Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters

Complementary Medicines
Country/Region as subject
Publication year range
1.
Inflamm Res ; 70(7): 753-764, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34212215

ABSTRACT

Janus kinase/signal transduction and transcriptional activator (JAK/STAT) signaling pathway is a transport hub for cytokine secretion and exerts its effects. The activation of JAK/STAT signaling pathway is essential for the regulation of inflammatory responses. Inappropriate activation or deletion of JAK/STAT signaling pathway is the initiator of the inflammatory response. JAK/STAT signaling pathway has been demonstrated to be involved in the process of innate and adaptive immune response to inflammatory bowel disease (IBD). In this review, we discuss the role of the JAK/STAT signaling pathway in the regulation of different cells in IBD, as well as new findings on the involvement of the JAK/STAT signaling pathway in the regulation of the intestinal immune response. The current status of JAK inhibitors in the treatment of IBD is summarized as well. This review highlights natural remedies that can serve as potential JAK inhibitors. These phytochemicals may be useful in the identification of precursor compounds in the process of designing and developing novel JAK inhibitors.


Subject(s)
Inflammatory Bowel Diseases/drug therapy , Janus Kinase Inhibitors/therapeutic use , Janus Kinases/metabolism , STAT Transcription Factors/metabolism , Animals , Biological Products/therapeutic use , Humans , Inflammatory Bowel Diseases/metabolism , Janus Kinase Inhibitors/pharmacology , Janus Kinases/antagonists & inhibitors , STAT Transcription Factors/antagonists & inhibitors , Signal Transduction/drug effects
2.
Int Immunopharmacol ; 97: 107720, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33945918

ABSTRACT

Presently, postmenopausal osteoporosis mainly caused by excessive activation of in vivo osteoclasts has become a global public health burden. Natural compounds have gradually become the potential drugs for the treatment of postmenopausal osteoporosis. Aloperine is a new alkaloid extracted from the leaves and seeds of sophora bean. The current studies have proved that aloperine has many biological activities, including anti-inflammatory, antiviral and anticancer activities. This study shows that aloperine can inhibit activity and formation of osteoclast mediated by RANKL in a dose-dependent manner without affecting the activity of bone marrow macrophages (BMM). In addition, it is found that aloperine can inhibit the expression of osteoclast specific marker genes, including nuclear factor of activated T cells cytoplasmic 1 (NFATc1), tartrate resistant acid phosphatase (TRAcP), matrix metallopeptidase 9 (MMP9), cathepsin K (Ctsk), V-ATPase d2 and calcitonin receptor. The in vitro experiment of aloperine proved that aloperine can inhibit the degradation of IκBα and the phosphorylation of P65, ERK and JNK. Additionally, aloperine improves bone loss in ovariectomized (OVX) mice by inhibiting osteoclast activity. This project proved that aloperine can affect the formation of osteoclasts by inhibiting RANKL signaling channel, and it is indicated that aloperine has the potential to be developed as a new drug for the prevention and treatment of postmenopausal osteoporosis.


Subject(s)
Osteogenesis/drug effects , Osteoporosis, Postmenopausal/drug therapy , Quinolizidines/pharmacology , Animals , Disease Models, Animal , Drug Evaluation, Preclinical , Female , Femur/drug effects , Femur/pathology , Humans , Janus Kinases/antagonists & inhibitors , Janus Kinases/metabolism , MAP Kinase Signaling System/drug effects , Mice , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Osteoclasts/drug effects , Osteoporosis, Postmenopausal/diagnosis , Osteoporosis, Postmenopausal/etiology , Osteoporosis, Postmenopausal/pathology , Ovariectomy , Quinolizidines/therapeutic use , RANK Ligand/antagonists & inhibitors , RANK Ligand/metabolism , X-Ray Microtomography
3.
Sci Adv ; 7(1)2021 01.
Article in English | MEDLINE | ID: mdl-33187978

ABSTRACT

Using AI, we identified baricitinib as having antiviral and anticytokine efficacy. We now show a 71% (95% CI 0.15 to 0.58) mortality benefit in 83 patients with moderate-severe SARS-CoV-2 pneumonia with few drug-induced adverse events, including a large elderly cohort (median age, 81 years). An additional 48 cases with mild-moderate pneumonia recovered uneventfully. Using organotypic 3D cultures of primary human liver cells, we demonstrate that interferon-α2 increases ACE2 expression and SARS-CoV-2 infectivity in parenchymal cells by greater than fivefold. RNA-seq reveals gene response signatures associated with platelet activation, fully inhibited by baricitinib. Using viral load quantifications and superresolution microscopy, we found that baricitinib exerts activity rapidly through the inhibition of host proteins (numb-associated kinases), uniquely among antivirals. This reveals mechanistic actions of a Janus kinase-1/2 inhibitor targeting viral entry, replication, and the cytokine storm and is associated with beneficial outcomes including in severely ill elderly patients, data that incentivize further randomized controlled trials.


Subject(s)
Antiviral Agents/pharmacology , Azetidines/pharmacology , COVID-19/mortality , Enzyme Inhibitors/pharmacology , Janus Kinases/antagonists & inhibitors , Liver/virology , Purines/pharmacology , Pyrazoles/pharmacology , SARS-CoV-2/pathogenicity , Sulfonamides/pharmacology , Adult , Aged , Aged, 80 and over , COVID-19/metabolism , COVID-19/virology , Cytokine Release Syndrome , Cytokines/metabolism , Drug Evaluation, Preclinical , Female , Gene Expression Profiling , Humans , Interferon alpha-2/metabolism , Italy , Janus Kinases/metabolism , Liver/drug effects , Male , Middle Aged , Patient Safety , Platelet Activation , Proportional Hazards Models , RNA-Seq , Spain , Virus Internalization/drug effects , COVID-19 Drug Treatment
4.
J Mol Model ; 26(9): 235, 2020 Aug 17.
Article in English | MEDLINE | ID: mdl-32808204

ABSTRACT

Rheumatoid arthritis is an autoimmune disorder causing joint deformity and work disability. Several drugs are available to deal with the disease including conventional drugs; biological drugs such as TNFα inhibitors, B cell-targeted drugs, T cell co-stimulation inhibitors, interleukin-6 inhibitors, and interleukin-1 inhibitors; and kinase inhibitory drugs. In spite of the broad spectrum of drugs available, the disease remains uncontrolled in a number of patients and there is a need for new drugs with better efficacy and universal response rate. The failure of the available drugs to control the disease can be owed to the complex pathogenesis with complementary pathways of disease progression. The blockade of one pathway cannot supersede pathogenesis through other complementary pathways. Janus kinase (JAK) and Bruton's tyrosine kinase (BTK) are the two important mediators of disease which control a number of signaling pathways involved in rheumatoid arthritis pathogenesis. In this study, using the computer-aided drug designing techniques (virtual screening, molecular docking, and molecular dynamics studies), we have designed piperidinyl dipyrrolopyridine-based dual inhibitors of Janus kinase and Bruton's tyrosine kinase. Dual JAK and BTK inhibitors seem promising to fight the complex pathogenesis of the disease at multiple fronts and can be the future drug for patients unresponsive to current remedies.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/chemistry , Janus Kinase Inhibitors/chemistry , Janus Kinases/chemistry , Protein Kinase Inhibitors/chemistry , Pyridines/chemistry , Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Arthritis, Rheumatoid/drug therapy , Binding Sites , Drug Design , Drug Development , Humans , Janus Kinase Inhibitors/pharmacology , Janus Kinases/antagonists & inhibitors , Molecular Docking Simulation , Molecular Dynamics Simulation , Molecular Structure , Protein Binding , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Structure-Activity Relationship
5.
Invest Ophthalmol Vis Sci ; 61(3): 8, 2020 03 09.
Article in English | MEDLINE | ID: mdl-32150250

ABSTRACT

Purpose: Thymic stromal lymphopoietin (TSLP) is a pro-allergic cytokine that initiates allergic inflammatory reaction between epithelial and dendritic cells (DCs). miR-19b was reported to suppress TSLP expression. The present study aimed to examine miR-19b expression, regulation, and function in allergic conjunctivitis (AC). Methods: A murine model of experimental AC was induced in BALB/c mice by short ragweed pollen. The serum, eye balls, conjunctiva, and cervical lymph nodes (CLN) were used for the study. Gene expression was determined by RT-PCR, whereas protein production and activation were evaluated by immunostaining, ELISA, and Western blotting. Results: In the murine AC model, miR-19b was aberrantly downregulated, whereas the levels of TSLP and p-STAT3, as well as the number of CD11c+ pSTAT3+ DCs were increased. Moreover, Th2 inflammatory cytokine expression was significantly increased. These severe phenotypes could be counteracted by either applying exogenous miR-19b mimic microRNAs or the JAK/STAT inhibitor CYT387. Moreover, overexpression of miR-19b repressed p-STAT3 expression and the number of CD11c+ cells in AC eye and CLN tissues. Conclusions: These findings suggested that miR-19b reduced ocular surface inflammation by inhibiting Stat3 signaling via TSLP downregulation in a murine AC model. Moreover, the present study further demonstrated the clinical potential of applying miR-19b and anti-JAK/STAT therapies in the treatment of AC.


Subject(s)
Conjunctivitis, Allergic/genetics , Janus Kinases/physiology , MicroRNAs/genetics , STAT Transcription Factors/physiology , Animals , Antigens, Plant , CD11 Antigens/metabolism , Cervical Vertebrae , Conjunctiva/metabolism , Conjunctivitis, Allergic/immunology , Conjunctivitis, Allergic/metabolism , Cornea/metabolism , Cytokines/biosynthesis , Disease Models, Animal , Down-Regulation , Female , Janus Kinases/antagonists & inhibitors , Lymph Nodes/metabolism , Mice, Inbred BALB C , MicroRNAs/biosynthesis , Phenotype , Plant Extracts , STAT Transcription Factors/antagonists & inhibitors , STAT3 Transcription Factor/metabolism , Signal Transduction , Thymic Stromal Lymphopoietin
6.
Lancet Gastroenterol Hepatol ; 5(9): 850-861, 2020 09.
Article in English | MEDLINE | ID: mdl-32171056

ABSTRACT

The incidence and prevalence of ulcerative colitis are increasing globally. Although the exact cause and pathogenesis of this disease is unclear, research has led to a better understanding of the condition and to identification of new targets for therapy, which in turn has encouraged the development of new therapies. As well as biologic therapies, which have changed the way inflammatory bowel disease is managed, small molecules have been developed for the treatment of ulcerative colitis. These small molecule treatments are orally administered and are likely to bring a substantial shift in the way this chronic disease is treated. Oral therapies offer many advantages over infusion therapies, such as ease of use, increased acceptability by patients, and reduction of cost. This Review focuses not only on oral therapies that have been approved for use in ulcerative colitis, but also on those that are in development, providing a comprehensive overview for clinicians of available oral therapies and drugs that are likely to become available. We have also reviewed drugs that have shown promise in preclinical studies and could be effective future therapies.


Subject(s)
Colitis, Ulcerative/drug therapy , Inflammatory Bowel Diseases/drug therapy , Molecular Targeted Therapy/methods , Administration, Oral , Aged , Chronic Disease/drug therapy , Clinical Trials as Topic , Colitis, Ulcerative/epidemiology , Cyclic Nucleotide Phosphodiesterases, Type 4/drug effects , Drug Evaluation, Preclinical/methods , Humans , Immunomodulation , Incidence , Janus Kinase Inhibitors/pharmacology , Janus Kinases/antagonists & inhibitors , Janus Kinases/drug effects , Middle Aged , Phosphodiesterase 4 Inhibitors/pharmacology , Prevalence , Sphingosine-1-Phosphate Receptors/agonists , Sphingosine-1-Phosphate Receptors/drug effects
7.
J Surg Res ; 248: 98-108, 2020 04.
Article in English | MEDLINE | ID: mdl-31877436

ABSTRACT

BACKGROUND: This study aimed to investigate the role of Janus kinase/signal transducers and activators of transcription (JAK/STAT) signaling pathway in protection by peritoneal resuscitation (PR) using pyruvate-peritoneal dialysis solution (PY-PDS) against intestinal injury from hemorrhagic shock (HS) in rats. MATERIALS AND METHODS: Sixty-four rats were assigned to eight groups: group SHAM; group intravenous resuscitation (VR); groups NS, LA, and PY in which the rats were subjected to HS and PR with normal saline (NS), lactate-peritoneal dialysis solution (LA-PDS), and PY-PDS, respectively, combined with VR; and groups DMSO, RPM, and AG490 in which the rats were subjected to HS and VR with pretreatment of dimethyl sulfoxide (DMSO), rapamycin (RPM), and tyrphostin B42 (AG490). RESULTS: At 2 h after HS and resuscitation, the levels of diamine oxidase, 15-F2t-isoprostane, thromboxane B2, and endothelin-1, in the blood and the intestinal mucosal apoptotic index and caspase-3 were lower in groups PY, RPM, and AG490 than in groups VR, NS, LA, and DMSO. Group PY showed lower levels of malondialdehyde and myeloperoxidase and a higher level of superoxide dismutase than groups VR, NS, and LA. Phosphorylated JAK2 and phosphorylated STAT3 levels were lower in groups PY, RPM, AG490, and LA than in groups VR, NS, and DMSO. CONCLUSIONS: The protection mechanism of PR with PY-PDS combined with VR was related to the inhibition of the JAK/STAT signaling pathway during HS and resuscitation. The process might include suppression of oxidative stress, reduction of neutrophil infiltration, regulation of microcirculation, and inhibition of apoptosis.


Subject(s)
Intestinal Diseases/prevention & control , Pyruvic Acid/therapeutic use , Resuscitation/methods , Shock, Hemorrhagic/therapy , Animals , Dialysis Solutions , Drug Evaluation, Preclinical , Intestinal Diseases/etiology , Janus Kinases/antagonists & inhibitors , Janus Kinases/metabolism , Male , Pyruvic Acid/pharmacology , Rats , Rats, Sprague-Dawley , STAT Transcription Factors/antagonists & inhibitors , STAT Transcription Factors/metabolism , Shock, Hemorrhagic/complications , Signal Transduction/drug effects
8.
J Med Chem ; 62(8): 3898-3923, 2019 04 25.
Article in English | MEDLINE | ID: mdl-30901208

ABSTRACT

Concurrent inhibition of Janus kinase (JAK) and histone deacetylase (HDAC) could potentially improve the efficacy of the HDAC inhibitors in the treatment of cancers and resolve the problem of HDAC inhibitor resistance in some tumors. Here, a novel series of pyrimidin-2-amino-pyrazol hydroxamate derivatives as JAK and HDAC dual inhibitors was designed, synthesized, and evaluated, among which 8m possessed potent and balanced activities against both JAK2 and HDAC6 with half-maximal inhibitory concentration at the nanomolar level. 8m exhibited improved antiproliferative and proapoptotic activities over SAHA and ruxolitinib in several hematological cell lines. Remarkably, 8m exhibited more potent antiproliferation effect than the combination of SAHA and ruxolitinib in HEL cells bearing JAK2V617F mutation. Pharmacokinetic studies in mice showed that 8m possessed good bioavailability after intraperitoneal administration. Finally, 8m showed antitumor efficacy with no significant toxicity in a HEL xenograft model. Collectively, the results confirm the therapeutic potential of JAK and HDAC dual inhibitors in hematological malignancies and provide valuable leads for further structural optimization and antitumor mechanism study.


Subject(s)
Histone Deacetylase Inhibitors/chemistry , Histone Deacetylases/chemistry , Janus Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Animals , Binding Sites , Catalytic Domain , Drug Evaluation, Preclinical , Half-Life , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/pathology , Histone Deacetylase Inhibitors/metabolism , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Histone Deacetylases/metabolism , Humans , Janus Kinases/metabolism , Male , Mice , Mice, Nude , Molecular Docking Simulation , Nitriles , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Pyrazoles/chemistry , Pyrazoles/metabolism , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Pyrimidines , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Xenograft Model Antitumor Assays
9.
J Drugs Dermatol ; 18(3): s115-s116, 2019 Mar 01.
Article in English | MEDLINE | ID: mdl-30909355

ABSTRACT

Hypopigmentation and depigmentation of the skin can be due to multiple causes and has a broad differential diagnosis. The most common cause of depigmentation worldwide is vitiligo. This disorder affects 1-2% of the world's population and is seen in all races. Vitiligo is an autoimmune disorder in which the predominant cause is an attack by CD8+ cytotoxic T cells on melanocytes in the epidermis. This condition can have a significant negative impact on the quality of life of affected individuals. Treatment options currently include psychological counseling, topical therapy, systemic therapy, phototherapy, surgical therapy, and depigmentation. In patients with stable, refractory disease, successful repigmentation has been achieved using mini-punch grafting, blister grafting, and non-cultured epidermal suspension (NCES) grafting. Emerging therapies include the Janus kinase (JAK) inhibitors ruxolitinib and tofacitinib. Further studies exploring the pathogenesis of vitiligo are warranted in order to optimize treatment for affected patients. J Drugs Dermatol. 2019;18(3 Suppl):s115-116.


Subject(s)
Autoimmune Diseases/therapy , Quality of Life , Vitiligo/therapy , Administration, Cutaneous , Administration, Oral , Autoimmune Diseases/diagnosis , Autoimmune Diseases/immunology , Autoimmune Diseases/psychology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Counseling/methods , Dermatologic Agents/therapeutic use , Diagnosis, Differential , Epidermis/transplantation , Humans , Janus Kinases/antagonists & inhibitors , Janus Kinases/immunology , Melanocytes/transplantation , Mycosis Fungoides/diagnosis , Nitriles , Phototherapy/methods , Piperidines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use , Pyrroles/therapeutic use , Skin Pigmentation/drug effects , Skin Pigmentation/immunology , Tinea Versicolor/diagnosis , Vitiligo/diagnosis , Vitiligo/immunology , Vitiligo/psychology
10.
Bioorg Med Chem ; 27(12): 2592-2597, 2019 06 15.
Article in English | MEDLINE | ID: mdl-30926315

ABSTRACT

The Janus kinase (JAK) family of tyrosine kinases has been proven to provide targeted immune modulation. Orally available JAK inhibitors have been used for the treatment of immune-mediated inflammatory diseases, such as rheumatoid arthritis (RA). Here, we report the design, synthesis and biological evaluation of 4-(4,5,6,7-tetrahydrofuro[3,2-c]pyridin-2-yl) pyrimidin-2-amino derivatives as JAK inhibitors. Systematic structure-activity relationship studies led to the discovery of compound 7j, which strongly inhibited the four isoforms of JAK kinases. Molecular modeling rationalized the importance of cyanoacetyl and phenylmorpholine moieties. The in vivo investigation indicated that compound 7j possessed favorable pharmacokinetic properties and displayed slightly better anti-inflammatory efficacy than tofacitinib at the same dosage. Accordingly, compound 7j was advanced into preclinical development.


Subject(s)
Amines/chemistry , Anti-Inflammatory Agents/chemistry , Janus Kinase Inhibitors/chemistry , Janus Kinases/antagonists & inhibitors , Administration, Oral , Amines/metabolism , Animals , Anti-Inflammatory Agents/metabolism , Anti-Inflammatory Agents/pharmacokinetics , Anti-Inflammatory Agents/therapeutic use , Arthritis/drug therapy , Arthritis/pathology , Binding Sites , Disease Models, Animal , Drug Evaluation, Preclinical , Half-Life , Humans , Janus Kinase Inhibitors/metabolism , Janus Kinase Inhibitors/pharmacokinetics , Janus Kinase Inhibitors/therapeutic use , Janus Kinases/metabolism , Molecular Docking Simulation , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/metabolism , Pyrimidines/chemistry , Rats , Structure-Activity Relationship
11.
Ann Dermatol Venereol ; 146(12S3): 12S76-12S84, 2019 Dec.
Article in French | MEDLINE | ID: mdl-31997736

ABSTRACT

The treatment of atopic dermatitis is based on the use of topical steroids and emollients. When AD is resistant to a well-conducted topical treatment, phototherapy or systemic therapeutics can be used: ciclosporin, methotrexate, azathioprine or mycophenolate mofetil. However, the therapeutic landscape of AD is changing dramatically because of the approval of dupilumab (an anti-IL4/IL13 biologic therapy) and the possible future arrival of other biologicals (anti-IL13, anti-IL31…), and JAK inhibitors. © 2019 Elsevier Masson SAS. All rights reserved.


Subject(s)
Dermatitis, Atopic/drug therapy , Dermatologic Agents/therapeutic use , Adrenal Cortex Hormones/therapeutic use , Adult , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/therapeutic use , Biological Products/adverse effects , Biological Products/therapeutic use , Clinical Trials as Topic , Dermatitis, Atopic/therapy , Dermatologic Agents/administration & dosage , Dermatologic Agents/adverse effects , Histamine H1 Antagonists/therapeutic use , Humans , Immunosuppressive Agents/adverse effects , Immunosuppressive Agents/therapeutic use , Janus Kinases/antagonists & inhibitors , Multicenter Studies as Topic , Phosphodiesterase 4 Inhibitors/therapeutic use , Phototherapy , Protein Kinase Inhibitors/therapeutic use , Randomized Controlled Trials as Topic
12.
Bull Exp Biol Med ; 165(5): 665-668, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30225700

ABSTRACT

JAK/STAT signaling pathway was examined comparatively during realization of growth potential of mesenchymal progenitor cells stimulated with diterpene alkaloid songorine or fibroblast growth factor. The stimulating role of JAKs and STAT3 on the mitotic activity and differentiation of progenitor cells cultured with songorine was revealed. Under these conditions, the study demonstrated suppression of fibroblast colony formation against the background of reduced number of actively proliferating CFU-fibroblasts and a drop of differentiation index of progenitor cells induced by pan-JAKs and STAT3 inhibitors. The observed changes were in almost complete agreement with the character of functional reactions of the progenitor elements in response to blockade of JAKs and STAT3 with fibroblast growth factor. In addition, blockade of JAKs with this factor enhanced the differentiation rate of the progenitor cells.


Subject(s)
Aconitum/chemistry , Alkaloids/pharmacology , Janus Kinases/genetics , Mesenchymal Stem Cells/drug effects , STAT3 Transcription Factor/genetics , Alkaloids/isolation & purification , Animals , Anthraquinones/pharmacology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Fibroblast Growth Factors/pharmacology , Gene Expression Regulation , Janus Kinases/antagonists & inhibitors , Janus Kinases/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Nitriles , Phosphorylation/drug effects , Plant Extracts/chemistry , Primary Cell Culture , Pyrazoles/pharmacology , Pyrimidines , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/metabolism , Signal Transduction , Sulfonamides/pharmacology
13.
Dermatol Online J ; 24(7)2018 Jul 15.
Article in Spanish | MEDLINE | ID: mdl-30261562

ABSTRACT

La alopecia areata constituye un reto terapéutico, sobre todo en sus formas extensas. Antes de iniciar cualquier tratamiento es necesario tener en cuenta algunas consideraciones. Se trata de una enfermedad que no afecta de forma directa a la salud del paciente y que puede presentar resolución espontánea. Las formas extensas, las que se inician en la infancia y las de larga evolución son muy rebeldes a los tratamientos y asocian recaídas. Todos los tratamientos tienen efectos secundarios. Ningún tratamiento ha demostrado alterar el curso de la enfermedad, muy pocos han demostrado eficacia en ensayos clínicos aleatorizados y no existen guías terapéuticas salvo la publicada en 2003 y actualizada en 2012 en el British Journal of Dermatology. Por todo ello, es necesario elaborar un plan de tratamiento individualizado en cada paciente. Se debe comenzar con los fármacos más seguros e inocuos, y pasar al siguiente escalón terapéutico cuando el actual haya demostrado su ineficacia durante un periodo de 6 meses. Se revisan las principales propuestas farmacológicas para alopecia areata, aportando datos sobre su mecanismo de acción, efectos secundarios y posicionamiento terapéutico en función de los estudios disponibles. Finalmente, se propone un algoritmo terapéutico como guía en el manejo de esta patología.


Subject(s)
Adrenal Cortex Hormones/therapeutic use , Alopecia Areata/drug therapy , Biological Products/therapeutic use , Immunologic Factors/therapeutic use , Adrenal Cortex Hormones/administration & dosage , Alopecia Areata/therapy , Humans , Janus Kinases/antagonists & inhibitors , Laser Therapy , Minoxidil/therapeutic use , Phototherapy , Platelet-Rich Plasma , Prostaglandins/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Vasodilator Agents/therapeutic use
14.
Int J Oncol ; 53(4): 1681-1690, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30066853

ABSTRACT

Adult T­cell leukemia/lymphoma (ATLL) constitutes an aggressive malignancy caused by human T­cell leukemia virus type 1 (HTLV­1) that is resistant to available chemotherapeutics. The constitutive activation of Janus kinase (JAK)-signal transducer and activator of transcription (STAT) signaling is an important feature of ATLL, and spleen tyrosine kinase (SYK) is overexpressed in HTLV­1-transformed T­cell lines. In this study, we evaluated the effects of SYK- (PRT060318) or JAK- (JAK inhibitor 1) selective inhibitors and the dual SYK/JAK inhibitor, cerdulatinib, on the viability of HTLV­1-transformed and ATLL-derived T­cell lines. Cell proliferation, viability, cell cycle, apoptosis and intracellular signaling cascades were analyzed by the water-soluble tetrazolium-8 assay, flow cytometry and western blot analysis. HTLV­1-infected T­cell lines were sensitive to both SYK-selective and pan-JAK inhibitors, whereas cerdulatinib more potently suppressed cell proliferation and reduced cell viability than either of these agents alone. By contrast, the cytotoxic effects of cerdulatinib on uninfected T­cell lines and peripheral blood mononuclear cells from a healthy donor were less pronounced. Cerdulatinib induced cell cycle arrest in the G2/M phase, which was associated with a decreased cyclin-dependent kinase 1 and cyclin B1, and an increased p21 and p27 expression. Hoechst staining revealed chromatin condensation and nuclear fragmentation in the cells treated with cerdulatinib, and an increased fraction of apoptotic APO2.7-stained cells was detected by flow cytometry. This corresponded to the activation of caspase-8, -9 and -3, and decreased levels of the anti-apoptotic factors, Bcl-xL, survivin, X-linked inhibitor of apoptosis (XIAP) and c­FLIP. The cerdulatinib-induced decrease in cell viability was partly reversed by the caspase inhibitor, z­VAD­FMK. These anti-ATLL effects were associated with the suppression of SYK and JAK/STAT signaling, along with that of the downstream factors, AKT, ERK, activator protein­1 and nuclear factor-κB. Finally, oral dosing with cerdulatinib lowered the tumor burden in a murine model of ATLL. Thus, our findings indicate that the simultaneous inhibition of therapeutically relevant targets, such as SYK and JAK is a more effective approach than single-agent therapy for the treatment of ATLL.


Subject(s)
Janus Kinases/antagonists & inhibitors , Leukemia-Lymphoma, Adult T-Cell/drug therapy , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacokinetics , Sulfones/pharmacokinetics , Syk Kinase/antagonists & inhibitors , Animals , Benzimidazoles/pharmacology , Benzimidazoles/therapeutic use , Cell Line , Cell Survival/drug effects , Cyclohexylamines/pharmacology , Cyclohexylamines/therapeutic use , Drug Evaluation, Preclinical , Female , Human T-lymphotropic virus 1/pathogenicity , Humans , Leukemia-Lymphoma, Adult T-Cell/virology , Mice, Inbred ICR , Mice, SCID , Protein Kinase Inhibitors/therapeutic use , Pyridones/pharmacology , Pyridones/therapeutic use , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Signal Transduction/drug effects , Sulfones/pharmacology , Sulfones/therapeutic use , T-Lymphocytes , Treatment Outcome , Xenograft Model Antitumor Assays
15.
Molecules ; 23(6)2018 Jun 19.
Article in English | MEDLINE | ID: mdl-29921758

ABSTRACT

Aberrant activations of the STAT3 (signal transducer and activator of transcription 3) signaling pathway are associated with cancer and inflammatory diseases. Three of the four Janus kinases, JAK1, JAK2, and Tyk2, are the major upstream kinases of STAT3 in responses to cytokine stimulations. Among them, JAK2 is the key kinase in the IL-6-induced STAT3 phosphorylation. Here we report the mechanisms of a natural compound parthenolide from the medicinal herb Feverfew in regulating the JAK/STAT3 signaling. We found that parthenolide was a potent inhibitor of JAKs. It covalently modified the Cys178, Cys243, Cys335, and Cys480 of JAK2 and suppressed its kinase activity. It also interacted with other JAKs in a similar fashion. The binding of parthenolide to JAKs was selective. It preferentially bound to the JAKs, but not to the abundant proteins, such as tubulin and actin. Parthenolide also induced reactive oxygen species (ROS), but the increased ROS did not seem to contribute to the inhibition of JAK/STAT3 signaling. Furthermore, parthenolide inhibited the IL-6-induced cancer cell migration and preferentially inhibited the growth of cancer cells that had constitutively activated STAT3. Our study suggests a novel strategy to inactivate JAKs and provides a promising anti-inflammation and anticancer drug candidate.


Subject(s)
Janus Kinases/antagonists & inhibitors , STAT3 Transcription Factor/metabolism , Sesquiterpenes/pharmacology , Signal Transduction/drug effects , HEK293 Cells , HeLa Cells , Hep G2 Cells , Humans , Interleukin-6/metabolism , Janus Kinases/metabolism , Reactive Oxygen Species/metabolism
16.
PLoS One ; 13(6): e0198611, 2018.
Article in English | MEDLINE | ID: mdl-29897957

ABSTRACT

BACKGROUND: Recent studies have highlighted the JAK/STAT signaling pathway in the regulation of muscle satellite cell behavior. Herein we report preclinical studies designed to characterize the effects of a novel JAK/STAT inhibitor on plantar flexor skeletal muscle function, morphology, and satellite cell content. METHODS: The compound, SGI-1252, was administered orally (400mg/kg) in a 10% dextrose solution to wild type mice (n = 6) 3 times per week for 8 weeks. A control group (n = 6) received only the dextrose solution. RESULTS: SGI-1252 was well tolerated, as animals displayed similar weight gain over the 8-week treatment period. Following treatment, fatigue in the gastrocnemius-soleus-plantaris complex was greater in the SGI-1252 mice during a 300 second tetanic contraction bout (p = 0.035), though both the rate of fatigue and maximal force production were similar. SGI-1252 treated mice had increased type II myofiber cross-sectional area (1434.8 ± 225.4 vs 1754.7 ± 138.5 µm2), along with an increase in wet muscle mass (125.45 ± 5.46 vs 139.6 ± 12.34 mg, p = 0.032) of the gastrocnemius relative to vehicle treated mice. SGI-1252 treatment reduced gastrocnemius STAT3 phosphorylation 53% (94.79 ± 45.9 vs 44.5 ± 6.1 MFI) and significantly increased the concentration of Pax7+ satellite cells (2589.2 ± 105.5 vs 2859.4 ± 177.5 SC/mm3) in the gastrocnemius. SGI-1252 treatment suppressed MyoD (p = 0.013) and Myogenin (p<0.0001) expression in human primary myoblasts, resulting in reduced myogenic differentiation (p = 0.039). CONCLUSIONS: Orally delivered SGI-1252 was well tolerated, attenuates skeletal muscle STAT3 activity, and increases satellite cell content in mouse gastrocnemius muscle, likely by inhibiting myogenic progression.


Subject(s)
Diamines/pharmacology , Janus Kinases/metabolism , Muscle, Skeletal/drug effects , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , STAT Transcription Factors/metabolism , Administration, Oral , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Drug Administration Schedule , Drug Evaluation, Preclinical , Humans , Janus Kinases/antagonists & inhibitors , Mice , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , MyoD Protein/metabolism , Myoblasts/cytology , Myoblasts/drug effects , Myoblasts/metabolism , Myogenin/metabolism , PAX7 Transcription Factor/metabolism , Phosphorylation/drug effects , STAT Transcription Factors/antagonists & inhibitors , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/drug effects , Satellite Cells, Skeletal Muscle/metabolism
17.
Hautarzt ; 68(11): 876-884, 2017 Nov.
Article in German | MEDLINE | ID: mdl-29030646

ABSTRACT

Systemic drug treatment of vitiligo is currently limited to predominantly adjuvant measures for increasing the effectiveness of UV light therapy. We here present new approaches for the systemic treatment of vitiligo currently under clinical investigation. These include the α­MSH-analogue afamelatonide and oral immunosuppressants such as the Janus kinase (JAK) inhibitors which target interferon-α-dependent autotoxic inflammatory reactions. In 2015 the first publications on the successful systemic use of Janus kinase (JAK) inhibitors in vitiligo appeared. The effectiveness was experimentally supported by animal models of vitiligo and by the characterization of new biomarkers in the serum of vitiligo patients. This may significantly expand the range of treatment options for vitiligo. Topical antiinflammatory and UV therapies are still the main components of vitiligo treatment, often in combination. The main outcome parameters include the extent and duration of repigmentation, cessation of spreading, avoidance of side effects and improvement in the quality of life of patients.


Subject(s)
Immunosuppressive Agents/therapeutic use , Janus Kinases/antagonists & inhibitors , Piperidines/therapeutic use , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use , Pyrroles/therapeutic use , Ultraviolet Therapy , Vitiligo/diagnosis , Vitiligo/therapy , alpha-MSH/analogs & derivatives , Animals , Biomarkers/blood , Combined Modality Therapy , Disease Models, Animal , Humans , Immunosuppressive Agents/adverse effects , Interferon-alpha/antagonists & inhibitors , Nitriles , Piperidines/adverse effects , Pyrazoles/adverse effects , Pyrimidines/adverse effects , Pyrroles/adverse effects , Ultraviolet Therapy/adverse effects , Vitiligo/physiopathology , alpha-MSH/adverse effects , alpha-MSH/therapeutic use
18.
J Ethnopharmacol ; 204: 107-117, 2017 May 23.
Article in English | MEDLINE | ID: mdl-28438564

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Artemisia argyi is a herbal medicine traditionally used in Asia for the treatment of bronchitis, dermatitis and arthritis. Recent studies revealed the anti-inflammatory effect of essential oil in this plant. However, the mechanisms underlying the therapeutic potential have not been well elucidated. The present study is aimed to verify its anti-inflammatory effect and investigate the probable mechanisms. MATERIALS AND METHODS: The essential oil from Artemisia argyi (AAEO) was initially tested against LPS-induced production of inflammatory mediators and cytokines in RAW264.7 macrophages. Protein and mRNA expressions of iNOS and COX-2 were determined by Western blotting and RT-PCR analysis, respectively. The effects on the activation of MAPK/NF-κB/AP-1 and JAK/STATs pathway were also investigated by western blot. Meanwhile, in vivo anti-inflammatory effect was examined by histologic and immunohistochemical analysis in TPA-induced mouse ear edema model. RESULTS: The results of in vitro experiments showed that AAEO dose-dependently suppressed the release of pro-inflammatory mediators (NO, PGE2 and ROS) and cytokines (TNF-α, IL-6, IFN-ß and MCP-1) in LPS-induced RAW264.7 macrophages. It down-regulated iNOS and COX-2 protein and mRNA expression but did not affect the activity of these two enzymes. AAEO significantly inhibited the phosphorylation of JAK2 and STAT1/3, but not the activation of MAPK and NF-κB cascades. In animal model, oral administration of AAEO significantly attenuated TPA-induced mouse ear edema and decreased the protein level of COX-2. CONCLUSION: AAEO suppresses inflammatory responses via down-regulation of the JAK/STATs signaling and ROS scavenging, which could contribute, at least in part, to the anti-inflammatory effect of AAEO.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Artemisia , Janus Kinase Inhibitors/pharmacology , Oils, Volatile/pharmacology , Animals , Anti-Inflammatory Agents/therapeutic use , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Cytokines/metabolism , Dinoprostone/metabolism , Edema/chemically induced , Edema/drug therapy , Edema/pathology , Janus Kinase Inhibitors/therapeutic use , Janus Kinases/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Oils, Volatile/therapeutic use , RAW 264.7 Cells , Reactive Oxygen Species/metabolism , STAT Transcription Factors/antagonists & inhibitors , Tetradecanoylphorbol Acetate
19.
Retrovirology ; 13(1): 88, 2016 Dec 20.
Article in English | MEDLINE | ID: mdl-27998278

ABSTRACT

BACKGROUND: Despite the durable viral suppression afforded by antiretroviral therapy, HIV-1 eradication will require strategies to target latently infected cells that persist in infected individuals. Protein kinase C (PKC) activation is a promising strategy to reactivate latent proviruses and allow for subsequent recognition and clearance of infected cells by the immune system. Ingenol derivatives are PKC agonists that induce latency reversal but also lead to T cell activation and the release of pro-inflammatory cytokines, which would be undesirable in vivo. In this work, we sought to identify compounds that would suppress pro-inflammatory cytokine production in the context of PKC activation. DESIGN AND METHODS: We performed an in vitro screen to identify compounds that could dampen pro-inflammatory cytokine release associated with T cell activation, using IL-6 as a model cytokine. We then tested the ability of the most promising screening hit, the FDA-approved Janus Kinase (JAK) inhibitor ruxolitinib, to diminish release of multiple cytokines and its effect on latency reversal using cells from HIV-1-positive, aviremic participants. RESULTS: We demonstrate that co-administration of ruxolitinib with ingenol-3,20-dibenzoate significantly reduces pro-inflammatory cytokine release without impairing latency reversal ex vivo. CONCLUSION: The combination of ingenol compounds and JAK inhibition represents a novel strategy for HIV-1 eradication.


Subject(s)
CD4-Positive T-Lymphocytes/virology , Cytokines/metabolism , Diterpenes/pharmacology , HIV-1/physiology , Janus Kinases/antagonists & inhibitors , Pyrazoles/pharmacology , Virus Latency , Anti-HIV Agents/therapeutic use , CD4-Positive T-Lymphocytes/drug effects , Drug Evaluation, Preclinical , HIV Infections/drug therapy , HIV-1/drug effects , High-Throughput Screening Assays , Humans , Interleukin-6/analysis , Lymphocyte Activation , Nitriles , Protein Kinase C/metabolism , Pyrimidines , Virus Activation
20.
Semin Cutan Med Surg ; 35(5 Suppl): S92-6, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27525671

ABSTRACT

The newer and emerging treatments for atopic dermatitis (AD) focus on blockade of inflammatory cytokines, especially those that derive from T helper cell type 2 (TH2) and are associated with a pathway of immunoglobulin E (IgE) sensitization. Among the proinflammatory cytokines that have been identified as promising therapeutic targets are chemoattractant receptor-homologous molecule expressed on TH2 cells (CRTH2), IgE, thymic stromal lymphopoietin (TSLP), and several monoclonal antibodies that block key cytokine pathways in the innate immune response. Two agents that have been studied in phase III clinical trials are the boronbased phosphodiesterase-4 (PDE-4) inhibitor, crisaborole, and dupilumab, an antibody that inhibits the interleukin-4/ IL-13 receptor α chain. Semin Cutan Med Surg 35(supp5):S92-S96.


Subject(s)
Dermatitis, Atopic/drug therapy , Child , Dermatitis, Atopic/complications , Dermatitis, Atopic/prevention & control , Dietary Supplements , Humans , Interleukin-4 Receptor alpha Subunit/antagonists & inhibitors , Interleukins/antagonists & inhibitors , Janus Kinases/antagonists & inhibitors , Melatonin/therapeutic use , Phosphodiesterase 4 Inhibitors/therapeutic use , Receptors, Cytokine/antagonists & inhibitors , Vitamin D/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL