Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Orphanet J Rare Dis ; 19(1): 175, 2024 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-38671465

RESUMEN

Polycystic liver disease (PLD) is a rare condition observed in three genetic diseases, including autosomal dominant polycystic liver disease (ADPLD), autosomal dominant polycystic kidney disease (ADPKD), and autosomal recessive polycystic kidney disease (ARPKD). PLD usually does not impair liver function, and advanced PLD becomes symptomatic when the enlarged liver compresses adjacent organs or increases intra-abdominal pressure. Currently, the diagnosis of PLD is mainly based on imaging, and genetic testing is not required except for complex cases. Besides, genetic testing may help predict patients' prognosis, classify patients for genetic intervention, and conduct early treatment. Although the underlying genetic causes and mechanisms are not fully understood, previous studies refer to primary ciliopathy or impaired ciliogenesis as the main culprit. Primarily, PLD occurs due to defective ciliogenesis and ineffective endoplasmic reticulum quality control. Specifically, loss of function mutations of genes that are directly involved in ciliogenesis, such as Pkd1, Pkd2, Pkhd1, and Dzip1l, can lead to both hepatic and renal cystogenesis in ADPKD and ARPKD. In addition, loss of function mutations of genes that are involved in endoplasmic reticulum quality control and protein folding, trafficking, and maturation, such as PRKCSH, Sec63, ALG8, ALG9, GANAB, and SEC61B, can impair the production and function of polycystin1 (PC1) and polycystin 2 (PC2) or facilitate their degradation and indirectly promote isolated hepatic cystogenesis or concurrent hepatic and renal cystogenesis. Recently, it was shown that mutations of LRP5, which impairs canonical Wnt signaling, can lead to hepatic cystogenesis. PLD is currently treated by somatostatin analogs, percutaneous intervention, surgical fenestration, resection, and liver transplantation. In addition, based on the underlying molecular mechanisms and signaling pathways, several investigational treatments have been used in preclinical studies, some of which have shown promising results. This review discusses the clinical manifestation, complications, prevalence, genetic basis, and treatment of PLD and explains the investigational methods of treatment and future research direction, which can be beneficial for researchers and clinicians interested in PLD.


Asunto(s)
Quistes , Hepatopatías , Humanos , Hepatopatías/genética , Quistes/genética , Mutación/genética
2.
Horm Metab Res ; 2024 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-38471571

RESUMEN

Obesity, non-alcoholic fatty liver disease (NAFLD), and atherosclerotic cardiovascular diseases are common and growing public health concerns. Previous epidemiological studies unfolded the robust correlation between obesity, NAFLD, and atherosclerotic cardiovascular diseases. Obesity is a well-known risk factor for NAFLD, and both of them can markedly increase the odds of atherosclerotic cardiovascular diseases. On the other hand, significant weight loss achieved by lifestyle modification, bariatric surgery, or medications, such as semaglutide, can concomitantly improve NAFLD and atherosclerotic cardiovascular diseases. Therefore, certain pathophysiological links are involved in the development of NAFLD in obesity, and atherosclerotic cardiovascular diseases in obesity and NAFLD. Moreover, recent studies indicated that simultaneously targeting several mechanisms by tirzepatide and retatrutide leads to greater weight loss and markedly improves the complications of metabolic syndrome. These findings remind the importance of a mechanistic viewpoint for breaking the association between obesity, NAFLD, and atherosclerotic cardiovascular diseases. In this review article, we mainly focus on shared pathophysiological mechanisms, including insulin resistance, dyslipidemia, GLP1 signaling, inflammation, oxidative stress, mitochondrial dysfunction, gut dysbiosis, renin-angiotensin-aldosterone system (RAAS) overactivity, and endothelial dysfunction. Most of these pathophysiological alterations are primarily initiated by obesity. The development of NAFLD further exacerbates these molecular and cellular alterations, leading to atherosclerotic cardiovascular disease development or progression as the final manifestation of molecular perturbation. A better insight into these mechanisms makes it feasible to develop new multi-target approaches to simultaneously unhinge the deleterious chain of events linking obesity and NAFLD to atherosclerotic cardiovascular diseases.

3.
Photobiomodul Photomed Laser Surg ; 41(11): 622-631, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37890115

RESUMEN

Background: Oral mucositis (OM) is a common adverse effect of radiation to the head and neck. Recent research has shown that extra oral photobiomodulation (EO-PBM) reduces the severity of OM. However, appropriate EO-PBM therapy parameters for OM severity reduction have not been documented. Objective: This work aims to optimize EO-PBM radiation parameters for lowering the severity of radiation-induced OM in rats by establishing a photobiomodulation (PBM) treatment system based on light-emitting diode arrays with top-hat beam profile. Methods: The 36 rats are separated into 2 control groups and 4 groups receiving PBM treatment. The PBM groups are exposed to irradiance between 4 and 24 J/cm2 at 660 nm. The cheek pouch mucosa is removed after scarification for biochemical and histological examination. Student's t-test, and one-way analysis of variance (ANOVA) followed by Tukey's Multiple were applied to compare the statistical significance of differences between control groups and PBM treatment groups. Results: Statistical analysis reveals that PBM irradiation at 12 J/cm2 (200 sec) with a flatness of 0.8 and a diameter of 3 cm substantially decreased the level of inflammatory cytokines compared with the positive control group. Conclusions: Our results indicate that the designed treatment PBM system is capable of delivering the optical parameters necessary for therapeutic treatment.


Asunto(s)
Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Terapia por Luz de Baja Intensidad , Estomatitis , Ratas , Animales , Estomatitis/etiología , Estomatitis/radioterapia , Terapia por Luz de Baja Intensidad/métodos , Citocinas
4.
Parkinsons Dis ; 2023: 2893662, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37664790

RESUMEN

Introduction: Despite remarkable progress in identifying Parkinson's disease (PD) genetic risk loci, the genetic basis of PD remains largely unknown. With the help of the endophenotype approach and using data from dopamine transporter single-photon emission computerized tomography (DaTscan), we identified potentially involved genes in PD. Method: We conducted an imaging genetic study by performing exome-wide association study (EWAS) and genome-wide association study (GWAS) on the specific binding ratio (SBR) of six DaTscan anatomical areas between 489 and 559 subjects of Parkinson's progression markers initiative (PPMI) cohort and 83,623 and 36,845 single-nucleotide polymorphisms (SNPs)/insertion-deletion mutations (INDELs). We also investigated the association of cerebrospinal fluid (CSF) protein concentration of our significant genes with PD progression using PPMI CSF proteome data. Results: Among 83,623 SNPs/INDELs in EWAS, one SNP (rs201465075) on 1 q32.1 locus was significantly (P value = 4.03 × 10-7) associated with left caudate DaTscan SBR, and 33 SNPs were suggestive. Among 36,845 SNPs in GWAS, one SNP (rs12450112) on 17 p.12 locus was significantly (P value = 1.34 × 10-6) associated with right anterior putamen DaTscan SBR, and 39 SNPs were suggestive among which 8 SNPs were intergenic. We found that rs201465075 and rs12450112 are most likely related to IGFN1 and MAP2K4 genes. The protein level of MAP2K4 in the CSF was significantly associated with PD progression in the PPMI cohort; however, proteomic data were not available for the IGFN1 gene. Conclusion: We have shown that particular variants of IGFN1 and MAP2K4 genes may be associated with PD. Since DaTscan imaging could be positive in other Parkinsonian syndromes, caution should be taken when interpreting our results. Future experimental studies are also needed to verify these findings.

5.
Front Endocrinol (Lausanne) ; 14: 1202560, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37476491

RESUMEN

Background: Male infertility is a multifaceted issue that has gained scientific interest due to its increasing rate. Studies have demonstrated that oxidative stress is involved in male infertility development. Furthermore, metabolic disorders, including obesity, diabetes, hypo- and hyperthyroidism, are risk factors for male infertility, and oxidative stress is believed to contribute to this association. Melatonin, functioning as an oxidative scavenger, may represent a promising therapeutic approach for the prevention and treatment of metabolic disorder-associated male infertility. Material and methods: We systematically searched three online databases (PubMed, Scopus, and Web of Science) for studies that evaluated the effects of melatonin therapy on metabolic disorders-induce infertility in male rodents. The favorable outcomes were histopathological parameters of testicular tissue, reproductive hormones, and markers of oxidative stress. Then, meta-analyses were done for each outcome. The results are reported as standardized mean difference (Cohen's d) and 95% confidence interval. Results: 24 studies with 31 outcomes were included. Rats and mice were the subjects. Studies have employed obesity, diabetes, hypothyroidism, hyperthyroidism, hyperlipidemia, and food deprivation as metabolic disorders. To induce these disorders, a high-fat diet, high-fructose diet, leptin, streptozotocin, alloxan, carbimazole, and levothyroxine were used. The outcomes included histopathologic characteristics (abnormal sperm morphology, apoptotic cells, apoptotic index, Johnsen's testicular biopsy score, seminiferous epithelial height, tubular basement membrane thickness, tubular diameter, sperm count, and motility), weight-related measurements (absolute epididymis, testis, and body weight, body weight gain, epididymal adipose tissue weight, and relative testis to body weight), hormonal characteristics (androgen receptor expression, serum FSH, LH, and testosterone level), markers of oxidative stress (tissue and serum GPx and MDA activity, tissue CAT, GSH, and SOD activity), and exploratory outcomes (serum HDL, LDL, total cholesterol, triglyceride, and blood glucose level). The overall pooled effect sizes were statistically significant for all histopathological characteristics and some markers of oxidative stress. Conclusions: Melatonin can reduce damage to male rodents' gonadal tissue and improve sperm count, motility, and morphology in metabolic diseases. Future clinical studies and randomized controlled trials are needed to evaluate the safety and effectiveness of melatonin for male infertility in patients with metabolic diseases.


Asunto(s)
Diabetes Mellitus , Hipertiroidismo , Infertilidad Masculina , Melatonina , Enfermedades Metabólicas , Animales , Masculino , Ratones , Ratas , Peso Corporal , Diabetes Mellitus/metabolismo , Hipertiroidismo/metabolismo , Infertilidad Masculina/tratamiento farmacológico , Infertilidad Masculina/etiología , Infertilidad Masculina/prevención & control , Melatonina/farmacología , Melatonina/uso terapéutico , Melatonina/metabolismo , Enfermedades Metabólicas/etiología , Enfermedades Metabólicas/complicaciones , Obesidad/complicaciones , Obesidad/tratamiento farmacológico , Estrés Oxidativo , Roedores , Semen , Testículo/metabolismo
6.
Sci Rep ; 13(1): 12401, 2023 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-37524849

RESUMEN

The term "geriatric giants" refers to the chronic disabilities of senescence leading to adverse health outcomes. This study aimed to investigate the prevalence and predictors of geriatric giants in Southern Iran. The participants were selected from Bushehr city using a multistage cluster random sampling method. Demographic data were collected through interviews. Frailty, incontinence, immobility, depression, cognitive impairment, and malnutrition were measured by questionnaires and instruments. Finally, data from 2392 participants were analyzed. The prevalence of fecal incontinence was less than 1% among all participants and similar in men and women. In contrast, compared with men, women had higher prevalence of urinary incontinence (36.44% vs. 17.65%), depression (39.05% vs. 12.89%), anorexia and malnutrition (2.35% vs. 0.82%), immobility (8.00% vs. 2.5%), frailty (16.84 vs. 7.34), and pre-frailty (54.19 vs. 38.63%). The prevalence of dependence and cognitive impairment was also higher in women and considerably increased with the age of participants. In total, 12.07% of subjects were frail, and 46.76% were pre-frail. The prevalence of frailty exponentially increased in older age, ranging from 4.18% among those aged 60-64 years to 57.35% in those aged ≥ 80 years. Considering 95% confidence interval (CI), multivariate logistic regression revealed that low physical activity [odds ratio (OR) 31.73 (18.44-54.60)], cancer (OR 3.28 (1.27-8.44)), depression [OR 2.42 (1.97-2.98)], age [OR 1.11 (1.08-1.14)], waist circumference [OR 1.03 (1.01-1.06)], BMI [OR 1.07 (1.01-1.14)], MNA score [OR 0.85 (0.79-0.92)], polypharmacy [OR 2.26 (1.30-3.95)] and male gender [OR 0.63 (0.42-0.93)] were independently associated with frailty. White blood cell count (WBC), smoking, marital status, and number of comorbidities were not independently associated with frailty. Low physical activity was the strongest predictor of frailty, which may need more attention in geriatric care. Frailty, its predictors, and other components of geriatric giants were considerably more common among women and older ages.


Asunto(s)
Fragilidad , Desnutrición , Anciano , Humanos , Masculino , Femenino , Fragilidad/complicaciones , Estudios Transversales , Prevalencia , Vida Independiente , Evaluación Geriátrica/métodos , Desnutrición/epidemiología , Medio Oriente
7.
Biomed Pharmacother ; 165: 115104, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37393866

RESUMEN

Hepatocellular carcinoma (HCC) is the second most lethal cancer and a leading cause of cancer-related mortality worldwide. Immune checkpoint inhibitors (ICIs) significantly improved the prognosis of HCC; however, the therapeutic response remains unsatisfactory in a substantial proportion of patients or needs to be further improved in responders. Herein, other methods of immunotherapy, including vaccine-based immunotherapy, adoptive cell therapy, cytokine delivery, kynurenine pathway inhibition, and gene delivery, have been adopted in clinical trials. Although the results were not encouraging enough to expedite their marketing. A major proportion of human genome is transcribed into non-coding RNAs (ncRNAs). Preclinical studies have extensively investigated the roles of ncRNAs in different aspects of HCC biology. HCC cells reprogram the expression pattern of numerous ncRNAs to decrease the immunogenicity of HCC, exhaust the cytotoxic and anti-cancer function of CD8 + T cells, natural killer (NK) cells, dendritic cells (DCs), and M1 macrophages, and promote the immunosuppressive function of T Reg cells, M2 macrophages, and myeloid-derived suppressor cells (MDSCs). Mechanistically, cancer cells recruit ncRNAs to interact with immune cells, thereby regulating the expression of immune checkpoints, functional receptors of immune cells, cytotoxic enzymes, and inflammatory and anti-inflammatory cytokines. Interestingly, prediction models based on the tissue expression or even serum levels of ncRNAs could predict response to immunotherapy in HCC. Moreover, ncRNAs markedly potentiated the efficacy of ICIs in murine models of HCC. This review article first discusses recent advances in the immunotherapy of HCC, then dissects the involvement and potential application of ncRNAs in the immunotherapy of HCC.


Asunto(s)
Antineoplásicos , Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Ratones , Animales , Carcinoma Hepatocelular/terapia , Carcinoma Hepatocelular/tratamiento farmacológico , Neoplasias Hepáticas/terapia , Neoplasias Hepáticas/tratamiento farmacológico , Inmunoterapia/métodos , ARN no Traducido/genética , ARN no Traducido/metabolismo , Antineoplásicos/uso terapéutico , Citocinas/metabolismo
8.
Ageing Res Rev ; 89: 101980, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37302757

RESUMEN

Mesenchymal stem cells (MSCs) are multipotent stromal cells with regenerative, anti-inflammatory, and immunomodulatory properties. MSCs and their exosomes significantly improved structural and functional alterations after myocardial infarction (MI) in preclinical studies and clinical trials. By reprograming intracellular signaling pathways, MSCs attenuate inflammatory response, oxidative stress, apoptosis, pyroptosis, and endoplasmic reticulum (ER) stress and improve angiogenesis, mitochondrial biogenesis, and myocardial remodeling after MI. MSC-derived exosomes contain a mixture of non-coding RNAs, growth factors, anti-inflammatory mediators, and anti-fibrotic factors. Although primary results from clinical trials were promising, greater efficacies can be achieved by controlling several modifiable factors. The optimum timing of transplantation, route of administration, origin of MSCs, number of doses, and number of cells per dose need to be further investigated by future studies. Newly, highly effective MSC delivery systems have been developed to improve the efficacy of MSCs and their exosomes. Moreover, MSCs can be more efficacious after being pretreated with non-coding RNAs, growth factors, anti-inflammatory or inflammatory mediators, and hypoxia. Similarly, viral vector-mediated overexpression of particular genes can augment the protective effects of MSCs on MI. Therefore, future clinical trials must consider these advances in preclinical studies to properly reflect the efficacy of MSCs or their exosomes for MI.


Asunto(s)
Exosomas , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Infarto del Miocardio , Humanos , Exosomas/metabolismo , Trasplante de Células Madre Mesenquimatosas/métodos , Infarto del Miocardio/terapia , Infarto del Miocardio/metabolismo , Miocardio/metabolismo , Células Madre Mesenquimatosas/metabolismo
9.
Obes Rev ; 24(6): e13563, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36951144

RESUMEN

Nonalcoholic fatty liver disease (NAFLD) is a growing health concern that is closely related to obesity and metabolic syndrome. In particular, NAFLD has been increasingly reported in adolescents and young adults in recent years. Cardiovascular diseases (CVDs) such as cardiac remodeling, heart failure, myocardial infarction, valvular heart diseases, and arrhythmia are more common in patients with NAFLD. CVD are the major cause of mortality in NAFLD. Although NAFLD often affects patients with obesity/overweight, it can also affect subjects with normal body mass index (BMI), known as lean NAFLD, which has a strong correlation with CVD. Obesity imposes a considerably increased risk of NAFLD and CVD. Consistently, weight-lowering approaches that can pronouncedly decrease body weight and maintain it in the long term, such as bariatric surgery and treatment with semaglutide and tirzepatide, have been promising in alleviating both CVD and NAFLD. Interestingly, compared with patients with NAFLD and obesity, a minimal amount of weight loss resolves NAFLD in lean patients. Besides the widespread use of bariatric surgery, the development of new GLP-1 agonists and GLP-1 GIP agonists revolutionized the treatment of obesity in recent years. Here, we discuss the interwoven correlation between obesity, NAFLD, and CVD and the benefits of weight-lowering approaches.


Asunto(s)
Enfermedades Cardiovasculares , Enfermedad del Hígado Graso no Alcohólico , Adolescente , Adulto Joven , Humanos , Enfermedad del Hígado Graso no Alcohólico/complicaciones , Enfermedad del Hígado Graso no Alcohólico/epidemiología , Enfermedades Cardiovasculares/epidemiología , Enfermedades Cardiovasculares/etiología , Obesidad/complicaciones , Pérdida de Peso , Péptido 1 Similar al Glucagón
10.
Pharmacol Rep ; 75(3): 623-633, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-36920684

RESUMEN

BACKGROUND: Mesenteric ischemia has remained without effective pharmacological management for many years. Sumatriptan, an abortive medication for migraine and cluster headaches, has potent anti-inflammatory properties and ameliorated organ ischemia in previous animal studies. Similarly, inhibition of the kynurenine pathway ameliorated renal and myocardial ischemia/reperfusion (I/R) in many preclinical studies. Herein, we assessed the effect of sumatriptan on experimental mesenteric I/R and investigated whether kynurenine pathway inhibition is a mechanism underlying its action. METHODS: Ischemia was induced by ligating the origin of the superior mesenteric artery (SMA) and its anastomosis with the inferior mesenteric artery (IMA) with bulldog clamps for 30 min. Ischemia was followed by 1 h of reperfusion. Sumatriptan (0.1, 0.3, and 1 mg/kg ip) was injected 5 min before the reperfusion phase, 1-methyltryptophan (1-MT) (100 mg/kg iv) was used to inhibit kynurenine production. At the end of the reperfusion phase, samples were collected from the jejunum of rats for H&E staining and molecular assessments. RESULTS: Sumatriptan improved the integrity of intestinal mucosa after I/R, and 0.1 mg/kg was the most effective dose of sumatriptan in this study. Sumatriptan decreased the increased levels of TNF-α, kynurenine, and p-ERK but did not change the decreased levels of NO. Furthermore, sumatriptan significantly increased the decreased ratio of Bcl2/Bax. Similarly, 1-MT significantly decreased TNF-α and kynurenine and protected against mucosal damage. CONCLUSIONS: This study demonstrated that sumatriptan has protective effects against mesenteric ischemia and the kynurenine inhibition is potentially involved in this process. Therefore, it can be assumed that sumatriptan has the potential to be repurposed as a treatment for acute mesenteric ischemia.


Asunto(s)
Isquemia Mesentérica , Daño por Reperfusión , Ratas , Animales , Isquemia Mesentérica/tratamiento farmacológico , Sumatriptán/farmacología , Sumatriptán/uso terapéutico , Quinurenina , Daño por Reperfusión/metabolismo , Factor de Necrosis Tumoral alfa , Isquemia
11.
J Diabetes Res ; 2023: 8776878, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36818747

RESUMEN

Sestrin2 regulates cell homeostasis and is an upstream signaling molecule for several signaling pathways. Sestrin2 leads to AMP-activated protein kinase- (AMPK-) and GTPase-activating protein activity toward Rags (GATOR) 1-mediated inhibition of mammalian target of rapamycin complex 1 (mTORC1), thereby enhancing autophagy. Sestrin2 also improves mitochondrial biogenesis via AMPK/Sirt1/peroxisome proliferator-activated receptor-gamma coactivator 1 alpha (PGC-1α) signaling pathway. Blockade of ribosomal protein synthesis and augmentation of autophagy by Sestrin2 can prevent misfolded protein accumulation and attenuate endoplasmic reticulum (ER) stress. In addition, Sestrin2 enhances P62-mediated autophagic degradation of Keap1 to release nuclear factor erythroid 2-related factor 2 (Nrf2). Nrf2 release by Sestrin2 vigorously potentiates antioxidant defense in diabetic nephropathy. Impaired autophagy and mitochondrial biogenesis, severe oxidative stress, and ER stress are all deeply involved in the development and progression of diabetic nephropathy. It has been shown that Sestrin2 expression is lower in the kidney of animals and patients with diabetic nephropathy. Sestrin2 knockdown aggravated diabetic nephropathy in animal models. In contrast, upregulation of Sestrin2 enhanced autophagy, mitophagy, and mitochondrial biogenesis and suppressed oxidative stress, ER stress, and apoptosis in diabetic nephropathy. Consistently, overexpression of Sestrin2 ameliorated podocyte injury, mesangial proliferation, proteinuria, and renal fibrosis in animal models of diabetic nephropathy. By suppressing transforming growth factor beta (TGF-ß)/Smad and Yes-associated protein (YAP)/transcription enhancer factor 1 (TEF1) signaling pathways in experimental models, Sestrin2 hindered epithelial-mesenchymal transition and extracellular matrix accumulation in diabetic kidneys. Moreover, modulation of the downstream molecules of Sestrin2, for instance, augmentation of AMPK or Nrf2 signaling and inhibition of mTORC1, has been protective in diabetic nephropathy. Regarding the beneficial effects of Sestrin2 on diabetic nephropathy and its interaction with several signaling molecules, it is worth targeting Sestrin2 in diabetic nephropathy.


Asunto(s)
Diabetes Mellitus , Nefropatías Diabéticas , Podocitos , Animales , Proteínas Quinasas Activadas por AMP/metabolismo , Biología , Diabetes Mellitus/metabolismo , Nefropatías Diabéticas/metabolismo , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Mamíferos/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Podocitos/metabolismo , Transducción de Señal , Sestrinas/metabolismo , Humanos
12.
J Int Med Res ; 50(10): 3000605221117146, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36314792

RESUMEN

The gastrointestinal tract is an uncommon site for metastasis. Gastrointestinal metastasis of melanoma is usually asymptomatic, often affects the small intestine (81.1%) and the colon (15.1%), and rarely affects the stomach. Our patient was a 40-year-old man presenting with gradually worsening dyspepsia of a few weeks' duration. He did not mention other gastrointestinal symptoms, and he was not anemic. He had a history of auricular melanoma, which was resected. A black lesion with a diameter of approximately 20 mm was detected in the body of his stomach during upper gastrointestinal endoscopy. Biopsies were taken, and the diagnosis of metastatic melanoma was confirmed by immunohistochemistry (IHC). A gastrointestinal work-up, computed tomography (CT), and positron-emission tomography (PET) did not reveal additional lesions. The lesion in the stomach was resected, and a new course of chemotherapy was initiated. A lower threshold should be considered for gastrointestinal work-up in patients with melanoma.


Asunto(s)
Dispepsia , Neoplasias Gastrointestinales , Melanoma , Neoplasias Primarias Secundarias , Masculino , Humanos , Adulto , Dispepsia/complicaciones , Melanoma/tratamiento farmacológico , Neoplasias Gastrointestinales/patología , Tomografía Computarizada por Rayos X/métodos , Tomografía de Emisión de Positrones
13.
Int J Tryptophan Res ; 15: 11786469221096643, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35784899

RESUMEN

Kynurenine pathway is the main route of tryptophan metabolism and produces several metabolites with various biologic properties. It has been uncovered that several cardiovascular diseases are associated with the overactivation of kynurenine pathway and kynurenine and its metabolites have diagnostic and prognostic value in cardiovascular diseases. Furthermore, it was found that several kynurenine metabolites can differently affect cardiovascular health. For instance, preclinical studies have shown that kynurenine, xanthurenic acid and cis-WOOH decrease blood pressure; kynurenine and 3-hydroxyanthranilic acid prevent atherosclerosis; kynurenic acid supplementation and kynurenine 3-monooxygenase (KMO) inhibition improve the outcome of stroke. Indoleamine 2,3-dioxygenase (IDO) overactivity and increased kynurenine levels improve cardiac and vascular transplantation outcomes, whereas exacerbating the outcome of myocardial ischemia, post-ischemic myocardial remodeling, and abdominal aorta aneurysm. IDO inhibition and KMO inhibition are also protective against viral myocarditis. In addition, dysregulation of kynurenine pathway is observed in several conditions such as senescence, depression, diabetes, chronic kidney disease (CKD), cirrhosis, and cancer closely connected to cardiovascular dysfunction. It is worth defining the exact effect of each metabolite of kynurenine pathway on cardiovascular health. This narrative review is the first review that separately discusses the involvement of kynurenine pathway in different cardiovascular diseases and dissects the underlying molecular mechanisms.

14.
J Cell Mol Med ; 26(16): 4556-4565, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35810384

RESUMEN

Radiation-induced oral mucositis is a common and dose-limiting complication of head and neck radiotherapy with no effective treatment. Previous studies revealed that sildenafil, a phosphodiesterase 5 inhibitor, has anti-inflammatory and anti-cancer effects. In this study, we investigated the effect of sildenafil on radiation-induced mucositis in rats. Two doses of radiation (8 and 26 Gy X-ray) were used to induce low-grade and high-grade oral mucositis, separately. A control group and three groups of sildenafil citrate-treated rats (5, 10, and 40 mg/kg/day) were used for each dose of radiation. Radiation increased MDA and activated NF-κB, ERK and JNK signalling pathways. Sildenafil significantly decreased MDA level, nitric oxide (NO) level, IL1ß, IL6 and TNF-α. The most effective dose of sildenafil was 40 mg/kg/day in this study. Sildenafil also significantly inhibited NF-κB, ERK and JNK signalling pathways and increased bcl2/bax ratio. In addition, high-dose radiation severely destructed the mucosal layer in histopathology and led to mucosal cell apoptosis in the TUNEL assay. Sildenafil significantly improved mucosal structure and decreased inflammatory cell infiltration after exposure to high-dose radiation and reduced apoptosis in the TUNEL assay. These findings show that sildenafil can improve radiation-induced oral mucositis and decrease the apoptosis of mucosal cells via attenuation of inflammation and oxidative stress.


Asunto(s)
FN-kappa B , Estomatitis , Animales , Apoptosis , FN-kappa B/metabolismo , Estrés Oxidativo , Ratas , Citrato de Sildenafil/farmacología , Citrato de Sildenafil/uso terapéutico , Estomatitis/tratamiento farmacológico , Estomatitis/etiología , Estomatitis/metabolismo
15.
Biomark Res ; 10(1): 29, 2022 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-35527284

RESUMEN

Sestrin2 is a conserved antioxidant, metabolism regulator, and downstream of P53. Sestrin2 can suppress oxidative stress and inflammation, thereby preventing the development and progression of cancer. However, Sestrin2 attenuates severe oxidative stress by activating nuclear factor erythroid 2-related factor 2 (Nrf2), thereby enhancing cancer cells survival and chemoresistance. Sestrin2 inhibits endoplasmic reticulum stress and activates autophagy and apoptosis in cancer cells. Attenuation of endoplasmic reticulum stress and augmentation of autophagy hinders cancer development but can either expedite or impede cancer progression under specific conditions. Furthermore, Sestrin2 can vigorously inhibit oncogenic signaling pathways through downregulation of mammalian target of rapamycin complex 1 (mTORC1) and hypoxia-inducible factor 1-alpha (HIF-1α). Conversely, Sestrin2 decreases the cytotoxic activity of T cells and natural killer cells which helps tumor cells immune evasion. Sestrin2 can enhance tumor cells viability in stress conditions such as glucose or glutamine deficiency. Cancer cells can also upregulate Sestrin2 during chemotherapy or radiotherapy to attenuate severe oxidative stress and ER stress, augment autophagy and resist the treatment. Recent studies unveiled that Sestrin2 is involved in the development and progression of several types of human cancer. The effect of Sestrin2 may differ depending on the type of tumor, for instance, several studies revealed that Sestrin2 protects against colorectal cancer, whereas results are controversial regarding lung cancer. Furthermore, Sestrin2 expression correlates with metastasis and survival in several types of human cancer such as colorectal cancer, lung cancer, and hepatocellular carcinoma. Targeted therapy for Sestrin2 or regulation of its expression by new techniques such as non-coding RNAs delivery and vector systems may improve cancer chemotherapy and overcome chemoresistance, metastasis and immune evasion that should be investigated by future trials.

16.
Cell Physiol Biochem ; 56(S1): 24-35, 2022 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-35263537

RESUMEN

BACKGROUND/AIMS: Colitis is a main presentation of inflammatory bowel disease (IBD) and yet, has no definitive cure. Currently, corticosteroids, anti-tumor necrosis factor (anti-TNF) agents and 5-aminosalicylic acid derivatives are prescribed for management of colitis. Except their failure rate, they are not always tolerated because of their severe adverse effects. Additive formulas with fewer adverse effects may improve the treatment of colitis. METHODS: In this study, colitis was induced with intra-rectal injection of three concentrations of acetic acid (4, 6 and 8 v/v). Each group received sodium selenite (0.5 mg/kg) or saline, gavaged on days 0 and 1 for treatment. Two days after induction of colitis, rats were sacrificed and the end part of their colons were resected for macroscopic and microscopic evaluation and molecular measurement. RESULTS: Sodium selenite improved macroscopic and microscopic view of the colon, decreased cryptitis, crypt abscess and inflammatory cells infiltration and partly maintained mucosal structure. Sodium selenite markedly reduced tissue levels of malondialdehyde (MDA), TNF-α and interferon γ (INF-γ) and decreased myeloperoxidase (MPO) activity. Treatment with sodium selenite also significantly downregulated IL17, IL22, indoleamine 2,3-dioxygenase (IDO1), and kynurenine levels. Western blotting revealed that sodium selenite prevented apoptosis by increasing bcl2/Bax ratio. Furthermore, our findings showed that sodium selenite significantly downregulated the upstream inflammatory molecules such as nuclear factor kappa B (NF-κB) and toll-like receptor 4 (TLR4) in colitis. CONCLUSION: These findings show that sodium selenite alleviates inflammatory response and oxidative stress and protects against colitis.


Asunto(s)
Colitis , FN-kappa B , Ácido Acético/toxicidad , Animales , Colitis/inducido químicamente , Colitis/tratamiento farmacológico , Colitis/metabolismo , Colon/metabolismo , Quinurenina/metabolismo , Quinurenina/farmacología , Quinurenina/uso terapéutico , FN-kappa B/metabolismo , Ratas , Transducción de Señal , Selenito de Sodio/metabolismo , Selenito de Sodio/farmacología , Selenito de Sodio/uso terapéutico , Receptor Toll-Like 4/metabolismo , Inhibidores del Factor de Necrosis Tumoral
17.
Oxid Med Cell Longev ; 2022: 1197061, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35126806

RESUMEN

BACKGROUND: Recent meta-analyses have shown that sodium-glucose cotransporter 2 (SGLT-2) inhibitors alleviate chronic kidney disease and acute kidney injury in diabetic patients. In this study, we aimed to investigate the effect of empagliflozin on renal ischemia/reperfusion (I/R) in nondiabetic rats and find the possible mechanisms. Experimental Approach. Eighteen male Wistar rats were randomly divided into three groups, including healthy control, ischemic control, and empagliflozin-treated group. Thirty minutes of bilateral renal ischemia was induced by clamping the renal hilum. Forty-eight hours after reopening the clamps, rats' blood samples and tissue specimens were collected. Empagliflozin 10 mg/kg was administered by gavage, 2 hours before ischemia and 24 hours after the first dose. RESULTS: I/R injury led to a significant rise in serum creatinine and blood urea nitrogen which was significantly decreased after treatment with empagliflozin. Empagliflozin also alleviated tubulointerstitial and glomerular damage and significantly decreased tissue histology scores. Empagliflozin decreased the increased levels of malondialdehyde, interleukin 1ß, and tumor necrosis factor α. SGLT2 inhibition increased the decreased expression of nuclear factor erythroid 2-related factor 2 and PPARG coactivator 1 alpha that conduct antioxidant defense and mitochondrial biogenesis, respectively. Furthermore, empagliflozin markedly increased LC3-II/LC3-I and bcl2/bax ratios, showing its beneficial effect on activation of autophagy and inhibition of apoptosis. Despite its effects on diabetic nephropathy, empagliflozin did not activate the Sestrin2/AMP-activated protein kinase pathway in this study. CONCLUSION: Empagliflozin improved renal I/R injury in nondiabetic rats in this study by promoting autophagy and mitochondrial biogenesis and attenuation of oxidative stress, inflammation, and apoptosis.


Asunto(s)
Antioxidantes/administración & dosificación , Autofagia/efectos de los fármacos , Compuestos de Bencidrilo/administración & dosificación , Glucósidos/administración & dosificación , Enfermedades Renales/complicaciones , Enfermedades Renales/tratamiento farmacológico , Biogénesis de Organelos , Daño por Reperfusión/complicaciones , Daño por Reperfusión/tratamiento farmacológico , Inhibidores del Cotransportador de Sodio-Glucosa 2/administración & dosificación , Animales , Apoptosis/efectos de los fármacos , Nitrógeno de la Urea Sanguínea , Creatinina/sangre , Modelos Animales de Enfermedad , Interleucina-1beta/metabolismo , Enfermedades Renales/sangre , Masculino , Malondialdehído/metabolismo , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Wistar , Daño por Reperfusión/sangre , Transducción de Señal/efectos de los fármacos , Resultado del Tratamiento , Factor de Necrosis Tumoral alfa/metabolismo
18.
Cancer Biol Ther ; 23(1): 34-50, 2022 12 31.
Artículo en Inglés | MEDLINE | ID: mdl-34978469

RESUMEN

C-Myc overexpression is a common finding in pancreatic cancer and predicts the aggressive behavior of cancer cells. It binds to the promoter of different genes, thereby regulating their transcription. C-Myc is downstream of KRAS and interacts with several oncogenic and proliferative pathways in pancreatic cancer. C-Myc enhances aerobic glycolysis in cancer cells and regulates glutamate biosynthesis from glutamine. It provides enough energy for cancer cells' metabolism and sufficient substrate for the synthesis of organic molecules. C-Myc overexpression is associated with chemoresistance, intra-tumor angiogenesis, epithelial-mesenchymal transition (EMT), and metastasis in pancreatic cancer. Despite its title, c-Myc is not "undruggable" and recent studies unveiled that it can be targeted, directly or indirectly. Small molecules that accelerate c-Myc ubiquitination and degradation have been effective in preclinical studies. Small molecules that hinder c-Myc-MAX heterodimerization or c-Myc/MAX/DNA complex formation can functionally inhibit c-Myc. In addition, c-Myc can be targeted through transcriptional, post-transcriptional, and translational modifications.


Asunto(s)
Neoplasias Pancreáticas , Proteínas Proto-Oncogénicas c-myc , Línea Celular Tumoral , Humanos , Oncogenes , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Neoplasias Pancreáticas
19.
Curr Diabetes Rev ; 18(8): e051121197762, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34749618

RESUMEN

Metformin is an old, inexpensive, and relatively safe anti-diabetic medication that can decrease the risk of several types of cancer in patients with diabetes. Recent meta-analyses have revealed that metformin markedly decreased the incidence of colorectal adenoma, advanced adenoma, and colorectal cancer (CRC) among patients with diabetes. Potential mechanisms by which metformin may decrease colorectal cancer risk include its effects on ameliorating intestinal inflammation and dysbiosis, suppressing major proliferative pathways, preventing DNA replication, accelerating tumor cells apoptosis, inhibiting intra-tumor angiogenesis and epithelial-mesenchymal transition (EMT), increasing tumor-infiltrating lymphocytes and CD68+ tumor-associated macrophages, and enhancing T cells cytotoxic activity. It is well-known that metformin can improve overall survival and CRC-specific survival among patients with diabetes and CRC. Interestingly, metformin decreases the incidence of colonic adenoma in patients with acromegaly and reduces the incidence of inflammatory bowel disease (IBD) among patients with diabetes, which can indirectly lower the risk of CRC. Results of phase II clinical trials have revealed that metformin can enhance the anti-cancer effects of chemotherapeutic agents, such as 5-Fluorouracil (5-FU) and irinotecan on refractory CRC. Furthermore, metformin decreases the risk of new polyps and adenomas in patients without diabetes. Regarding the results of previous preclinical and clinical studies, it is rational to assess the effect of metformin in normoglycemic patients with CRC and expand its clinical application for treating CRC or preventing it in a high-risk population.


Asunto(s)
Adenoma , Neoplasias Colorrectales , Metformina , Adenoma/epidemiología , Adenoma/patología , Adenoma/prevención & control , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/prevención & control , Fluorouracilo/farmacología , Fluorouracilo/uso terapéutico , Humanos , Hipoglucemiantes/farmacología , Hipoglucemiantes/uso terapéutico , Incidencia , Metformina/farmacología , Metformina/uso terapéutico
20.
Int Rev Immunol ; 41(3): 326-345, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34289794

RESUMEN

Tryptophan is an essential amino acid, going through three different metabolic pathways in the intestines. Indole pathway in the gut microbiota, serotonin system in the enterochromaffin cells and kynurenine pathway in the immune cells and intestinal lining are the three arms of tryptophan metabolism in the intestines. Clinical, in vivo and in vitro studies showed that each one of these arms has a significant impact on IBD. This review explains how different metabolites of tryptophan are involved in the pathophysiology of IBD and colorectal cancer, as a major complication of IBD. Indole metabolites alleviate colitis and protect against colorectal cancer while serotonin arm follows a more complicated and receptor-specific pattern. Indole metabolites and kynurenine interact with aryl hydrocarbon receptor (AHR) to induce T regulatory cells differentiation, confine Th17 and Th1 response and produce anti-inflammatory mediators. Kynurenine decreases tumor-infiltrating CD8+ cells and mediates tumor cells immune evasion. Serotonin system also increases colorectal cancer cells proliferation and metastasis while, indole metabolites can profoundly decrease colorectal cancer growth. Targeted therapy for tryptophan metabolites may improve the management of IBD and colorectal cancer, e.g. supplementation of indole metabolites such as indole-3-carbinol (I3C), inhibition of kynurenine monooxygenase (KMO) and selective stimulation or inhibition of specific serotonergic receptors can mitigate colitis. Furthermore, it will be explained how indole metabolites supplementation, inhibition of indoleamine 2,3-dioxygenase 1 (IDO1), KMO and serotonin receptors can protect against colorectal cancer. Additionally, extensive molecular interactions between tryptophan metabolites and intracellular signaling pathways will be thoroughly discussed.


Asunto(s)
Colitis , Neoplasias Colorrectales , Enfermedades Inflamatorias del Intestino , Colitis/tratamiento farmacológico , Colitis/metabolismo , Humanos , Indoles/metabolismo , Indoles/uso terapéutico , Enfermedades Inflamatorias del Intestino/metabolismo , Quinurenina/metabolismo , Quinurenina/uso terapéutico , Serotonina/metabolismo , Serotonina/uso terapéutico , Células Th17 , Triptófano/metabolismo , Triptófano/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA