Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 374
Filter
1.
Molecules ; 26(19)2021 Oct 08.
Article in English | MEDLINE | ID: mdl-34641629

ABSTRACT

Psoriasis is a chronic inflammatory skin disease accompanied by excessive keratinocyte proliferation. Corticosteroids, vitamin D3 analogs, and calcineurin inhibitors, which are used to treat psoriasis, have diverse adverse effects, whereas natural products are popular due to their high efficiency and relatively low toxicity. The roots of the Cudrania tricuspidata (C. tricuspidata) are known to have diverse pharmacological effects, among which the anti-inflammatory effect is reported as a potential therapeutic agent in skin cells. Nevertheless, its effectiveness against skin diseases, especially psoriasis, is not fully elucidated. Here, we investigated the effect of cudraxanthone D (CD), extracted from the roots the C. tricuspidata Bureau, on psoriasis using an imiquimod (IMQ)-induced mouse model and the tumor necrosis factor (TNF)-α/interferon (IFN)-γ-activated keratinocytes. IMQ was topically applied to the back skin of C57BL/6 mice for seven consecutive days, and the mice were orally administered with CD. This resulted in reduced psoriatic characteristics, such as the skin thickness and Psoriasis Area Severity Index score, and the infiltration of neutrophils in IMQ-induced skin. CD inhibited the serum levels of TNF-α, immunoglobulin G2a, and myeloperoxidase, and the expression of Th1/Th17 cells in splenocytes. In TNF-α/IFN-γ-activated keratinocytes, CD reduced the expressions of CCL17, IL-1ß, IL-6, and IL-8 by inhibiting the phosphorylation of STAT1 and the nuclear translocation of NF-kB. Taken together, these results suggest that CD could be a potential drug candidate for the treatment of psoriasis.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Imiquimod/adverse effects , Keratinocytes/cytology , Moraceae/chemistry , Psoriasis/drug therapy , Xanthones/administration & dosage , Administration, Oral , Animals , Anti-Inflammatory Agents/pharmacology , Cell Line , Disease Models, Animal , Female , Humans , Interferon-gamma/adverse effects , Keratinocytes/drug effects , Keratinocytes/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , NF-kappa B/pharmacology , Plant Extracts/chemistry , Plant Roots/chemistry , Psoriasis/chemically induced , Psoriasis/immunology , Treatment Outcome , Tumor Necrosis Factor-alpha/pharmacology , Xanthones/pharmacology
2.
Sci Adv ; 7(4)2021 01.
Article in English | MEDLINE | ID: mdl-33523949

ABSTRACT

Chronic inflammatory diseases often lead to muscle wasting and contractile deficit. While exercise can have anti-inflammatory effects, the underlying mechanisms remain unclear. Here, we used an in vitro tissue-engineered model of human skeletal muscle ("myobundle") to study effects of exercise-mimetic electrical stimulation (E-stim) on interferon-γ (IFN-γ)-induced muscle weakness. Chronic IFN-γ treatment of myobundles derived from multiple donors induced myofiber atrophy and contractile loss. E-stim altered the myobundle secretome, induced myofiber hypertrophy, and attenuated the IFN-γ-induced myobundle wasting and weakness, in part by down-regulating JAK (Janus kinase)/STAT1 (signal transducer and activator of transcription 1) signaling pathway amplified by IFN-γ. JAK/STAT inhibitors fully prevented IFN-γ-induced myopathy, confirming the critical roles of STAT1 activation in proinflammatory action of IFN-γ. Our results reveal a previously unknown mechanism of the cell-autonomous anti-inflammatory effects of muscle exercise and establish the utility of human myobundle platform for studies of inflammatory muscle disease and therapy.


Subject(s)
Exercise , Interferon-gamma , Janus Kinase Inhibitors , Muscle, Skeletal , Electric Stimulation , Exercise/physiology , Humans , Interferon-gamma/adverse effects , Interferon-gamma/metabolism , Janus Kinase Inhibitors/pharmacology , Janus Kinases , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiology , Signal Transduction , Tissue Engineering/methods
3.
Thorac Cancer ; 12(6): 775-782, 2021 03.
Article in English | MEDLINE | ID: mdl-33491334

ABSTRACT

BACKGROUND: Immune checkpoint inhibitors targeting the programmed cell death-1 (PD-1)/PD-1 ligand 1 (PD-L1) axis have shown promising results in patients with nonsmall cell lung cancer (NSCLC). One major PD-L1 inducer is IFNγ, which is secreted by T cells and NK cells. Importantly, IFNγ-induced PD-L1 is one of the major mechanisms by which cancer cells escape host immunity. METHODS: Here, we found that the NSCLC cell line, LC-2/ad, has a unique character; the PD-L1 expression in these cells is up-regulated by both IFNγ and epidermal growth factor (EGF). RESULTS: Comparative analysis of the cell signaling pathway showed that IFNγ activates STAT1 signaling, while EGF activates AKT, MAPK, and ribosomal protein S6 kinase in LC-2/ad cells. IFNγ-induced PD-L1, but not EGF-induced PD-L1, was clearly blocked by the JAK-STAT inhibitor tofacitinib. Interestingly, IFNγ decreased the expression of NK cell-activating ligands while increasing the expression of MHC class I molecules, resulting in a phenotype that can easily escape from NK cells, theoretically. Finally, we showed that IFNγ stimuli attenuated NK cell-mediated cytotoxicity in LC-2/ad cells, which was, however, blocked by tofacitinib. CONCLUSIONS: Taken together, our study shows that tofacitinib blocks the IFNγ-induced transformation from an NK cell-sensitive phenotype to an NK cell-resistant one in IFNγ-reacted LC-2/ad cells, thereby implicating that tofacitinib may be a promising agent to overcome IFNγ-induced tumor immune escape, although it may be adapted to the limited number of NSCLC patients.


