Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
J Autoimmun ; 141: 103114, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37748979

ABSTRACT

The gastrointestinal tract is home to the largest microbial population in the human body. The gut microbiota plays significant roles in the development of the gut immune system and has a substantial impact on the maintenance of immune tolerance beginning in early life. These microbes interact with the immune system in a dynamic and interdependent manner. They generate immune signals by presenting a vast repertoire of antigenic determinants and microbial metabolites that influence the development, maturation and maintenance of immunological function and homeostasis. At the same time, both the innate and adaptive immune systems are involved in modulating a stable microbial ecosystem between the commensal and pathogenic microorganisms. Hence, the gut microbial population and the host immune system work together to maintain immune homeostasis synergistically. In susceptible hosts, disruption of such a harmonious state can greatly affect human health and lead to various auto-inflammatory and autoimmune disorders. In this review, we discuss our current understanding of the interactions between the gut microbiota and immunity with an emphasis on: a) important players of gut innate and adaptive immunity; b) the contribution of gut microbial metabolites; and c) the effect of disruption of innate and adaptive immunity as well as alteration of gut microbiome on the molecular mechanisms driving autoimmunity in various autoimmune diseases.


Subject(s)
Autoimmune Diseases , Gastrointestinal Microbiome , Humans , Ecosystem , Immune System , Adaptive Immunity , Immune Tolerance , Dysbiosis
2.
Nutrients ; 14(18)2022 Sep 09.
Article in English | MEDLINE | ID: mdl-36145091

ABSTRACT

BACKGROUND AND AIMS: A compromise in intestinal mucosal functions is associated with several chronic inflammatory diseases. Previously, we reported that obese humans have a reduced expression of intestinal Janus kinase-3 (Jak3), a non-receptor tyrosine kinase, and a deficiency of Jak3 in mice led to predisposition to obesity-associated metabolic syndrome. Since meta-analyses show cognitive impairment as co-morbidity of obesity, the present study demonstrates the mechanistic role of Jak3 in obesity associated cognitive impairment. Our data show that high-fat diet (HFD) suppresses Jak3 expression both in intestinal mucosa and in the brain of wild-type mice. METHODOLOGY: Recapitulating these conditions using global (Jak3-KO) and intestinal epithelial cell-specific conditional (IEC-Jak3-KO) mice and using cognitive testing, western analysis, flow cytometry, immunofluorescence microscopy and 16s rRNA sequencing, we demonstrate that HFD-induced Jak3 deficiency is responsible for cognitive impairments in mice, and these are, in part, specifically due to intestinal epithelial deficiency of Jak3. RESULTS: We reveal that Jak3 deficiency leads to gut dysbiosis, compromised TREM-2-functions-mediated activation of microglial cells, increased TLR-4 expression and HIF1-α-mediated inflammation in the brain. Together, these lead to compromised microglial-functions-mediated increased deposition of ß-amyloid (Aß) and hyperphosphorylated Tau (pTau), which are responsible for cognitive impairments. Collectively, these data illustrate how the drivers of obesity promote cognitive impairment and demonstrate the underlying mechanism where HFD-mediated impact on IEC-Jak3 deficiency is responsible for Jak3 deficiency in the brain, reduced microglial TREM2 expression, microglial activation and compromised clearance of Aß and pTau as the mechanism during obesity-associated cognitive impairments. CONCLUSION: Thus, we not only demonstrate the mechanism of obesity-associated cognitive impairments but also characterize the tissue-specific role of Jak3 in such conditions through mucosal tolerance, gut-brain axis and regulation of microglial functions.


Subject(s)
Cognitive Dysfunction , Toll-Like Receptor 4 , Animals , Cognitive Dysfunction/etiology , Diet, High-Fat , Humans , Janus Kinase 3 , Membrane Glycoproteins , Mice , Mice, Inbred C57BL , Obesity/metabolism , RNA, Ribosomal, 16S , Receptors, Immunologic
3.
Environ Sci Pollut Res Int ; 29(23): 34288-34301, 2022 May.
Article in English | MEDLINE | ID: mdl-35038087

ABSTRACT

Bioleaching is considered an eco-friendly technique for leaching metals from spent hydroprocessing catalysts; however, the low bioleaching yield of some valuable metals (Mo and V) is a severe bottleneck to its successful implementation. The present study reported the potential of an integrated bioleaching-chemical oxidation process in improved leaching of valuable metals (Mo and V) from refinery spent hydroprocessing catalysts. The first stage bioleaching of a spent catalyst (coked/decoked) was conducted using sulfur-oxidizing microbes. The results suggested that after 72 h of bioleaching, 85.7% Ni, 86.9% V, and 72.1% Mo were leached out from the coked spent catalyst. Bioleaching yield in decoked spent catalyst was relatively lower (86.8% Ni, 79.8% V, and 59.8% Mo). The low bioleaching yield in the decoked spent catalyst was attributed to metals' presence in stable fractions (residual + oxidizable). After first stage bioleaching, the integration of a second stage chemical oxidation process (1 M H2O2) drastically improved the leaching of Ni, Mo, and V (94.2-100%) from the coked spent catalyst. The improvement was attributed to the high redox potential (1.77 V) of the H2O2, which led to the transformation of low-valence metal sulfides into high-valence metallic ions more conducive to acidic bioleaching. In the decoked spent catalyst, the increment in the leaching yield after second stage chemical oxidation was marginal (<5%). The results suggested that the integrated bioleaching-chemical oxidation process is an effective method for the complete leaching of valuable metals from the coked spent catalyst.


Subject(s)
Hydrogen Peroxide , Metals , Catalysis , Feasibility Studies , Oxidation-Reduction
4.
Mediators Inflamm ; 2022: 9621668, 2022.
Article in English | MEDLINE | ID: mdl-35082553

ABSTRACT

Inflammatory bowel disease (IBD) is a chronic inflammatory condition of gastrointestinal (GI) tract with dysregulated mucosal immune functions and disturbed commensal ecosystem of the intestinal lumen. IBD is categorized into two major subsets: Crohn's disease (CD) and ulcerative colitis (UC). Though advent of biologics has shifted the treatment with relatively longer remission compared to small molecule pharmaceuticals, patients still suffer from long-term complications. Since gut-microbiome is now accepted as another human organ holding potential for long-lasting human health, probiotics, and its engineering hold great promises to treat several previously untreatable chronic inflammatory conditions including IBD. Several emerging biological engineering tools have unlimited potential to manipulate probiotic bacterial system. These can produce useful therapeutic biologics with a goal to either ameliorate and/or treat previously untreatable chronic inflammatory conditions. As gut-microbiome is diverse and vary in different ethnic, geographic, and cultural human population, it will be important to develop vision for personalized probiotic treatment and develop the technology thereof to make personalized probiotic options a reality. The aim of this review paper is to present an overview of the current knowledge on both pharmacological and nonpharmacological IBD treatment modalities with a special emphasis on probiotic strains that are developed through the probiotic engineering. These engineered probiotics contain the most anti-inflammatory cytokines found within the human immune response and are currently being used to treat the intestinal inflammation in IBD for the IBD treatment.


Subject(s)
Colitis, Ulcerative , Crohn Disease , Inflammatory Bowel Diseases , Probiotics , Colitis, Ulcerative/drug therapy , Crohn Disease/drug therapy , Ecosystem , Humans , Inflammatory Bowel Diseases/therapy , Probiotics/therapeutic use
5.
Life Sci ; 278: 119544, 2021 Aug 01.
Article in English | MEDLINE | ID: mdl-33945827

ABSTRACT

Drug-induced liver injury (DILI) is one of the most frequent sources of liver failure and the leading cause of liver transplant. Common non-prescription medications such as non-steroidal anti-inflammatory drugs (NSAIDs), acetaminophen, and other prescription drugs when taken at more than the recommended doses may lead to DILI. The severity of DILI is affected by factors such as age, ethnicity, race, gender, nutritional status, on-going liver diseases, renal function, pregnancy, alcohol consumption, and drug-drug interactions. Characteristics of DILI-associated inflammation include apoptosis and necrosis of hepatocytes and hepatic infiltration of pro-inflammatory immune cells. If untreated or if the inflammation continues, DILI and associated hepatic inflammation may lead to development of hepatocarcinoma. The therapeutic approach for DILI-associated hepatic inflammation depends on whether the inflammation is acute or chronic. Discontinuing the causative medication, vaccination, and special dietary supplementation are some of the conventional approaches to treat DILI. In this review, we discuss a concise overview of DILI-associated liver complications, and current therapeutic options with special emphasis on biologics including the scope of cytokine therapy in hepatic repair and resolution of inflammation caused by over- the-counter (OTC) or prescription drugs.


Subject(s)
Chemical and Drug Induced Liver Injury/drug therapy , Cytokines/therapeutic use , Interleukin-2/therapeutic use , Liver/drug effects , Analgesics, Non-Narcotic/adverse effects , Animals , Chemical and Drug Induced Liver Injury/pathology , Cytokines/pharmacology , Humans , Inflammation/drug therapy , Inflammation/pathology , Interleukin-2/pharmacology , Liver/pathology
6.
Mediators Inflamm ; 2021: 6618924, 2021.
Article in English | MEDLINE | ID: mdl-33814980

ABSTRACT

Janus kinases (Jaks) are a family of nonreceptor tyrosine kinase that include four different members, viz., Jak1, Jak2, Jak3, and Tyk2. Jaks play critical roles in immune cells functions; however, recent studies suggest they also play essential roles in nonimmune cell physiology. This review highlights the significance of epithelial Jaks in understanding the molecular basis of some of the diseases through regulation of epithelial-mesenchymal transition, cell survival, cell growth, development, and differentiation. Growth factors and cytokines produced by the cells of hematopoietic origin use Jak kinases for signal transduction in both immune and nonimmune cells. Among Jaks, Jak3 is widely expressed in both immune cells and in intestinal epithelial cells (IECs) of both humans and mice. Mutations that abrogate Jak3 functions cause an autosomal severe combined immunodeficiency disease (SCID) while activating Jak3 mutations lead to the development of hematologic and epithelial cancers. A selective Jak3 inhibitor CP-690550 (Xeljanz) approved by the FDA for certain chronic inflammatory conditions demonstrates immunosuppressive activity in rheumatoid arthritis, psoriasis, and organ transplant rejection. Here, we also focus on the consequences of Jak3-directed drugs on adverse effects in light of recent discoveries in mucosal epithelial functions of Jak3 with some information on other Jaks. Lastly, we brief on structural implications of Jak3 domains beyond the immune cells. As information about the roles of Jak3 in gastrointestinal functions and associated diseases are only just emerging, in the review, we summarize its implications in gastrointestinal wound repair, inflammatory bowel disease, obesity-associated metabolic syndrome, and epithelial cancers. Lastly, we shed lights on identifying potential novel targets in developing therapeutic interventions of diseases associated with dysfunctional IEC.


Subject(s)
Janus Kinases , Animals , Humans , Janus Kinases/metabolism , Mice , Mutation , Signal Transduction , TYK2 Kinase/metabolism
7.
J Biol Chem ; 294(48): 18337-18348, 2019 11 29.
Article in English | MEDLINE | ID: mdl-31653704

ABSTRACT

Breast cancer resistance protein (BCRP) is a member of ATP-binding cassette (ABC) transporter proteins whose primary function is to efflux substrates bound to the plasma membrane. Impaired intestinal barrier functions play a major role in chronic low-grade inflammation (CLGI)-associated obesity, but the regulation of BCRP during obesity and its role in maintaining the intestinal barrier function during CLGI-associated obesity are unknown. In the present study, using several approaches, including efflux assays, immunoprecipitation, immunoblotting, immunohistochemistry, paracellular permeability assay, FACS, cytokine assay, and immunofluorescence microscopy, we report that obese individuals have compromised intestinal BCRP functions and that diet-induced obese mice recapitulate these outcomes. We demonstrate that the compromised BCRP functions during obesity are because of loss of Janus kinase 3 (JAK3)-mediated tyrosine phosphorylation of BCRP. Our results indicate that JAK3-mediated phosphorylation of BCRP promotes its interactions with membrane-localized ß-catenin essential not only for BCRP expression and surface localization, but also for the maintenance of BCRP-mediated intestinal drug efflux and barrier functions. We observed that reduced intestinal JAK3 expression during human obesity or JAK3 knockout in mouse or siRNA-mediated ß-catenin knockdown in human intestinal epithelial cells all result in significant loss of intestinal BCRP expression and compromised colonic drug efflux and barrier functions. Our results uncover a mechanism of BCRP-mediated intestinal drug efflux and barrier functions and establish a role for BCRP in preventing CLGI-associated obesity both in humans and in mice.


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Epithelial Cells/metabolism , Intestinal Mucosa/metabolism , Janus Kinase 3/metabolism , Neoplasm Proteins/metabolism , Obesity/metabolism , Tissue Inhibitor of Metalloproteinase-1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , Animals , Biological Transport , Colon/metabolism , HT29 Cells , Humans , Insulin/metabolism , Intestinal Mucosa/cytology , Janus Kinase 3/antagonists & inhibitors , Janus Kinase 3/genetics , Mice , Mice, Knockout , Neoplasm Proteins/genetics , Obesity/genetics , Phosphorylation/drug effects , Piperidines/pharmacology , Pyrimidines/pharmacology , Pyrroles/pharmacology , Tissue Inhibitor of Metalloproteinase-1/genetics , beta Catenin/metabolism
8.
J Biol Chem ; 292(40): 16406-16419, 2017 10 06.
Article in English | MEDLINE | ID: mdl-28821617

ABSTRACT

Compromise in adherens junctions (AJs) is associated with several chronic inflammatory diseases. We reported previously that Janus kinase 3, a non-receptor tyrosine kinase, plays a crucial role in AJ formation through its interaction with ß-catenin. In this report, we characterize the structural determinants responsible for Jak3 interactions with ß-catenin and determine the functional implications of previously unknown tyrosine residues on ß-catenin phosphorylated by Jak3. We demonstrate that Jak3 autophosphorylation was the rate-limiting step during Jak3 trans-phosphorylation of ß-catenin, where Jak3 directly phosphorylated three tyrosine residues, viz. Tyr30, Tyr64, and Tyr86 in the N-terminal domain (NTD) of ß-catenin. However, prior phosphorylation of ß-catenin at Tyr654 was essential for further phosphorylation of ß-catenin by Jak3. Interaction studies indicated that phosphorylated Jak3 bound to phosphorylated ß-catenin with a dissociation constant of 0.28 µm, and although both the kinase and FERM (Band 41, ezrin, radixin, and moesin) domains of Jak3 interacted with ß-catenin, the NTD domain of ß-catenin facilitated its interactions with Jak3. Physiologically, Jak3-mediated phosphorylation of ß-catenin suppressed EGF-mediated epithelial-mesenchymal transition and facilitated epithelial barrier functions by AJ localization of phosphorylated ß-catenin through its interactions with α-catenin. Moreover, loss of Jak3-mediated phosphorylation sites in ß-catenin abrogated its AJ localization and compromised epithelial barrier functions. Thus, we not only characterize Jak3 interaction with ß-catenin but also demonstrate the mechanism of molecular interplay between AJ dynamics and EMT by Jak3-mediated NTD phosphorylation of ß-catenin.


Subject(s)
Adherens Junctions/metabolism , Epithelial-Mesenchymal Transition/physiology , Janus Kinase 3/metabolism , beta Catenin/metabolism , Adherens Junctions/genetics , Cell Line , Humans , Janus Kinase 3/genetics , Phosphorylation/physiology , Protein Domains , Protein Transport/physiology , Tyrosine/genetics , Tyrosine/metabolism , alpha Catenin/genetics , alpha Catenin/metabolism , beta Catenin/genetics
9.
J Biol Chem ; 290(49): 29301-12, 2015 Dec 04.
Article in English | MEDLINE | ID: mdl-26451047

ABSTRACT

Obesity, a worldwide epidemic, is a major risk factor for the development of metabolic syndrome (MetS) including diabetes and associated health complications. Recent studies indicate that chronic low-grade inflammation (CLGI) plays a key role in metabolic deterioration in the obese population. Previously, we reported that Jak3 was essential for mucosal differentiation and enhanced colonic barrier functions and its loss in mice resulted in basal CLGI and predisposition to DSS induced colitis. Since CLGI is associated with diabetes, obesity, and metabolic syndrome, present studies determined the role of Jak3 in development of such conditions. Our data show that loss of Jak3 resulted in increased body weight, basal systemic CLGI, compromised glycemic homeostasis, hyperinsulinemia, and early symptoms of liver steatosis. Lack of Jak3 also resulted in exaggerated symptoms of metabolic syndrome by western high-fat diet. Mechanistically, Jak3 was essential for reduced expression and activation of Toll-like receptors (TLRs) in murine intestinal mucosa and human intestinal epithelial cells where Jak3 interacted with and activated p85, the regulatory subunit of the PI3K, through tyrosine phosphorylation of adapter protein insulin receptor substrate (IRS1). These interactions resulted in activation of PI3K-Akt axis, which was essential for reduced TLR expression and TLR associated NFκB activation. Collectively, these results demonstrate the essential role of Jak3 in promoting mucosal tolerance through suppressed expression and limiting activation of TLRs thereby preventing intestinal and systemic CLGI and associated obesity and MetS.


Subject(s)
Janus Kinase 3/metabolism , Metabolic Syndrome/genetics , Obesity/genetics , Animals , Body Weight , Caco-2 Cells , Cytokines/blood , Diet, High-Fat , Disease Models, Animal , Genetic Predisposition to Disease , Glucose Tolerance Test , Humans , Immunity, Innate , Inflammation , Insulin/chemistry , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , Organ Size , Piperidines/chemistry , Pyrimidines/chemistry , Pyrroles/chemistry , Risk Factors , Signal Transduction , Toll-Like Receptors/metabolism
10.
J Biol Chem ; 289(23): 15951-6, 2014 Jun 06.
Article in English | MEDLINE | ID: mdl-24795043

ABSTRACT

Although constitutive activation of Janus kinase 3 (Jak3) leads to different cancers, the mechanism of trans-molecular regulation of Jak3 activation is not known. Previously we reported that Jak3 interactions with adapter protein p52ShcA (Shc) facilitate mucosal homeostasis. In this study, we characterize the structural determinants that regulate the interactions between Jak3 and Shc and demonstrate the trans-molecular mechanism of regulation of Jak3 activation by Shc. We show that Jak3 autophosphorylation was the rate-limiting step during Jak3 trans-phosphorylation of Shc where Jak3 directly phosphorylated two tyrosine residues in Src homology 2 (SH2) domain and one tyrosine residue each in calponin homology 1 (CH1) domain and phosphotyrosine interaction domain (PID) of Shc. Direct interactions between mutants of Jak3 and Shc showed that although FERM domain of Jak3 was sufficient for binding to Shc, CH1 and PID domains of Shc were responsible for binding to Jak3. Functionally Jak3 was autophosphorylated under IL-2 stimulation in epithelial cells. However, Shc recruited tyrosine phosphatases SHP2 and PTP1B to Jak3 and thereby dephosphorylated Jak3. Thus we not only characterize Jak3 interaction with Shc, but also demonstrate the molecular mechanism of intracellular regulation of Jak3 activation where Jak3 interactions with Shc acted as regulators of Jak3 dephosphorylation through direct interactions of Shc with both Jak3 and tyrosine phosphatases.


Subject(s)
Janus Kinase 3/metabolism , Shc Signaling Adaptor Proteins/metabolism , Blotting, Western , HT29 Cells , Humans , Immunoprecipitation , Intestinal Mucosa/cytology , Intestinal Mucosa/enzymology , Intestinal Mucosa/metabolism , Microscopy, Fluorescence , Phosphorylation , Protein Binding , Protein Tyrosine Phosphatases/metabolism
11.
J Biol Chem ; 288(44): 31795-806, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24045942

ABSTRACT

Janus kinase 3 (Jak3) is a nonreceptor tyrosine kinase expressed in both hematopoietic and nonhematopoietic cells. Previously, we characterized the functions of Jak3 in cytoskeletal remodeling, epithelial wound healing, and mucosal homeostasis. However, the role of Jak3 in mucosal differentiation and inflammatory bowel disease was not known. In this report, we characterize the role of Jak3 in mucosal differentiation, basal colonic inflammation, and predisposition toward colitis. Using the Jak3 knock-out (KO) mouse model, we show that Jak3 is expressed in colonic mucosa of mice, and the loss of mucosal expression of Jak3 resulted in reduced expression of differentiation markers for the cells of both enterocytic and secretory lineages. Jak3 KO mice showed reduced expression of colonic villin, carbonic anhydrase, secretory mucin muc2, and increased basal colonic inflammation reflected by increased levels of pro-inflammatory cytokines IL-6 and IL-17A in colon along with increased colonic myeloperoxidase activity. The inflammations in KO mice were associated with shortening of colon length, reduced cecum length, decreased crypt heights, and increased severity toward dextran sulfate sodium-induced colitis. In differentiated human colonic epithelial cells, Jak3 redistributed to basolateral surfaces and interacted with adherens junction (AJ) protein ß-catenin. Jak3 expression in these cells was essential for AJ localization of ß-catenin and maintenance of epithelial barrier functions. Collectively, these results demonstrate the essential role of Jak3 in the colon where it facilitated mucosal differentiation by promoting the expression of differentiation markers and enhanced colonic barrier functions through AJ localization of ß-catenin.


Subject(s)
Cell Differentiation , Colitis/enzymology , Genetic Predisposition to Disease , Intestinal Mucosa/enzymology , Janus Kinase 3/metabolism , Adherens Junctions/enzymology , Adherens Junctions/pathology , Animals , Antigens, Differentiation/biosynthesis , Antigens, Differentiation/genetics , Cecum/enzymology , Cecum/pathology , Cell Line , Colitis/genetics , Colitis/pathology , Colon/enzymology , Colon/pathology , Epithelial Cells/enzymology , Epithelial Cells/pathology , Gene Expression Regulation/genetics , Humans , Intestinal Mucosa/pathology , Janus Kinase 3/genetics , Mice , Mice, Knockout , beta Catenin/genetics , beta Catenin/metabolism
12.
Crit Rev Oncol Hematol ; 86(3): 232-50, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23098684

ABSTRACT

Colorectal cancer is the leading cause of cancer-related mortality in the western world. It is also the third most common cancer diagnosed in both men and women in the United States with a recent estimate for new cases of colorectal cancer in the year 2012 being around 103,170. Various risk factors for colorectal cancer include life-style, diet, age, personal and family history, and racial and ethnic background. While a few cancers are certainly preventable but this does not hold true for colon cancer as it is often detected in its advanced stage and generally not diagnosed until symptoms become apparent. Despite the fact that several options are available for treating this cancer through surgery, chemotherapy, radiation therapy, immunotherapy, and nutritional-supplement therapy, but the success rates are not very encouraging when used alone where secondary complications appear in almost all these therapies. To maximize the therapeutic-effects in patients, combinatorial approaches are essential. In this review we have discussed the therapies previously and currently available to patients diagnosed with colorectal-cancer, focus on some recent developments in basic research that has shaded lights on new therapeutic-concepts utilizing macrophages/dendritic cells, natural killer cells, gene delivery, siRNA-, and microRNA-technology, and specific-targeting of tyrosine kinases that are either mutated or over-expressed in the cancerous cell to treat these cancer. Potential strategies are discussed where these concepts could be applied to the existing therapies under a comprehensive approach to enhance the therapeutic effects.


Subject(s)
Colonic Neoplasms/therapy , Colonic Neoplasms/etiology , Colonic Neoplasms/mortality , Colonic Neoplasms/pathology , Combined Modality Therapy , Humans , Neoplasm Staging , Research
13.
J Biol Chem ; 287(49): 41386-91, 2012 Nov 30.
Article in English | MEDLINE | ID: mdl-23012362

ABSTRACT

Janus kinase 3 (Jak3) is a nonreceptor tyrosine kinase expressed in both hematopoietic and nonhematopoietic cells. Although mutations that abrogate Jak3 functions cause different immunological disorders, its constitutive activation leads to various types of cancer. Previously, we demonstrated that Jak3 interacted with actin-binding protein villin, thereby facilitating cytoskeletal remodeling and wound repair. In this study, we characterize the structural determinants that regulate the interactions between Jak3 and cytoskeletal proteins of the villin/gelsolin family. Functional reconstitution of kinase activity by recombinant full-length (wt) Jak3 using Jak3-wt or villin/gelsolin-wt as substrate showed that Jak3 autophosphorylation was the rate-limiting step during interactions between Jak3 and cytoskeletal proteins. Determination of kinetic parameters showed that phosphorylated (P) Jak3-wt binds to P-villin-wt with a dissociation constant (K(d)) of 23 nM and a Hill's coefficient of 3.7. Pairwise binding between Jak3 mutants and P-villin-wt showed that the FERM domain of Jak3 was sufficient for binding to P-villin-wt with a K(d) of 40.0 nM. However, the SH2 domain of Jak3 prevented P-villin-wt from binding to the FERM domain of nonphosphorylated protein. We demonstrate that the intramolecular interaction between the FERM and SH2 domains of nonphosphorylated Jak3 prevented Jak3 from binding to villin and that tyrosine autophosphorylation of Jak3 at the SH2 domain decreased these intramolecular interactions and facilitated binding of the FERM domain to villin. Thus we demonstrate the molecular mechanism of interactions between Jak3 and cytoskeletal proteins where tyrosine phosphorylation of the SH2 domain acted as an intramolecular switch for the interactions between Jak3 and cytoskeletal proteins.


Subject(s)
Cytoskeleton/metabolism , Gene Expression Regulation, Enzymologic , Janus Kinase 3/metabolism , Cell Line, Tumor , Cell Movement , Humans , Kinetics , Microfilament Proteins/chemistry , Mutagenesis , Neoplasm Metastasis , Phosphorylation , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Tyrosine/metabolism , Wound Healing , src Homology Domains
14.
Am J Physiol Cell Physiol ; 302(5): C735-47, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22116305

ABSTRACT

Sustained damage to the mucosal lining in patients with inflammatory bowel disease (IBD) facilitates translocation of intestinal microbes to submucosal immune cells leading to chronic inflammation. Previously, we demonstrated the role of Jak3 in IL-2-induced intestinal epithelial cell (IEC) migration, one of the early events during intestinal wound repair. In this study, we demonstrate that IL-2 also plays a role in IEC homeostasis through concentration-dependent regulation of IEC proliferation and cell death. At lower concentrations (≤50 U/ml), IL-2 promoted proliferation, while at higher concentrations (100 U/ml), it promoted apoptosis. Activation by IL-2 led to tyrosine phosphorylation-dependent interactions between Jak3 and p52ShcA only at lower concentrations. Phosphatase SHP1 dephosphorylated IL-2-induced phosphorylated p52ShcA. Higher concentrations of IL-2 decreased the phosphorylation of Jak3 and p52ShcA, disrupted their interactions, redistributed Jak3 to the nucleus, and induced apoptosis in IEC. IL-2 also induced dose-dependent upregulation of p52shcA and downregulation of jak3-mRNA. Constitutive overexpression and mir-shRNA-mediated knockdown studies showed that expression of both Jak3 and p52ShcA were necessary for IL-2-induced proliferation of IEC. Doxycycline-regulated sh-RNA expression demonstrated that IL-2-induced downregulation of jak3-mRNA was responsible for higher IL-2-induced apoptosis in IEC. Collectively, these data demonstrate a novel mechanism of IL-2-induced mucosal homeostasis through posttranslational and transcriptional regulation of Jak3 and p52ShcA.


Subject(s)
Inflammatory Bowel Diseases/metabolism , Interleukin-2/physiology , Janus Kinase 3/metabolism , Shc Signaling Adaptor Proteins/metabolism , Apoptosis/drug effects , Caco-2 Cells , Cell Proliferation/drug effects , Gene Expression Regulation/drug effects , HEK293 Cells , HT29 Cells , Homeostasis/drug effects , Homeostasis/physiology , Humans , Inflammatory Bowel Diseases/immunology , Interleukin-2/pharmacology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Janus Kinase 3/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Shc Signaling Adaptor Proteins/genetics , Src Homology 2 Domain-Containing, Transforming Protein 1
16.
Am J Biomed Sci ; 4(1): 14-25, 2012.
Article in English | MEDLINE | ID: mdl-25285182

ABSTRACT

A new series of isoxazole derivatives, N-phenyl-5-carboxamidyl isoxazoles, was investigated for their anticancer activity with solid tumor selectivity. Six N-phenyl-5-carboxamidylisoxazoles were chemically synthesized and evaluated by the in vitro disk-diffusion assay and IC50 cytotoxicity determination. The results showed that one of the derivatives, compound 3,N-(4-chlorophenyl)-5-carboxamidyl isoxazole, was the most active against colon 38 and CT-26 mouse colon tumor cells with an IC50 of 2.5 µg/mL for both cell lines. Western blot analysis showed that compound 3 significantly down-regulated the expression of phosphorylated STAT3 in both human and mouse colon cancer cells indicating that the mechanism of action for compound 3 may involve the inhibition of JAK3/STAT3 signaling pathways. Flow cytometric analysis with Annexin V staining showed that the death induced by compound 3 is mediated through cell necrosis and not apoptotic pathway. In summary, our results show that compound 3 is a new N-phenyl-5-carboxamidyl isoxazole with potential anticancer activity. Compound 3 inhibits the phosphorylation of STAT3, a novel target for chemotherapeutic drugs, and is worthy of further investigation as a potential chemotherapeutic agent for treating colon cancer.

17.
Drug Metab Dispos ; 36(10): 2145-9, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18687802

ABSTRACT

Increasingly, it is recognized that commensal microflora regulate epithelial cell processes through the dynamic interaction of pathogen-associated molecular patterns and host pattern recognition receptors such as Toll-like receptor 4 (TLR4). We therefore investigated the effects of bacterial lipopolysaccharide (LPS) on intestinal P-glycoprotein (P-gp) expression and function. Human SW480 (P-gp+/TLR4+) and Caco-2 (P-gp+/TLR4-) cells were treated with medium control or LPS (100 ng/ml) for 24 h prior to study. P-gp function was assessed by measuring the intracellular concentration of rhodamine 123 (Rh123). To confirm P-gp-specific effects, breast cancer resistance protein (BCRP/ABCG2) and multidrug resistance-associated protein 2 (MRP-2/ABCC2) were also analyzed. Treatment of SW480 cells with LPS led to diminished P-gp activity, which could be prevented with polymyxin B (control: 207+/-16 versus LPS: 402+/-22 versus LPS+polymyxin B: 238+/-26 pmoles Rh123/mg protein, p<0.05 control versus LPS). These effects could be blocked by using polymyxin B and were not seen in the P-gp+/TLR4--Caco-2 cell line (control: 771+/-28 versus LPS: 775+/-59 pmoles Rh123/mg protein). Total cellular levels of P-gp did not change in LPS-treated SW480 cells; however, a significant increase in cell surface P-gp was detected. No change in activity, total protein, or apically located MRP-2 was detected following LPS treatment. Sequence analysis confirmed wild-type status of SW480 cells. These data suggest that activation of TLR4 in intestinal epithelial cells leads to an increase in plasma membrane P-gp that demonstrates a diminished capacity to transport substrate.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Lipopolysaccharides/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/physiology , Base Sequence , Blotting, Western , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , DNA Primers , Fluorescent Dyes/metabolism , Humans , Immunoprecipitation , Intestinal Mucosa , Microscopy, Confocal , Multidrug Resistance-Associated Protein 2
18.
J Biol Chem ; 282(42): 30341-5, 2007 Oct 19.
Article in English | MEDLINE | ID: mdl-17537734

ABSTRACT

Janus kinase 3 (Jak3) is a non-receptor tyrosine kinase known to be expressed in hematopoietic cells. Studies of whole organ homogenates show that Jak3 is also expressed in the intestines of both human and mice. However, neither its expression nor its function has been defined in intestinal epithelial enterocytes. The present studies demonstrate that functional Jak3 is expressed in human intestinal enterocytes HT-29 Cl-19A and Caco-2 and plays an essential role in the intestinal epithelial wound repair process in response to interleukin 2 (IL-2). Exogenous IL-2 enhanced the wound repair of intestinal enterocytes in a dose-dependent manner. Activation by IL-2 led to rapid tyrosine phosphorylation and redistribution of Jak3. IL-2-stimulated redistribution of Jak3 was inhibited by the Jak3-specific inhibitor WHI-P131. IL-2 also induced Jak3-dependent redistribution of the actin cytoskeleton in migrating cells. In these cells Jak3 interacted with the intestinal and renal epithelial cell-specific cytoskeletal protein villin in an IL-2-dependent manner. Inhibition of Jak3 activation resulted in loss of tyrosine phosphorylation of villin and a significant decrease in wound repair of the intestinal epithelial cells. Previously, we had shown that tyrosine phosphorylation of villin is important for cytoskeletal remodeling and cell migration. The present study demonstrates a novel pathway in intestinal enterocytes in which IL-2 enhances intestinal wound repair through mechanisms involving Jak3 and its interactions with villin.


Subject(s)
Enterocytes/enzymology , Interleukin-2/pharmacology , Intestinal Mucosa/injuries , Janus Kinase 3/metabolism , Microfilament Proteins/metabolism , Wound Healing/drug effects , Actins/metabolism , Animals , Caco-2 Cells , Cell Movement/drug effects , Cytoskeleton/metabolism , Dose-Response Relationship, Drug , Enterocytes/pathology , Enzyme Activation/drug effects , Hematopoietic Stem Cells/enzymology , Humans , Intestinal Mucosa/enzymology , Intestinal Mucosa/pathology , Mice , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Protein Transport/drug effects , Quinazolines/pharmacology , Tyrosine/metabolism
19.
Biochem Biophys Res Commun ; 324(2): 679-85, 2004 Nov 12.
Article in English | MEDLINE | ID: mdl-15474481

ABSTRACT

Degenerate primers were designed from the conserved zone of hydA structural gene encoding for catalytic subunit of [Fe]-hydrogenase of different hydrogen producing bacteria. A 750 bp of PCR product was amplified by using the above-mentioned degenerate primers and genomic DNA of Enterobacter cloacae IIT-BT 08 as template. The amplified PCR product was cloned and sequenced. The sequence showed the presence of an ORF of 450 bp with significant similarity (40%) with C-terminal end of the conserved zone (H-cluster) of [Fe]- hydrogenase. hydA ORF was then amplified and cloned in-frame with GST in pGEX4T-1 and overexpressed in a non-hydrogen producing Escherichia coli BL-21 to produce a GST-fusion protein of a calculated molecular mass of about 42.1 kDa. Recombinant protein was purified and specifically recognized by anti-GST monoclonal antibody through Western blot. Southern hybridization confirmed the presence of this gene in E. cloacae IIT-BT 08 genome. In vitro hydrogenase assay with the overexpressed hydrogenase enzyme showed that it is catalytically active upon anaerobic adaptation. In vivo hydrogenase assay confirmed the presence of H2 gas in the gas mixture obtained from the batch culture of recombinant E. coli BL-21. A tentative molecular mechanism has been proposed about the transfer of electron from electron donor to H-cluster without the mediation of the F-cluster.


Subject(s)
Enterobacter cloacae/enzymology , Hydrogen/chemistry , Hydrogenase/chemistry , Hydrogenase/genetics , Iron-Sulfur Proteins/chemistry , Iron-Sulfur Proteins/genetics , Amino Acid Sequence , Blotting, Southern , Cloning, Molecular , DNA/chemistry , DNA/metabolism , Electrophoresis, Polyacrylamide Gel , Escherichia coli/metabolism , Genome , Glutathione Transferase/metabolism , Hydrogenase/metabolism , Molecular Sequence Data , Open Reading Frames , Polymerase Chain Reaction , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...