Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Med ; 30(5): 1309-1319, 2024 May.
Article in English | MEDLINE | ID: mdl-38627559

ABSTRACT

Cancer of unknown primary (CUP) site poses diagnostic challenges due to its elusive nature. Many cases of CUP manifest as pleural and peritoneal serous effusions. Leveraging cytological images from 57,220 cases at four tertiary hospitals, we developed a deep-learning method for tumor origin differentiation using cytological histology (TORCH) that can identify malignancy and predict tumor origin in both hydrothorax and ascites. We examined its performance on three internal (n = 12,799) and two external (n = 14,538) testing sets. In both internal and external testing sets, TORCH achieved area under the receiver operating curve values ranging from 0.953 to 0.991 for cancer diagnosis and 0.953 to 0.979 for tumor origin localization. TORCH accurately predicted primary tumor origins, with a top-1 accuracy of 82.6% and top-3 accuracy of 98.9%. Compared with results derived from pathologists, TORCH showed better prediction efficacy (1.677 versus 1.265, P < 0.001), enhancing junior pathologists' diagnostic scores significantly (1.326 versus 1.101, P < 0.001). Patients with CUP whose initial treatment protocol was concordant with TORCH-predicted origins had better overall survival than those who were administrated discordant treatment (27 versus 17 months, P = 0.006). Our study underscores the potential of TORCH as a valuable ancillary tool in clinical practice, although further validation in randomized trials is warranted.


Subject(s)
Deep Learning , Neoplasms, Unknown Primary , Humans , Neoplasms, Unknown Primary/pathology , Female , Male , Aged , Middle Aged , ROC Curve , Adult , Cytodiagnosis/methods , Aged, 80 and over , Ascites/pathology , Cytology
2.
Dermatology ; 240(1): 1-12, 2024.
Article in English | MEDLINE | ID: mdl-37939679

ABSTRACT

INTRODUCTION: Psoriasis is a chronic immune-mediated skin disease. Several clinical trials have studied some topical drugs aiming at new therapeutic targets. However, the comparative efficacy and safety of different concentrations and frequencies of newer topical drugs for psoriasis remain unclear. The aim of our study is to assess the comparative efficacy and safety of some newer topical treatments in patients with psoriasis. METHODS: A systematic review and network meta-analysis (NMA) was conducted using eligible randomized controlled trials (RCTs). Treatments included topical therapeutic aryl hydrocarbon receptor (AhR)-modulating agent (TAMA), topical phosphodiesterase type 4 (PDE-4) inhibitors, and topical janus kinase-signal transducer and activator of transcription (JAK-STAT) inhibitors. The primary efficacy assessment criterion was the proportion of patients' achieving Physician's Global Assessment 0/1 (PGA response). Secondary criterion was ≥75% reductions in the Psoriasis Area and Severity Index (PASI75). Adverse events (AEs) to represent the safety were also summarized. RESULTS: Among 6 including newer topical drugs, odds of achieving both PGA response and PASI75 were higher with all regimens of TAMA and roflumilast cream versus vehicle. In terms of safety outcomes, odds of AEs were also higher with all regimens of TAMA. There were no statistically significant differences between topical JAK-STAT inhibitors and vehicle for any outcome, except ruxolitinib ointment 1% once daily (QD). CONCLUSION: TAMA had a good therapeutic effect on plaque psoriasis but a relatively low treatment safety. Roflumilast cream had both promising efficacy and higher safety.


Subject(s)
Aminopyridines , Psoriasis , Humans , Network Meta-Analysis , Benzamides/therapeutic use , Psoriasis/drug therapy , Chronic Disease , Treatment Outcome , Cyclopropanes
3.
Aging (Albany NY) ; 13(10): 14001-14014, 2021 05 24.
Article in English | MEDLINE | ID: mdl-34030135

ABSTRACT

Vascular dementia (VaD) is the second most common type of dementia worldwide. Although there are five FDA-approved drugs for the treatment of Alzheimer's disease (AD), none of them have been applied to treat VaD. Adalimumab is a TNF-α inhibitor that is used for the treatment of autoimmune diseases such as rheumatoid arthritis. In a recent retrospective case-control study, the application of adalimumab for rheumatoid or psoriasis was shown to decrease the risk of AD. However, whether adalimumab can be used for the treatment of VaD is not clear. In this study, we used 2VO surgery to generate a VaD rat model and treated the rats with adalimumab or vehicle. We demonstrated that VaD rats treated with adalimumab exhibited significant improvements in memory. In addition, adalimumab treatment significantly alleviated neuronal loss in the hippocampi of VaD rats. Moreover, adalimumab significantly reduced microglial activation and reversed M1/M2 polarization in VaD rats. Furthermore, adalimumab treatment suppressed the activity of NF-κB, an important neuroinflammatory transcription factor. Finally, adalimumab displayed a protective role against oxidative stress in VaD rats. Our results indicate that adalimumab may be applied for the treatment of human patients with VaD.


Subject(s)
Adalimumab/therapeutic use , Brain/pathology , Cerebrovascular Circulation/physiology , Inflammation/pathology , Memory Disorders/drug therapy , Adalimumab/pharmacology , Animals , Biomarkers/metabolism , Cerebrovascular Circulation/drug effects , Chronic Disease , Memory Disorders/physiopathology , Microglia/drug effects , Microglia/pathology , NF-kappa B/metabolism , Neurons/drug effects , Neurons/pathology , Oxidative Stress/drug effects , Rats, Sprague-Dawley , Signal Transduction/drug effects
4.
Leuk Res ; 105: 106566, 2021 06.
Article in English | MEDLINE | ID: mdl-33848709

ABSTRACT

Angiogenesis is an integral part of the multiple myeloma (MM) microenvironment, and affects tumorigenesis, progression, invasion, and metastasis. Exosomes are essential for cell-cell communication and help in regulating the bone marrow microenvironment. Herein, we investigated macrophage polarization and angiogenesis in MM in vitro via exosome-derived miR-let-7c. We observed that exosomal miR-let-7c secreted by mesenchymal stem cells promoted M2 macrophage polarization, thereby enhancing angiogenesis in the bone marrow microenvironment. Suppressing miR-let-7c expression significantly inhibited vascular endothelial cell function in myeloma. Thus, exosomal miR-let-7c may be a reliable biomarker for early prediction of tumor progression and a promising therapeutic target for MM.


Subject(s)
Macrophages/pathology , MicroRNAs/metabolism , Multiple Myeloma/pathology , Neovascularization, Pathologic/pathology , Aged , Bone Marrow/metabolism , Bone Marrow/pathology , Exosomes/genetics , Exosomes/metabolism , Female , Gene Expression Regulation, Neoplastic/physiology , Humans , Macrophage Activation , Macrophages/metabolism , Male , MicroRNAs/genetics , Middle Aged , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Tumor Microenvironment/physiology
5.
Med Oncol ; 37(11): 99, 2020 Oct 10.
Article in English | MEDLINE | ID: mdl-33040185

ABSTRACT

Angiogenesis is a prerequisite for multiple myeloma development. Tumor cells can stimulate angiogenesis by secreting vascular endothelial growth factor A (VEGFA), but we previously reported that tumor angiogenesis was not significantly reduced when VEGFA expression was inhibited in myeloma cells. Tumor-associated macrophages (TAMs) are important components of the tumor microenvironment and have been reported to be involved in the regulation of angiogenesis. In this study, we performed in vitro macrophage coculture studies and studies with RPMI 8226 and TAMs cell-conditioned media to explore their effects on the proliferation, migration, and tube formation of human umbilical vein endothelial cells (HUVECs). Our results showed that M2 macrophages and RPMI 8226 cells could synergistically promote HUVEC proliferation, migration, and tube formation, and that VEGFA depletion in both cell types suppressed HUVEC tube formation ability. Conversely, M1 macrophages inhibited the tube formation in HUVECs. Mechanistically, M2 macrophage secretion of VEGFA may affect vascular endothelial growth factor receptor 1 signaling to regulate angiogenesis. In summary, our results suggest that macrophage clearance or inducing of transformation of M2 macrophages into M1 macrophages are potential treatment strategies for multiple myeloma.


Subject(s)
Multiple Myeloma/metabolism , Neovascularization, Pathologic/pathology , Tumor-Associated Macrophages/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Coculture Techniques , Culture Media, Conditioned/metabolism , Culture Media, Conditioned/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/pathology , Humans , Neovascularization, Pathologic/metabolism , Signal Transduction , THP-1 Cells , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism
6.
Biomed Res Int ; 2020: 9875636, 2020.
Article in English | MEDLINE | ID: mdl-32685551

ABSTRACT

PURPOSE: The incidence of papillary thyroid cancer (PTC) is increasing, and traditional diagnostic methods are unsatisfactory. Therefore, identifying novel prognostic markers is very important. ciRS-7 has been found to play an important role in many cancers, but its role in PTC has not been reported. This study was performed to evaluate the biological role and mechanism of ciRS-7 in PTC. Material and Methods. The expression of ciRS-7 in PTC tissues and the matched adjacent tissues was determined by quantitative reverse transcription polymerase chain reaction (qRT-PCR). The PTC cell lines (TPC-1 and BCPAP) were used to evaluate the role of ciRS-7. ciRS-7-siRNA and overexpression plasmid were constructed and transfected into PTC cells. A CCK-8 assay and colony formation assay were performed to explore the effects of ciRS-7 on cell proliferation. Annexin V/PI staining and FACS detection were used to detect cell apoptosis. Wound healing assay was performed to detect cell migration. A transwell assay was conducted to explore the effects of ciRS-7 on invasion and migration. Western blotting was performed to evaluate protein expression. The luciferase reporter system was used to determine the underlying mechanism of miR-7. RESULT: ciRS-7 was highly expressed in PTC tissues and cell lines compared with the corresponding controls. In vitro study showed that ciRS-7 silencing suppressed proliferation, migration, and invasion of TPC-1 and BCPAP. Mechanistically, the effects of ciRS-7 on invasion and migration may be related to epithelial-mesenchymal transition (EMT). ciRS-7 silencing could attenuate effects on PTC cells induced by miR-7 knockdown. Epidermal growth factor receptor (EGFR), which was demonstrated to be a target of miR-7, decreased significantly in ciRS-7-siRNA PTC cells. Overexpression of EGFR also attenuated effects of PTC cells induced by silencing ciRS-7. CONCLUSION: ciRS-7 was significantly upregulated in PTC tissues, and it promoted the progression of PTC by regulating the miR-7/EGFR axis. ciRS-7 is a promising prognostic biomarker and therapeutic target in PTC.


Subject(s)
Cell Movement , Cell Proliferation , MicroRNAs , Neoplasm Proteins , RNA, Circular , RNA, Neoplasm , Signal Transduction , Thyroid Cancer, Papillary , Thyroid Neoplasms , Cell Line, Tumor , ErbB Receptors/genetics , ErbB Receptors/metabolism , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , RNA, Circular/genetics , RNA, Circular/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Thyroid Cancer, Papillary/genetics , Thyroid Cancer, Papillary/metabolism , Thyroid Cancer, Papillary/pathology , Thyroid Neoplasms/genetics , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology
7.
J Alzheimers Dis ; 71(1): 97-108, 2019.
Article in English | MEDLINE | ID: mdl-31322570

ABSTRACT

Vascular dementia (VaD) is caused by chronic decreases in brain blood flow and accounts for 15-20% of dementia cases worldwide. In contrast to Alzheimer's disease (AD), no effective drug treatments are currently available for VaD. Previous studies have suggested that oxidative stress and neuroinflammation in the brain play important roles in the pathogenesis of VaD. Honokiol (HKL) is a well-known bioactive and nutraceutical compound that can act as an antioxidant and anti-inflammatory molecule. HKL can protect against memory impairments in AD mouse models. In this study, we explored whether the application of HKL was also protective against the insult of chronic cerebral hypoperfusion (CCH) in rats. We found that HKL supplementation prevented the memory impairments in the inhibitory avoidance step-down and Morris water maze tasks in CCH rats. HKL also suppressed the levels of oxidative stress and inflammation in CCH rats. Moreover, HKL prevented dendritic spines abnormalities in CCH rats. We also found that HKL inhibited the activity of GSK-3ß, which may be critical for the neuroprotective activity of HKL. Thus, our study demonstrated the protective role of HKL in VaD.


Subject(s)
Biphenyl Compounds/therapeutic use , Dementia, Vascular/drug therapy , Glycogen Synthase Kinase 3 beta/metabolism , Inflammation/prevention & control , Lignans/therapeutic use , Memory Disorders/prevention & control , Neuroprotective Agents/therapeutic use , Oxidative Stress/drug effects , Animals , Brain/drug effects , Brain/metabolism , Dementia, Vascular/metabolism , Dementia, Vascular/psychology , Disease Models, Animal , Enzyme Activation/drug effects , Maze Learning , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...