Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Pharmacol Res ; 201: 107092, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38311014

RESUMO

AP endonuclease-1/Redox factor-1 (APE1/Ref-1 or Ref-1) is a multifunctional protein that is overexpressed in most aggressive cancers and impacts various cancer cell signaling pathways. Ref-1's redox activity plays a significant role in activating transcription factors (TFs) such as NFκB, HIF1α, STAT3 and AP-1, which are crucial contributors to the development of tumors and metastatic growth. Therefore, development of potent, selective inhibitors to target Ref-1 redox function is an appealing approach for therapeutic intervention. A first-generation compound, APX3330 successfully completed phase I clinical trial in adults with progressing solid tumors with favorable response rate, pharmacokinetics (PK), and minimal toxicity. These positive results prompted us to develop more potent analogs of APX3330 to effectively target Ref-1 in solid tumors. In this study, we present structure-activity relationship (SAR) identification and validation of lead compounds that exhibit a greater potency and a similar or better safety profile to APX3330. In order to triage and characterize the most potent and on-target second-generation Ref-1 redox inhibitors, we assayed for PK, mouse and human S9 fraction metabolic stability, in silico ADMET properties, ligand-based WaterLOGSY NMR measurements, pharmacodynamic markers, cell viability in multiple cancer cell types, and two distinct 3-dimensional (3D) cell killing assays (Tumor-Microenvironment on a Chip and 3D spheroid). To characterize the effects of Ref-1 inhibition in vivo, global proteomics was used following treatment with the top four analogs. This study identified and characterized more potent inhibitors of Ref-1 redox function (that outperformed APX3330 by 5-10-fold) with PK studies demonstrating efficacious doses for translation to clinic.


Assuntos
DNA Liase (Sítios Apurínicos ou Apirimidínicos) , Neoplasias , Adulto , Humanos , Animais , Camundongos , Inibidores da Angiogênese , Apoptose , Bioensaio , Neoplasias/tratamento farmacológico , Microambiente Tumoral
2.
Immun Ageing ; 19(1): 64, 2022 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-36517853

RESUMO

Obesity is characterized by an elevated amount of fat and energy storage in the adipose tissue (AT) and is believed to be the root cause of many metabolic diseases (MDs). Obesity is associated with low-grade chronic inflammation in AT. Like obesity, chronic inflammation and MDs are prevalent in the elderly. The resident immune microenvironment is not only responsible for maintaining AT homeostasis but also plays a crucial role in stemming obesity and related MDs. Mounting evidence suggests that obesity promotes activation in resident T cells and macrophages. Additionally, inflammatory subsets of T cells and macrophages accumulated into the AT in combination with other immune cells maintain low-grade chronic inflammation. microRNAs (miRs) are small non-coding RNAs and a crucial contributing factor in maintaining immune response and obesity in AT. AT resident T cells, macrophages and adipocytes secrete various miRs and communicate with other cells to create a potential effect in metabolic organ crosstalk. AT resident macrophages and T cells-associated miRs have a prominent role in regulating obesity by targeting several signaling pathways. Further, miRs also emerged as important regulators of cellular senescence and aging. To this end, a clear link between miRs and longevity has been demonstrated that implicates their role in regulating lifespan and the aging process. Hence, AT and circulating miRs can be used as diagnostic and therapeutic tools for obesity and related disorders. In this review, we discuss how miRs function as biomarkers and impact obesity, chronic inflammation, and aging.

3.
Anal Bioanal Chem ; 411(24): 6475-6485, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31375853

RESUMO

Wilson's disease (WD), which might lead to acute liver failure, is an inherited disorder characterized by accumulation of copper (Cu2+) in the brain, the liver, and other vital organs. In the clinic, decreased serum alkaline phosphatase (ALP) concentration is used for WD diagnosis. But to the best of our knowledge, using a fluorescent probe to simultaneously detect multiple factors in WD (e.g., Cu2+, pyrophosphate (PPi), and ALP) has not been reported. Herein, we rationally designed a fluorescent switch (E)-8-((4-methylbenzylidene)amino)napthalen-1-amine (L) and successfully applied it for sequential and selective detections of Cu2+, PPi, and ALP in vitro, in living cells and synovial fluid samples with "Off," "On," and "Off" fluorescence signals, respectively. Considering the obvious correlations among Cu2+, PPi, and ALP in WD, we envision that our fluorescent probe L could be applied to in vitro diagnosing WD in the near future.


Assuntos
Fosfatase Alcalina/análise , Cobre/análise , Difosfatos/análise , Corantes Fluorescentes/química , Células HeLa , Humanos
4.
Nanomedicine ; 15(1): 264-273, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30442595

RESUMO

Tissue specific T1/T2 dual contrast abilities for magnetic resonance imaging (MRI) have great significance in initial detection of cancer lesions. Herein, we developed a novel kind of Magnus nano-bullets (Mn-DTPA-F-MSNs) distinguished by magnetic (Fe3O4-NPs) head combined with mesoporous (SiO2) persist body, respectively. Subsequently, modify mesoporous SiO2 group and finally loaded with Mn2+. These Magnus nano-bullets have relaxivity value (r1 = 5.12 mM-1 s-1) and relaxivity value (r2 = 265.32 mM-1 s-1); they were > 2 folds in comparison to control at 3.0 T. Meanwhile, Magnus nano-bullets also offered significant enhancements for the detection of Glutathione (GSH), a biomarker that has been showed a redox responsive T1-weighted MRI effect in vitro and in vivo evaluations with good biocompatibility. Therefore, our finding endorses that Magnus nano-bullets offer a "smart" and tremendous strategy for greater GSH responsive T1/T2 dual MRI image probes for future biomedical applications.


Assuntos
Carcinoma Hepatocelular/diagnóstico , Neoplasias Hepáticas/diagnóstico , Imageamento por Ressonância Magnética/métodos , Compostos de Manganês/química , Nanopartículas/química , Animais , Apoptose , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Proliferação de Células , Meios de Contraste/metabolismo , Glutationa/metabolismo , Humanos , Técnicas In Vitro , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Imagens de Fantasmas , Dióxido de Silício/química , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Bioact Mater ; 36: 301-316, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38496035

RESUMO

Natural killer (NK) cells are cytotoxic immune cells that can eliminate target cells without prior stimulation. Human induced pluripotent stem cells (iPSCs) provide a robust source of NK cells for safe and effective cell-based immunotherapy against aggressive cancers. In this in vitro study, a feeder-free iPSC differentiation was performed to obtain iPSC-NK cells, and distinct maturational stages of iPSC-NK were characterized. Mature cells of CD56bright CD16bright phenotype showed upregulation of CD56, CD16, and NK cell activation markers NKG2D and NKp46 upon IL-15 exposure, while exposure to aggressive atypical teratoid/rhabdoid tumor (ATRT) cell lines enhanced NKG2D and NKp46 expression. Malignant cell exposure also increased CD107a degranulation markers and stimulated IFN-γ secretion in activated NK cells. CD56bright CD16bright iPSC-NK cells showed a ratio-dependent killing of ATRT cells, and the percentage lysis of CHLA-05-ATRT was higher than that of CHLA-02-ATRT. The iPSC-NK cells were also cytotoxic against other brain, kidney, and lung cancer cell lines. Further NK maturation yielded CD56-ve CD16bright cells, which lacked activation markers even after exposure to interleukins or ATRT cells - indicating diminished cytotoxicity. Generation and characterization of different NK phenotypes from iPSCs, coupled with their promising anti-tumor activity against ATRT in vitro, offer valuable insights into potential immunotherapeutic strategies for brain tumors.

6.
Life Sci ; 336: 122317, 2024 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-38040245

RESUMO

Interstitial cystitis (IC), also called painful bladder syndrome (PBS), is 2 to 5 times more common in women than in men, yet its cause and pathogenesis remain unclear. In our study using the cyclophosphamide (CYP)-induced mouse model of cystitis, histological evaluation of the urinary bladder (UB) lamina propria (LP) showed immune cell infiltrations, indicating moderate to severe inflammation. In this study, we noticed a differential expression of a subset of microRNAs (miRs) in the UB cells (UBs) of CYP-induced cystitis as compared to the control. UB inflammatory scores and inflammatory signaling were also elevated in CYP-induced cystitis as compared to control. We identified eight UBs miRs that exhibited altered expression after CYP induction and are predicted to have a role in inflammation and smooth muscle function (miRs-34c-5p, -34b-3p, -212-3p, -449a-5p, -21a-3p, -376b-3p, -376b-5p and - 409-5p). Further analysis using ELISA for inflammatory markers and real-time PCR (RT-PCR) for differentially enriched miRs identified miR-34c as a potential target for the suppression of UB inflammation in cystitis. Blocking miR-34c by antagomir ex vivo reduced STAT3, TGF-ß1, and VEGF expression in the UBs, which was induced during cystitis as compared to control. Interestingly, miR-34c inhibition also downregulated ROCK2 but elevated ROCK1 expression in bladder and detrusor cells. Thus, the present study shows that targeting miR-34c can mitigate the STAT3, TGF-ß, and VEGF, inflammatory signaling in UB, and suppress ROCK2 expression in UBs to effectively suppress the inflammatory response in cystitis. This study highlights miR-34c as a potential biomarker and/or serves as the basis for new therapies for the treatment of cystitis.