Subject(s)
Gene Expression Regulation, Neoplastic/drug effects , Interferon-gamma/adverse effects , Killer Cells, Natural/immunology , Peptide Fragments/adverse effects , Piperidines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/therapeutic use , Humans , Piperidines/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology
4.
Sci Rep ; 11(1): 545, 2021 01 12.
Article in English | MEDLINE | ID: mdl-33436962

ABSTRACT

Doxorubicin (DOX) chemotherapy is associated with the release of inflammatory cytokines from macrophages. This has been suggested to be, in part, due to DOX-mediated leakage of endotoxins from gut microflora, which activate Toll-like receptor 4 (TLR4) signaling in macrophages, causing severe inflammation. However, the direct function of DOX on macrophages is still unknown. In the present study, we tested the hypothesis that DOX alone is incapable of stimulating inflammatory response in macrophages. Then, we compared the anti-inflammatory effects of curcumin (CUR), resveratrol (RES) and sulforaphane (SFN) against lipopolysaccharide/interferon-gamma (LPS/IFN-γ)-mediated inflammation in the absence or presence of DOX. For this purpose, RAW 264.7 cells were stimulated with LPS/IFN-γ (10 ng/mL/10 U/mL) in the absence or presence of DOX (0.1 µM). Our results showed that DOX alone is incapable of stimulating an inflammatory response in RAW 264.7 macrophages. Furthermore, after 24 h of incubation with LPS/IFN-γ, a significant increase in tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and inducible nitric oxide synthase (iNOS) mRNA levels was observed. Similarly, nitric oxide (NO) production and TNF-α and IL-6 protein levels were significantly upregulated. Moreover, in LPS/IFN-γ-treated macrophages, the microRNAs (miRNAs) miR-146a, miR-155, and miR-21 were significantly overexpressed. Interestingly, upon testing CUR, RES, and SFN against LPS/IFN-γ-mediated inflammation, only SFN was able to significantly reverse the LPS/IFN-γ-mediated induction of iNOS, TNF-α and IL-6 and attenuate miR-146a and miR-155 levels. In conclusion, SFN, at the transcriptional and posttranscriptional levels, exhibits potent immunomodulatory action against LPS/IFN-γ-stimulated macrophages, which may indicate SFN as a potential treatment for DOX-associated inflammation.


Subject(s)
Antibiotics, Antineoplastic/adverse effects , Curcumin/pharmacology , Doxorubicin/adverse effects , Immunologic Factors , Inflammation Mediators/metabolism , Isothiocyanates/pharmacology , Macrophages/immunology , Macrophages/metabolism , Resveratrol/pharmacology , Sulfoxides/pharmacology , Animals , Inflammation/chemically induced , Inflammation/drug therapy , Interferon-gamma/adverse effects , Interleukin-6/metabolism , Isothiocyanates/therapeutic use , Lipopolysaccharides/adverse effects , Mice , Molecular Targeted Therapy , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , RAW 264.7 Cells , RNA, Messenger/metabolism , Sulfoxides/therapeutic use , Tumor Necrosis Factor-alpha/metabolism
5.
J Cutan Pathol ; 48(5): 689-693, 2021 May.
Article in English | MEDLINE | ID: mdl-33442885

ABSTRACT

In mycosis fungoides (MF), cutaneous granuloma formation is unusual. Furthermore, MF showing interstitial granuloma, a rare type, after combination therapy with interferon-gamma (IFN-γ) and narrowband UVB (nbUVB) has not been previously reported. A 77-year-old man was referred to our hospital with a 2-month history of erythroderma. Biopsied specimens revealed infiltration of atypical lymphocytes and eosinophils. A diagnosis of an erythrodermic variant of MF was made. He was treated with combination therapy of IFN-γ and nbUVB. After the therapy, papules newly appeared and a histopathological specimen revealed interstitial granuloma. There were several CXCR3-positive cells around the granuloma. We speculated that the combination therapy made T-helper 1 cells migrate to the cutaneous lesion and resulted in the granuloma formation. Furthermore, judging from the disappearance of elastic fibers around the interstitial granuloma, we considered that IFN-γ may induce the infiltration of histiocytes interstitially after damage of elastic fibers caused by nbUVB therapy, and both IFN-γ and nbUVB may thus play an important role in the histogenesis. Not only histopathology but also immunological observations are needed to elucidate the mechanisms underlying the development of different types of granuloma in MF.


