groups, control, CILO (100 mg/kg, p.o for 14 days), MTX (7.5 mg/kg for 4 successive days), and CILO + MTX. The improving effect of CILO on the morphological structure was confirmed by an upturn in the histopathological and transition electron microscope examinations evidenced by the increased jejunal villus height/width and the crypt depth besides the maintenance of tight junctions. These findings were verified biochemically; on the molecular level, CILO reduced the MTX-induced lipid peroxidation, cleaved caspase-3, p53, and the inflammatory parameters (TLR-2, NF-κB, IL-23, TNF-α, IL-1ß), while increasing the anti-inflammatory marker IL-10 and the antioxidant enzyme SOD. Moreover, CILO decreased the injurious axis AKT/GSK-3ß/cyclin-D1, and CD44+, but increased the immunoexpression of the cell proliferating marker PCNA. CILO also upheld the intestinal barrier by enhancing the tight junction molecules (ZO-1, claudin-4) and the E-cadherin/ß-catenin complex while abating the mesenchymal marker vimentin. In conclusion, CILO protected gut integrity by reducing the epithelial-mesenchymal transition process, the MTX-induced oxidative, apoptotic, and inflammatory mediators, and turning off the CD44/AKT/GSK-3ß/cyclin D1 trajectory and intensifying the expression of PCNA.


Subject(s)
Cyclin D1 , Glycogen Synthase Kinase 3 beta , Methotrexate , Mucositis , NF-kappa B , Proto-Oncogene Proteins c-akt , Rats, Wistar , Toll-Like Receptor 2 , Animals , Glycogen Synthase Kinase 3 beta/metabolism , NF-kappa B/metabolism , Methotrexate/toxicity , Methotrexate/pharmacology , Rats , Toll-Like Receptor 2/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Mucositis/chemically induced , Mucositis/pathology , Mucositis/metabolism , Male , Cyclin D1/metabolism , Signal Transduction/drug effects , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Disease Models, Animal
7.
Article in English | MEDLINE | ID: mdl-38331651

ABSTRACT

OBJECTIVE: Oral plasma cell mucositis (PCM) or localized plasma cell gingivitis (PCG) is an idiopathic inflammatory condition often associated with hypersensitivity reactions. This study aimed to evaluate the frequency and features of PCM/PCG in a large biopsy service over a time period of more than 20 years. STUDY DESIGN: The biopsy archives at University of Florida College of Dentistry were searched from 2000 through the first quarter of 2023 for cases of oral PCM or PCG. Case data were aggregated and analyzed. RESULTS: A total of 107 cases were included. Between 2000 and 2019, PCM/PCG was diagnosed in 0.03% of all biopsy cases. Starting in 2020 through 2023, the percentage of biopsies diagnosed as PCM/PCG increased threefold to 0.10% of all biopsy cases, and the mean patient age increased by 3 years. There were no statistically significant differences between cases diagnosed from 2000 to 2019 and those from 2020 to 2023 regarding age, sex, location, or histology. CONCLUSIONS: A significant increase in PCM/PCG was identified in this study at one institution coinciding with the start of the COVID-19 pandemic. Further investigation is recommended to determine if this is a widespread phenomenon and identify possible etiologic mechanisms.


Subject(s)
COVID-19 , Gingivitis , Mucositis , Stomatitis , Humans , COVID-19/epidemiology , COVID-19 Testing , Gingivitis/etiology , Gingivitis/pathology , Mucositis/pathology , Pandemics , Plasma Cells/pathology , Retrospective Studies , Stomatitis/etiology
8.
JAMA Dermatol ; 160(3): 320-327, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38265770

ABSTRACT

Importance: Plasma cell orificial mucositis (PCOM) associated with cocaine use is an emerging, rare condition that has become a concern in Spain in recent years. Limited knowledge exists regarding this novel condition. Objectives: To delineate the clinicopathologic characteristics of this emerging entity and establish a novel approach in the differential diagnosis of cocaine-associated lesions. Design, Setting, and Participants: A descriptive, retrospective, multicenter case series of 10 patients diagnosed with cocaine-associated PCOM was conducted in Spain from April 2020 to March 2023. Main Outcomes and Measures: Patient demographic, clinical, histopathologic, and treatment data were collected. Results: A total of 10 patients (6 [60%] male; median [range] age, 45.5 [36-66] years) presenting with exudative ulcerated plaques were identified for this study. The lesions had raised and erythematous edges over the nostril and a median (range) evolution time of 9 (2-24) months. Septal or palate perforations were observed in 4 (40%) of the patients. Biopsies revealed a dense inflammatory infiltrate of plasma cells in the dermis without atypia and with eosinophils. All patients reported recent cocaine use. Three urine tests detected cocaine but found no presence of amphetamines or opiates. Six patients improved with corticosteroid therapy. Up to 60% of patients were lost to follow-up. Conclusions and Relevance: This case series describes the clinicopathologic characteristics of PCOM, an emerging entity associated with cocaine use in Spain, and demonstrates a novel approach in the differential diagnosis of cocaine-associated lesions. To date, cocaine-associated skin lesions have been reported as neutrophilic dermatoses and vasculitis. The appearance of a plasma cell infiltrate changes what has been described in the medical literature so far. PCOM is a benign condition of unknown cause characterized by a proliferative polyclonal plasma cell infiltrate. A comprehensive differential diagnosis workup is required to reach this exclusionary diagnosis. Several irritants have been documented in cases of PCOM, and a hypersensitivity mechanism has been proposed. Since the initial report of cocaine-associated PCOM in Spain, its incidence has experienced a surge in the country. The cause of this phenomenon may be attributed to newly unidentified adulterants. The administration of corticosteroids and discontinuation of cocaine use are the sole treatments that have demonstrated efficacy. Clinicians should be vigilant regarding this emerging condition and conduct inquiries into cocaine use. Additional research is required to clarify the pathophysiology of this emerging condition.


Subject(s)
Cocaine , Mucositis , Humans , Male , Middle Aged , Female , Mucositis/pathology , Plasma Cells/pathology , Retrospective Studies , Erythema/pathology , Inflammation/pathology , Cocaine/adverse effects
9.
Inflammation ; 47(4): 1185-1203, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38289578

ABSTRACT

Methotrexate (MTX)-induced intestinal mucositis (IM) is a common side effect in cancer treatment that impairs the immune system and gut microbes, resulting in loss of mucosal integrity and gut barrier dysfunction. The quality of life and outcomes of treatment are compromised by IM. The present study was designed to investigate the mucoprotective potential of the benzimidazole derivative N-{4-[2-(4-methoxyphenyl)-1H-benzimidazole-1-sulfonyl] phenyl} acetamide (B8) on MTX-induced IM in mice. IM was induced by a single dose of MTX in mice and assessed by physical manifestations as well as biochemical, oxidative, histological, and inflammatory parameters. B8 (1, 3, 9 mg/kg) significantly reduced diarrhea score, mitigated weight loss, increased feed intake and, survival rate in a dose-dependent manner. Notably, B8 exhibited a mucoprotective effect evident through the mitigation of villus atrophy, crypt hypoplasia, diminished crypt mitotic figures, mucin depletion, and oxidative stress markers (GSH, SOD, MDA, and catalase concentration). Gene expression analysis revealed that B8 downregulated the mRNA expression of tumor necrosis factor-α (TNF-α), cyclooxygenase-2 (COX-2), interleukin-6 (IL-6), IL-1ß, and nuclear factor-κB (NF-κB) and concurrently upregulated IL-10 expression in contrast to the MTX group. Further, B8 significantly improved the luminal microflora profile by augmenting the growth of Lactobacillus spp. and reducing the number of pathogenic bacteria (E. coli). Additionally, the enzyme-linked immunoassay showed that B8 decreased the levels of pro-inflammatory cytokines. Our findings suggest that B8 had mucoprotective effects against MTX-induced IM and could be used as an adjunct in chemotherapy to deter this side effect.


Subject(s)
Benzimidazoles , Methotrexate , Mucositis , Oxidative Stress , Animals , Mucositis/chemically induced , Mucositis/drug therapy , Mucositis/metabolism , Mucositis/pathology , Mice , Oxidative Stress/drug effects , Benzimidazoles/pharmacology , Benzimidazoles/therapeutic use , Methotrexate/pharmacology , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/chemically induced , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Male , Biomarkers/metabolism , Cytokines/metabolism , Inflammation Mediators/metabolism , Inflammation Mediators/antagonists & inhibitors , Antimetabolites, Antineoplastic/toxicity
10.
J Med Life ; 16(5): 712-718, 2023 May.
Article in English | MEDLINE | ID: mdl-37520490