Assuntos
Cistite Intersticial , Cistite , MicroRNAs , Masculino , Camundongos , Animais , Humanos , Feminino , Fator A de Crescimento do Endotélio Vascular/metabolismo , Cistite/induzido quimicamente , Bexiga Urinária/metabolismo , Cistite Intersticial/genética , Cistite Intersticial/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Ciclofosfamida/efeitos adversos , Inflamação/metabolismo , Quinases Associadas a rho/genética , Quinases Associadas a rho/metabolismo
7.
Front Immunol ; 14: 1213415, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37334370

RESUMO

Background: Obesity is a multifactorial disease characterized by an enhanced amount of fat and energy storage in adipose tissue (AT). Obesity appears to promote and maintain low-grade chronic inflammation by activating a subset of inflammatory T cells, macrophages, and other immune cells that infiltrate the AT. Maintenance of AT inflammation during obesity involves regulation by microRNAs (miRs), which also regulate the expression of genes implicated in adipocyte differentiation. This study aims to use ex vivo and in vitro approaches to evaluate the role and mechanism of miR-10a-3p in adipose inflammation and adipogenesis. Methods: Wild-type BL/6 mice were placed on normal (ND) and high-fat diet (HFD) for 12 weeks and their obesity phenotype, inflammatory genes, and miRs expression were examined in the AT. We also used differentiated 3T3-L1 adipocytes for mechanistic in vitro studies. Results: Microarray analysis allowed us to identify an altered set of miRs in the AT immune cells and Ingenuity pathway analysis (IPA) prediction demonstrated that miR-10a-3p expression was downregulated in AT immune cells in the HFD group as compared to ND. A molecular mimic of miR-10a-3p reduced expression of inflammatory M1 macrophages, cytokines, and chemokines, including transforming growth factor-beta 1 (TGF-ß1), transcription factor Krüppel-like factor 4 (KLF4), and interleukin 17F (IL-17F) and induced expression of forkhead box P3 (FoxP3) in the immune cells isolated from AT of HFD-fed mice as compared to ND. In differentiated 3T3-L1 adipocytes, the miR-10a-3p mimics also reduced expression of proinflammatory genes and lipid accumulation, which plays a role in the dysregulation of AT function. In these cells, overexpression of miR-10a-3p reduced the expression of TGF-ß1, Smad3, CHOP-10, and fatty acid synthase (FASN), relative to the control scramble miRs. Conclusion: Our findings suggest that miR-10a-3p mimic mediates the TGF-ß1/Smad3 signaling to improve metabolic markers and adipose inflammation. This study provides a new opportunity for the development of miR-10a-3p as a novel therapeutic for adipose inflammation, and its associated metabolic disorders.


Assuntos
Adiposidade , MicroRNAs , Animais , Camundongos , Adiposidade/genética , Inflamação/genética , Inflamação/metabolismo , MicroRNAs/metabolismo , Obesidade/genética , Obesidade/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo
8.
Biomed Pharmacother ; 161: 114514, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-36921534

RESUMO

The beneficial effects of the polyphenolic compound piceatannol (PC) has been reported for metabolic diseases, antiproliferative, antioxidant, and anti-cancer properties. Despite its beneficial effects on inflammatory diseases, little is known about how PC regulates inflammatory responses and adipogenesis. Therefore, this study was designed to determine the effects of PC on the inflammatory response and adipogenesis. The effect of PC on splenocytes, 3T3-L1 adipocytes, and RAW264.7 macrophages was analyzed by flow cytometry, qRT-PCR, morphometry, and western blot analysis. PC induced apoptosis in activated T cells in a dose-dependent manner using stimulated splenocytes and reduced the activation of T cells, altered T cell frequency, and interestingly induced the frequency of regulatory T (Treg) cells as compared to controls. PC suppressed the expression of TNF-α, iNOS, IL-6R, and NF-κB activation in RAW264.7 macrophages after lipopolysaccharides (LPS)-induction as compared to the control. Interestingly, PC altered the cell morphology of 3T3-L1 adipocytes with a concomitant decrease in cell volume, lipid deposition, and TNF-α expression, but upregulation of leptin and IL-1ß. Our findings suggested that PC induced apoptosis in activated T cells, decreased immune cell activation and inflammatory response, and hindered adipogenesis. This new set of data provides promising hope as a new therapeutic to treat both inflammatory disease and obesity.


Assuntos
Adipogenia , Fator de Necrose Tumoral alfa , Camundongos , Animais , Fator de Necrose Tumoral alfa/metabolismo , Linfócitos T Reguladores/metabolismo , Transdução de Sinais , Células 3T3-L1 , Lipopolissacarídeos/farmacologia , NF-kappa B/metabolismo , Inflamação/tratamento farmacológico , Inflamação/metabolismo
9.
Front Immunol ; 13: 903834, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35720309

RESUMO

Interstitial cystitis (IC)/bladder pain syndrome (BPS), hereafter referred together as IC, is a clinical syndrome characterized by sterile inflammation in the bladder. While the etiology and pathophysiology of IC remain unclear, it may involve autoimmunity in light of the significant role played by the NLRP3 inflammasome. However, the effect of NLRP3 inhibitors including dapansutrile (Dap) on IC had not been explored previously. Here, we investigated the effect of Dap in the cyclophosphamide (CYP)-induced experimental mouse model of IC, which results in functional and histological alterations confined to the urinary bladder (UB) comparable to that of clinical IC. CYP-induced mice treated with Dap exhibited improved UB pathology and reductions in inflammation scores and the frequency and the number of mast cells and neutrophils, relative to mice that received CYP alone. Dap- and CYP-treated mice also exhibited infiltration of T cells in the spleen and iliac lymph nodes (ILNs) and a concurrent significant decrease (p<0.01) in CXCR3+CD8+ T cells in the UB, induction of systemic and mucosal dendritic cells (DCs), and reduced levels of systemic proinflammatory cytokines, as compared to CYP alone. We also observed decreases in the expression of several signaling pathways regulators, including interleukin-1 beta (IL-1ß), NLRP3, caspase-1, nuclear factor kappa B (NF-κB), and inducible nitric oxide synthase (iNOS) in the UB of CYP- and Dap-treated mice, relative to those receiving CYP alone. Taken together, these results suggest that Dap suppresses IC through the reduction of CXCR3+T cells, mast cells, and neutrophils in the UB and induces DCs as a protective measure. The present study identifies the mechanisms underlying the amelioration of IC by the NLRP3 inhibitor Dap and may provide an avenue for a potential therapeutic agent for the treatment of IC.


Assuntos
Cistite Intersticial , Animais , Linfócitos T CD8-Positivos/metabolismo , Ciclofosfamida/efeitos adversos , Cistite Intersticial/induzido quimicamente , Cistite Intersticial/tratamento farmacológico , Cistite Intersticial/metabolismo , Inflamassomos/uso terapêutico , Inflamação , Camundongos , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Nitrilas , Sulfonas
10.
Int Rev Immunol ; 41(1): 30-44, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34423733

RESUMO

Obesity is characterized by low-grade, chronic inflammation, which promotes insulin resistance and diabetes. Obesity can lead to the development and progression of many autoimmune diseases, including inflammatory bowel disease, psoriasis, psoriatic arthritis, rheumatoid arthritis, thyroid autoimmunity, and type 1 diabetes mellitus (T1DM). These diseases result from an alteration of self-tolerance by promoting pro-inflammatory immune response by lowering numbers of regulatory T cells (Tregs), increasing Th1 and Th17 immune responses, and inflammatory cytokine production. Therefore, understanding the immunological changes that lead to this low-grade inflammatory milieu becomes crucial for the development of therapies that suppress the risk of autoimmune diseases and other immunological conditions. Cells generate extracellular vesicles (EVs) to eliminate cellular waste as well as communicating the adjacent and distant cells through exchanging the components (genetic material [DNA or RNA], lipids, and proteins) between them. Immune cells and adipocytes from individuals with obesity and a high basal metabolic index (BMI) produce also release exosomes (EXOs) and microvesicles (MVs), which are collectively called EVs. These EVs play a crucial role in the development of autoimmune diseases. The current review discusses the immunological dysregulation that leads to inflammation, inflammatory diseases associated with obesity, and the role played by EXOs and MVs in the induction and progression of this devastating conditi8on.


Assuntos
Doenças Autoimunes , Exossomos , Vesículas Extracelulares , Doenças Autoimunes/metabolismo , Exossomos/metabolismo , Vesículas Extracelulares/metabolismo , Humanos , Inflamação/metabolismo , Obesidade/metabolismo
11.
Pharmaceutics ; 14(5)2022 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-35631522

RESUMO

The causes of Crohn's disease (CD) and ulcerative colitis (UC), the two most common forms of inflammatory bowel disease (IBD), are multi-factorial and include dysregulation of immune cells in the intestine. Cannabinoids mediate protection against intestinal inflammation by binding to the G-protein coupled cannabinoid receptors 1 and 2 (CB1 and CB2). Here, we investigate the effects of the CB2 inverse agonist SMM-189 on dextran sodium sulfate (DSS)-induced experimental colitis. We observed that SMM-189 effectively attenuated the overall clinical score, reversed colitis-associated pathogenesis, and increased both body weight and colon length. Treatment with SMM-189 also increased the expression of CB2 and protein kinase A (PKA) in colon lamina propria lymphocytes (LPLs). We noticed alterations in the percentage of Th17, neutrophils, and natural killer T (NKT) cells in the spleen, mesenteric lymph nodes (MLNs), and LPLs of mice with DSS-induced colitis after treatment with SMM-189 relative to DSS alone. Further, myeloid-derived suppressor cells (MDSCs) during colitis progression increased with SMM-189 treatment as compared to DSS alone or with control cohorts. These findings suggest that SMM-189 may ameliorate experimental colitis by inducing the expression of endogenous CB2 and PKA in LPLs, increasing numbers of MDSCs in the spleen, and reducing numbers of Th17 cells and neutrophils in the spleen, MLNs, and LPLs. Taken together, these data support the idea that SMM-189 may be developed as a safe novel therapeutic target for IBD.

12.
J Hazard Mater ; 435: 128884, 2022 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-35483261

RESUMO

Plastics have been part of our ecosystem for about a century and their degradation by different environmental factors produce secondary microplastics (MPs). To date, the impact of MPs on human health has not been well investigated. To understand the possible effects of polystyrene-MPs (PS-MPs) on the human brain, a 3D model of human forebrain cortical spheroids has been derived, which mimics early development of human cerebral cortex. The spheroids were exposed to 100, 50, and 5 µg/mL of 1 µm and 10 µm PS-MPs during day 4-10 and day 4-30. The short-term MP exposure showed the promoted proliferation and high gene expression of Nestin, PAX6, ATF4, HOXB4 and SOD2. For long-term exposure, reduced cell viability was observed. Moreover, changes in size and concentration of PS-MPs altered the gene expression of DNA damage and neural tissue patterning. In particular, ß-tubulin III, Nestin, and TBR1/TBR2 gene expression decreased in PS-MP treated conditions compare to the untreated control. The results of this study suggest that the size- and concentration-dependent exposure to PS-MPs can adversely affect embryonic brain-like tissue development in forebrain cerebral spheroids. This study has significance in assessing environmental factors in neurotoxicity and degeneration in human.


Assuntos
Células-Tronco Pluripotentes , Poluentes Químicos da Água , Córtex Cerebral , Ecossistema , Humanos , Microplásticos , Nestina/genética , Plásticos , Células-Tronco Pluripotentes/química , Poliestirenos , Poluentes Químicos da Água/análise
13.
Cells ; 11(8)2022 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-35456006

RESUMO

Obesity is a complex disease associated with various metabolic abnormalities, cardiovascular diseases, and low-grade chronic inflammation. Inflammation associated with T helper 1 (Th1) immune cells is dominant in adipose tissue (AT) and exerts metabolically deleterious impacts. The precise mechanism of alteration in AT immune system and its effect on metabolic homeostasis remains unclear. In this study, we investigated how a high-fat diet (HFD) alters the AT immune response and influences inflammation during obesity. HFD consumption amends the metabolic parameters, including body weight, glucose, and insulin levels. We observed increased infiltration of Th17 cells, a subset of dendritic cells (CD103+), and M1 macrophages in AT of mice fed HFD compared to those fed a normal diet (ND). In mice that were fed HFD, we also observed a reduction in regulatory T cells (Tregs) relative to the numbers of these cells in mice fed ND. Corresponding with this, mice in the HFD group exhibited higher levels of proinflammatory cytokines and chemokines than those in the ND group. We also observed alterations in signaling pathways, including increased protein expression of IRF3, TGFß1, and mRNA expression of IL-6, KLF4, and STAT3 in the AT of the mice fed HFD as compared to those fed ND. Further, HFD-fed mice exhibited decreased protein expression of peroxisome proliferator-activated receptor-gamma (PPAR-γ) compared to mice fed ND, suggesting that PPAR-γ functions as a negative regulator of Th17 cell differentiation. These results suggest that HFD induces increased levels of inflammatory cytokines and key immune cells, including Th17, M1 macrophages, and CD103+ dendritic cells, and reduces levels of PPAR-γ and Tregs to sustain AT inflammation. This study supports the notion that dysregulation of Th17/Tregs, which polarizes macrophages towards M1 phenotypes in part through TGFß1-IRF3-STAT3 and negatively regulates PPAR-γ mediated pathways, results in AT inflammation during obesity.


Assuntos
Tecido Adiposo , Dieta Hiperlipídica , Tecido Adiposo/metabolismo , Animais , Citocinas/metabolismo , Dieta Hiperlipídica/efeitos adversos , Inflamação/metabolismo , Macrófagos/metabolismo , Camundongos , Obesidade/metabolismo , PPAR gama/metabolismo , Fenótipo
14.
Cells ; 11(11)2022 05 26.
Artigo em Inglês | MEDLINE | ID: mdl-35681445

RESUMO

The lymphatic system is pivotal for immunosurveillance and the maintenance of tissue homeostasis. Lymphangiogenesis, the formation of new lymphatic vessels from pre-existing vessels, has both physiological and pathological roles. Recent advances in the molecular mechanisms regulating lymphangiogenesis have opened a new area of research on reparative lymphangiogenesis for the treatment of various pathological disorders comprising neurological disorders, cardiac repair, autoimmune disease, obesity, atherosclerosis, etc. Reactive oxygen species (ROS) produced by the various cell types serve as signaling molecules in several cellular mechanisms and regulate various aspects of growth-factor-mediated responses, including lymphangiogenesis. The ROS, including superoxide anion, hydrogen peroxide, and nitric oxide, play both beneficial and detrimental roles depending upon their levels and cellular microenvironment. Low ROS levels are essential for lymphangiogenesis. On the contrary, oxidative stress due to enhanced ROS generation and/or reduced levels of antioxidants suppresses lymphangiogenesis via promoting lymphatic endothelial cell apoptosis and death. In this review article, we provide an overview of types and sources of ROS, discuss the role of ROS in governing lymphangiogenesis and lymphatic function, and summarize the role of lymphatics in various diseases.


Assuntos
Linfangiogênese , Vasos Linfáticos , Células Endoteliais/metabolismo , Sistema Linfático , Espécies Reativas de Oxigênio/metabolismo
15.
ACS Biomater Sci Eng ; 8(2): 801-813, 2022 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-35072448

RESUMO

The environmental nanoscale iron magnetite may contribute to the risk of developing neurodegenerative diseases. In addition, iron oxides can be used as the contrast agents in magnetic resonance imaging of neural tissues. The potential long-term impact of nanoscale iron oxides on cellular stress and neuro-inflammation remains unknown. The objective of this study is to evaluate the long-term effects of nanoscale iron oxide exposure on human pluripotent stem cell-derived cortical spheroids that mimic human forebrain-like tissue development. In particular, the cortical spheroids were treated with 8 nm and 15-20 nm magnetite at 0.023, 2.3, and 23 µg/mL for 4-30 days. The cell viability did not show significant differences among different test groups. The neuronal marker ß-tubulin III, cell proliferation marker Ki67, and antioxidant enzyme SOD2 did not show significant changes either. The molecular levels of cellular stress, inflammation, cell apoptosis, DNA damage and repair, and the reactive oxygen species (ROS) response were measured. A negative effect (i.e., increased inflammation and ROS response genes) of 8 nm iron oxide exposure and a positive effect (i.e., decreased inflammation, apoptosis, and ROS response and clean up genes) for 15-20 nm iron oxide exposure were observed. It is postulated that the intracellular iron content and the aggregation of iron oxides contribute to the observed differential response. Although our results demonstrate similar intracellular iron content for 8 nm and 15-20 nm groups, the aggregation is more severe for the 8 nm group (∼500 nm) than the 15 nm group (∼220-250 nm). Therefore, our data indicate an iron oxide aggregate size-dependent effects on cellular stress, inflammation, cell apoptosis, DNA damage, and the ROS response in the developing human forebrain-like tissue.


Assuntos
Óxido Ferroso-Férrico , Células-Tronco , Sobrevivência Celular , Óxido Ferroso-Férrico/farmacologia , Humanos , Prosencéfalo , Espécies Reativas de Oxigênio/farmacologia
16.
Sci Rep ; 12(1): 12880, 2022 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-35896708

RESUMO

Human cerebellum consists of high density and complexity of neurons. Thus, it is challenging to differentiate cerebellar-like organoids with similar cellular markers and function to the human brain. Our previous study showed that the combination of retinoic acid (RA), Wingless/integrated (Wnt) activator, and Sonic Hedgehog (SHH) activator promotes cerebellar differentiation from human induced pluripotent stem cells (hiPSCs). This study examined phenotypic, metabolic, and biogenesis in early cerebellar development. Cerebellum spheroids were differentiated from human iPSK3 cells. During day 7-14, RA and Wnt activator CHIR99021 were used and SHH activator purmorphamine (PMR) was added later to promote ventralization. Gene expression for early cerebellar layer markers, metabolism, and extracellular vesicle (EV) biogenesis were characterized. Zinc-induced neurotoxicity was investigated as a proof-of-concept of neurotoxicity study. Flow cytometry results showed that there was no significant difference in NEPH3, PTF1A, OLIG2, and MATH1 protein expression between RCP (RA-CHIR-PMR) versus the control condition. However, the expression of cerebellar genes for the molecular layer (BHLE22), the granule cell layer (GABRB2, PAX6, TMEM266, KCNIP4), the Bergmann glial cells (QK1, DAO), and the Purkinje cell layer (ARHGEF33, KIT, MX1, MYH10, PPP1R17, SCGN) was significantly higher in the RCP condition than the control. The shift in metabolic pathways toward glycolysis was observed for RCP condition. The EV biogenesis marker expression was retained. Mild zinc-induced neurotoxicity may exist when zinc exposure exceeds 1.0 µM. RCP treatment can promote specific cerebellar-like differentiation from hiPSCs indicated by gene expression of early cerebellar markers and regionally enriched genes. The higher cerebellar marker expression is accompanied by the elevated glycolysis with the retained EV biogenesis. This study should advance the understanding of biomarkers during early cerebellar development for cerebellum organoid engineering and neurotoxicity study.


Assuntos
Cerebelo , Proteínas Hedgehog , Células-Tronco Pluripotentes Induzidas , Esferoides Celulares , Cerebelo/citologia , Proteínas Hedgehog/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Neurônios/metabolismo , Esferoides Celulares/metabolismo , Tretinoína/metabolismo , Zinco/metabolismo
17.
Front Immunol ; 12: 716564, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34335632

RESUMO

Interstitial cystitis (IC)/bladder pain syndrome (BPS) primarily affects women. It varies in its severity and currently has no effective treatment. The symptoms of IC include pelvic pain, urgency and frequency of urination, and discomfort or pain in the bladder and lower abdomen. The bladders of IC patients exhibit infiltration by immune cells, which lends credence to the hypothesis that immune mechanisms also play a role in the etiology and pathophysiology of IC. The Differentially expressed microRNAs (miRs) in immune cells may serve as crucial immunoregulators in the IC. Therefore, we sought to determine whether miRs might play a regulatory role in the progression and pathogenesis of IC, using experimental autoimmune cystitis (EAC) model. In the present study, we observed differential expression of a specific subset of miRs in iliac lymph nodes (ILNs) and urinary bladders (UB) of IC mice compared to that in control mice. Microarray analysis of 96 miRs from the bladder and 135 miRs from ILNs allowed us to identify 50 that exhibited at least a 1.5-fold greater difference in expression in EAC mice compared to control mice. Hierarchical cluster analysis of the microarray data was used to search available databases to predict molecular pathways with which the miRs might interact. Four miRs from each organ that exhibited altered expression in EAC mice and that were predicted to have roles in inflammation (miR-146a, -181, -1931, and -5112) were selected for further analysis by reverse transcription-polymerase chain reaction (RT-PCR). All were confirmed to be elevated in EAC mice. Histological inflammatory scores, systemic chemokines, and cytokines expressed by T helper type 1 (Th1) lymphocytes were also elevated in EAC mice as compared to control animals. We hypothesize that the mechanism of EAC induction might involve the modulation of specific miRs that increase local and systemic levels of chemokines and cytokines. The present study identifies novel miRs expressed in UB and ILNs that will allow us to highlight mechanisms of EAC pathogenesis and may provide potential biomarkers and/or serve as the basis of new therapies for the treatment of IC.


Assuntos
Cistite Intersticial/diagnóstico , Cistite Intersticial/etiologia , Suscetibilidade a Doenças , Regulação da Expressão Gênica , MicroRNAs/genética , Animais , Doenças Autoimunes/diagnóstico , Doenças Autoimunes/etiologia , Doenças Autoimunes/metabolismo , Biomarcadores , Cistite Intersticial/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Perfilação da Expressão Gênica , Humanos , Mediadores da Inflamação/metabolismo , Camundongos , Índice de Gravidade de Doença
18.
Front Immunol ; 12: 680944, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34248964

RESUMO

Obesity in the United States and worldwide reached epidemic proportions within the last 20 years. Obesity is a very powerful health determinant or indicator that facilitates the development and progression of several metabolic diseases, insulin resistance, and low-grade chronic inflammation. Low-grade chronic inflammation in adipose tissue (AT) is marked by the accumulation of T cells, macrophages, and other immune cells and increased production of proinflammatory cytokines. During the onset of obesity but before the influx of macrophages, the AT is infiltrated by T cells that are strongly implicated in the initiation of obesity-associated inflammation. In comparing mice fed a high-fat diet (HFD) with those fed a normal diet (ND), we observed in HFD epididymal AT induction and infiltration of activated T cells, an accumulation and polarization of macrophages, and an increase in populations of activated CD4+ T cells and CD8+ T cells that express CXCR3 or killer cell lectin-like receptor subfamily G member 1 (KLRG1). Levels of inflammatory cytokines and leptin and the results of in vitro co-culture experiments revealed interactions among HFD- and ND-induced CD8+ T cells, macrophages, and adipocytes. Our findings suggest that obese tissues activate and induce both CD4+ and CD8+ CD69+ T cells and augment the expression of CXCR3 receptors, which promotes the recruitment and numbers of pro-inflammatory M1 macrophages to maintain low-grade chronic inflammation. The results support the hypothesis that CXCR3-expressing CD8+T cells play an essential role in the initiation and maintenance of adipose tissue inflammation.


Assuntos
Tecido Adiposo/imunologia , Tecido Adiposo/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Dieta Hiperlipídica , Macrófagos/imunologia , Macrófagos/metabolismo , Animais , Biomarcadores , Peso Corporal , Células Cultivadas , Doença Crônica , Citocinas/metabolismo , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Metabolismo Energético , Expressão Gênica , Imunofenotipagem , Inflamação/etiologia , Inflamação/metabolismo , Inflamação/patologia , Contagem de Linfócitos , Masculino , Camundongos , Receptores CXCR3/genética , Receptores CXCR3/metabolismo
19.
Cells ; 10(8)2021 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-34440785

RESUMO

Volumetric muscle loss (VML) is defined as a condition in which a large volume of skeletal muscle is lost due to physical insult. VML often results in a heightened immune response, resulting in significant long-term functional impairment. Estimates indicate that ~250,000 fractures occur in the US alone that involve VML. Currently, there is no active treatment to fully recover or repair muscle loss in VML patients. The health economics burden due to VML is rapidly increasing around the world. Immunologists, developmental biologists, and muscle pathophysiologists are exploring both immune responses and biomaterials to meet this challenging situation. The inflammatory response in muscle injury involves a non-specific inflammatory response at the injured site that is coordination between the immune system, especially macrophages and muscle. The potential role of biomaterials in the regenerative process of skeletal muscle injury is currently an important topic. To this end, cell therapy holds great promise for the regeneration of damaged muscle following VML. However, the delivery of cells into the injured muscle site poses a major challenge as it might cause an adverse immune response or inflammation. To overcome this obstacle, in recent years various biomaterials with diverse physical and chemical nature have been developed and verified for the treatment of various muscle injuries. These biomaterials, with desired tunable physicochemical properties, can be used in combination with stem cells and growth factors to repair VML. In the current review, we focus on how various immune cells, in conjunction with biomaterials, can be used to promote muscle regeneration and, most importantly, suppress VML pathology.


Assuntos
Materiais Biocompatíveis/uso terapêutico , Imunomodulação/efeitos dos fármacos , Músculo Esquelético/efeitos dos fármacos , Doenças Musculares/terapia , Regeneração/efeitos dos fármacos , Animais , Humanos , Imunomodulação/imunologia , Músculo Esquelético/imunologia , Músculo Esquelético/patologia , Doenças Musculares/imunologia , Doenças Musculares/fisiopatologia , Regeneração/imunologia , Regeneração/fisiologia , Transplante de Células-Tronco/métodos , Engenharia Tecidual/métodos , Alicerces Teciduais/química
20.
Cells ; 10(5)2021 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-33923175

RESUMO

Obesity is characterized as a complex and multifactorial excess accretion of adipose tissue (AT) accompanied with alterations in the immune response that affects virtually all age and socioeconomic groups around the globe. The abnormal accumulation of AT leads to several metabolic diseases, including nonalcoholic fatty liver disorder (NAFLD), low-grade inflammation, type 2 diabetes mellitus (T2DM), cardiovascular disorders (CVDs), and cancer. AT is an endocrine organ composed of adipocytes and immune cells, including B-Cells, T-cells and macrophages. These immune cells secrete various cytokines and chemokines and crosstalk with adipokines to maintain metabolic homeostasis and low-grade chronic inflammation. A novel form of adipokines, microRNA (miRs), is expressed in many developing peripheral tissues, including ATs, T-cells, and macrophages, and modulates the immune response. miRs are essential for insulin resistance, maintaining the tumor microenvironment, and obesity-associated inflammation (OAI). The abnormal regulation of AT, T-cells, and macrophage miRs may change the function of different organs including the pancreas, heart, liver, and skeletal muscle. Since obesity and inflammation are closely associated, the dysregulated expression of miRs in inflammatory adipocytes, T-cells, and macrophages suggest the importance of miRs in OAI. Therefore, in this review article, we have elaborated the role of miRs as epigenetic regulators affecting adipocyte differentiation, immune response, AT browning, adipogenesis, lipid metabolism, insulin resistance (IR), glucose homeostasis, obesity, and metabolic disorders. Further, we will discuss a set of altered miRs as novel biomarkers for metabolic disease progression and therapeutic targets for obesity.


Assuntos
Adipócitos/patologia , Linfócitos/imunologia , Macrófagos/imunologia , Doenças Metabólicas/patologia , MicroRNAs/genética , Obesidade/patologia , Animais , Humanos , Doenças Metabólicas/genética , Doenças Metabólicas/imunologia , Obesidade/genética , Obesidade/imunologia , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA