Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Mol Cell ; 51(4): 409-22, 2013 Aug 22.
Article in English | MEDLINE | ID: mdl-23973372

ABSTRACT

The individuals carrying melanocortin-1 receptor (MC1R) variants, especially those associated with red hair color, fair skin, and poor tanning ability (RHC trait), are more prone to melanoma; however, the underlying mechanism is poorly defined. Here, we report that UVB exposure triggers phosphatase and tensin homolog (PTEN) interaction with wild-type (WT), but not RHC-associated MC1R variants, which protects PTEN from WWP2-mediated degradation, leading to AKT inactivation. Strikingly, the biological consequences of the failure of MC1R variants to suppress PI3K/AKT signaling are highly context dependent. In primary melanocytes, hyperactivation of PI3K/AKT signaling leads to premature senescence; in the presence of BRAF(V600E), MC1R deficiency-induced elevated PI3K/AKT signaling drives oncogenic transformation. These studies establish the MC1R-PTEN axis as a central regulator for melanocytes' response to UVB exposure and reveal the molecular basis underlying the association between MC1R variants and melanomagenesis.


Subject(s)
Gene Expression Regulation/radiation effects , Melanocytes/metabolism , Melanoma, Experimental/pathology , PTEN Phosphohydrolase/metabolism , Receptor, Melanocortin, Type 1/metabolism , Skin Pigmentation/physiology , Ultraviolet Rays , Animals , Blotting, Western , Cells, Cultured , Humans , Immunoenzyme Techniques , Melanocytes/radiation effects , Melanoma, Experimental/genetics , Melanoma, Experimental/metabolism , Mice , Mutation/genetics , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptor, Melanocortin, Type 1/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Skin Pigmentation/radiation effects , alpha-MSH/genetics , alpha-MSH/metabolism
2.
J Immunol ; 206(11): 2763, 2021 06 01.
Article in English | MEDLINE | ID: mdl-34045319
3.
IUBMB Life ; 66(8): 530-7, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25130181

ABSTRACT

Prolonged perturbation of the endoplasmic reticulum (ER) leads to ER stress and unfolded protein response (UPR) and contributes to the pathogenesis of various chronic disorders. This review focuses on the role of ER stress and UPR in endothelial cells and the relevance of these processes to vascular diseases. Chronic activation of ER stress and UPR pathways in endothelial cells leads to increased oxidative stress and inflammation and often results in cell death. Because endothelial cells play a pivotal role in maintaining vascular homeostasis, various pathological conditions interfering with this homeostasis including homocysteinemia, hyperlipidemia, high glucose, insulin resistance, disturbed blood flow, and oxidative stress can lead to endothelial dysfunction in part through the activation of ER stress. We discuss recently discovered aspects of the role of ER stress/UPR in those pathological conditions. We also summarize recent findings implicating ER stress and UPR in systemic hypertension as well as pulmonary arterial hypertension. Finally, this review will highlight a novel role of UPR mediators in the process of angiogenesis.


Subject(s)
Endoplasmic Reticulum Stress/physiology , Endothelial Cells/physiology , Inflammation/physiopathology , Models, Biological , Oxidative Stress/physiology , Unfolded Protein Response/physiology , Vascular Diseases/physiopathology , Endothelial Cells/pathology , Humans , Hyperhomocysteinemia/physiopathology , Hyperlipidemias/physiopathology , Hypertension/physiopathology , Insulin Resistance/physiology , Neovascularization, Pathologic/physiopathology
4.
Arthritis Rheum ; 65(5): 1357-66, 2013 May.
Article in English | MEDLINE | ID: mdl-23400395

ABSTRACT

OBJECTIVE: Pulmonary arterial hypertension (PAH), a common complication of limited cutaneous systemic sclerosis (lcSSc), is associated with alterations of markers of inflammation and vascular damage in peripheral blood mononuclear cells (PBMCs). Endoplasmic reticulum (ER) stress and the unfolded protein response (UPR) have been implicated in autoimmune and inflammatory diseases. The goal of this study was to assess whether markers of ER stress and the UPR are present in PBMCs from lcSSc patients with PAH. METHODS: PBMCs were purified from 36 healthy controls, 32 lcSSc patients with PAH, and 34 lcSSc patients without PAH. Gene expression in healthy control PBMCs stimulated with thapsigargin was analyzed by DNA microarray. Genes were validated by quantitative real-time reverse transcription-polymerase chain reaction in PBMCs from healthy controls and lcSSc patients. RESULTS: Several ER stress/UPR genes, including BiP, activating transcription factor 4 (ATF-4), ATF-6, and a spliced form of X-box binding protein 1, were up-regulated in PBMCs from lcSSc patients, with the highest levels in patients with PAH. Thapsigargin up-regulated heat-shock proteins (HSPs) and interferon (IFN)-regulated genes in PBMCs from healthy controls. Selected HSP genes (particularly DnaJB1) and IFN-related genes were also found at significantly elevated levels in PBMCs from lcSSc patients, while IFN regulatory factor 4 expression was significantly decreased. There was a positive correlation between DnaJB1 and severity of PAH (measured by pulmonary artery pressure) (r = 0.56, P < 0.05) and between ER stress markers and interleukin-6 levels (r = 0.53, P < 0.0001) in PBMCs from lcSSc patients. CONCLUSION: This study demonstrates an association between select ER stress/UPR markers and lcSSc with PAH, suggesting that ER stress and the UPR may contribute to the altered function of circulating immune cells in lcSSc.


Subject(s)
Endoplasmic Reticulum Stress/genetics , Hypertension, Pulmonary/genetics , Leukocytes, Mononuclear/metabolism , Scleroderma, Limited/genetics , Unfolded Protein Response/genetics , Familial Primary Pulmonary Hypertension , Gene Expression Regulation , Humans , Hypertension, Pulmonary/blood , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/physiopathology , Leukocytes, Mononuclear/drug effects , Oligonucleotide Array Sequence Analysis , Scleroderma, Limited/blood , Scleroderma, Limited/complications , Scleroderma, Limited/physiopathology , Severity of Illness Index , Thapsigargin/pharmacology , Up-Regulation
5.
ACS Biomater Sci Eng ; 10(3): 1646-1660, 2024 03 11.
Article in English | MEDLINE | ID: mdl-38350651

ABSTRACT

Osteosarcoma (OS) is the most common primary malignant bone tumor, and the current standard of care for OS includes neoadjuvant chemotherapy, followed by an R0 surgical resection of the primary tumor, and then postsurgical adjuvant chemotherapy. Bone reconstruction following OS resection is particularly challenging due to the size of the bone voids and because patients are treated with adjuvant and neoadjuvant systemic chemotherapy, which theoretically could impact bone formation. We hypothesized that an osteogenic material could be used in order to induce bone regeneration when adjuvant or neoadjuvant chemotherapy is given. We utilized a biomimetic, biodegradable magnesium-doped hydroxyapatite/type I collagen composite material (MHA/Coll) to promote bone regeneration in the presence of systemic chemotherapy in a murine critical size defect model. We found that in the presence of neoadjuvant or adjuvant chemotherapy, MHA/Coll is able to enhance and increase bone formation in a murine critical size defect model (11.16 ± 2.55 or 13.80 ± 3.18 versus 8.70 ± 0.81 mm3) for pre-op cisplatin + MHA/Coll (p-value = 0.1639) and MHA/Coll + post-op cisplatin (p-value = 0.1538), respectively, at 12 weeks. These findings indicate that neoadjuvant and adjuvant chemotherapy will not affect the ability of a biomimetic scaffold to regenerate bone to repair bone voids in OS patients. This preliminary data demonstrates that bone regeneration can occur in the presence of chemotherapy, suggesting that there may not be a necessity to modify the current standard of care concerning neoadjuvant and adjuvant chemotherapy for the treatment of metastatic sites or micrometastases.


Subject(s)
Bone Neoplasms , Osteosarcoma , Humans , Animals , Mice , Cisplatin/pharmacology , Cisplatin/therapeutic use , Disease Models, Animal , Osteosarcoma/drug therapy , Bone Regeneration , Bone Neoplasms/drug therapy , Bone Neoplasms/pathology , Bone Neoplasms/surgery
6.
J Cell Physiol ; 228(8): 1774-83, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23359533

ABSTRACT

Systemic sclerosis (SSc) is a complex disease characterized by vascular alterations, activation of the immune system and tissue fibrosis. Previous studies have implicated activation of the interferon pathways in the pathogenesis of SSc. The goal of this study was to determine whether interferon type I and/or type II could play a pathogenic role in SSc vasculopathy. Human dermal microvascular endothelial cells (HDMVECs) and fibroblasts were obtained from foreskins of healthy newborns. The RT Profiler PCR Array System was utilized to screen for EndoMT genes. Treatment with IFN-α or IFN-γ downregulated Fli1 and VE-cadherin. In contrast, IFN-α and IFN-γ exerted opposite effects on the expression of α-SMA, CTGF, ET-1, and TGFß2, with IFN-α downregulating and IFN-γ upregulating this set of genes. Blockade of TGFß signaling normalized IFN-γ-mediated changes in Fli1, VE-cadherin, CTGF, and ET-1 levels, whereas upregulation of α-SMA and TGFß2 was not affected. Bosentan treatment was more effective than TGFß blockade in reversing the actions of IFN-γ, including downregulation of α-SMA and TGFß2, suggesting that activation of the ET-1 pathway plays a main role in the IFN-γ responses in HDMECs. IFN-γ induced expression of selected genes related to endothelial-to-mesenchymal transition (EndoMT), including Snail1, FN1, PAI1, TWIST1, STAT3, RGS2, and components of the WNT pathway. The effect of IFN-γ on EndoMT was mediated via TGFß2 and ET-1 signaling pathways. This study demonstrates distinct effects of IFN-α and IFN-γ on the biology of vascular endothelial cells. IFN-γ may contribute to abnormal vascular remodeling and fibrogenesis in SSc, partially via induction of EndoMT.


Subject(s)
Endothelial Cells/metabolism , Endothelin-1/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Interferon-gamma/physiology , Transforming Growth Factor beta2/metabolism , Cells, Cultured , Endothelin-1/genetics , Fibrosis , Humans , Scleroderma, Systemic/etiology , Scleroderma, Systemic/pathology , Scleroderma, Systemic/physiopathology , Transforming Growth Factor beta2/genetics , Up-Regulation/drug effects
7.
Sci Rep ; 13(1): 6593, 2023 04 22.
Article in English | MEDLINE | ID: mdl-37087509

ABSTRACT

Pulmonary arterial hypertension (PAH) is a life-threatening condition characterized by a progressive increase in pulmonary vascular resistance leading to right ventricular failure and often death. Here we report that deficiency of transcription factor GATA6 is a shared pathological feature of PA endothelial (PAEC) and smooth muscle cells (PASMC) in human PAH and experimental PH, which is responsible for maintenance of hyper-proliferative cellular phenotypes, pulmonary vascular remodeling and pulmonary hypertension. We further show that GATA6 acts as a transcription factor and direct positive regulator of anti-oxidant enzymes, and its deficiency in PAH/PH pulmonary vascular cells induces oxidative stress and mitochondrial dysfunction. We demonstrate that GATA6 is regulated by the BMP10/BMP receptors axis and its loss in PAECs and PASMC in PAH supports BMPR deficiency. In addition, we have established that GATA6-deficient PAEC, acting in a paracrine manner, increase proliferation and induce other pathological changes in PASMC, supporting the importance of GATA6 in pulmonary vascular cell communication. Treatment with dimethyl fumarate resolved oxidative stress and BMPR deficiency, reversed hemodynamic changes caused by endothelial Gata6 loss in mice, and inhibited proliferation and induced apoptosis in human PAH PASMC, strongly suggesting that targeting GATA6 deficiency may provide a therapeutic advance for patients with PAH.


Subject(s)
Bone Morphogenetic Proteins , GATA6 Transcription Factor , Oxidative Stress , Pulmonary Arterial Hypertension , Animals , Mice , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Cell Proliferation , Cells, Cultured , Familial Primary Pulmonary Hypertension/pathology , GATA6 Transcription Factor/genetics , GATA6 Transcription Factor/metabolism , Myocytes, Smooth Muscle/metabolism , Pulmonary Arterial Hypertension/genetics , Pulmonary Arterial Hypertension/metabolism , Pulmonary Arterial Hypertension/pathology , Pulmonary Artery/pathology , Vascular Remodeling
8.
Curr Opin Rheumatol ; 24(6): 663-8, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22918530

ABSTRACT

PURPOSE OF REVIEW: To review the present knowledge of the role of endoplasmic reticulum (ER) stress and unfolded protein response (UPR) in the pathogenesis of fibrotic diseases. RECENT FINDINGS: ER stress and UPR occur in a number of diseases associated with organ fibrosis; however, the contribution of these pathways to the fibrotic process has not been systematically investigated. Current studies suggest that prolonged ER stress may lead to fibrosis through activation of CCAAT/enhancer-binding homologous protein-mediated apoptosis, followed by an inflammatory response and release of profibrotic cytokines. A direct profibrotic role of UPR mediators in activation of TGF-ß signaling has been shown in lung fibroblasts. In addition, activation of ER stress and UPR pathways in immune cells contributes to increased production of proinflammatory cytokines. SUMMARY: Although limited in scope, current studies strongly suggest that ER stress and UPR may play an important role during development of fibrosis. Further studies are warranted to gain additional insights into the relationship between these processes.


Subject(s)
Endoplasmic Reticulum Stress/physiology , Unfolded Protein Response/physiology , Animals , Apoptosis/physiology , CCAAT-Enhancer-Binding Proteins , Disease Models, Animal , Fibroblasts/metabolism , Fibrosis/metabolism , Humans , Kidney/metabolism , Kidney Diseases/complications , Kidney Diseases/metabolism , Kidney Diseases/pathology , Lung/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology
9.
J Immunol ; 184(9): 4654-61, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20335527

ABSTRACT

The presence of the HLA-B35 allele has emerged as an important risk factor for the development of isolated pulmonary hypertension in patients with scleroderma, however the mechanisms underlying this association have not been fully elucidated. The goal of our study was to determine the molecular mechanisms that mediate the biological effects of HLA-B35 in endothelial cells (ECs). Our data demonstrate that HLA-B35 expression at physiological levels via adenoviral vector resulted in significantly increased endothelin-1 (ET-1) and a significantly decreased endothelial NO synthase (eNOS), mRNA, and protein levels. Furthermore, HLA-B35 greatly upregulated expression of chaperones, including heat shock proteins (HSPs) HSP70 (HSPA1A and HSPA1B) and HSP40 (DNAJB1 and DNAJB9), suggesting that HLA-B35 induces the endoplasmic reticulum (ER) stress and unfolded protein response in ECs. Examination of selected mediators of the unfolded protein response, including H chain binding protein (BiP; GRP78), C/Ebp homologous protein (CHOP; GADD153), endoplasmic reticulum oxidase, and protein disulfide isomerase has revealed a consistent increase of BiP expression levels. Accordingly, thapsigargin, a known ER stress inducer, stimulated ET-1 mRNA and protein levels in ECs. This study suggests that HLA-B35 could contribute to EC dysfunction via ER stress-mediated induction of ET-1 in patients with pulmonary hypertension.


Subject(s)
Down-Regulation/immunology , Endoplasmic Reticulum/immunology , Endothelin-1/biosynthesis , Endothelium, Vascular/immunology , HLA-B35 Antigen/physiology , Hypertension, Pulmonary/immunology , Hypertension, Pulmonary/metabolism , Nitric Oxide Synthase Type III/antagonists & inhibitors , Stress, Physiological/immunology , Up-Regulation/immunology , Cell Line , Cells, Cultured , Endoplasmic Reticulum/enzymology , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Chaperone BiP , Endothelium, Vascular/cytology , Endothelium, Vascular/enzymology , Endothelium, Vascular/metabolism , Humans , Hypertension, Pulmonary/enzymology , Nitric Oxide Synthase Type III/biosynthesis
10.
Stem Cells Dev ; 31(21-22): 672-683, 2022 11.
Article in English | MEDLINE | ID: mdl-36039931

ABSTRACT

Due to their immunosuppressive potential and ability to differentiate into multiple musculoskeletal cell lineages, mesenchymal stromal cells (MSCs) became popular in clinical trials for the treatment of musculoskeletal disorders. The aim of this study was to isolate and characterize native populations of MSCs from human cortical and cancellous bone from the posterior elements of the lumbar spine and determine what source of MSCs yields better quality and quantity of cells to be potentially used for spinal fusion repair. We were able to show that MSCs from trabecular and cortical spine had the typical MSC morphology and expression markers; the ability to differentiate in adipocyte, chondrocyte, or osteoblast but they did not have a consistent pattern in the expression of the specific differentiation lineage genes. Moreover, MSCs from both sites demonstrated an immune suppression profile suggesting that these cells may have a more promising success in applications related to immunomodulation more than exploring their ability to drive osteogenesis to prevent nonunion in spine fusion procedures.


Subject(s)
Mesenchymal Stem Cells , Humans , Osteogenesis , Cell Differentiation , Osteoblasts , Cell Lineage , Cells, Cultured
11.
Cells ; 11(5)2022 02 26.
Article in English | MEDLINE | ID: mdl-35269446

ABSTRACT

The mechanical homeostasis of tissues can be altered in response to trauma or disease, such as cancer, resulting in altered mechanotransduction pathways that have been shown to impact tumor development, progression, and the efficacy of therapeutic approaches. Specifically, ovarian cancer progression is parallel to an increase in tissue stiffness and fibrosis. With in vivo models proving difficult to study, tying tissue mechanics to altered cellular and molecular properties necessitate advanced, tunable, in vitro 3D models able to mimic normal and tumor mechanic features. First, we characterized normal human ovary and high-grade serous (HGSC) ovarian cancer tissue stiffness to precisely mimic their mechanical features on collagen I-based sponge scaffolds, soft (NS) and stiff (MS), respectively. We utilized three ovarian cancer cell lines (OVCAR-3, Caov-3, and SKOV3) to evaluate changes in viability, morphology, proliferation, and sensitivity to doxorubicin and liposomal doxorubicin treatment in response to a mechanically different microenvironment. High substrate stiffness promoted the proliferation of Caov-3 and SKOV3 cells without changing their morphology, and upregulated mechanosensors YAP/TAZ only in SKOV3 cells. After 7 days in culture, both OVCAR3 and SKOV3 decreased the MS scaffold storage modulus (stiffness), suggesting a link between cell proliferation and the softening of the matrix. Finally, high matrix stiffness resulted in higher OVCAR-3 and SKOV3 cell cytotoxicity in response to doxorubicin. This study demonstrates the promise of biomimetic porous scaffolds for effective inclusion of mechanical parameters in 3D cancer modeling. Furthermore, this work establishes the use of porous scaffolds for studying ovarian cancer cells response to mechanical changes in the microenvironment and as a meaningful platform from which to investigate chemoresistance and drug response.


Subject(s)
Apoptosis , Ovarian Neoplasms , Cell Line, Tumor , Doxorubicin , Extracellular Matrix/metabolism , Female , Humans , Mechanotransduction, Cellular , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Tumor Microenvironment
12.
Macromol Biosci ; 22(4): e2100450, 2022 04.
Article in English | MEDLINE | ID: mdl-35014177

ABSTRACT

Fibrosis and solid tumor progression are closely related, with both involving pathways associated with chronic wound dysregulation. Fibroblasts contribute to extracellular matrix (ECM) remodeling in these processes, a crucial step in scarring, organ failure, and tumor growth, but little is known about the biophysical evolution of remodeling regulation during the development and progression of matrix-related diseases including fibrosis and cancer. A 3D collagen-based scaffold model is employed here to mimic mechanical changes in normal (2 kPa, soft) versus advanced pathological (12 kPa, stiff) tissues. Activated fibroblasts grown on stiff scaffolds show lower migration and increased cell circularity compared to those on soft scaffolds. This is reflected in gene expression profiles, with cells cultured on stiff scaffolds showing upregulated DNA replication, DNA repair, and chromosome organization gene clusters, and a concomitant loss of ability to remodel and deposit ECM. Soft scaffolds can reproduce biophysically meaningful microenvironments to investigate early stage processes in wound healing and tumor niche formation, while stiff scaffolds can mimic advanced fibrotic and cancer stages. These results establish the need for tunable, affordable 3D scaffolds as platforms for aberrant stroma research and reveal the contribution of physiological and pathological microenvironment biomechanics to gene expression changes in the stromal compartment.


Subject(s)
Biomimetics , Neoplasms , Extracellular Matrix/metabolism , Fibroblasts , Fibrosis , Humans , Neoplasms/genetics , Neoplasms/metabolism , Phenotype , Tissue Scaffolds , Tumor Microenvironment
13.
J Tissue Eng ; 13: 20417314221138945, 2022.
Article in English | MEDLINE | ID: mdl-36451687

ABSTRACT

Osteosarcoma (OS) is the most common bone tumor in pediatrics. After resection, allografts or metal endoprostheses reconstruct bone voids, and systemic chemotherapy is used to prevent recurrence. This urges the development of novel treatment options for the regeneration of bone after excision. We utilized a previously developed biomimetic, biodegradable magnesium-doped hydroxyapatite/type I collagen composite material (MHA/Coll) to promote bone regeneration in the presence of chemotherapy. We also performed experiments to determine if human mesenchymal stem cells (hMSCs) seeded on MHA/Coll scaffold migrate less toward OS cells, suggesting that hMSCs will not contribute to tumor growth and therefore the potential of oncologic safety in vitro. Also, hMSCs seeded on MHA/Coll had increased expression of osteogenic genes (BGLAP, SPP1, ALP) compared to hMSCs in the 2D condition, even when exposed to chemotherapeutics. This is the first study to demonstrate that a highly osteogenic scaffold can potentially be oncologically safe because hMSCs on MHA/Coll tend to differentiate and lose the ability to migrate toward tumor cells. Therefore, hMSCs on MHA/Coll could potentially be utilized for bone regeneration after OS excision.

14.
Clin Transl Med ; 11(10): e551, 2021 10.
Article in English | MEDLINE | ID: mdl-34709744

ABSTRACT

BACKGROUND: Ovarian cancer (OC) is typically diagnosed late, associated with high rates of metastasis and the onset of ascites during late stage disease. Understanding the tumor microenvironment and how it impacts the efficacy of current treatments, including immunotherapies, needs effective in vivo models that are fully characterized. In particular, understanding the role of immune cells within the tumor and ascitic fluid could provide important insights into why OC fails to respond to immunotherapies. In this work, we comprehensively described the immune cell infiltrates in tumor nodules and the ascitic fluid within an optimized preclinical model of advanced ovarian cancer. METHODS: Green Fluorescent Protein (GFP)-ID8 OC cells were injected intraperitoneally into C57BL/6 mice and the development of advanced stage OC monitored. Nine weeks after tumor injection, mice were sacrificed and tumor nodules analyzed to identify specific immune infiltrates by immunohistochemistry. Ascites, developed in tumor bearing mice over a 10-week period, was characterized by mass cytometry (CyTOF) to qualitatively and quantitatively assess the distribution of the immune cell subsets, and their relationship to ascites from ovarian cancer patients. RESULTS: Tumor nodules in the peritoneal cavity proved to be enriched in T cells, antigen presenting cells and macrophages, demonstrating an active immune environment and cell-mediated immunity. Assessment of the immune landscape in the ascites showed the predominance of CD8+ , CD4+ , B- , and memory T cells, among others, and the coexistance of different immune cell types within the same tumor microenvironment. CONCLUSIONS: We performed, for the first time, a multiparametric analysis of the ascitic fluid and specifically identify immune cell populations in the peritoneal cavity of mice with advanced OC. Data obtained highlights the impact of CytOF as a diagnostic tool for this malignancy, with the opportunity to concomitantly identify novel targets, and define personalized therapeutic options.


Subject(s)
Ovarian Neoplasms/immunology , Tumor Microenvironment/immunology , Animals , Ascites/immunology , Disease Models, Animal , Female , Mice , Mice, Inbred C57BL
15.
RSC Adv ; 10(47): 28041-28048, 2020 Jul 27.
Article in English | MEDLINE | ID: mdl-35519100

ABSTRACT

The outbreak of new coronavirus disease (COVID-19) has quickly spread all over the world. Real time reverse transcriptase polymerase chain reaction (rRT-PCR) for nucleic acid detection has become the standard method for clinical diagnosis of COVID-19 infection. But these rRT-PCR tests have many inherent limitations, and carry a high false negative rate. It is an urgent to develop a method to accurately identify the vast infected patients and asymptomatic viral carriers from the population. In this article, we present the principle and procedure of developing a colloidal gold immunochromatographic assay (GICA) for rapid detection of COVID-19-specific antibodies. The detection kit can be used to detect immunoglobulin M (IgM) and IgG of COVID-19 in human blood samples within 15 minutes, and to identify different stages of viral infection. Test results can be digitalized using an office scanner and a FiJi software with appropriate confidence interval (CI) setting. Based on analysis from 375 samples, we calculated that overall sensitivity and specificity of the assay were 95.85% and 97.47%, respectively. Compared with rRT-PCR, this assay has many advantages including convenience and rapid detection. The detection kit can be widely used in hospitals, clinics and laboratories for rapid screening of both symptomatic and asymptomatic COVID-19 carriers in large scale.

16.
ACS Appl Bio Mater ; 3(10): 6737-6745, 2020 Oct 19.
Article in English | MEDLINE | ID: mdl-35019338

ABSTRACT

Ponatinib (Pon) is a multi-tyrosine kinase inhibitor that demonstrated high efficiency for treating cancer. However, severe side effects caused by Pon off-targeting effects prevent its extensive use. Using our understanding into the mechanisms by which Pon is transported by bovine serum albumin in the blood, we have successfully encapsulated Pon into a biomimetic nanoparticle (NP). This lipid NP (i.e., "leukosomes") incorporates membrane proteins purified from activated leukocytes that enable immune evasion, and enhanced targeting of inflamed endothelium NPs have been characterized for their size, charge, and encapsulation efficiency. Membrane proteins enriched on the NP surface enabled modulation of Pon release. These NP formulations showed promising dose-response results on two different murine osteosarcoma cell lines, F420 and RF379. Our results indicate that our fabrication method is reproducible, nonuser-dependent, efficient in loading Pon, and applicable toward repurposing numerous therapeutic agents previously shelved due to toxicity profiles.

17.
J Exp Clin Cancer Res ; 39(1): 40, 2020 Feb 22.
Article in English | MEDLINE | ID: mdl-32087737

ABSTRACT

BACKGROUND: Osteosarcoma (OS) is an aggressive malignant neoplasm that still suffers from poor prognosis in the case of distal metastases or occurrence of multi-drug resistance. It is therefore crucial to find novel therapeutic options able to go beyond these limitations and improve patients' survival. The objective of this study is to exploit the intrinsic properties of mesenchymal stromal cells (MSCs) to migrate and infiltrate the tumor stroma to specifically deliver therapeutic agents directly to cancer cells. In particular, we aimed to test the efficacy of the photoactivation of MSCs loaded with nanoparticles in vitro and in a murine in vivo ectopic osteosarcoma model. METHODS: AlPcS4@FNPs were produced by adding tetra-sulfonated aluminum phthalocyanine (AlPcS4) to an aqueous solution of positively charged poly-methyl methacrylate core-shell fluorescent nanoparticles (FNPs). The photodynamic therapy (PDT) effect is achieved by activation of the photosensitizer AlPcS4 in the near-infrared light with an LED source. Human MSCs were isolated from the bone marrow of five donors to account for inter-patients variability and used in this study after being evaluated for their clonogenicity, multipotency and immunophenotypic profile. MSC lines were then tested for the ability to internalize and retain the nanoparticles, along with their migratory properties in vitro. Photoactivation effect was evaluated both in a monolayer (2D) co-culture of AlPcS4@FNPs loaded MSCs with human OS cells (SaOS-2) and in tridimensional (3D) multicellular spheroids (AlPcS4@FNPs loaded MSCs with human OS cells, MG-63). Cell death was assessed by AnnexinV/PI and Live&Dead CalceinAM/EthD staining in 2D, while in the 3D co-culture, the cell killing effect was measured through ATP content, CalceinAM/EthD staining and TEM imaging. We also evaluated the effectiveness of AlPcS4@FNPs loaded MSCs as delivery systems and the ability of the photodynamic treatment to kill cancer cells in a subcutaneous mouse model of OS by bioluminescence imaging (BLI) and histology. RESULTS: MSCs internalized AlPcS4@FNPs without losing or altering their motility and viability in vitro. Photoactivation of AlPcS4@FNPs loaded MSCs induced high level of OS cells death in the 2D co-culture. Similarly, in the 3D co-culture (MSCs:OS ratios 1:1 or 1:3), a substantial decrease of both MSCs and OS cells viability was observed. Notably, when increasing the MSCs:OS ratio to 1:7, photoactivation still caused more than 40% cells death. When tested in an in vivo ectopic OS model, AlPcS4@FNPs loaded MSCs were able to decrease OS growth by 68% after two cycles of photoactivation. CONCLUSIONS: Our findings demonstrate that MSCs can deliver functional photosensitizer-decorated nanoparticles in vitro and in vivo and inhibit OS tumor growth. MSCs may be an effective platform for the targeted delivery of therapeutic nanodrugs in a clinical scenario, alone or in combination with other osteosarcoma treatment modalities.


Subject(s)
Bone Neoplasms/therapy , Indoles/administration & dosage , Mesenchymal Stem Cells/cytology , Organometallic Compounds/administration & dosage , Osteosarcoma/therapy , Photosensitizing Agents/administration & dosage , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Coculture Techniques , Humans , Indoles/pharmacology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/chemistry , Mice , Nanoparticles , Organometallic Compounds/pharmacology , Photochemotherapy , Photosensitizing Agents/pharmacology , Xenograft Model Antitumor Assays
18.
Clin Ther ; 30(5): 924-9, 2008 May.
Article in English | MEDLINE | ID: mdl-18555939

ABSTRACT

BACKGROUND: Vasculopathy is one of the hallmarks of systemic sclerosis (SSc), characterized by endothelial activation and over expression of adhesion molecules. A preliminary in vitro study has suggested that aminaftone, a naphtohydrochinone used in the treatment of capillary disorders, may downregulate the expression of adhesion molecules in endothelial cells. OBJECTIVE: This study investigated the ex vivo effects of aminaftone on soluble adhesion molecule concentrations in patients with SSc. METHODS: This randomized, open-label pilot study was conducted in patients with SSc. Patients received baseline treatment for Raynaud's phenomenon (eg, calcium channel blockers and IV cyclic iloprost) with (test) or without (control) aminaftone 75 mg or placebo TID for 12 weeks. Standard treatment for Raynaud's phenomenon was allowed as long as the dose was stable for >or=3 months prior to randomization. Concentrations of soluble E-selectin adhesion molecule 1 (sELAM-1), soluble vascular cell adhesion molecule 1 (sVCAM-1), and soluble intracellular adhesion molecule 1 (sICAM-1) were measured at baseline and 12 weeks, and their variation was tested using the analysis of variance for repeated measures with statistical correction. Laboratory analyses were performed by experienced personnel blinded to treatment assignment. RESULTS: A total of 24 patients were enrolled (21 women, 3 men; mean age, 53.4 years; aminaftone, 12 patients; control, 12 patients). Decreases in mean (SD) sELAM-1 and sVCAM-1 concentrations were significantly greater in treated patients (sELAM-1, from 17.0 [7.8] to 11.9 [9.0] pg/mL; sVCAM-1, from 51.2 [12.9] to 40.8 [13.8] ng/mL) compared with controls (sELAM-1, from 20.3 [9.9] to 20.4 [10.5] pg/mL; sVCAM-1, from 56.8 [49.6] to 62.7 [40.6] ng/mL) (both, P < 0.05 [analysis of variance or repeated measures after Bonferroni correction]). No significant changes in sICAM-1 concentrations versus controls were observed. CONCLUSIONS: In this small pilot study in this select group of patients with SSc, aminaftone was associated with downregulation of sELAM-1 and sVCAM-1 concentrations. Studies evaluating the potential role of aminaftone in the treatment of vascular sclerodermal disease and SSc are warranted.


Subject(s)
Cell Adhesion Molecules/biosynthesis , Scleroderma, Systemic/metabolism , para-Aminobenzoates , 4-Aminobenzoic Acid/administration & dosage , 4-Aminobenzoic Acid/pharmacology , E-Selectin/biosynthesis , Female , Humans , Intercellular Adhesion Molecule-1/biosynthesis , Male , Middle Aged , Pilot Projects , Prospective Studies , Solubility , Vascular Cell Adhesion Molecule-1/biosynthesis
19.
Drugs R D ; 9(4): 251-7, 2008.
Article in English | MEDLINE | ID: mdl-18588356

ABSTRACT

BACKGROUND AND OBJECTIVE: Endothelin-1 (ET-1) plays a central role in the pathogenesis of several vascular diseases. Aminaftone is a drug used for the treatment of capillary disorders but which has a mechanism of action that is not fully understood. We investigated whether aminaftone may exert its effect by interfering with the production of ET-1. METHODS: Human ECV304 endothelial cells were incubated with interleukin-1beta (IL-1beta) 100 IU/mL with or without the addition of increasing concentrations of aminaftone (2, 4 or 6 microg/mL). ET-1 concentrations in surnatants were quantified by enzyme immunoassay kit at 3, 6 and 12 hours. Pre-pro-endothelin-1 (PPET-1) gene expressions were also analysed by real-time polymerase chain reaction (RT-PCR) at the same time points. Endothelin-converting enzyme (ECE) activity was also determined. RESULTS: Incubation with IL-1beta increased concentrations of ET-1 and PPET-1 relative gene expression. Incubation with aminaftone significantly reduced production of ET-1 in a concentration-dependent manner. A strong direct correlation was found between ET-1 concentrations and PPET-1 relative gene expression, but aminaftone did not influence ECE activity. CONCLUSION: Aminaftone inhibits ET-1 production in cell cultures by interfering with transcription of the PPET-1 gene. These findings may account for the clinical efficacy of aminaftone in the treatment of capillary disorders and may encourage conduct of further clinical trials.


Subject(s)
Endothelin-1/biosynthesis , Endothelium, Vascular/metabolism , Hemostatics/pharmacology , para-Aminobenzoates , 4-Aminobenzoic Acid/pharmacology , Aspartic Acid Endopeptidases/biosynthesis , Aspartic Acid Endopeptidases/genetics , Cell Line , Down-Regulation/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelin-Converting Enzymes , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Enzyme-Linked Immunosorbent Assay , Humans , Interleukin-1beta/blood , Metalloendopeptidases/biosynthesis , Metalloendopeptidases/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
20.
Hum Immunol ; 68(3): 181-91, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17349873

ABSTRACT

Human leukocyte antigen (HLA) class I antigens can act as signal-transducing molecules that influence individual reactivity to external stimuli and the existence of haplotype-specific cell signal regulation has been suggested. In this article, we provide definite experimental evidence for the existence of a HLA-B35 haplotype-specific regulation of cell apoptosis in different experimental models. First, we demonstrated that HLA-B35, but not other HLA-class I antigens, was associated with an increased cell susceptibility to apoptosis in human peripheral mononuclear cells (PBMCs) exposed in vitro to thapsigargin. Second, we confirmed this association in human ECV 304 cells transfected with HLA-B35 or with HLA-B8, an antigen that did not appear to influence the apoptosis rate in the thapsigargin-treated PBMCs. Third, we confirmed the specific influence of HLA-B35 on cell apoptosis in non human cells (i.e., HLA-B35-transfected NIH3T3 murine fibroblasts). Our data show the existence of HLA-B35 haplotype-specific regulation of cell apoptosis and open new perspectives on the role of HLA class I genes in cell activation and disease susceptibility.


Subject(s)
Apoptosis/physiology , HLA-B35 Antigen/physiology , Leukocytes, Mononuclear/cytology , Animals , Apoptosis/drug effects , HLA-B35 Antigen/genetics , HLA-B8 Antigen/genetics , HLA-B8 Antigen/physiology , Haplotypes/genetics , Humans , Leukocytes, Mononuclear/drug effects , Mice , NIH 3T3 Cells/cytology , NIH 3T3 Cells/drug effects , Recombinant Fusion Proteins/physiology , Thapsigargin/pharmacology , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL