Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Clin Transl Med ; 14(3): e1605, 2024 03.
Article in English | MEDLINE | ID: mdl-38445456

ABSTRACT

BACKGROUND: Bone or brain metastases may develop in 20-40% of individuals with late-stage non-small-cell lung cancer (NSCLC), resulting in a median overall survival of only 4-6 months. However, the primary lung cancer tissue's distinctions between bone, brain and intrapulmonary metastases of NSCLC at the single-cell level have not been underexplored. METHODS: We conducted a comprehensive analysis of 14 tissue biopsy samples obtained from treatment-naïve advanced NSCLC patients with bone (n = 4), brain (n = 6) or intrapulmonary (n = 4) metastasis using single-cell sequencing originating from the lungs. Following quality control and the removal of doublets, a total of 80 084 cells were successfully captured. RESULTS: The most significant inter-group differences were observed in the fraction and function of fibroblasts. We identified three distinct cancer-associated fibroblast (CAF) subpopulations: myofibroblastic CAF (myCAF), inflammatory CAF (iCAF) and antigen-presenting CAF (apCAF). Notably, apCAF was prevalent in NSCLC with bone metastasis, while iCAF dominated in NSCLC with brain metastasis. Intercellular signalling network analysis revealed that apCAF may play a role in bone metastasis by activating signalling pathways associated with cancer stemness, such as SPP1-CD44 and SPP1-PTGER4. Conversely, iCAF was found to promote brain metastasis by activating invasion and metastasis-related molecules, such as MET hepatocyte growth factor. Furthermore, the interaction between CAFs and tumour cells influenced T-cell exhaustion and signalling pathways within the tumour microenvironment. CONCLUSIONS: This study unveils the direct interplay between tumour cells and CAFs in NSCLC with bone or brain metastasis and identifies potential therapeutic targets for inhibiting metastasis by disrupting these critical cell-cell interactions.


Subject(s)
Brain Neoplasms , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Brain , Fibroblasts , Tumor Microenvironment
2.
Cereb Cortex ; 33(24): 11570-11581, 2023 12 09.
Article in English | MEDLINE | ID: mdl-37851710

ABSTRACT

Facial palsy therapies based on cortical plasticity are in development, but facial synkinesis progress is limited. Studying neural plasticity characteristics, especially network organization and its constitutive elements (nodes/edges), is the key to overcome the bottleneck. We studied 55 participants (33 facial synkinesis patients, 22 healthy controls) with clinical assessments, functional magnetic resonance imaging (fMRI), and diffusion tensor imaging (DTI). We analyzed rich-club organization and metrics of structural brain networks (rich-club coefficients, strength, degree, density, and efficiency). Functional brain network metrics, including functional connectivity and its coupling with the structural network, were also computed. Patients displayed reduced strength and density of rich-club nodes and edges, as well as decreased global efficiency. All nodes exhibited decreased nodal efficiency in patients. Patients had significantly increased functional connectivity and decreased structural-functional coupling strength in rich-club nodes, rich-club edges, and feeder edges. Our study indicates that facial synkinesis patients have weakened structural connections but enhanced functional transmission from rich-club nodes. The loss of connections and efficiency in structural network may trigger compensatory increases in functional connectivity of rich-club nodes. Two potential biomarkers, rich-club edge density and structural-functional coupling strength, may serve as indicators of disease outcome. These findings provide valuable insights into synkinesis mechanisms and offer potential targets for cortical intervention.


Subject(s)
Diffusion Tensor Imaging , Synkinesis , Humans , Synkinesis/diagnostic imaging , Synkinesis/pathology , Brain , Magnetic Resonance Imaging , Neural Pathways/diagnostic imaging
3.
Semin Cancer Biol ; 95: 52-74, 2023 10.
Article in English | MEDLINE | ID: mdl-37473825

ABSTRACT

Head and neck tumors (HNTs) constitute a multifaceted ensemble of pathologies that primarily involve regions such as the oral cavity, pharynx, and nasal cavity. The intricate anatomical structure of these regions poses considerable challenges to efficacious treatment strategies. Despite the availability of myriad treatment modalities, the overall therapeutic efficacy for HNTs continues to remain subdued. In recent years, the deployment of artificial intelligence (AI) in healthcare practices has garnered noteworthy attention. AI modalities, inclusive of machine learning (ML), neural networks (NNs), and deep learning (DL), when amalgamated into the holistic management of HNTs, promise to augment the precision, safety, and efficacy of treatment regimens. The integration of AI within HNT management is intricately intertwined with domains such as medical imaging, bioinformatics, and medical robotics. This article intends to scrutinize the cutting-edge advancements and prospective applications of AI in the realm of HNTs, elucidating AI's indispensable role in prevention, diagnosis, treatment, prognostication, research, and inter-sectoral integration. The overarching objective is to stimulate scholarly discourse and invigorate insights among medical practitioners and researchers to propel further exploration, thereby facilitating superior therapeutic alternatives for patients.


Subject(s)
Artificial Intelligence , Head and Neck Neoplasms , Humans , Machine Learning , Neural Networks, Computer , Head and Neck Neoplasms/diagnosis , Head and Neck Neoplasms/therapy , Diagnostic Imaging/methods
4.
J Clin Med ; 12(6)2023 Mar 10.
Article in English | MEDLINE | ID: mdl-36983174

ABSTRACT

Surgery with the assistance of conventional radiotherapy, chemotherapy and immunotherapy is the basis for head and neck squamous cell carcinoma (HNSCC) treatment. However, with these treatment modalities, the recurrence and metastasis of tumors remain at a high level. Increasingly, the evidence indicates an excellent anti-tumor effect of chimeric antigen receptor T (CAR-T) cells in hematological malignancy treatment, and this novel immunotherapy has attracted researchers' attention in HNSCC treatment. Although several clinical trials have been conducted, the weak anti-tumor effect and the side effects of CAR-T cell therapy against HNSCC are barriers to clinical translation. The limited choices of targeting proteins, the barriers of CAR-T cell infiltration into targeted tumors and short survival time in vivo should be solved. In this review, we introduce barriers of CAR-T cell therapy in HNSCC. The limitations and current promising strategies to overcome barriers in solid tumors, as well as the applications for HNSCC treatment, are covered. The perspectives of CAR-T cell therapy in future HNSCC treatment are also discussed.

5.
J Nanobiotechnology ; 20(1): 277, 2022 Jun 14.
Article in English | MEDLINE | ID: mdl-35701847

ABSTRACT

Cancer immunotherapy is a novel therapeutic regimen because of the specificity and durability of immune modulations to treat cancers. Current cancer immunotherapy is limited by some barriers such as poor response rate, low tumor specificity and systemic toxicities. Porous nanomaterials (PNMs) possess high loading capacity and tunable porosity, receiving intense attention in cancer immunotherapy. Recently, novel PNMs based drug delivery systems have been employed in antitumor immunotherapy to enhance tissue or organ targeting and reduce immune-related adverse events. Herein, we summarize the recent progress of PNMs including inorganic, organic, and organic-inorganic hybrid ones for cancer immunotherapy. The design of PNMs and their performance in cancer immunotherapy are discussed in detail, with a focus on how those designs can address the challenges in current conventional immunotherapy. Lastly, we present future directions of PNMs for cancer immunotherapy including the challenges and research gaps, providing new insights about the design of PNMs for efficient cancer immunotherapy with better performance as powerful weapons against tumors. Finally, we discussed the relevant challenges that urgently need to be addressed in clinical practice, coupled with corresponding solutions to these problems.


Subject(s)
Nanostructures , Neoplasms , Drug Delivery Systems , Humans , Immunologic Factors , Immunotherapy , Nanostructures/therapeutic use , Neoplasms/drug therapy , Porosity
6.
MedComm (2020) ; 3(2): e124, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35356799