ABSTRACT

Intestinal mucositis (IM) is a common side effect of several anticancer medications, including 5-fluorouracil (5-FU), and can lead to treatment disruptions and compromised outcomes. IM has severe clinical effects such as diarrhea, erythematous mucosal lesions, and the development of ulcers accompanied by excruciating pain. This study aimed to evaluate the mucoprotective effects of ellagic acid on 5-FU-induced IM in mice. Mice were administered normal saline intraperitoneally for six days, followed by intraperitoneal injection of 5-FU for four days at a dose of 50 mg per kilogram. Ellagic acid was orally administered to the mice in groups III and IV in two doses (5 mg and 10 mg), with a one-hour time separation from 5-FU for ten days. At the end of the experiment, small intestine tissue was collected to measure the levels of antioxidant enzymes superoxide dismutase (SOD), glutathione (GSH), malondialdehyde (MDA), and inflammatory cytokines (IL-6, IL-B, TNF) using ELISA assay. Pre-treatment with ellagic acid led to a significant decrease in pro-inflammatory cytokines and improved antioxidant enzyme levels compared to the 5-FU group. Histopathological analysis demonstrated the mucoprotective effect of ellagic acid against 5-FU-induced intestinal changes, including villi atrophy, damage to stem cells, infiltration of inflammatory cells in the mucosal layer, edema, damage to muscular mucosa, and decreased oxidative stress production, such as MDA. These results suggest that ellagic acid may be a potential candidate for treating IM induced by antineoplastic drugs.


Subject(s)
Antineoplastic Agents , Mucositis , Mice , Animals , Fluorouracil/adverse effects , Ellagic Acid/adverse effects , Intestinal Mucosa/pathology , Antioxidants/pharmacology , Mucositis/chemically induced , Mucositis/drug therapy , Mucositis/pathology , Antineoplastic Agents/pharmacology , Cytokines , Glutathione
11.
Rhinology ; 61(5): 470-480, 2023 Oct 01.
Article in English | MEDLINE | ID: mdl-37454274

ABSTRACT

BACKGROUND: Radiotherapy (RT) is one of the main methods used in the treatment of head and neck cancers but may cause mucosal side effects in the tumor area and surrounding structures. These include nasal mucosal disorders and chronic rhinosinusitis due to disruption of the mucociliary system. This situation seriously affects the quality of life of the patients and there is no accepted effective method for its treatment yet. In our study, we aimed to examine the side effects of RT on the nasal mucosa and mucociliary system and to investigate histopathologically and immunohistochemically the effectiveness of N-acetyl cysteine (NAC) in preventing these side effects of RT. METHODOLOGY: The study was carried out with 30 female Sprague Dawley rats devided in three groups. No intervention was made in the control group. On the second day of the experiment, 30 Gy radiotherapy was applied to the head area in the RT group. NAC was administered intraperitoneally at a dose of 1 g/kg/day for 14 days from the first day of the study to the RT+ NAC group. On the second day, 30 Gy of radiotherapy was applied to the head area 1 hour after the NAC application. On the 14th day, 1 hour after NAC was applied to the RT+NAC group, all animals were sacrificed. The nasal mucosa samples were stained with hematoxylin-eosin, and the intensity and extent of staining sentan in the nasopharyngeal tissue samples were evaluated by immunohistochemical staining using anti-SNTN antibody. RESULTS: The loss of cilia in the nasal tissue was lower in the RT+NAC group than in the RT group. The intensity and extent of staining in the nasopharyngeal tissue of Sentan was higher in the RT+NAC group than in the RT group. Mucosal neutrophil and mononuclear inflammatory cell infiltration in the nasal tissue, vascular dilatation, hyperemia and hemorrhage, erosion and shedding of the mucosal epithelium, mucosal ulceration were found to be similar in the RT+NAC group and the control group. It was milder in the RT+NAC group than in the RT group, but not statistically significant. CONCLUSIONS: Radiotherapy caused pathological changes in the nasal mucosa, caused loss of cilia and a decrease in the level of Sentan, the cilia apical protein. The results of our study showed that NAC treatment can reduce the side effects of RT on the nasal mucosa. It also showed that NAC was effective in preventing the loss of cilia, which is the building block of the mucociliary system, and improving the expression of Sentan.


Subject(s)
Mucositis , Rats , Animals , Humans , Female , Mucositis/etiology , Mucositis/prevention & control , Mucositis/pathology , Quality of Life , Rats, Sprague-Dawley , Nasal Mucosa , Acetylcysteine/pharmacology , Acetylcysteine/therapeutic use
12.
Article in English | MEDLINE | ID: mdl-37328328

ABSTRACT

OBJECTIVE: Plasma cell mucositis (PCM) is a non-neoplastic plasma cell disorder of the upper aerodigestive tract with a high impact on life quality. Less than 70 cases were reported in the literature. The objective of this study was to report 2 cases of PCM. A concise review of the literature is also presented. STUDY DESIGN: Two cases of PCM that presented during the COVID-19 quarantine are reported. The inclusion criteria for the literature review were English-indexed case reports of the last 20 years. RESULTS: Cases were treated with meprednisone. As mechanical trauma was proposed as a triggering factor, its control was also considered. Patients were followed with no relapses. There were 29 studies included. The mean age was 57 years, with a male predominance, different clinical phenotypes, and intensely erythematous mucosa as a classical finding. The most frequent site was the lip, followed by the buccal mucosa. The final diagnosis is clinicopathologic. CD138 expression is a hallmark of plasma cells, frequently aiding PCM diagnosis. Plasma cell mucositis treatment is mostly symptomatic, and several therapeutic modalities have been mostly unsuccessful. CONCLUSIONS: Diagnosing plasma cell mucositis becomes challenging as many lesions may mimic other conditions. Consequently, in these cases, the diagnostic process should gather clinical, histopathologic, and immunohistochemical data.


Subject(s)
COVID-19 , Mucositis , Male , Female , Humans , Mucositis/pathology , Mouth Mucosa/pathology , Plasma Cells/pathology
13.
Biomed Pharmacother ; 164: 114902, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37209628

ABSTRACT

BACKGROUND: Intestinal mucositis (IM) is characterized by damage to the intestinal mucosa resulting from inhibition of epithelial cell division and loss of renewal capacity following anticancer chemotherapy and radiotherapy. Cytarabine (Ara-C), the main chemotherapy drug for the treatment of leukemia and lymphoma, is a frequent cause of IM. Guiqi Baizhu prescription (GQBZP) is a traditional Chinese medicine with anti-cancer and anti-inflammatory effects. PURPOSE: To determine if GQBZP can ameliorate Ara-C induced IM and identify and characterize the pharmacologic and pharmacodynamic mechanisms. STUDY DESIGN AND METHODS: IM was induced in mice with Ara-C and concurrently treated with orally administered GQBZP. Body weight and food intake was monitored, with HE staining to calculate ileal histomorphometric scoring and villus length/crypt depth. Immunoblotting was used to detect intestinal tissue inflammatory factors. M1 macrophages (M1) were labeled with CD86 by flow cytometry and iNOS + F4/80 by immunofluorescence. Virtual screening was used to find potentially active compounds in GQBZP that targeted JAK2. In vitro, RAW264.7 cells were skewed to M1 macrophage polarization by lipopolysaccharide (LPS) and interferon-γ (INF-γ) and treated orally with GQBZP or potential active compounds. M1 was labeled with CD86 by flow cytometry and iNOS by immunofluorescence. ELISA was used to detect inflammatory factor expression. Active compounds against JAK2, p-JAK2, STAT1 and p-STAT1 were identified by western blotting and HCS fluorescence. Molecular dynamics simulations and pharmacokinetic predictions were carried out on representative active compounds. RESULTS: Experimental results with mice in vivo suggest that GQBZP significantly attenuated Ara-C-induced ileal damage and release of pro-inflammatory factors by inhibiting macrophage polarization to M1. Molecular docking was used to identify potentially active compounds in GQBZP that targeted JAK2, a key factor in macrophage polarization to M1. By examining the main components of each herb and applying Lipinski's rules, ten potentially active compounds were identified. In vitro experimental results suggested that all 10 compounds of GQBZP targeted JAK2 and could inhibit M1 polarization in RAW264.7 cells treated with LPS and INF-γ. Among them, acridine and senkyunolide A down-regulated the expression of JAK2 and STAT1. MD simulations revealed that acridine and senkyunolide A were stable in the active site of JAK2 and exhibited good interactions with the surrounding amino acids. CONCLUSIONS: GQBZP can ameliorate Ara-C-induced IM by reducing macrophage polarization to M1, and acridine and senkyunolide A are representative active compounds in GQBZP that target JAK2 to inhibit M1 polarization. Targeting JAK2 to regulate M1 polarization may be a valuable therapeutic strategy for IM.


Subject(s)
Mucositis , Mice , Animals , Mucositis/pathology , Cytarabine/pharmacology , Lipopolysaccharides/pharmacology , Lipopolysaccharides/metabolism , Molecular Docking Simulation , Macrophages/metabolism , Interferon-gamma/metabolism
14.
Int J Mol Sci ; 24(9)2023 May 05.
Article in English | MEDLINE | ID: mdl-37175980

ABSTRACT

Oral mucositis (OM) is a common and impactful toxicity of standard cancer therapy, affecting up to 80% of patients. Its aetiology centres on the initial destruction of epithelial cells and the increase in inflammatory signals. These changes in the oral mucosa create a hostile environment for resident microbes, with oral infections co-occurring with OM, especially at sites of ulceration. Increasing evidence suggests that oral microbiome changes occur beyond opportunistic infection, with a growing appreciation for the potential role of the microbiome in OM development and severity. This review collects the latest articles indexed in the PubMed electronic database which analyse the bacterial shift through 16S rRNA gene sequencing methodology in cancer patients under treatment with oral mucositis. The aims are to assess whether changes in the oral and gut microbiome causally contribute to oral mucositis or if they are simply a consequence of the mucosal injury. Further, we explore the emerging role of a patient's microbial fingerprint in OM development and prediction. The maintenance of resident bacteria via microbial target therapy is under constant improvement and should be considered in the OM treatment.


Subject(s)
Microbiota , Mucositis , Neoplasms , Stomatitis , Humans , RNA, Ribosomal, 16S/genetics , Stomatitis/pathology , Mouth Mucosa/pathology , Neoplasms/pathology , Bacteria , Mucositis/pathology
15.
Int J Mol Sci ; 24(6)2023 Mar 14.
Article in English | MEDLINE | ID: mdl-36982601

ABSTRACT

Inflammatory bowel diseases (IBDs) consist of a group of chronic inflammatory disorders with a complex etiology, which represent a clinical challenge due to their often therapy-refractory nature. In IBD, inflammation of the intestinal mucosa is characterized by strong and sustained leukocyte infiltration, resulting in the loss of epithelial barrier function and subsequent tissue destruction. This is accompanied by the activation and the massive remodeling of mucosal micro-vessels. The role of the gut vasculature in the induction and perpetuation of mucosal inflammation is receiving increasing recognition. While the vascular barrier is considered to offer protection against bacterial translocation and sepsis after the breakdown of the epithelial barrier, endothelium activation and angiogenesis are thought to promote inflammation. The present review examines the respective pathological contributions of the different phenotypical changes observed in the microvascular endothelium during IBD, and provides an overview of potential vessel-specific targeted therapy options for the treatment of IBD.


Subject(s)
Inflammatory Bowel Diseases , Mucositis , Humans , Inflammatory Bowel Diseases/pathology , Inflammation/metabolism , Intestinal Mucosa/metabolism , Mucositis/pathology , Leukocytes/metabolism
16.
PLoS One ; 18(3): e0283626, 2023.
Article in English | MEDLINE | ID: mdl-36996098

ABSTRACT

BACKGROUND: Daikenchuto (DKT) has positive therapeutic effects on improving various gastrointestinal disorders. The present study investigated whether or not DKT has a potential therapeutic effect on chemotherapy-induced acute small intestinal mucositis (CIM) in a rat model. METHODS: Intraperitoneal injection of 10 mg/kg methotrexate (MTX) every 3 days for a total of 3 doses was used for induction of CIM in a rat model. The MTX and DKT-MTX groups were injected with MTX as above from the first day, and the DKT-MTX and DKT groups were administered 2.7% DKT via the diet at the same time. The rats were euthanized on day 15. RESULTS: The DKT-MTX group showed an improvement in the body weight and conditions of gastrointestinal disorders as well as increased levels of diamine oxidase in plasma and in the small intestinal villi. The pathology results showed that small intestinal mucosal injury in the DKT-MTX group was less severe than that in the MTX group. Immunohistochemistry for myeloperoxidase and malondialdehyde and quantitative real-time polymerase chain reaction (RT-qPCR) for TGF-ß1 and HIF-1α showed that DKT attenuated peroxidative damage. The crypts in the DKT-MTX group contained more Ki-67-positive cells than MTX group. The zonula occluden-1 and claudin-3 results showed that DKT promoted repair of the mucosal barrier. RT-qPCR for the amino acid transporters EAAT3 and BO+AT also confirmed that DKT promoted mucosal repair and thus promoted nutrient absorption. CONCLUSION: DKT protected against MTX-induced CIM in a rat model by reducing inflammation, stimulating cell proliferation, and stabilizing the mucosal barrier.


Subject(s)
Enteritis , Mucositis , Panax , Rats , Animals , Methotrexate/toxicity , Mucositis/chemically induced , Mucositis/drug therapy , Mucositis/pathology , Intestinal Mucosa/metabolism , Enteritis/pathology
17.
Sci Rep ; 13(1): 1396, 2023 01 25.
Article in English | MEDLINE | ID: mdl-36697446

ABSTRACT

Oral and intestinal mucositis are debilitating inflammatory diseases observed in cancer patients undergoing chemo-radiotherapy. These are devastating clinical conditions which often lead to treatment disruption affecting underlying malignancy management. Although alimentary tract mucositis involves the entire gastrointestinal tract, oral and intestinal mucositis are often studied independently utilizing distinct organ-specific pre-clinical models. This approach has however hindered the development of potentially effective whole-patient treatment strategies. We now characterize a murine model of alimentary tract mucositis using 5-Fluorouracil (5-FU). Mice were given 5-FU intravenously (50 mg/kg) or saline every 48 h for 2 weeks. Post initial injection, mice were monitored clinically for weight loss and diarrhea. The incidence and extent of oral mucositis was assessed macroscopically. Microscopical and histomorphometric analyses of the tongue and intestinal tissues were conducted at 3 interim time points during the experimental period. Repeated 5-FU treatment caused severe oral and intestinal atrophy, including morphological damage, accompanied by body weight loss and mild to moderate diarrhea in up to 77.8% of mice. Oral mucositis was clinically evident throughout the observation period in 88.98% of mice. Toluidine blue staining of the tongue revealed that the ulcer size peaked at day-14. In summary, we have developed a model reproducing the clinical and histologic features of both oral and intestinal mucositis, which may represent a useful in vivo pre-clinical model for the study of chemotherapy-induced alimentary tract mucositis and the development of preventative therapies.


Subject(s)
Mucositis , Stomatitis , Animals , Mice , Mucositis/pathology , Intestinal Mucosa/pathology , Antimetabolites, Antineoplastic/toxicity , Disease Models, Animal , Fluorouracil/toxicity , Diarrhea/drug therapy , Stomatitis/drug therapy
18.
Probiotics Antimicrob Proteins ; 15(2): 424-440, 2023 04.
Article in English | MEDLINE | ID: mdl-36631616

ABSTRACT

Mucositis is defined as inflammatory and ulcerative lesions along of the gastrointestinal tract that leads to the imbalance of the intestinal microbiota. The use of compounds with action on the integrity of the intestinal epithelium and their microbiota may be a beneficial alternative for the prevention and/or treatment of mucositis. So, the aim of this study was to evaluate the effectiveness of the association of fructo-oligosaccharides (FOS) and arginine on intestinal damage in experimental mucositis. BALB/c mice were randomized into five groups: CTL (without mucositis + saline), MUC (mucositis + saline), MUC + FOS (mucositis + supplementation with FOS-1st until 10th day), MUC + ARG (mucositis + supplementation with arginine-1st until 10th day), and MUC + FOS + ARG (mucositis + supplementation with FOS and arginine-1st until 10th day). On the 7th day, mucositis was induced with an intraperitoneal injection of 300 mg/kg 5-fluorouracil (5-FU), and after 72 h, the animals were euthanized. The results showed that association of FOS and arginine reduced weight loss and oxidative stress (P < 0.05) and maintained intestinal permeability and histological score at physiological levels. The supplementation with FOS and arginine also increased the number of goblet cells, collagen area, and GPR41 and GPR43 gene expression (P < 0.05). Besides these, the association of FOS and arginine modulated intestinal microbiota, leading to an increase in the abundance of the genera Bacteroides, Anaerostipes, and Lactobacillus (P < 0.05) in relation to increased concentration of propionate and acetate. In conclusion, the present results show that the association of FOS and arginine could be important adjuvants in the prevention of intestinal mucositis probably due to modulated intestinal microbiota.


Subject(s)
Gastrointestinal Microbiome , Mucositis , Mice , Animals , Mucositis/drug therapy , Mucositis/metabolism , Mucositis/pathology , Arginine/metabolism , Intestines , Intestinal Mucosa/metabolism , Fluorouracil , Oligosaccharides/pharmacology
19.
In Vivo ; 37(1): 218-224, 2023.
Article in English | MEDLINE | ID: mdl-36593024

ABSTRACT

BACKGROUND/AIM: This study aimed to develop a reliable chemotherapy-induced oral mucositis (CIOM) rat model by intraperitoneally administering a single dosage of 5-fluorouracil (5-FU) combined with a chemical stimulus. MATERIALS AND METHODS: The 5-FU dosage for CIOM development was determined by the survival rate of rats administrated 160 mg/kg, 200 mg/kg, and 240 mg/kg of 5-FU. Thirty rats were assigned to normal control (NC) and three experimental groups: i) ulcer formation without 5-FU administration (PBS/U+), ii) 5-FU administration without ulcer formation (5-FU/U-), and iii) ulcer formation after 5-FU administration (5-FU/U+). White blood cell count and weight were measured at the day of 5-FU administration (D0), ulcer formation (D2), and two days after ulcer formation (D4). The oral mucosa for histologic evaluations was obtained two (D4) and five days (D7) after ulcer formation. RESULTS: The 5-FU dosage for CIOM development was 200 mg/kg. White blood cell count (WBC) counts and weight of rats were significantly lower in 5-FU/U- (WBC, p<0.001; weight, p=0.002) and 5-FU/U+ (WBC, p<0.001; weight, p<0.001) groups compared to those in the NC group at D4. The number of Ki-67 positive cells in the oral epithelium was lower in 5-FU/U+ group compared to that in NC (p<0.001) and PBS/U+ (p=0.047) groups at D7. CONCLUSION: Single administration of 200 mg/kg of 5-FU combined with a chemical stimulus can lead to an immune-suppressive status, failure of weight gain, and impairment of epithelium regeneration as observed in a CIOM rat model.


Subject(s)
Mucositis , Stomatitis , Rats , Animals , Fluorouracil/adverse effects , Mucositis/pathology , Ulcer/pathology , Stomatitis/chemically induced , Stomatitis/drug therapy , Stomatitis/pathology , Mouth Mucosa/pathology , Intestinal Mucosa/pathology
20.
Mini Rev Med Chem ; 23(3): 298-306, 2023.
Article in English | MEDLINE | ID: mdl-35980046

ABSTRACT

Intestinal mucositis is characterized by inflammation and ulceration of the mucosa that affects the gastrointestinal tract and is associated with administering some drugs, such as 5- Fluorouracil (5-FU), conventional chemotherapy used in clinics for cancer therapy. Inside intestinal mucosa, the 5-FU acts, leading to oxidative stress, stimulating the production/release of proinflammatory cytokines, local accumulation of neutrophils and consequent tissue damage. These alterations favor bacterial proliferation, triggering secondary infections, and are responsible for undesired effects such as myelosuppression and diarrhea. These factors negatively impact oncological patients' quality of life and explain why they commonly interrupt their treatment prematurely. Currently, there is no specific drug with the ability to completely avoid this condition, so the search for new molecules with pharmacological properties that can be used for preventing or ameliorating intestinal mucositis is important. Plumeria pudica is a plant that produces latexcontaining molecules with therapeutic potential. A protein fraction obtained from this latex (LPPp), which comprises a well-defined mixture of chitinases, proteinases proteinase inhibitors, was demonstrated to have antioxidant and anti-inflammatory activities, preserving tissue glutathione and malondialdehyde concentration, reducing superoxide dismutase and myeloperoxidase activity, and reducing the level of proinflammatory cytokines in different experimental models. Given this scenario, inflammation and oxidative stress are directly involved in the pathogenesis of intestinal mucositis promoted by 5-FU. So, the hypothesis is that LPPp could inhibit these factors to attenuate the cytotoxicity of this pathology associated with 5-FU-treatment. This article brings new insights into the potential of the laticifer proteins extracted from the latex of P. pudica and opens new perspectives for the treatment of this type of intestinal mucositis with LPPp.


Subject(s)
Apocynaceae , Mucositis , Humans , Fluorouracil/therapeutic use , Mucositis/chemically induced , Mucositis/drug therapy , Mucositis/pathology , Latex/metabolism , Quality of Life , Intestinal Mucosa , Inflammation/metabolism , Cytokines/metabolism , Apocynaceae/metabolism , Antioxidants/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL