Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 373
Filter
1.
Oncol Rep ; 48(5)2022 11.
Article in English | MEDLINE | ID: mdl-36177901

ABSTRACT

Colorectal cancer (CRC) is one of the most common malignancies worldwide. Patients with CRC may need chemotherapy (CTx) in a neoadjuvant, adjuvant or palliative setting through the course of the disease. Unfortunately, its effect is limited by chemoresistance and chemotoxicity. Novel more effective and non­toxic CTx regimens are needed to further improve CRC treatment outcomes. Thus, the present study was designed to test the hypothesis that non­toxic sulforaphane (SF) is effective against CRC and has additive effects in combination with conventional 5­fluorouracil, oxaliplatin and folinic acid (FOLFOX) CTx in vitro. Highly metastatic human colon cancer cells, CX­1, and fibroblasts were treated with FOLFOX ± SF. Cell viability was assessed using an MTT assay. The level of apoptosis and the expression of apoptotic proteins were measured by TUNEL assay and quantitative PCR analysis. Aldehyde dehydrogenase isoform 1 (ALDH1) and multidrug resistance protein 2 (MRP2) levels were evaluated. The ability of cells to form spheroids was measured in three­dimensional cell culture. SF alone and in combination with FOLFOX effectively decreased the viability of the CX­1 cells, promoted apoptosis within the CX­1 cells, prevented cellular spheroid formation and decreased ALDH1 activity. However, SF promoted MRP2 expression and protein levels. In conclusion, SF together with conventional FOLFOX has additive anticancer effects against highly metastatic human CRC in vitro.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Carcinoma , Colonic Neoplasms , Isothiocyanates , Sulfoxides , Aldehyde Dehydrogenase 1 Family , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma/drug therapy , Colonic Neoplasms/drug therapy , Fluorouracil/therapeutic use , Humans , Isothiocyanates/therapeutic use , Leucovorin/therapeutic use , Organoplatinum Compounds/therapeutic use , Oxaliplatin/therapeutic use , Sulfoxides/therapeutic use
2.
Cell Stress Chaperones ; 27(5): 499-511, 2022 09.
Article in English | MEDLINE | ID: mdl-35779187

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is a major health concern. Endoplasmic reticulum (ER) stress, inflammation, and metabolic dysfunctions may be targeted to prevent the progress of nonalcoholic fatty liver disease. Sulforaphane (SFN), a sulfur-containing compound that is abundant in broccoli florets, seeds, and sprouts, has been reported to have beneficial effects on attenuating metabolic diseases. In light of this, the present study was designed to elucidate the mechanisms by which SFN ameliorated ER stress, inflammation, lipid metabolism, and insulin resistance - induced by a high-fat diet and ionizing radiation (IR) in rats. In our study, the rats were randomly divided into five groups: control, HFD, HFD + SFN, HFD + IR, and HFD + IR + SFN groups. After the last administration of SFN, liver and blood samples were taken. As a result, the lipid profile, liver enzymes, glucose, insulin, IL-1ß, adipokines (leptin and resistin), and PI3K/AKT protein levels, as well as the mRNA gene expression of ER stress markers (IRE-1, sXBP-1, PERK, ATF4, and CHOP), fatty acid synthase (FAS), peroxisome proliferator-activated receptor-α (PPAR-α). Interestingly, SFN treatment modulated the levels of proinflammatory cytokine including IL-1ß, metabolic indices (lipid profile, glucose, insulin, and adipokines), and ER stress markers in HFD and HFD + IR groups. SFN also increases the expression of PPAR-α and AMPK genes in the livers of HFD and HFD + IR groups. Meanwhile, the gene expression of FAS and CHOP was significantly attenuated in the SFN-treated groups. Our results clearly show that SFN inhibits liver toxicity induced by HFD and IR by ameliorating the ER stress events in the liver tissue through the upregulation of AMPK and PPAR-α accompanied by downregulation of FAS and CHOP gene expression.


Subject(s)
Endoplasmic Reticulum Stress , Insulins , Isothiocyanates , Non-alcoholic Fatty Liver Disease , Sulfoxides , AMP-Activated Protein Kinases/genetics , Animals , Cytokines/metabolism , Diet, High-Fat , Endoplasmic Reticulum Stress/drug effects , Fatty Acid Synthases/genetics , Fatty Acid Synthases/metabolism , Fatty Acid Synthases/pharmacology , Glucose/metabolism , Insulins/genetics , Insulins/metabolism , Insulins/pharmacology , Isothiocyanates/pharmacology , Isothiocyanates/therapeutic use , Leptin , Lipids/blood , Liver/drug effects , Liver/metabolism , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Peroxisome Proliferator-Activated Receptors/genetics , Peroxisome Proliferator-Activated Receptors/metabolism , Peroxisome Proliferator-Activated Receptors/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/metabolism , Rats , Resistin/genetics , Resistin/metabolism , Resistin/pharmacology , Sulfoxides/pharmacology , Sulfoxides/therapeutic use , Up-Regulation
3.
J Immunother Cancer ; 10(7)2022 07.
Article in English | MEDLINE | ID: mdl-35790315

ABSTRACT

BACKGROUND: Targeting the DNA damage repair (DDR) pathways is an attractive strategy for boosting cancer immunotherapy. Ceralasertib (AZD6738) is an oral kinase inhibitor of ataxia telangiectasia and Rad3 related protein, which is a master regulator of DDR. We conducted a phase II trial of ceralasertib plus durvalumab in patients with previously treated advanced gastric cancer (AGC) to demonstrate the safety, tolerability, and clinical activity of the combination. METHODS: This phase II, open-label, single-center, non-randomized study was designed to evaluate the efficacy and safety of ceralasertib in combination with durvalumab in patients with AGC. The study drug regimen was ceralasertib (240 mg two times a day) days 15-28 in a 28-day cycle in combination with durvalumab (1500 mg) at day 1 every 4 weeks. The primary end point was overall response rate (ORR) by Response Evaluation Criteria in Solid Tumors (V.1.1). Exploratory biomarker analysis was performed using fresh tumor biopsies in all enrolled patients. RESULTS: Among 31 patients, the ORR, disease control rate, median progression-free survival (PFS), and overall survival were 22.6% (95% CI 9.6% to 41.1%), 58.1% (95% CI 39.1% to 75.5%), 3.0 (95% CI 2.1 to 3.9) months, and 6.7 (95% CI 3.8 to 9.6) months, respectively. Common adverse events were manageable with dose modification. A subgroup of patients with a loss of ataxia telangiectasia mutated (ATM) expression and/or high proportion of mutational signature attributable to homologous repair deficiency (sig. HRD) demonstrated a significantly longer PFS than those with intact ATM and low sig. HRD (5.60 vs 1.65 months; HR 0.13, 95% CI 0.045 to 0.39; long-rank p<0.001). During the study treatment, upregulation of the innate immune response by cytosolic DNA, activation of intratumoral lymphocytes, and expansion of circulating tumor-reactive CD8 +T cell clones were identified in responders. Enrichment of the tumor vasculature signature was associated with treatment resistance. CONCLUSIONS: Ceralasertib plus durvalumab has promising antitumor activity, with durable responses in patients with refractory AGC. Thus, a biomarker-driven trial is required. TRIAL REGISTRATION: NCT03780608.


Subject(s)
Antibodies, Monoclonal , Antineoplastic Combined Chemotherapy Protocols , Protein Kinase Inhibitors , Stomach Neoplasms , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Agents, Immunological/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Ataxia Telangiectasia Mutated Proteins/genetics , Humans , Indoles/administration & dosage , Indoles/therapeutic use , Morpholines/administration & dosage , Morpholines/therapeutic use , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/administration & dosage , Pyrimidines/therapeutic use , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Sulfonamides/administration & dosage , Sulfonamides/therapeutic use , Sulfoxides/administration & dosage , Sulfoxides/therapeutic use
4.
Biomed Pharmacother ; 152: 113273, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35709656

ABSTRACT

Insulin resistance is a characteristic feature of type 2 diabetes. Sulforaphane (SFN) is a natural antioxidant extracted from the cruciferous vegetables. Recent study reported that SFN exhibits excellent anti-diabetic effects, however, the underlying mechanism is still unclear. This study aimed to investigate the therapeutic effects of SFN on a high-fat diet (HFD)-induced insulin resistance and potential mechanism. SFN was found to effectively reduce body weight, fasting blood glucose and hyperlipidemia, and improve liver function in HFD-fed mice. Furthermore, SFN effectively increased glucose uptake and improved insulin signaling in palmitic acid (PA)-induced HepG2 cells. SFN also led to increased expression of antioxidant genes downstream of Nrf2 and decreased accumulation of lipid peroxides MDA and 4-HNE, both in vivo and in vitro. Further studies revealed that SFN significantly reduced glutathione peroxidase 4 (GPx4) inactivation-mediated oxidative stress by activating the AMPK and Nrf2 signaling pathways. In PA-induced HepG2 cells and flies, the alleviation of insulin resistance by SFN was diminished by GPx4 inhibitor. Taken together, SFN ameliorated HFD-induced insulin resistance by activating the AMPK-Nrf2-GPx4 pathway, providing new insights into SFN as a therapeutic compound for the alleviation of insulin resistance.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin Resistance , Isothiocyanates , Sulfoxides , Animals , Mice , AMP-Activated Protein Kinases/metabolism , Antioxidants/pharmacology , Diabetes Mellitus, Type 2/drug therapy , Diet, High-Fat/adverse effects , Isothiocyanates/pharmacology , Isothiocyanates/therapeutic use , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Sulfoxides/pharmacology , Sulfoxides/therapeutic use
5.
Commun Biol ; 5(1): 242, 2022 03 18.
Article in English | MEDLINE | ID: mdl-35304580

ABSTRACT

Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), the cause of coronavirus disease 2019 (COVID-19), has incited a global health crisis. Currently, there are limited therapeutic options for the prevention and treatment of SARS-CoV-2 infections. We evaluated the antiviral activity of sulforaphane (SFN), the principal biologically active phytochemical derived from glucoraphanin, the naturally occurring precursor present in high concentrations in cruciferous vegetables. SFN inhibited in vitro replication of six strains of SARS-CoV-2, including Delta and Omicron, as well as that of the seasonal coronavirus HCoV-OC43. Further, SFN and remdesivir interacted synergistically to inhibit coronavirus infection in vitro. Prophylactic administration of SFN to K18-hACE2 mice prior to intranasal SARS-CoV-2 infection significantly decreased the viral load in the lungs and upper respiratory tract and reduced lung injury and pulmonary pathology compared to untreated infected mice. SFN treatment diminished immune cell activation in the lungs, including significantly lower recruitment of myeloid cells and a reduction in T cell activation and cytokine production. Our results suggest that SFN should be explored as a potential agent for the prevention or treatment of coronavirus infections.


Subject(s)
Antiviral Agents/therapeutic use , Common Cold/drug therapy , Coronavirus Infections/drug therapy , Coronavirus OC43, Human , Isothiocyanates/therapeutic use , SARS-CoV-2 , Sulfoxides/therapeutic use , Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/therapeutic use , Alanine/analogs & derivatives , Alanine/therapeutic use , Animals , Caco-2 Cells , Chlorocebus aethiops , Common Cold/virology , Coronavirus Infections/immunology , Coronavirus Infections/virology , Cytokines/immunology , Drug Synergism , Humans , Lung/immunology , Lung/virology , Macrophages, Alveolar/immunology , Male , Mice, Transgenic , Spleen/immunology , T-Lymphocytes/immunology , Vero Cells , Viral Load , COVID-19 Drug Treatment
6.
Eur J Pharmacol ; 919: 174824, 2022 Mar 15.
Article in English | MEDLINE | ID: mdl-35157913

ABSTRACT

Sulforaphane (SFN), the main ingredient in broccoli/broccoli sprouts, has a good anticancer effect in a wide variety of tumors, but whether SFN affects acute leukemia is not elucidated. Due to the self-renewal capability for leukemia stem cells, acute leukemia has a high relapse rate. This study explored the effects and related molecular mechanisms of SFN on the proliferation of leukemia stem-like cells in acute myeloid leukemia cells. We found that SFN inhibited the proliferation of leukemia stem-like cells in vitro and in vivo. Meanwhile, we observed that SFN could regulate the stem characteristic of leukemia cells. After SFN treatment, the expression of the key players in the Sonic Hedgehog (Shh) signaling pathway was significantly decreased at the transcriptional and protein levels. To further determine the contribution of the Shh signaling molecular mechanism to SFN-mediated self-renewal capability of LSCs, we then manipulated the Shh gene in the leukemia cells to either overexpress the gene using lentiviral vector transduction or knockdown the gene via siRNA. The results demonstrated that SFN suppressed proliferation in Shh-overexpressed cells more than in Shh-downregulated cells, suggesting that SFN negatively modulates proliferation of leukemia stem-like cells via affecting the Shh signaling pathway. Altogether, these results suggest that SFN is a potent anti-leukemia agent that has inhibitory effects on leukemia stem-like cells' proliferation by regulating the Shh signaling pathway.


Subject(s)
Anticarcinogenic Agents/pharmacology , Isothiocyanates/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Signal Transduction/drug effects , Sulfoxides/pharmacology , Animals , Anticarcinogenic Agents/chemistry , Anticarcinogenic Agents/therapeutic use , Cell Line, Tumor/drug effects , Cell Proliferation/drug effects , Hedgehog Proteins/metabolism , Humans , Isothiocyanates/chemistry , Isothiocyanates/therapeutic use , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/drug effects , Sulfoxides/chemistry , Sulfoxides/therapeutic use , Xenograft Model Antitumor Assays
7.
J Food Biochem ; 46(3): e13886, 2022 03.
Article in English | MEDLINE | ID: mdl-34350614

ABSTRACT

Sulforaphane belongs to the active class of isothiocyanates capable of delivering various biological benefits for health promotion and disease prevention. This compound is considered vital to curtail numerous metabolic disorders. Various studies have proven its beneficial effects against cancer prevention and its possible utilization as a therapeutic agent in cancer treatment. Understanding the mechanistic pathways and possible interactions at cellular and subcellular levels is key to design and develop cancer therapeutics for humans. In this respect, a number of mechanisms such as modulation of carcinogen metabolism & phase II enzymatic activities, cell cycle arrest, activation of Nrf2, cytotoxic, proapoptotic and apoptotic pathways have been reported to be involved in cancer prevention. This article provides sufficient information by critical analysis to understand the mechanisms involved in cancer prevention attributed to sulforaphane. Furthermore, various clinical studies have also been included for design and development of novel therapies for cancer prevention and cure. PRACTICAL APPLICATIONS: Diet and dietary components are potential tools to address various lifestyle-related disorders. Due to plenty of environmental and cellular toxicants, the chances of cancer prevalence are quite large which are worsen by adopting unhealthy lifestyles. Cancer can be treated with various therapies but those are acquiring side effects causing the patients to suffer the treatment regime. Nutraceuticals and functional foods provide safer options to prevent or delay the onset of cancer. In this regard, sulforaphane is a pivotal compound to be targeted as a potential agent for cancer treatment both in preventive and therapeutic regimes. This article provides sufficient evidence via discussing the underlying mechanisms of positive effects of sulforaphane to further the research for developing anticancer drugs that will help assuage this lethal morbidity.


Subject(s)
Antineoplastic Agents , Neoplasms , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Cycle Checkpoints , Humans , Isothiocyanates/pharmacology , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/prevention & control , Sulfoxides/pharmacology , Sulfoxides/therapeutic use
8.
Biomed Pharmacother ; 146: 112598, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34959120

ABSTRACT

Colorectal cancer (CRC) is a significant global health burden that ranks as the third most diagnosed and second most common cause of cancer related deaths worldwide. New therapeutic strategies include chemoprevention and use of molecules which could prevent, suppress or reverse CRC progression such as sulforaphane (SFN). However, evidences about its safety in CRC patients are still lacking. The aim of this in silico investigation was to predict SFN-induced adverse effects in CRC patients by computational analysis. The study showed that 334 genes were consistently dysregulated in CRC (223 downregulated and 111 upregulated), while 38 were recognized as significant and might be used as predictive biomarkers for overall survival and metastasis (TCGA, GEO, R studio). Among them, SFN interacted with 86 genes, out of which 11 were marked as significant (correlate with overall prognosis and metastasis). Sulforaphane potentiates the overexpression of TIMP1, AURKA, and CEP55, and promotes inhibition of CRYAB, PLCE1, and MMP28, that might lead to the progression of CRC (CTD). Pathway enrichment analysis revealed that SFN stimulated Transcriptional activation of RUNX2, AURKA activation by TPX2, IL-10 signaling, while inhibited Differentiation of White and Brown Adipocyte process, an underlying pathway which inactivation led to obesity (Cytoscape ClueGo + CluePedia, DAVID). Thus, genome signature of CRC patients could serve as important factor when addressing the risk-to-benefit profile of SFN. Patients with colon cancer and increased expression of TIMP1, CCL20, SPP1, AURKA, CEP55, NEK2, SOX9 and CDK1, or downregulation of CRYAB, PLCE1, MMP28, BMP2 and PLAC8 may not be ideal candidates for SFN chemoprevention.


Subject(s)
Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , Isothiocyanates/adverse effects , Sulfoxides/adverse effects , Adipocytes/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Regulatory Networks , Humans , Isothiocyanates/therapeutic use , Neoplasm Metastasis/genetics , Prognosis , Protein Array Analysis , Sulfoxides/therapeutic use , Survival Analysis , Transcriptional Activation/drug effects
9.
Eur J Pharmacol ; 909: 174412, 2021 Oct 15.
Article in English | MEDLINE | ID: mdl-34375671

ABSTRACT

Due to the efficacy and tolerability of the available drugs, the current treatment for cancer-induced bone pain (CIBP) is not considered ideal, and new drugs are required for better treatment results. This study investigated whether intrathecal injection of sulforaphane (SFN) can modulates the noxious behavior associated with CIBP and enhances the analgesic effects of morphine and the possible mechanisms related to these effects were investigated. Walker256 breast cancer cells were injected into the bone marrow cavity of rats to establish the CIBP model. When CIBP rats began to exhibit painful behavior (CIBP 6 days), SFN was injected intrathecally for 7 days. The results showed that SFN alleviated the painful behavioral hypersensitivity caused by cancer, accompanied by nuclear factor, erythroid 2 like 2 (Nrf2), Haem oxygenase 1 (HO-1) activation, nuclear factor kappa B (NF-κB) inhibition and inflammation-related factors (tumor necrosis factor-alpha (TNF-α), interleukin-1ß (IL-ß), interleukin-6 (IL-6), and inducible nitric oxide synthase (iNOS) reduction. In addition, SFN treatment inhibited the proliferation of Walker 256 cells in a dose-dependent manner, promoted mu-opioid receptor (MOR) expression in SH-SY5Y cells and enhanced the antihyperalgesic effects of morphine on CIBP rats by restoring the downregulation of MOR expression in the spinal cord. Interestingly, the antihyperalgesic effects of SFN were partially blocked by opioid receptor antagonists. This study showed that SFN combined with morphine might be a new way to treat CIBP.


Subject(s)
Bone Neoplasms/complications , Cancer Pain/drug therapy , Hyperalgesia/drug therapy , Isothiocyanates/pharmacology , Morphine/pharmacology , Sulfoxides/pharmacology , Animals , Bone Neoplasms/secondary , Cancer Pain/etiology , Cell Line, Tumor , Disease Models, Animal , Drug Synergism , Drug Therapy, Combination , Female , Humans , Hyperalgesia/etiology , Isothiocyanates/therapeutic use , Morphine/therapeutic use , Rats , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/metabolism , Spinal Cord Dorsal Horn/drug effects , Spinal Cord Dorsal Horn/metabolism , Sulfoxides/therapeutic use
10.
Metab Brain Dis ; 36(7): 1641-1671, 2021 10.
Article in English | MEDLINE | ID: mdl-34338974

ABSTRACT

Early diagnosis and treatment for autism spectrum disorder (ASD) pose challenges. The current diagnostic approach for ASD is mainly clinical assessment of patient behaviors. Biomarkers-based identification of ASD would be useful for pediatricians. Currently, there is no specific treatment for ASD, and evidence for the efficacy of alternative treatments remains inconclusive. The prevalence of ASD is increasing, and it is becoming more urgent to find the pathogenesis of such disorder. Metabolomic studies have been used to deeply investigate the alteration of metabolic pathways, including those associated with ASD. Metabolomics is a promising tool for identifying potential biomarkers and possible pathogenesis of ASD. This review comprehensively summarizes and discusses the abnormal metabolic pathways in ASD children, as indicated by evidence from metabolomic studies in urine and blood. In addition, the targeted interventions that could correct the metabolomic profiles relating to the improvement of autistic behaviors in affected animals and humans have been included. The results revealed that the possible underlying pathophysiology of ASD were alterations of amino acids, reactive oxidative stress, neurotransmitters, and microbiota-gut-brain axis. The potential common pathways shared by animal and human studies related to the improvement of ASD symptoms after pharmacological interventions were mammalian-microbial co-metabolite, purine metabolism, and fatty acid oxidation. The content of this review may contribute to novel biomarkers for the early diagnosis of ASD and possible therapeutic paradigms.


Subject(s)
Autism Spectrum Disorder/metabolism , Metabolomics , Animals , Autism Spectrum Disorder/diagnosis , Autism Spectrum Disorder/drug therapy , Biomarkers/blood , Biomarkers/urine , Humans , Isothiocyanates/therapeutic use , Metabolic Networks and Pathways , Sulfoxides/therapeutic use , Suramin/therapeutic use
11.
Int J Immunopathol Pharmacol ; 35: 20587384211034086, 2021.
Article in English | MEDLINE | ID: mdl-34344210

ABSTRACT

INTRODUCTION: Cruciferous vegetables are a rich source of sulforaphane (SFN), which acts as a natural HDAC inhibitor (HDACi). Our previous study found that HDACi could restore histone acetyltransferase/histone deacetylase (HAT/HDAC) balance in the cochlea and attenuate gentamicin-induced hearing loss in guinea pigs. Here, we investigated the protective effect of SFN on cisplatin-induced hearing loss (CIHL). METHODS: Thirty rats were randomly divided into 3 equal groups: the control group, cisplatin group, and SFN+cisplatin group. Rats were injected with SFN (30 mg/kg once a day) and cisplatin (7 mg/kg twice a day) for 7 days to investigate the protective role of SFN on CIHL. We observed auditory brainstem response (ABR) threshold shifts and immunostained cochlear basilar membranes of rats. For in vitro experiments, we treated HEI-OC1 cells and rat cochlear organotypic cultures with SFN (5, 10, and 15 µM) and cisplatin (10 µM). Immunofluorescence, cell viability, and protein analysis were performed to further analyze the protective mechanism of SFN on CIHL. RESULTS: SFN (30 mg/kg once a day) decreased cisplatin (7 mg/kg twice a day)-induced ABR threshold shifts and outer hair cell loss. CCK-8 assay showed that cisplatin (10 µM) reduced the viability of HEI-OC1 cells to 42%, and SFN had a dose-dependent protective effect. In cochlear organotypic cultures, we found that SFN (10 and 15 µM) increased cisplatin (10 µM)-induced myosin 7a+ cell count and restored ciliary morphology. SFN (5, 10, and 15 µM) reversed the cisplatin (10 µM)-induced increase in HDAC2, -4, and -5 and SFN (15 µM) reversed the cisplatin (10 µM)-induced decrease in H3-Ack9 [acetyl-histone H3 (Lys9)] protein expression in HEI-OC1 cells. Neither cisplatin nor cisplatin combined with SFN affected the expression of HDAC7, or HDAC9. CONCLUSION: SFN prevented disruption of the HAT/HDAC balance, protecting against CIHL in rats.


Subject(s)
Antineoplastic Agents , Cisplatin , Hearing Loss/chemically induced , Hearing Loss/drug therapy , Histone Deacetylase Inhibitors/therapeutic use , Isothiocyanates/therapeutic use , Sulfoxides/therapeutic use , Animals , Cell Count , Cilia/pathology , Cochlea/pathology , Dose-Response Relationship, Drug , Evoked Potentials, Auditory, Brain Stem/drug effects , Hair Cells, Auditory, Outer/pathology , Histone Deacetylases/biosynthesis , Histone Deacetylases/drug effects , Histone Deacetylases/genetics , Rats , Rats, Wistar
12.
Carbohydr Polym ; 269: 118294, 2021 Oct 01.
Article in English | MEDLINE | ID: mdl-34294320

ABSTRACT

Breast cancer stem cell (BCSC) properties are correlated with the malignancy of tumor cells. Sulforaphane (SFN), a natural isothiocyanate, has anti-cancer effects. However, SFN is an oil-like, hydrophobic and unstable substance. To enhance the inhibitory effect of SFN on BCSC-like properties, the mineralized hyaluronic acid-SS-tetradecyl nano-carriers (M-HA-SS-TA) were prepared. The nano-carriers possessed high SFN entrapment rate (92.36%) and drug-loading efficiency (33.64%). The carriers were responsive to the high reducing and mild acidic tumor micro-environment, leading to rapid SFN releasing from SFN-loaded nano-drug (SFN/M-HA-SS-TA). Through the specific recognition of breast cancer cells bearing CD44+ by HA, M-HA-SS-TA nano-carriers showed excellent tumor-targeting ability. Moreover, compared with free SFN, SFN/M-HA-SS-TA showed much stronger inhibition on the BCSC-like properties (invasiveness, self-renewal and tumor growth) both in vitro and in vivo. Together, these results suggested M-HA-SS-TA nano-carriers were promising platforms for tumor-targeted delivery of SFN, enhancing the therapeutic efficacy against BCSC-like properties by SFN.


Subject(s)
Breast Neoplasms/drug therapy , Disulfides/chemistry , Drug Carriers/chemistry , Hyaluronic Acid/analogs & derivatives , Isothiocyanates/therapeutic use , Nanoparticles/chemistry , Sulfoxides/therapeutic use , Animals , Disulfides/chemical synthesis , Disulfides/metabolism , Drug Carriers/chemical synthesis , Drug Carriers/metabolism , Drug Liberation , Female , Glutathione/metabolism , Humans , Hyaluronic Acid/metabolism , Hydrogen-Ion Concentration , Isothiocyanates/chemistry , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/metabolism , Sulfoxides/chemistry , Xenograft Model Antitumor Assays
13.
Int J Mol Sci ; 22(11)2021 May 31.
Article in English | MEDLINE | ID: mdl-34073079

ABSTRACT

Sulforaphane (SFN) is a natural glucosinolate found in cruciferous vegetables that acts as a chemopreventive agent, but its mechanism of action is not clear. Due to antioxidative mechanisms being thought central in preventing cancer progression, SFN could play a role in oxidative processes. Since redox imbalance with increased levels of reactive oxygen species (ROS) is involved in the initiation and progression of bladder cancer, this mechanism might be involved when chemoresistance occurs. This review summarizes current understanding regarding the influence of SFN on ROS and ROS-related pathways and appraises a possible role of SFN in bladder cancer treatment.


Subject(s)
Anticarcinogenic Agents , Antioxidants , Carcinoma/drug therapy , Isothiocyanates , Sulfoxides , Urinary Bladder Neoplasms/drug therapy , Animals , Anticarcinogenic Agents/pharmacology , Anticarcinogenic Agents/therapeutic use , Antioxidants/pharmacology , Antioxidants/therapeutic use , Cell Line, Tumor , Humans , Isothiocyanates/pharmacology , Isothiocyanates/therapeutic use , Kelch-Like ECH-Associated Protein 1/metabolism , NF-E2-Related Factor 2/metabolism , Reactive Oxygen Species/metabolism , Sulfoxides/pharmacology , Sulfoxides/therapeutic use
14.
Mol Med Rep ; 24(2)2021 Aug.
Article in English | MEDLINE | ID: mdl-34184072

ABSTRACT

Osteoarthritis (OA), the most common form of human joint disease, is characterized by progressive degeneration of the articular cartilage, synovitis and subchondral osteoporosis. Chondrocyte apoptosis is the primary pathogenic mechanism of OA and is considered to be a potential therapeutic target. Sulforaphane (SFN), a dietary isothiocyanate obtained from cruciferous vegetables, has been reported to exert an anti­apoptotic effect by activating sirtuin 1 (SIRT1). To the best of our knowledge, however, the effects of SFN on apoptotic responses in OA have not been reported. In the present study, SFN was shown to significantly inhibit chondrocyte apoptosis while enhancing expression levels of SIRT1 in a H2O2­induced OA mouse model. The anti­apoptotic effect of SFN was reversed by SIRT1 small interfering RNA, implying that SIRT1 exerted a protective role against the effect of SFN on chondrocytes. The expression levels of C/EBP homologous protein, 78­kDa glucose regulated protein, Bax, Bcl­2 and cleaved caspase 3 were found to be downregulated in SFN­treated mice. Furthermore, SFN ameliorated cartilage degradation in the OA mouse model. These findings indicate that SFN exerted an anti­apoptotic effect on chondrocytes and ameliorated OA in vivo by activating the SIRT1 signaling pathway.


Subject(s)
Isothiocyanates/pharmacology , Isothiocyanates/therapeutic use , Osteoarthritis/prevention & control , Protective Agents/pharmacology , Protective Agents/therapeutic use , Sirtuin 1/metabolism , Sulfoxides/pharmacology , Sulfoxides/therapeutic use , Animals , Apoptosis/drug effects , Cell Survival/drug effects , Chondrocytes/cytology , Chondrocytes/drug effects , Chondrocytes/metabolism , Disease Models, Animal , Endoplasmic Reticulum Stress/drug effects , Female , Gene Silencing , Hydrogen Peroxide/toxicity , Male , Mice, Inbred C57BL , Osteoarthritis/etiology , Osteoarthritis/metabolism , Oxidative Stress/drug effects , Primary Cell Culture , Sirtuin 1/genetics , Tibial Meniscus Injuries/complications
15.
Psychiatry Clin Neurosci ; 75(8): 250-255, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34033171

ABSTRACT

AIM: Depression has been recognized as one of the disorders associated with cardiac interventions such as percutaneous coronary intervention (PCI) or coronary artery bypass graft surgery (CABG). In the present study, we evaluated the efficacy and safety of sulforaphane in treatment of depression induced by cardiac interventions. METHODS: After initial screening, 66 patients with previous history of at least one cardiac intervention and current mild to moderate depression were randomly assigned to two parallel groups receiving either sulforaphane (n = 33) or placebo (n = 33) for six successive weeks. Efficacy was assessed using the Hamilton Rating Scale for Depression (HAM-D) at baseline and week 2, 4, and 6. Safety of the treatments was checked during the trial period. RESULTS: Sixty participants completed the clinical trial (n = 30 in each group). Baseline demographic and clinical parameters were all similar among groups. Repeated measures analysis indicated that the sulforaphane group exhibited greater improvement in HAM-D scores throughout the trial (P < 0.001). Response to treatment (≥50% reduction in the HAM-D score) rate was higher in the sulforaphane group at trial endpoint (30% vs 6.67%, P = 0.042). Remission (HAM-D score ≤ 7) rate was also higher in the sulforaphane group; however, the difference was not significant (23.33% vs 3.33%, P = 0.052). Finally, no significant difference was observed between the two groups in terms of frequency of side effects. CONCLUSIONS: Sulforaphane could safely improve depressive symptoms induced by cardiac interventions. Further clinical trials with larger sample sizes and longer follow-up periods are warranted to confirm our results.


Subject(s)
Depression/drug therapy , Isothiocyanates/adverse effects , Isothiocyanates/therapeutic use , Percutaneous Coronary Intervention , Sulfoxides/adverse effects , Sulfoxides/therapeutic use , Adult , Aged , Double-Blind Method , Female , Humans , Male , Middle Aged , Treatment Outcome
16.
Front Immunol ; 12: 660179, 2021.
Article in English | MEDLINE | ID: mdl-33936094

ABSTRACT

The complex interplay between the gut microbiota, the intestinal barrier, the immune system and the liver is strongly influenced by environmental and genetic factors that can disrupt the homeostasis leading to disease. Among the modulable factors, diet has been identified as a key regulator of microbiota composition in patients with metabolic syndrome and related diseases, including the metabolic dysfunction-associated fatty liver disease (MAFLD). The altered microbiota disrupts the intestinal barrier at different levels inducing functional and structural changes at the mucus lining, the intercellular junctions on the epithelial layer, or at the recently characterized vascular barrier. Barrier disruption leads to an increased gut permeability to bacteria and derived products which challenge the immune system and promote inflammation. All these alterations contribute to the pathogenesis of MAFLD, and thus, therapeutic approaches targeting the gut-liver-axis are increasingly being explored. In addition, the specific changes induced in the intestinal flora may allow to characterize distinctive microbial signatures for non-invasive diagnosis, severity stratification and disease monitoring.


Subject(s)
Gastrointestinal Microbiome/immunology , Intestinal Mucosa/immunology , Liver/immunology , Metabolic Syndrome/immunology , Non-alcoholic Fatty Liver Disease/immunology , Animals , CCR5 Receptor Antagonists/therapeutic use , Dysbiosis/immunology , Dysbiosis/microbiology , Humans , Imidazoles/therapeutic use , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Liver/metabolism , Liver/pathology , Metabolic Syndrome/drug therapy , Metabolic Syndrome/metabolism , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/metabolism , Sulfoxides/therapeutic use
17.
Biochem Pharmacol ; 188: 114539, 2021 06.
Article in English | MEDLINE | ID: mdl-33819468

ABSTRACT

The antitumor properties of cruciferous vegetables are mainly due to their high content of isothiocyanates, and sulforaphane (SFA) is the most well-known compound. The aim of this study was to determine the mechanism of SFA inhibiting gastric cancer (GC) progression. After verifying SFA suppressing GC growth in vivo, we utilized the GSE79973 and GSE118916 datasets to identify the GC development signatures that overlap with the RNA-seq analysis in SFA-treated AGS cells. GSEA of the RNA-seq data indicated that SFA regulation of GC progression was related to extracellular matrix and collagens; thus, we identified COL3A1 and COL5A1 as the targets of SFA, which functioned as oncogenes. We found positive correlations between COL3A1 and COL5A1 expression in GC cells, and confirmed that miR-29a-3p is the common regulator of their expression. RNA immunoprecipitation assays based on Ago2, Dicer, and exportin-5 showed that SFA could promote mature miR-29a-3p generation. We also proved that SFA inactivated the Wnt/ß-catenin pathway in GC cells in a miR-29a-3p-dependent manner. Overall, SFA boosts miR-29a-3p maturation to downregulate COL3A1 and COL5A1 and inactivate the Wnt/ ß -catenin pathway to suppress GC progression.


Subject(s)
Anticarcinogenic Agents/therapeutic use , Collagen Type III/biosynthesis , Collagen Type V/biosynthesis , Isothiocyanates/therapeutic use , MicroRNAs/biosynthesis , Stomach Neoplasms/metabolism , Sulfoxides/therapeutic use , Animals , Anticarcinogenic Agents/pharmacology , Cell Line, Tumor , Collagen Type III/antagonists & inhibitors , Collagen Type III/genetics , Collagen Type V/antagonists & inhibitors , Collagen Type V/genetics , Disease Progression , Dose-Response Relationship, Drug , Female , Gene Expression Regulation, Neoplastic , Humans , Isothiocyanates/pharmacology , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Sulfoxides/pharmacology , Xenograft Model Antitumor Assays/methods
18.
Curr Med Sci ; 41(2): 250-269, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33877541

ABSTRACT

Tumorigenicity-inhibiting compounds have been identified in our daily diet. For example, isothiocyanates (ITCs) found in cruciferous vegetables were reported to have potent cancer-prevention activities. The best characterized ITC is sulforaphane (SF). SF can simultaneously modulate multiple cellular targets involved in carcinogenesis, including (1) modulating carcinogen-metabolizing enzymes and blocking the action of mutagens; (2) inhibition of cell proliferation and induction of apoptosis; and (3) inhibition of neo-angiogenesis and metastasis. SF targets cancer stem cells through modulation of nuclear factor kappa B (NF-κB), Sonic hedgehog (SHH), epithelial-mesenchymal transition, and Wnt/ß-catenin pathways. Conventional chemotherapy/SF combination was tested in several studies and resulted in favorable outcomes. With its favorable toxicological profile, SF is a promising agent in cancer prevention and/or therapy. In this article, we discuss the human metabolism of SF and its effects on cancer prevention, treatment, and targeting cancer stem cells, as well as providing a brief review of recent human clinical trials on SF.


Subject(s)
Biological Products/therapeutic use , Isothiocyanates/therapeutic use , Neoplasms/drug therapy , Neoplasms/prevention & control , Sulfoxides/therapeutic use , Animals , Brassica/chemistry , Chemoprevention , Clinical Trials as Topic , Humans , Isothiocyanates/metabolism , Isothiocyanates/pharmacology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Sulfoxides/metabolism , Sulfoxides/pharmacology
19.
Front Immunol ; 12: 632132, 2021.
Article in English | MEDLINE | ID: mdl-33815384

ABSTRACT

Diabetic retinopathy (DR) is one of the most common complications of Diabetes Mellitus (DM) and is directly associated with inflammatory processes. Currently, neuro-inflammation is considered an early event in DR and proceeds via microglia polarization. A hallmark of DR is the presence of retinal reactive gliosis. Here we report the beneficial effect of (SS,1R)-1-docecylsulfiny-5N,6O-oxomethylidenenojirimycin ((Ss)-DS-ONJ), a member of the sp2-iminosugar glycolipid (sp2-IGL) family, by decreasing iNOS and inflammasome activation in Bv.2 microglial cells exposed to pro-inflammatory stimuli. Moreover, pretreatment with (Ss)-DS-ONJ increased Heme-oxygenase (HO)-1 as well as interleukin 10 (IL10) expression in LPS-stimulated microglial cells, thereby promoting M2 (anti-inflammatory) response by the induction of Arginase-1. The results strongly suggest that this is the likely molecular mechanism involved in the anti-inflammatory effects of (SS)-DS-ONJ in microglia. (SS)-DS-ONJ further reduced gliosis in retinal explants from type 1 diabetic BB rats, which is consistent with the enhanced M2 response. In conclusion, targeting microglia polarization dynamics in M2 status by compounds with anti-inflammatory activities offers promising therapeutic interventions at early stages of DR.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Diabetic Retinopathy/drug therapy , Glycolipids/therapeutic use , Sulfoxides/therapeutic use , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Cytokines/metabolism , Diabetic Retinopathy/immunology , Diabetic Retinopathy/pathology , Gliosis , Glycolipids/chemistry , Glycolipids/pharmacology , Inflammasomes/drug effects , Inflammation , Lipopolysaccharides/adverse effects , Microglia/drug effects , Microglia/immunology , Nitric Oxide Synthase Type II/metabolism , Rats , Retina/drug effects , Retina/immunology , Retina/pathology , Signal Transduction/drug effects , Sulfoxides/chemistry , Sulfoxides/pharmacology
20.
Hepatol Commun ; 5(3): 478-490, 2021 03.
Article in English | MEDLINE | ID: mdl-33681680

ABSTRACT

Primary sclerosing cholangitis (PSC) is a chronic cholestatic disease with no approved treatments. C-C chemokine receptor types 2 and 5 (CCR2/CCR5) play an important role in inflammation and fibrosis and are potential therapeutic targets for PSC. We evaluated the efficacy and safety of cenicriviroc (CVC), a dual antagonist of CCR2 and CCR5, for the treatment of PSC. This was a single-arm, open-label, exploratory study of CVC in adults with a clinical diagnosis of PSC, serum alkaline phosphatase (ALP) ≥1.5 times the upper limit of normal (ULN), with or without inflammatory bowel disease, across eight sites in the United States and Canada. The primary endpoint was percent change in ALP over 24 weeks; key secondary efficacy endpoints were proportion of participants who achieved ALP normalization and overall response (decrease to <1.5 times the ULN or 50% decrease). Of the 24 participants, 20 completed the study. The mean age was 43 years, 50% were female, and the mean body mass index was 25 kg/m2. From a median ALP baseline of 369 U/L (range: 173, 1,377 U/L), a median absolute reduction of 49.5 U/L (range: -460, 416 U/L) was achieved at week 24, corresponding to a median reduction of 18.0% (range: -46%, 89%). No participant achieved ALP normalization or a 50% decrease; 2 participants (10%) achieved a reduction in ALP to < 1.5 times the ULN, and 4 had ≥25% increase. Twenty participants (83.3%) reported at least one adverse event; most were mild to moderate in severity. The most frequent events were rash, fatigue, and dizziness. Conclusion: After 24 weeks of CVC treatment, adults with PSC achieved a modest reduction (median 18%) in the surrogate endpoint of ALP. CVC was well tolerated, and no new safety signals were observed. ClinicalTrials.gov identifier: NCT02653625.


Subject(s)
Cholangitis, Sclerosing/drug therapy , Imidazoles/therapeutic use , Sulfoxides/therapeutic use , Adolescent , Adult , Aged , Alkaline Phosphatase/blood , Biomarkers/blood , Canada , Cholangitis, Sclerosing/blood , Female , Humans , Liver Function Tests , Male , Middle Aged , Treatment Outcome , United States , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...