ABSTRACT

Signal transducer and activator of transcription 3 (STAT3), a member of the STAT family, discovered in the cytoplasm of almost all types of mammalian cells, plays a significant role in biological functions. The duration of STAT3 activation in normal tissues is a transient event and is strictly regulated. However, in cancer tissues, STAT3 is activated in an aberrant manner and is induced by certain cytokines. The continuous activation of STAT3 regulates the expression of downstream proteins associated with the formation, progression, and metastasis of cancers. Thus, elucidating the mechanisms of STAT3 regulation and designing inhibitors targeting the STAT3 pathway are considered promising strategies for cancer treatment. This review aims to introduce the history, research advances, and prospects concerning the STAT3 pathway in cancer. We review the mechanisms of STAT3 pathway regulation and the consequent cancer hallmarks associated with tumor biology that are induced by the STAT3 pathway. Moreover, we summarize the emerging development of inhibitors that target the STAT3 pathway and novel drug delivery systems for delivering these inhibitors. The barriers against targeting the STAT3 pathway, the focus of future research on promising targets in the STAT3 pathway, and our perspective on the overall utility of STAT3 pathway inhibitors in cancer treatment are also discussed.

7.
J Extracell Vesicles ; 10(14): e12175, 2021 12.
Article in English | MEDLINE | ID: mdl-34918479

ABSTRACT

Extracellular vesicles (EVs) are lipid-bilayer membrane structures secreted by most cell types. EVs act as messengers via the horizontal transfer of lipids, proteins, and nucleic acids, and influence various pathophysiological processes in both parent and recipient cells. Compared to EVs obtained from body fluids or cell culture supernatants, EVs isolated directly from tissues possess a number of advantages, including tissue specificity, accurate reflection of tissue microenvironment, etc., thus, attention should be paid to tissue-derived EVs (Ti-EVs). Ti-EVs are present in the interstitium of tissues and play pivotal roles in intercellular communication. Moreover, Ti-EVs provide an excellent snapshot of interactions among various cell types with a common histological background. Thus, Ti-EVs may be used to gain insights into the development and progression of diseases. To date, extensive investigations have focused on the role of body fluid-derived EVs or cell culture-derived EVs; however, the number of studies on Ti-EVs remains insufficient. Herein, we summarize the latest advances in Ti-EVs for cancers and non-cancer diseases. We propose the future application of Ti-EVs in basic research and clinical practice. Workflows for Ti-EV isolation and characterization between cancers and non-cancer diseases are reviewed and compared. Moreover, we discuss current issues associated with Ti-EVs and provide potential directions.


Subject(s)
Extracellular Vesicles/metabolism , Neoplasms/pathology , Tumor Microenvironment/physiology , Humans
8.
J Nanobiotechnology ; 19(1): 379, 2021 Nov 21.
Article in English | MEDLINE | ID: mdl-34802438

ABSTRACT

Matrix metalloproteinase (MMP) 2 and 9 are the family members of proteases normally up-regulated in tumor to enhance the invasion and metastatic of tumor cells, and are associated with poor outcome of head and neck squamous cell carcinomas (HNSCCs). In the present work, MMPs-degradable gelatin nanoparticles (GNPs) are simultaneously loaded with photosensitizer indocyanine green (ICG) along with signal transducer activator of transcription 3 (STAT3) inhibitor NSC74859 (NSC, N) for efficient photothermal therapy (PTT) and immunotherapy of HNSCCs. In the tumor tissue, Gel-N-ICG nanoparticle was degraded and encapsulated ICG and NSC were effectively released. Under near-infrared (NIR) irradiation, the released ICG nanoparticles enabled effective photothermal destruction of tumors, and the STAT3 inhibitor NSC elicited potent antitumor immunity for enhanced cancer therapy. Based on two HNSCC mouse models, we demonstrated that Gel-N-ICG significantly delayed tumor growth without any appreciable body weight loss. Taken together, the strategy reported here may contribute that the stimuli-responsive proteases triggered nanoplatform could reduce tumor size more effectively in complex tumor microenvironment (TME) through combination of PTT and immunotherapy.


Subject(s)
Gelatinases/metabolism , Nanoparticles , Photosensitizing Agents , Protein Inhibitors of Activated STAT , Animals , Cell Line, Tumor , Cell Survival/drug effects , Immunotherapy , Indocyanine Green/chemistry , Indocyanine Green/pharmacokinetics , Mice , Nanoparticles/chemistry , Nanoparticles/metabolism , Photochemotherapy , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacokinetics , Photosensitizing Agents/pharmacology , Photothermal Therapy , Protein Inhibitors of Activated STAT/chemistry , Protein Inhibitors of Activated STAT/pharmacokinetics , Protein Inhibitors of Activated STAT/pharmacology , STAT3 Transcription Factor/antagonists & inhibitors
9.
J Neurosci Res ; 94(1): 50-61, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26423029

ABSTRACT

The midbrain ventrolateral periaqueductal gray (VL-PAG) is a key component that mediates pain modulation. Although spinal cord glial cells appear to play an important role in chronic pain development, the precise mechanisms involving descending facilitation pathways from the PAG following nerve injury are poorly understood. This study shows that cellular events that occur during glial activation in the VL-PAG may promote descending facilitation from the PAG during neuropathic pain. Chronic constriction nerve injury (CCI) was induced by ligature construction of the sciatic nerve in male Sprague-Dawley rats. Behavioral responses to noxious mechanical (paw withdrawal threshold; PWT) and thermal (paw withdrawal latency; PWL) stimuli were evaluated. After CCI, immunohistochemical and Western blot analysis of microglia and astrocytes in the VL-PAG showed morphological and quantitative changes indicative of activation in microglia and astrocytes. Intra-VL-PAG injection of microglial or astrocytic inhibitors attenuated PWT and PWL at days 7 and 14, respectively, following CCI. We also evaluated the effects of intra-VL-PAG administration of the phosphorylated p38 mitogen-activated protein kinase (p-p38 MAPK) inhibitor SB 203580 at day 7 after CCI. This treatment abolished microglial activation and produced a significant time-dependent attenuation of PWT and PWL. Western blot analysis showed localized expression of p-p38 in the VL-PAG after CCI. P-p38 was expressed in labeled microglia of the VL-PAG but was not present in astrocytes and neurons on day 7 after CCI. These results demonstrate that CCI-induced neuropathic pain is associated with glial activation in the VL-PAG, which likely participates in descending pain facilitation through the p38 MAPK signaling pathway.


Subject(s)
Neuroglia/pathology , Periaqueductal Gray/pathology , Sciatica/pathology , Sciatica/physiopathology , Signal Transduction/physiology , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Calcium-Binding Proteins/metabolism , Disease Models, Animal , Glial Fibrillary Acidic Protein/metabolism , Hyperalgesia/drug therapy , Hyperalgesia/physiopathology , Imidazoles/therapeutic use , Male , Microfilament Proteins/metabolism , Pain Measurement , Pain Threshold/drug effects , Phosphopyruvate Hydratase/metabolism , Pyridines/therapeutic use , Rats , Rats, Sprague-Dawley , Sciatica/drug therapy , Signal Transduction/drug effects , Time Factors
10.
Langmuir ; 31(11): 3422-30, 2015 Mar 24.
Article in English | MEDLINE | ID: mdl-25734470

ABSTRACT

This article describes the use of surface plasmon resonance (SPR) spectroscopy to study antibody-ligand interactions for hydrophobic charge-induction chromatography (HCIC) and its versatility in investigating the surface and solution factors affecting the interactions. Two density model surfaces presenting the HCIC ligand (mercapto-ethyl-pyridine, MEP) were prepared on Au using a self-assembly technique. The surface chemistry and structure, ionization, and protein binding of such model surfaces were characterized by X-ray photoelectron spectroscopy (XPS), near-edge X-ray absorption fine structure (NEXAFS), contact-angle titration, and SPR, respectively. The influences of the surface and solution factors, e.g., ligand density, salt concentration, and solution pH, on protein adsorption were determined by SPR. Our results showed that ligand density affects both equilibrium and dynamic aspects of the interactions. Specifically, a dense ligand leads to an increase in binding strength, rapid adsorption, slow desorption, and low specificity. In addition, both hydrophobic interactions and hydrogen bonding contribute significantly to the protein adsorption at neutral pH, while the electrostatic repulsion is overwhelmed under acidic conditions. The hydrophobic interaction at a high concentration of lyotropic salt would cause drastic conformational changes in the adsorbed protein. Combined with the self-assembly technique, SPR proves to be a powerful tool for studying the interactions between an antibody and a chromatographic ligand.


Subject(s)
Chromatography/methods , Surface Plasmon Resonance/methods , Hydrophobic and Hydrophilic Interactions , Ligands
SELECTION OF CITATIONS
SEARCH DETAIL
...