Subject(s)
Granuloma/immunology , Granuloma/pathology , Interferon-gamma/adverse effects , Mycosis Fungoides/pathology , Ultraviolet Therapy/adverse effects , Adult , Aged , Antiviral Agents/adverse effects , Antiviral Agents/therapeutic use , Biopsy/methods , Combined Modality Therapy/methods , Dermatitis, Exfoliative/diagnosis , Dermatitis, Exfoliative/pathology , Female , Granuloma/chemically induced , Granuloma/diagnosis , Histiocytes/drug effects , Histiocytes/pathology , Humans , Immunohistochemistry/methods , Interferon-gamma/therapeutic use , Male , Middle Aged , Mycosis Fungoides/diagnosis , Mycosis Fungoides/drug therapy , Mycosis Fungoides/radiotherapy , Receptors, CXCR3/metabolism , Skin Neoplasms/pathology , Th1 Cells/drug effects , Th1 Cells/pathology , Th1 Cells/radiation effects , Ultraviolet Therapy/methods
6.
mBio ; 11(5)2020 09 10.
Article in English | MEDLINE | ID: mdl-32913009

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the cause of coronavirus disease 2019 (COVID-19), is a recently emerged respiratory coronavirus that has infected >23 million people worldwide with >800,000 deaths. Few COVID-19 therapeutics are available, and the basis for severe infections is poorly understood. Here, we investigated properties of type I (ß), II (γ), and III (λ1) interferons (IFNs), potent immune cytokines that are normally produced during infection and that upregulate IFN-stimulated gene (ISG) effectors to limit virus replication. IFNs are already in clinical trials to treat COVID-19. However, recent studies highlight the potential for IFNs to enhance expression of host angiotensin-converting enzyme 2 (ACE2), suggesting that IFN therapy or natural coinfections could exacerbate COVID-19 by upregulating this critical virus entry receptor. Using a cell line model, we found that beta interferon (IFN-ß) strongly upregulated expression of canonical antiviral ISGs, as well as ACE2 at the mRNA and cell surface protein levels. Strikingly, IFN-λ1 upregulated antiviral ISGs, but ACE2 mRNA was only marginally elevated and did not lead to detectably increased ACE2 protein at the cell surface. IFN-γ induced the weakest ISG response but clearly enhanced surface expression of ACE2. Importantly, all IFN types inhibited SARS-CoV-2 replication in a dose-dependent manner, and IFN-ß and IFN-λ1 exhibited potent antiviral activity in primary human bronchial epithelial cells. Our data imply that type-specific mechanisms or kinetics shape IFN-enhanced ACE2 transcript and cell surface levels but that the antiviral action of IFNs against SARS-CoV-2 counterbalances any proviral effects of ACE2 induction. These insights should aid in evaluating the benefits of specific IFNs, particularly IFN-λ, as repurposed therapeutics.IMPORTANCE Repurposing existing, clinically approved, antiviral drugs as COVID-19 therapeutics is a rapid way to help combat the SARS-CoV-2 pandemic. Interferons (IFNs) usually form part of the body's natural innate immune defenses against viruses, and they have been used with partial success to treat previous new viral threats, such as HIV, hepatitis C virus, and Ebola virus. Nevertheless, IFNs can have undesirable side effects, and recent reports indicate that IFNs upregulate the expression of host ACE2 (a critical entry receptor for SARS-CoV-2), raising the possibility that IFN treatments could exacerbate COVID-19. Here, we studied the antiviral- and ACE2-inducing properties of different IFN types in both a human lung cell line model and primary human bronchial epithelial cells. We observed differences between IFNs with respect to their induction of antiviral genes and abilities to enhance the cell surface expression of ACE2. Nevertheless, all the IFNs limited SARS-CoV-2 replication, suggesting that their antiviral actions can counterbalance increased ACE2.


Subject(s)
Antiviral Agents/pharmacology , Coronavirus Infections/drug therapy , Interferon Type I/pharmacology , Interferon-gamma/pharmacology , Interferons/pharmacology , Peptidyl-Dipeptidase A/metabolism , Pneumonia, Viral/drug therapy , Aged , Angiotensin-Converting Enzyme 2 , Animals , Betacoronavirus/immunology , COVID-19 , Cell Line , Chlorocebus aethiops , Female , Humans , Immunotherapy/methods , Interferon Type I/adverse effects , Interferon-gamma/adverse effects , Interferons/adverse effects , Pandemics , Peptidyl-Dipeptidase A/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Virus/metabolism , Respiratory Mucosa/cytology , Respiratory Mucosa/virology , SARS-CoV-2 , Up-Regulation/drug effects , Vero Cells , Virus Replication/drug effects , Interferon Lambda
7.
Nutrients ; 12(5)2020 Apr 27.
Article in English | MEDLINE | ID: mdl-32349358

ABSTRACT

Pyropia yezoensis, a red alga, is popular and harvested a lot in East Asia and is famous for its medicinal properties attributable to its bioactive compounds including amino acids (porphyra-334 and shinorine, etc.), polysaccharides, phytosterols, and pigments, but its anti-inflammatory effect and mechanism of anti-atopic dermatitis (AD) have not been elucidated. In this study, we investigate the anti-AD effect of P. yezoensis extract (PYE) on mRNA and protein levels of the pro-inflammatory chemokines, thymus, and activation-regulated chemokine (TARC/CCL17) and macrophage-derived chemokine (MDC/CCL22), in human HaCaT keratinocyte cells treated to interferon (IFN)-γ or tumor necrosis factor (TNF)-α (10 ng/mL each). The effect of the PYE on extracellular signal-regulated kinase (ERK) and other mitogen-activated protein kinases (MAPKs) was related to its suppression of TARC and MDC production by blocking NF-κB activation in HaCaT cells. Furthermore, astaxanthin and xanthophyll from P. yezoensis were identified as anti-AD candidate compounds. These results suggest that the PYE may improve AD and contained two carotenoids by regulating pro-inflammatory chemokines.


Subject(s)
Chemokine CCL17/metabolism , Chemokine CCL22/metabolism , Down-Regulation/drug effects , Inflammation Mediators/metabolism , Interferon-gamma/adverse effects , NF-kappa B/metabolism , Plant Extracts/pharmacology , Porphyra/chemistry , Tumor Necrosis Factor-alpha/adverse effects , Anti-Inflammatory Agents , Dermatitis, Atopic/drug therapy , Extracellular Signal-Regulated MAP Kinases/metabolism , HaCaT Cells , Humans , Phytotherapy , Plant Extracts/isolation & purification , Plant Extracts/therapeutic use , Xanthophylls/isolation & purification , Xanthophylls/therapeutic use , p38 Mitogen-Activated Protein Kinases/metabolism
8.
J Immunother Cancer ; 8(1)2020 04.
Article in English | MEDLINE | ID: mdl-32269142

ABSTRACT

BACKGROUND: Adoptive cellular therapy (ACT) is a promising treatment for synovial sarcoma (SS) with reported response rates of over 50%. However, more work is needed to obtain deeper and more durable responses. SS has a 'cold' tumor immune microenvironment with low levels of major histocompatibility complex (MHC) expression and few T-cell infiltrates, which could represent a barrier toward successful treatment with ACT. We previously demonstrated that both MHC expression and T-cell infiltration can be increased using systemic interferon gamma (IFN-γ), which could improve the efficacy of ACT for SS. CASE PRESENTATION: We launched a phase I trial incorporating four weekly doses of IFN-γ in an ACT regimen of high-dose cyclophosphamide (HD Cy), NY-ESO-1-specific T cells, and postinfusion low-dose interleukin (IL)-2. Two patients were treated. While one patient had significant tumor regression and resultant clinical benefit, the other patient suffered a fatal histiocytic myocarditis. Therefore, this cohort was terminated for safety concerns. CONCLUSION: We describe a new and serious toxicity of immunotherapy from IFN-γ combined with HD Cy-based lymphodepletion and low-dose IL-2. While IFN-γ should not be used concurrently with HD Cy or with low dose IL-2, IFN-γ may still be important in sensitizing SS for ACT. Future studies should avoid using IFN-γ during the immediate period before/after cell infusion. TRIAL REGISTRATION NUMBERS: NCT04177021, NCT01957709, and NCT03063632.


Subject(s)
Cyclophosphamide/adverse effects , Histiocytes/pathology , Immunotherapy, Adoptive/methods , Interferon-gamma/adverse effects , Lymphocyte Depletion/adverse effects , Myocarditis/pathology , Sarcoma, Synovial/therapy , Adult , Antineoplastic Agents, Alkylating/adverse effects , Antiviral Agents/adverse effects , Clinical Trials, Phase I as Topic , Drug Therapy, Combination , Histiocytes/drug effects , Humans , Male , Myocarditis/chemically induced , Prognosis , Sarcoma, Synovial/immunology , Sarcoma, Synovial/pathology
9.
J Aerosol Med Pulm Drug Deliv ; 33(2): 108-115, 2020 04.
Article in English | MEDLINE | ID: mdl-31855492

ABSTRACT

Background: In volunteers with idiopathic pulmonary fibrosis (IPF), inhaled Interferon-γ (IFN-γ) is safe and may improve pulmonary function. However, coughing, associated with upper airway deposition, is often reported. To address this problem, a small-particle, breath-enhanced jet nebulizer (i-NEB Mini; InspiRx, Inc., Somerset, NJ) was developed. Using gamma scintigraphy, this device was tested in healthy individuals and subjects with IPF to determine efficiency and regional deposition in lung and airways. Methods: Four healthy individuals and nine subjects with IPF were enrolled. The nebulizer was filled with 2 mL of saline with 99m Tc bound to diethylenetriaminepentaacetic acid (DTPA) powered continuously with 3.4 L/min of compressed air. Mass median aerodynamic diameter (MMAD) was measured by cascade impactor. To maximize deposition in alveoli, inspiratory flow was limited by an inspiratory resistance incorporated into the nebulizer, resulting in a deep inspiration ∼6 seconds. The treatment was run to completion (10 minutes), and each subject underwent deposition imaging. Mass balance and regions of interest determined upper airway (measured by calibrated stomach activity) and regional lung deposition as a percent of pretreatment nebulizer charge. Results: Subjects tolerated the device with no complaints. MMAD (mean [geometric standard deviation]) = 1.04 [1.92] µm. Lung deposition (mean ± standard error, % nebulizer charge) in healthy subjects was 26.2% ± 1.83 and in IPF individuals 23.4% ± 1.60 (p = 0.414). Upper airway deposition was 1.4% ± 0.83 and 2.3% ± 0.48, respectively (p = 0.351), and 20.1% was lost during expiration. Central/Peripheral ratios were consistent in both groups, showing high peripheral deposition (1.32 ± 0.050, vs. 1.28 ± 0.046, p = 0.912). Conclusion: The i-NEB Mini jet nebulizer with breath enhancement produced small particles, resulting in minimal upper airway deposition. Using slow and deep breathing, more than half of the emitted dose deposited in the peripheral lung in normal subjects and individuals with IPF. These data indicate that, for future clinical trials, controlled lung doses of small particles, designed to avoid coughing, are possible even in subjects with advanced disease.


Subject(s)
Idiopathic Pulmonary Fibrosis/drug therapy , Interferon-gamma/administration & dosage , Lung/metabolism , Nebulizers and Vaporizers , Administration, Inhalation , Case-Control Studies , Cough/etiology , Equipment Design , Humans , Interferon-gamma/adverse effects , Interferon-gamma/pharmacokinetics , Particle Size , Tissue Distribution
10.
J Transl Med ; 17(1): 147, 2019 05 09.
Article in English | MEDLINE | ID: mdl-31072323

ABSTRACT

BACKGROUND: Interferon gamma (IFNγ) plays an important role in the development of chronic lung diseases via the production of inflammatory mediators, although the exact mechanism remains unclear. The present study aimed at investigating the potential mechanisms by which IFNγ induced over-production of interleukins through the interaction between carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) and phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K) pathway. METHODS: IFN-γ induced over-production of interleukin (IL) 6 and IL8, and RNA expression of CEACAM1 and its subtypes or PI3K and its subtypes in human bronchial epithelial cells (HBE). The production of IL6 and IL8 or cell proliferation and movement were also evaluated in cellCEACAM1- or cellCEACAM1+ after the induction of IFN-γ. Roles of PI3K subtype proteins, e.g. PI3Kp110α/δ, Akt, p110α/γ/δ/ß/mTOR, PI3Kp110α/δ/ß, PI3Kp110δ, or pan-PI3K in IFN-γ-induced CEACAM1 subtype alterations were furthermore validated using those proteins of PI3K subtypes. RESULTS: CEACAM1, especially CEACAM1-S isoforms, was significantly up-regulated in HBE cells after treatment with IFN-γ. CEACAM1 played roles in expression of IL-6 and IL-8, and facilitated cellular proliferation and migration. IFN-γ up-regulated the expression of CEACAM1 in airway epithelial cells, especially CEACAM1-S isoforms, promoting cellular proliferation, migration, and the production of inflammatory factors. PI3K (p110δ)/Akt/mTOR pathway was involved in the process of IFN-γ-upregulated CEACAM1, especially CEACAM1-S. On the other hand, CEACAM1 could promote the activation of PI3K/Akt/mTOR pathway. CONCLUSION: IFN-γ could induce inflammatory responses, cellular growth and proliferation through the interaction of CEACAM1 (especially CEACAM1-S isoforms) and PI3K(p110δ)/Akt/mTOR in airway epithelial cells, which might be new alternative of future therapies against epithelial transition from inflammation to cancer.


Subject(s)
Antigens, CD/metabolism , Bronchi/pathology , Cell Adhesion Molecules/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Inflammation/metabolism , Interferon-gamma/adverse effects , Phosphatidylinositol 3-Kinases/metabolism , Antigens, CD/genetics , Cell Adhesion Molecules/genetics , Cell Line , Epithelial Cells/drug effects , Humans , Inflammation/pathology , Models, Biological , Phosphatidylinositol 3-Kinases/genetics , Protein Binding/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects
11.
J Bone Miner Res ; 34(8): 1436-1445, 2019 08.
Article in English | MEDLINE | ID: mdl-30889272

ABSTRACT

In autosomal dominant osteopetrosis type 2 (ADO2) CLCN7 mutations cause impaired osteoclast function. Severe consequences include skeletal fragility despite high bone mass, osteomyelitis, osteonecrosis, bone marrow failure, and severe cranial nerve impingement. There is no effective medical treatment for ADO2. We recruited subjects with ADO2 into a 14-week, open-label, pilot clinical trial of interferon gamma-1b. Doses were titrated based on tolerability and if fasting serum C-telopeptide (CTX) was <25% above baseline at week 8, targeting doses of 100 µg/m2 three times a week. The primary outcomes were change from baseline in CTX and N-telopeptide/creatinine ratio (NTX/Cr) at week 14. Secondary outcomes included changes in urine calcium/creatinine ratio, bone formation markers and tolerability. Nine adults and three children were recruited. Severe manifestations of ADO2 included histories of fractures (100%), osteomyelitis (16.7%), vision loss (50%), and anemia (58.3%). Baseline CTX and NTX/Cr were generally low-normal. Procollagen type I N-terminal propeptide was elevated or in the upper-normal range in 11 of 12 (91.6%) subjects. Elevations of aspartate aminotransferase (AST) and lactate dehydrogenase (LDH) were common. One subject withdrew due to rash. Five subjects achieved doses of 50 µg/m2 3 days a week, while six reached the full dose of 100 µg/m2 3 days a week. Only 3 of 11 (27.3%) completing subjects achieved the primary outcome of increasing CTX ≥25% above baseline at week 14. The mean ± SD change from baseline in CTX at week 14 was +2.2% ± 43.2%, p = 0.86). Likewise, there was no significant change in NTX/Cr (mean change -2.1%, p = 0.81). Interferon gamma-1b was poorly tolerated. Most subjects had adverse events, and the Mental Health and Mental Component Scales of the SF-36v2 health survey declined slightly (p < 0.05). Over 14 weeks, interferon gamma-1b failed to significantly increase bone turnover markers in ADO2 and was poorly tolerated. Consequently, interferon gamma-1b is unlikely to be effective for decreasing bone mass in ADO2. © 2019 American Society for Bone and Mineral Research.


Subject(s)
Bone Resorption , Collagen Type I/blood , Creatinine/blood , Interferon-gamma/administration & dosage , Osteopetrosis , Peptides/blood , Adolescent , Adult , Aged , Biomarkers/blood , Bone Resorption/blood , Bone Resorption/drug therapy , Bone Resorption/pathology , Child , Child, Preschool , Female , Humans , Interferon-gamma/adverse effects , Male , Middle Aged , Osteopetrosis/blood , Osteopetrosis/drug therapy , Osteopetrosis/pathology , Recombinant Proteins/administration & dosage , Recombinant Proteins/adverse effects
12.
Gene ; 700: 38-46, 2019 Jun 05.
Article in English | MEDLINE | ID: mdl-30898705

ABSTRACT

Anti-inflammation is a key process to restore tissue integrity and function. CXCL12 is a homeostasis chemokine, which plays a coordinating role in organogenesis, tumorigenesis and regeneration. In the present study we found that the uterus of abortion mice showed different histo-morphological changes with the development of abortion. The expression of chemokine CXCL12 and its receptor CXCR4 in abortion uterus showed a time-dependent pattern. Compared with normal pregnancy, the expression of CXCL12 and CXCR4 did not change in the uterus of GD7 abortion mice, but increased significantly in the uterus of GD8 and GD10 abortion mice. However, the expression of IFN-γ increased significantly in the uterus of GD7 abortion mice, while there was no significant change detected in GD8 aborted mice uterus. Our further data show that the expression of CXCL12 is not regulated by IFN-γ in endometrial stromal cell culture system in vitro. The treatment of CXCL12 significantly inhibits the expression of IFN-γ in in vitro cultured stromal cells and splenic monocytes. This suggests that CXCL12 may play an anti-inflammatory role in the uterus of abortion mice to promote the process of endometrial restoration after abortion, rather than participate in the process of abortion as a response molecule of IFN-γ.


Subject(s)
Abortion, Induced/veterinary , Abortion, Veterinary/metabolism , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , Interferon-gamma/adverse effects , Up-Regulation , Abortion, Veterinary/chemically induced , Abortion, Veterinary/genetics , Animals , Cells, Cultured , Female , Mice , Pregnancy , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Stromal Cells/cytology , Stromal Cells/drug effects , Stromal Cells/metabolism , Time Factors , Uterus/cytology , Uterus/drug effects , Uterus/metabolism
13.
Sci Rep ; 8(1): 12518, 2018 08 21.
Article in English | MEDLINE | ID: mdl-30131581

ABSTRACT

Alopecia areata (AA) is a chronic, relapsing hair-loss disorder that is considered to be a T-cell-mediated autoimmune disease. Several animal models for AA have been created to investigate the pathophysiology and screen for effective therapeutic targets. As C3H/HeJ mice develop AA spontaneously in a low frequency, a novel animal model is needed to establish an AA-like condition faster and more conveniently. In this study, we present a novel non-invasive AA rodent model that avoids skin or lymph-node cell transfer. We simply injected C3H/HeJ mice subcutaneously with interferon-gamma (IFNγ) along with polyinosinic:polycytidylic acid (poly[I:C]), a synthetic dsRNA, to initiate innate immunity via inflammasome activation. Approximately 80% of the IFNγ and poly(I:C) co-injected mice showed patchy AA lesions after 8 weeks. None of the mice displayed hair loss in the IFNγ or poly(I:C) solely injection group. Immunohistochemical staining of the AA lesions revealed increased infiltration of CD4+ and CD8+ cells infiltration around the hair follicles. IFNγ and poly(I:C) increased the expression of NLRP3, IL-1ß, CXCL9, CXCL10, and CXCL11 in mouse skin. Taken together, these findings indicate a shorter and more convenient means of AA animal model induction and demonstrate that inflammasome-activated innate immunity is important in AA pathogenesis.


Subject(s)
Alopecia Areata/immunology , Disease Models, Animal , Interferon-gamma/adverse effects , Poly I-C/adverse effects , Alopecia Areata/chemically induced , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cells, Cultured , Chemokines, CXC/metabolism , Drug Synergism , Female , Humans , Injections, Subcutaneous , Interferon-gamma/administration & dosage , Interleukin-1beta/metabolism , Mice , Mice, Inbred C3H , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Poly I-C/administration & dosage
14.
An. bras. dermatol ; 91(5,supl.1): 76-78, Sept.-Oct. 2016. tab, graf
Article in English | LILACS | ID: biblio-837916

ABSTRACT

Abstract Cutaneous reactions associated with interferons (IFNs) treatment are either localized or generalized. The most common presentation of localized reactions at IFNs injection site is usually an erythematous patch or plaque. Local leukocytoclastic vasculitis presenting with cutaneous necrosis is extremely rare. We report a 19-year-old man with hepatitis B who had local leukocytoclastic vasculitis induced by interferon-gama injection at the injection site. After changing the injection sites and using the combined treatment of prednisone and colchicine, the previous lesion healed and no other cutaneous lesion occurred. We also made a mini review of such cases.


Subject(s)
Humans , Male , Young Adult , Skin/pathology , Interferon-gamma/adverse effects , Vasculitis, Leukocytoclastic, Cutaneous/chemically induced , Skin/drug effects , Prednisone/therapeutic use , Colchicine/therapeutic use , Treatment Outcome , Vasculitis, Leukocytoclastic, Cutaneous/pathology , Vasculitis, Leukocytoclastic, Cutaneous/drug therapy , Erythema/chemically induced , Erythema/pathology , Injections, Subcutaneous/adverse effects , Anti-Inflammatory Agents/therapeutic use , Necrosis/chemically induced , Necrosis/pathology
16.
An Bras Dermatol ; 91(5 suppl 1): 76-78, 2016.
Article in English | MEDLINE | ID: mdl-28300901

ABSTRACT

Cutaneous reactions associated with interferons (IFNs) treatment are either localized or generalized. The most common presentation of localized reactions at IFNs injection site is usually an erythematous patch or plaque. Local leukocytoclastic vasculitis presenting with cutaneous necrosis is extremely rare. We report a 19-year-old man with hepatitis B who had local leukocytoclastic vasculitis induced by interferon-gama injection at the injection site. After changing the injection sites and using the combined treatment of prednisone and colchicine, the previous lesion healed and no other cutaneous lesion occurred. We also made a mini review of such cases.


Subject(s)
Interferon-gamma/adverse effects , Skin/pathology , Vasculitis, Leukocytoclastic, Cutaneous/chemically induced , Anti-Inflammatory Agents/therapeutic use , Colchicine/therapeutic use , Erythema/chemically induced , Erythema/pathology , Humans , Injections, Subcutaneous/adverse effects , Male , Necrosis/chemically induced , Necrosis/pathology , Prednisone/therapeutic use , Skin/drug effects , Treatment Outcome , Vasculitis, Leukocytoclastic, Cutaneous/drug therapy , Vasculitis, Leukocytoclastic, Cutaneous/pathology , Young Adult
17.
Neurol Sci ; 37(3): 361-4, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26621361

ABSTRACT

Friedreich's ataxia is an autosomal recessive progressive degenerative disorder caused by deficiency of the protein frataxin. The most common genetic cause is a homozygotic expansion of GAA triplets within intron 1 of the frataxin gene leading to impaired transcription. Preclinical in vivo and in vitro studies have shown that interferon gamma (IFNγ) is able to up-regulate the expression of frataxin gene in multiple cell types. We designed a phase IIa clinical trial, the first in Italy, aimed at assessing both safety and tolerability of IFNγ in Friedreich's patients and ability to increase frataxin levels in peripheral blood mononuclear cells. Nine patients (6 female and 3 males aged 21-38 years) with genetically confirmed disease were given 3 subcutaneous escalating doses (100, 150 and 200 µg) of IFNγ (human recombinant interferon 1 b gamma, trade name IMUKIN(®)), over 4 weeks. The primary end-point was the assessment of the safety and tolerability of IFNγ by means of standard clinical and hematological criteria. The secondary end-point was the detection of changes of frataxin levels in peripheral blood mononuclear cells after each single escalating dose of the drug. IFNγ was generally well tolerated, the main adverse event was hyperthermia/fever. Although, increases in frataxin levels could be detected in a minority of patients, these changes were not significant. A large phase III multicenter, randomized clinical trial with IFNγ in Friedreich's ataxia patients is currently ongoing. This study is expected to conclusively address the clinical efficacy of IFNγ therapy in patients with Friedreich's ataxia.


Subject(s)
Friedreich Ataxia/drug therapy , Interferon-gamma/therapeutic use , Neuroprotective Agents/therapeutic use , Adult , Blood Chemical Analysis , Drug Administration Schedule , Female , Friedreich Ataxia/blood , Humans , Interferon-gamma/adverse effects , Iron-Binding Proteins/blood , Italy , Male , Neuroprotective Agents/adverse effects , Recombinant Proteins/adverse effects , Recombinant Proteins/therapeutic use , Treatment Outcome , Young Adult , Frataxin
18.
Neurodegener Dis Manag ; 5(6): 497-504, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26634868

ABSTRACT

IFN-γ-1b is currently US FDA approved as an orphan drug for the treatment of chronic granulomatous disease and severe malignant osteopetrosis. It is administered via subcutaneous injection and is a potential therapy for Friedreich ataxia (FRDA), a rare degenerative neurological condition. Ongoing Phase II and III trials in both adults and children with FRDA were preceded by a small Phase I, open-label trial in children that showed that IFN-γ-1b was reasonably well-tolerated and improved overall neurological function as measured by the Friedreich Ataxia Rating Scale after 12 weeks of treatment, though the primary outcome measure of frataxin level showed no improvement. Although there is an established dose of IFN-γ-1b prescribed for the current indications, the efficacy and tolerability of these dose levels in the FRDA population remains the subject of ongoing investigation.


Subject(s)
Friedreich Ataxia/drug therapy , Interferon-gamma/therapeutic use , Neuroprotective Agents/therapeutic use , Animals , Clinical Trials as Topic , Humans , Interferon-gamma/adverse effects , Interferon-gamma/pharmacokinetics , Interferon-gamma/pharmacology , Neuroprotective Agents/adverse effects , Neuroprotective Agents/pharmacokinetics , Neuroprotective Agents/pharmacology , Recombinant Proteins/adverse effects , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use
19.
Dermatol Clin ; 33(4): 731-45, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26433845

ABSTRACT

Interferons are polypeptides that naturally occur in the human body as a part of the innate immune response. By harnessing these immunomodulatory functions, synthetic interferons have shown efficacy in combating various diseases including cutaneous T-cell lymphoma. This article closely examines the qualities of interferon alfa and interferon gamma and the evidence behind their use in the 2 most common types of cutaneous T-cell lymphomas, namely, mycosis fungoides and Sézary syndrome.


Subject(s)
Interferon-alpha/therapeutic use , Interferon-gamma/therapeutic use , Mycosis Fungoides/drug therapy , Skin Neoplasms/drug therapy , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Combined Modality Therapy/methods , Humans , Interferon-alpha/adverse effects , Interferon-alpha/pharmacology , Interferon-beta/therapeutic use , Interferon-gamma/adverse effects , Interferon-gamma/pharmacology , Mycosis Fungoides/therapy , PUVA Therapy/methods , Retinoids/therapeutic use , Skin Neoplasms/therapy
20.
Diabetes ; 64(12): 4285-97, 2015 12.
Article in English | MEDLINE | ID: mdl-25858560

ABSTRACT

Activation of AMPK suppresses inflammation, but the underlying mechanisms remain poorly understood. This study was designed to characterize the molecular mechanisms by which AMPK suppresses vascular inflammation. In cultured human aortic smooth muscle cells, pharmacologic or genetic activation of AMPK inhibited the signal transducer and activator of transcription-1 (STAT1), while inhibition of AMPK had opposite effects. Deletion of AMPKα1 or AMPKα2 resulted in activation of STAT1 and in increases in proinflammatory mediators, both of which were attenuated by administration of STAT1 small interfering RNA or fludarabine, a selective STAT1 inhibitor. Moreover, AMPK activation attenuated the proinflammatory actions induced by STAT1 activators such as interferon-γ and angiotensin II (AngII). Mechanistically, we found that AMPK activation increased, whereas AMPK inhibition decreased, the levels of mitogen-activated protein kinase phosphatase-1 (MKP-1), an inducible nuclear phosphatase, by regulating proteasome-dependent degradation of MKP-1. Gene silencing of MKP-1 increased STAT1 phosphorylation and prevented 5-aminoimidazole-4-carboxyamide ribonucleoside-reduced STAT1 phosphorylation. Finally, we found that infusion of AngII caused a more severe inflammatory response in AMPKα2 knockout mouse aortas, all of which were suppressed by chronic administration of fludarabine. We conclude that AMPK activation suppresses STAT1 signaling and inhibits vascular inflammation through the upregulation of MKP-1.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Dual Specificity Phosphatase 1/metabolism , Muscle, Smooth, Vascular/metabolism , STAT1 Transcription Factor/metabolism , Vasculitis/metabolism , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/chemistry , AMP-Activated Protein Kinases/genetics , Angiotensin II/adverse effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Aorta, Thoracic , Cells, Cultured , Dual Specificity Phosphatase 1/antagonists & inhibitors , Dual Specificity Phosphatase 1/chemistry , Dual Specificity Phosphatase 1/genetics , Enzyme Activation/drug effects , Humans , Interferon-gamma/adverse effects , MAP Kinase Signaling System/drug effects , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/immunology , Muscle, Smooth, Vascular/pathology , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , RNA Interference , Random Allocation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , STAT1 Transcription Factor/agonists , STAT1 Transcription Factor/antagonists & inhibitors , STAT1 Transcription Factor/genetics , Vasculitis/chemically induced , Vasculitis/immunology , Vasculitis/pathology
SELECTION OF CITATIONS
SEARCH DETAIL