Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cells ; 13(11)2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38891089

RESUMEN

Inflammatory bowel disease (IBD) is a chronic gut disorder that also elevates the risk of colorectal cancer (CRC). The global incidence and severity of IBD are rising, yet existing therapies often lead to severe side effects. Curcumin offers potent anti-inflammatory and chemotherapeutic properties. However, its clinical translation is hindered by rapid metabolism, as well as poor water solubility and stability, which limits its bioavailability. To address these challenges, we developed OC-S, a water-soluble and colon-targeted curcumin formulation that protects against colitis in mice. The current study advances OC-S as a dietary supplement by establishing its stability and compatibility with various commercial dietary products. Further, OC-S exhibited specific binding to inflamed colon tissue, potentially aiding in targeted drug retention at the inflammation site in colitis with diarrhea symptoms. We further investigated its efficacy in vivo and in vitro using a murine model of colitis and tumoroids from APCmin mice. OC-S significantly reduced colitis severity and pro-inflammatory cytokine expression compared with curcumin, even at very low doses (5 mg/kg/day). It also demonstrated higher anti-proliferative activity in CRC cells and colon cancer tumoroids vs. curcumin. Overall, this study demonstrated that OC-S effectively targets and retains water-soluble curcumin at the inflamed colon sites, while showing promise in addressing both colitis and colorectal cancer, which potentially paves the way for OC-S to advance into clinical development as a dietary product for both IBD and CRC.


Asunto(s)
Colitis , Neoplasias Colorrectales , Curcumina , Animales , Curcumina/farmacología , Curcumina/uso terapéutico , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/tratamiento farmacológico , Colitis/tratamiento farmacológico , Colitis/patología , Colitis/inducido químicamente , Ratones , Humanos , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Proliferación Celular/efectos de los fármacos , Suplementos Dietéticos , Masculino , Sustancias Protectoras/farmacología
2.
Gut Microbes ; 15(2): 2282789, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38010872

RESUMEN

Dysregulation of both the gut barrier and microbiota (dysbiosis) promotes susceptibility to and severity of Inflammatory Bowel Diseases (IBD). Leaky gut and dysbiosis often coexist; however, potential interdependence and molecular regulation are not well understood. Robust expression of claudin-3 (CLDN3) characterizes the gut epithelium, and studies have demonstrated a positive association between CLDN3 expression and gut barrier maturity and integrity, including in response to probiotics. However, the exact status and causal role of CLDN3 in IBD and regulation of gut dysbiosis remain unknown. Analysis of mouse and human IBD cohorts helped examine CLDN3 expression in IBD. The causal role was determined by modeling CLDN3 loss of expression during experimental colitis. 16S sequencing and in silico analysis helped examine gut microbiota diversity between Cldn3KO and WT mice and potential host metabolic responses. Fecal microbiota transplant (FMT) studies were performed to assess the role of gut dysbiosis in the increased susceptibility of Cldn3KO mice to colitis. A significant decrease in CLDN3 expression characterized IBD and CLDN3 loss of expression promoted colitis. 16S sequencing analysis suggested gut microbiota changes in Cldn3KO mice that were capable of modulating fatty acid metabolism and oxidative stress response. FMT from naïve Cldn3KO mice promoted colitis susceptibility in recipient germ-free mice (GFM) compared with GFM-receiving microbiota from WT mice. Our data demonstrate a critical role of CLDN3 in maintaining normal gut microbiota and inflammatory responses, which can be harnessed to develop novel therapeutic opportunities for patients with IBD.


Asunto(s)
Claudina-3 , Colitis , Microbioma Gastrointestinal , Enfermedades Inflamatorias del Intestino , Humanos , Claudina-3/genética , Colitis/genética , Colitis/complicaciones , Disbiosis/complicaciones , Trasplante de Microbiota Fecal , Enfermedades Inflamatorias del Intestino/complicaciones , Animales , Ratones
3.
J Clin Invest ; 133(23)2023 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-37815870

RESUMEN

Patients with inflammatory bowel disease (IBD) are susceptible to colitis-associated cancer (CAC). Chronic inflammation promotes the risk for CAC. In contrast, mucosal healing predicts improved prognosis in IBD and reduced risk of CAC. However, the molecular integration among colitis, mucosal healing, and CAC remains poorly understood. Claudin-2 (CLDN2) expression is upregulated in IBD; however, its role in CAC is not known. The current study was undertaken to examine the role for CLDN2 in CAC. The AOM/DSS-induced CAC model was used with WT and CLDN2-modified mice. High-throughput expression analyses, murine models of colitis/recovery, chronic colitis, ex vivo crypt culture, and pharmacological manipulations were employed in order to increase our mechanistic understanding. The Cldn2KO mice showed significant inhibition of CAC despite severe colitis compared with WT littermates. Cldn2 loss also resulted in impaired recovery from colitis and increased injury when mice were subjected to intestinal injury by other methods. Mechanistic studies demonstrated a possibly novel role of CLDN2 in promotion of mucosal healing downstream of EGFR signaling and by regulation of Survivin expression. An upregulated CLDN2 expression protected from CAC and associated positively with crypt regeneration and Survivin expression in patients with IBD. We demonstrate a potentially novel role of CLDN2 in promotion of mucosal healing in patients with IBD and thus regulation of vulnerability to colitis severity and CAC, which can be exploited for improved clinical management.


Asunto(s)
Neoplasias Asociadas a Colitis , Colitis , Enfermedades Inflamatorias del Intestino , Animales , Humanos , Ratones , Claudina-2/genética , Claudina-2/metabolismo , Colitis/inducido químicamente , Colitis/complicaciones , Colitis/genética , Neoplasias Asociadas a Colitis/complicaciones , Neoplasias Asociadas a Colitis/metabolismo , Sulfato de Dextran/toxicidad , Modelos Animales de Enfermedad , Enfermedades Inflamatorias del Intestino/complicaciones , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/metabolismo , Mucosa Intestinal/metabolismo , Ratones Endogámicos C57BL , Survivin/metabolismo
4.
Cells ; 12(14)2023 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-37508547

RESUMEN

BACKGROUND: Despite significant progress in clinical management, colorectal cancer (CRC) remains the third most common cause of cancer-related deaths. A positive association between PYCR2 (pyrroline-5-carboxylate reductase-2), a terminal enzyme of proline metabolism, and CRC aggressiveness was recently reported. However, how PYCR2 promotes colon carcinogenesis remains ill understood. METHODS: A comprehensive analysis was performed using publicly available cancer databases and CRC patient cohorts. Proteomics and biochemical evaluations were performed along with genetic manipulations and in vivo tumor growth assays to gain a mechanistic understanding. RESULTS: PYCR2 expression was significantly upregulated in CRC and associated with poor patient survival, specifically among PYCR isoforms (PYCR1, 2, and 3). The genetic inhibition of PYCR2 inhibited the tumorigenic abilities of CRC cells and in vivo tumor growth. Coinciding with these observations was a significant decrease in cellular proline content. PYCR2 overexpression promoted the tumorigenic abilities of CRC cells. Proteomics (LC-MS/MS) analysis further demonstrated that PYCR2 loss of expression in CRC cells inhibits survival and cell cycle pathways. A subsequent biochemical analysis supported the causal role of PYCR2 in regulating CRC cell survival and the cell cycle, potentially by regulating the expression of MASTL, a cell-cycle-regulating protein upregulated in CRC. Further studies revealed that PYCR2 regulates Wnt/ß-catenin-signaling in manners dependent on the expression of MASTL and the cancer stem cell niche. CONCLUSIONS: PYCR2 promotes MASTL/Wnt/ß-catenin signaling that, in turn, promotes cancer stem cell populations and, thus, colon carcinogenesis. Taken together, our data highlight the significance of PYCR2 as a novel therapeutic target for effectively treating aggressive colon cancer.


Asunto(s)
Neoplasias del Colon , Proteínas Serina-Treonina Quinasas , Humanos , Proteínas Serina-Treonina Quinasas/metabolismo , beta Catenina/metabolismo , Cromatografía Liquida , Espectrometría de Masas en Tándem , Carcinogénesis , Vía de Señalización Wnt/genética , Microtúbulos/metabolismo , Prolina , Oxidorreductasas/metabolismo , Serina , Proteínas Asociadas a Microtúbulos/metabolismo
5.
Commun Biol ; 6(1): 740, 2023 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-37460613

RESUMEN

Impaired autophagy promotes Inflammatory Bowel Disease (IBD). Claudin-2 is upregulated in IBD however its role in the pathobiology remains uncertain due to its complex regulation, including by autophagy. Irrespective, claudin-2 expression protects mice from DSS colitis. This study was undertaken to examine if an interplay between autophagy and claudin-2 protects from colitis and associated epithelial injury. Crypt culture and intestinal epithelial cells (IECs) are subjected to stress, including starvation or DSS, the chemical that induces colitis in-vivo. Autophagy flux, cell survival, co-immunoprecipitation, proximity ligation assay, and gene mutational studies are performed. These studies reveal that under colitis/stress conditions, claudin-2 undergoes polyubiquitination and P62/SQSTM1-assisted degradation through autophagy. Inhibiting autophagy-mediated claudin-2 degradation promotes cell death and thus suggest that claudin-2 degradation promotes autophagy flux to promote cell survival. Overall, these data inform for the previously undescribed role for claudin-2 in facilitating IECs survival under stress conditions, which can be harnessed for therapeutic advantages.


Asunto(s)
Colitis , Enfermedades Inflamatorias del Intestino , Ratones , Animales , Claudina-2/metabolismo , Proteína Sequestosoma-1/genética , Proteína Sequestosoma-1/metabolismo , Mucosa Intestinal/metabolismo , Colitis/metabolismo , Autofagia/fisiología , Enfermedades Inflamatorias del Intestino/metabolismo
6.
Pharm Res ; 40(1): 107-122, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36271204

RESUMEN

Nucleic acid (NA) therapy has gained importance over the past decade due to its high degree of selectivity and minimal toxic effects over conventional drugs. Currently, intravenous (IV) or intramuscular (IM) formulations constitute majority of the marketed formulations containing nucleic acids. However, oral administration is traditionally preferred due to ease of administration as well as higher patient compliance. To leverage the benefits of oral delivery for NA therapy, the NA of interest must be delivered to the target site avoiding all degrading and inhibiting factors during its transition through the gastrointestinal tract. The oral route presents myriad of challenges to NA delivery, making formulation development challenging. Researchers in the last few decades have formulated various delivery systems to overcome such challenges and several reviews summarize and discuss these strategies in detail. However, there is a need to differentiate between the approaches based on target so that in future, delivery strategies can be developed according to the goal of the study and for efficient delivery to the desired site. The goal of this review is to summarize the mechanisms for target specific delivery, list and discuss the formulation strategies used for oral delivery of NA therapies and delineate the similarities and differences between local and systemic targeting oral delivery systems and current challenges.


Asunto(s)
Sistemas de Liberación de Medicamentos , Ácidos Nucleicos , Humanos , Administración Oral , Tracto Gastrointestinal
7.
Rep Biochem Mol Biol ; 10(4): 711-721, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-35291617

RESUMEN

Background: This study correlates the serum levels of sCD95 & TNF-α with a simple cell-based assay to evaluate the capacity of the serum sample to induce apoptosis in Jurkat cells. Interlinking of these parameters can be explored to design a minimum invasive diagnostic strategy for cervical cancer (CC). Methods: Sera samples were assessed to induce apoptosis in Jurkat cells through FACS. Serum levels of sCD95 and TNF-α were measured by ELISA. JNK phosphorylation was evaluated in sera incubated Jurkat cells. Data was scrutinized through statistical analysis. Results: Significantly higher serum levels of sCD95 and lower TNF-α levels were observed in CC patients; their sera samples inhibited induction of apoptosis in Jurkat cells through reduced JNK phosphorylation. Statistical analysis linked these three parameters for the early screening of CC. Conclusion: Distinct sera levels of sCD95 & TNF-α in CC patients showed an anti-apoptotic effect, which can be considered for early detection of CC.

8.
Biotechniques ; 71(3): 456-464, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34392706

RESUMEN

The conventional orthotopic/xenograft models or genetically engineered murine models of colon cancer (CRC) are limited in their scope for a true understanding of tumor growth, progression and eventual metastasis in its natural microenvironment. In the currently used murine models of CRC metastasis, the metastasis occurs primarily in the liver, though lung metastasis accounts for a significant proportion of CRC metastasis. There is an urgent need for a murine model of CRC, which not only allows tumor progression in the colonic mucosa but also metastasis of the lung. The authors describe a minimally invasive murine model of colon cancer progression that may be ideal for a wide range of applications, including evaluating gene function, microenvironment, cancer metastasis and therapeutic translational research.


Asunto(s)
Neoplasias del Colon , Neoplasias Pulmonares , Trasplante de Neoplasias , Animales , Línea Celular Tumoral , Neoplasias del Colon/patología , Colonoscopía , Modelos Animales de Enfermedad , Neoplasias Pulmonares/secundario , Ratones , Microambiente Tumoral
9.
Cancers (Basel) ; 13(9)2021 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-33946505

RESUMEN

Background: Despite recent advances in therapies, resistance to chemotherapy remains a critical problem in the clinical management of colorectal cancer (CRC). Cancer stem cells (CSCs) play a central role in therapy resistance. Thus, elimination of CSCs is crucial for effective CRC therapy; however, such strategies are limited. Autophagy promotes resistance to cancer therapy; however, whether autophagy protects CSCs to promote resistance to CRC-therapy is not well understood. Moreover, specific and potent autophagy inhibitors are warranted as clinical trials with hydroxychloroquine have not been successful. Methods: Colon cancer cells and tumoroids were used. Fluorescent reporter-based analysis of autophagy flux, spheroid and side population (SP) culture, and qPCR were done. We synthesized 36-077, a potent inhibitor of PIK3C3/VPS34 kinase, to inhibit autophagy. Combination treatments were done using 5-fluorouracil (5-FU) and 36-077. Results: The 5-FU treatment induced autophagy only in a subset of the treated colon cancer. These autophagy-enriched cells also showed increased expression of CSC markers. Co-treatment with 36-077 significantly improved efficacy of the 5-FU treatment. Mechanistic studies revealed that combination therapy inhibited GSK-3ß/Wnt/ß-catenin signaling to inhibit CSC population. Conclusion: Autophagy promotes resistance to CRC-therapy by specifically promoting GSK-3ß/Wnt/ß-catenin signaling to promote CSC survival, and 36-077, a PIK3C3/VPS34 inhibitor, helps promote efficacy of CRC therapy.

10.
J Exp Clin Cancer Res ; 40(1): 77, 2021 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-33622361

RESUMEN

BACKGROUND: Claudin-2 expression is upregulated in multiple cancers and promotes cancer malignancy. Remarkably, the regulation of claudin-2 expression in kidney cell lines contrasts its reported regulation in other organs. However, claudin-2 role in renal clear cell carcinoma (RCC) remains unknown despite its predominant expression in the proximal tubular epithelium (PTE), the site of RCC origin. METHODS: Publicly available and independent patient databases were examined for claudin-2 association with RCC. The novel protein function was validated in vitro and in vivo by gain or loss of function assays. Mechanistic results were concluded by Mass spectroscopy, immunoprecipitation and mutational studies, and functional evaluations. RESULTS: We show that the significant decrease in claudin-2 expression characterized PTE cells and Ex-vivo cultured mouse kidney subjected to dedifferentiation. Inhibition of claudin-2 was enough to induce mesenchymal plasticity and invasive mobility in these models. Further, a progressive loss of claudin-2 expression associated with the RCC progression and poor patient survival. Overexpression of claudin-2 in RCC-derived cancer cells inhibited tumorigenic abilities and xenograft tumor growth. These data supported a novel tumor-suppressive role of claudin-2 in RCC. Mechanistic insights further revealed that claudin-2 associates with YAP-protein and modulates its phosphorylation (S127) and nuclear expression. The tumor suppressive effects of claudin-2 expression were lost upon deletion of its PDZ-binding motif emphasizing the critical role of the PDZ-domain in claudin-2 interaction with YAP in regulating RCC malignancy. CONCLUSIONS: Our results demonstrate a novel kidney specific tumor suppressive role for claudin-2 protein and further demonstrate that claudin-2 co-operates with the YAP signaling in regulating the RCC malignancy.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Carcinoma de Células Renales/genética , Claudina-2/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , Animales , Carcinoma de Células Renales/mortalidad , Línea Celular Tumoral , Proliferación Celular , Modelos Animales de Enfermedad , Humanos , Ratones , Análisis de Supervivencia , Proteínas Señalizadoras YAP
11.
Respir Res ; 21(1): 97, 2020 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-32321514

RESUMEN

BACKGROUND: Environmental organic dust exposures enriched in Toll-like receptor (TLR) agonists can reduce allergic asthma development but are associated with occupational asthma and chronic bronchitis. The TLR adaptor protein myeloid differentiation factor88 (MyD88) is fundamental in regulating acute inflammatory responses to organic dust extract (ODE), yet its role in repetitive exposures is unknown and could inform future strategies. METHODS: Wild-type (WT) and MyD88 knockout (KO) mice were exposed intranasally to ODE or saline daily for 3 weeks (repetitive exposure). Repetitively exposed animals were also subsequently rested with no treatments for 4 weeks followed by single rechallenge with saline/ODE. RESULTS: Repetitive ODE exposure induced neutrophil influx and release of pro-inflammatory cytokines and chemokines were profoundly reduced in MyD88 KO mice. In comparison, ODE-induced cellular aggregates, B cells, mast cell infiltrates and serum IgE levels remained elevated in KO mice and mucous cell metaplasia was increased. Expression of ODE-induced tight junction protein(s) was also MyD88-dependent. Following recovery and then rechallenge with ODE, inflammatory mediators, but not neutrophil influx, was reduced in WT mice pretreated with ODE coincident with increased expression of IL-33 and IL-10, suggesting an adaptation response. Repetitively exposed MyD88 KO mice lacked inflammatory responsiveness upon ODE rechallenge. CONCLUSIONS: MyD88 is essential in mediating the classic airway inflammatory response to repetitive ODE, but targeting MyD88 does not reduce mucous cell metaplasia, lymphocyte influx, or IgE responsiveness. TLR-enriched dust exposures induce a prolonged adaptation response that is largely MyD88-independent. These findings demonstrate the complex role of MyD88-dependent signaling during acute vs. chronic organic dust exposures.


Asunto(s)
Adaptación Fisiológica/fisiología , Polvo , Exposición a Riesgos Ambientales/efectos adversos , Exposición por Inhalación/efectos adversos , Enfermedades Pulmonares/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Animales , Femenino , Enfermedades Pulmonares/inducido químicamente , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
12.
Sci Rep ; 9(1): 12187, 2019 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-31434922

RESUMEN

Cocaine use disorder (CUD), a major health crisis, has traditionally been considered a complication of the CNS; however, it is also closely associated with malnourishment and deteriorating gut health. In light of emerging studies on the potential role of gut microbiota in neurological disorders, we sought to understand the causal association between CUD and gut dysbiosis. Using a comprehensive approach, we confirmed that cocaine administration in mice resulted in alterations of the gut microbiota. Furthermore, cocaine-mediated gut dysbiosis was associated with upregulation of proinflammatory mediators including NF-κB and IL-1ß. In vivo and in vitro analyses confirmed that cocaine altered gut-barrier composition of the tight junction proteins while also impairing epithelial permeability by potentially involving the MAPK/ERK1/2 signaling. Taken together, our findings unravel a causal link between CUD, gut-barrier dysfunction and dysbiosis and set a stage for future development of supplemental strategies for the management of CUD-associated gut complications.


Asunto(s)
Cocaína/toxicidad , Microbioma Gastrointestinal/efectos de los fármacos , Mucosa Intestinal/efectos de los fármacos , Animales , Bacterias/genética , Bacterias/aislamiento & purificación , Factor de Transcripción CDX2/metabolismo , Células CACO-2 , Cocaína/administración & dosificación , Colon/microbiología , Disbiosis/inducido químicamente , Disbiosis/complicaciones , Disbiosis/patología , Humanos , Mucosa Intestinal/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Permeabilidad/efectos de los fármacos , Proteínas de Uniones Estrechas/metabolismo
13.
Life Sci ; 224: 263-273, 2019 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-30902545

RESUMEN

Human immunodeficiency type 1 virus accessory protein Nef is a key modulator of AIDS pathogenesis. With no enzymatic activity, Nef regulated functions in host cells largely depends on its ability to form multi-protein complex with the cellular proteins. Here, we identified Calcium (Ca2+)/Calmodulin dependent protein kinase II subunit delta (CAMKIIδ) as novel Nef interacting host protein. Further, we confirmed that Nef mediated [Ca2+]I promote formation of Nef-CAMKIIδ - apoptosis signal-regulating kinase (ASK-1) heterotrimeric complex. The assembly of Nef with CAMKIIδ - ASK-1 inhibits the downstream p38MAPK phosphorylation resulting in abrogation of apoptosis. Further, using competitive peptide inhibitors against Nef binding domains to CAMKIIδ, identified in the present study and ASK-1, individually blocked physical interaction of Nef with CAMKIIδ-ASK-1 complex and restored p38MAPK phosphorylation and apoptosis. Altogether, our study indicates that HIV-Nef modulates cytosolic [Ca2+]I and blocks CAMKIIδ - ASK-1 kinase activity to inhibit apoptosis of infected cells.


Asunto(s)
Apoptosis , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Infecciones por VIH/metabolismo , MAP Quinasa Quinasa Quinasa 5/metabolismo , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/química , Células HEK293 , Infecciones por VIH/virología , VIH-1/fisiología , Humanos , Células Jurkat , MAP Quinasa Quinasa Quinasa 5/química , Fosforilación , Unión Proteica , Conformación Proteica , Transducción de Señal , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/química , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
14.
Life Sci ; 214: 158-166, 2018 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-30391463

RESUMEN

AIMS: Human immunodeficiency virus -1 [HIV-1] Nef, localizes in different cellular compartments and modulates several cellular pathways. Nef promotes virus pathogenicity through alteration in cell surface receptor expression, apoptosis, protein trafficking etc. Nef regulates viral pathogenesis through interaction with different host proteins. Thus, molecular mechanisms of pathogenesis could be deciphered by identifying novel Nef interacting proteins. MAIN METHODS: HIV-1 Nef interacting proteins were identified by pull down assay and MALDI-TOF analysis. The interaction was further validated through mammalian two hybrid assay. Functional role of this interaction was identified by immunoprecipitation assay, cell invasion and cell migration studies. Fold Change in mRNA levels of CD163, CD206, CCL17 and CCL18 was analyzed using qPCR. KEY FINDINGS: In current study, C. elegans protein ACT4C and its human homolog POTEE was identified to be interacting with Nef. This interaction activates mTORC2 complex, which in-turn activates AKT and PKC-α. The activation of mTORC2 complex was found to be initiated by the interaction of Nef, mTORC2, Rictor to POTEE. The cellular phenotype and functions affected by Nef-POTEE interaction resulted in significant increase in cell invasion and migration of macrophages (MΦ). SIGNIFICANCE: MΦ is primary target of HIV-1 infection where HIV-1 replicates and polarizes immunosuppressive M2 phenotype. Combine effect of M2 phenotype and Viral-host protein interactions compromise the MΦ associated physiological functions. Infected MΦ dissemination into other system also leads to HIV-1 induced malignancies. Therefore, targeting POTEE-Nef interaction can lead to formulating better therapeutic strategy against HIV-1.


Asunto(s)
Antígenos de Neoplasias/metabolismo , VIH-1/patogenicidad , Interacciones Huésped-Patógeno/fisiología , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/metabolismo , Antígenos de Neoplasias/genética , Proteínas de Caenorhabditis elegans/metabolismo , Células HEK293 , Humanos , Macrófagos/virología , Fosforilación , Proteína Quinasa C-alfa/metabolismo , Serina/metabolismo , Transducción de Señal , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/genética
15.
Mol Cancer ; 17(1): 111, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-30068336

RESUMEN

BACKGROUND: Chemotherapeutic agents that modulate cell cycle checkpoints and/or tumor-specific pathways have shown immense promise in preclinical and clinical studies aimed at anti-cancer therapy. MASTL (Greatwall in Xenopus and Drosophila), a serine/threonine kinase controls the final G2/M checkpoint and prevents premature entry of cells into mitosis. Recent studies suggest that MASTL expression is highly upregulated in cancer and confers resistance against chemotherapy. However, the role and mechanism/s of MASTL mediated regulation of tumorigenesis remains poorly understood. METHODS: We utilized a large patient cohort and mouse models of colon cancer as well as colon cancer cells to determine the role of Mastl and associated mechanism in colon cancer. RESULTS: Here, we show that MASTL expression increases in colon cancer across all cancer stages compared with normal colon tissue (P < 0.001). Also, increased levels of MASTL associated with high-risk of the disease and poor prognosis. Further, the shRNA silencing of MASTL expression in colon cancer cells induced cell cycle arrest and apoptosis in vitro and inhibited xenograft-tumor growth in vivo. Mechanistic analysis revealed that MASTL expression facilitates colon cancer progression by promoting the ß-catenin/Wnt signaling, the key signaling pathway implicated in colon carcinogenesis, and up-regulating anti-apoptotic proteins, Bcl-xL and Survivin. Further studies where colorectal cancer (CRC) cells were subjected to 5-fluorouracil (5FU) treatment revealed a sharp increase in MASTL expression upon chemotherapy, along with increases in Bcl-xL and Survivin expression. Most notably, inhibition of MASTL in these cells induced chemosensitivity to 5FU with downregulation of Survivin and Bcl-xL expression. CONCLUSION: Overall, our data shed light on the heretofore-undescribed mechanistic role of MASTL in key oncogenic signaling pathway/s to regulate colon cancer progression and chemo-resistance that would tremendously help to overcome drug resistance in colon cancer treatment.


Asunto(s)
Neoplasias del Colon/patología , Resistencia a Antineoplásicos , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Regulación hacia Arriba , Células CACO-2 , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Progresión de la Enfermedad , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Células HCT116 , Células HT29 , Humanos , Estadificación de Neoplasias , Trasplante de Neoplasias , Pronóstico , Análisis de Supervivencia , Vía de Señalización Wnt
16.
Oncotarget ; 8(50): 87718-87736, 2017 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-29152115

RESUMEN

In normal colon, claudin-2 expression is restricted to the crypt bottom containing the undifferentiated and proliferative colonocytes. Claudin-2 expression is also upregulated in colorectal cancer (CRC) and promotes carcinogenesis. However, cellular mechanism/s regulated by increased claudin-2 expression during the CRC and mechanism/s regulating this increase remain poorly understood. Epigenetic mechanisms help regulate expression of cancer-associated genes and inhibition of Histone Deacetylases (HDACs) induces cell cycle arrest and differentiation. Accordingly, based on a comprehensive in vitro and in vivo analysis we here report that Histone Deacetylases regulate claudin-2 expression in causal association with colonocyte dedifferentiation to promote CRC. Detailed differentiation analyses using colon cancer cells demonstrated inverse association between claudin-2 expression and epithelial differentiation. Genetic manipulation studies revealed the causal role of HDAC-4 in regulating claudin-2 expression during this process. Further analysis identified transcriptional regulation as the underlying mechanism, which was dependent on HDAC-4 dependent modulation of the EGFR-ERK1/2 signaling. Accordingly, colon tumors demonstrated marked upregulation of the HDAC-4/ERK1/2/Claudin-2 signaling. Taken together, we demonstrate a novel role for HDAC-4/EGFR/ERK1/2 signaling in regulating claudin-2 expression to modulate colonocyte differentiation. These findings are of clinical significance and highlight epigenetic regulation as potential mechanism to regulate claudin-2 expression during mucosal pathologies including CRC.

17.
PLoS One ; 8(6): e67586, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23799149

RESUMEN

FasL mediated preferential apoptosis of bystander CTLs while protection of infected CD4(+)T cells remains one of the hallmarks of immune evasion during HIV infection. The property of infected host cells to evade cell-autonomous apoptosis emanates from ability of HIV-1Nef-protein to physically interact with ASK-1 and thereby inhibit its enzymatic activity. The specific domains of HIV-1Nef through which it may interact with ASK1 and thereby impair the ASK1 activity remain unidentified so far and represent a major challenge towards developing clear understanding about the dynamics of this interaction. Using mammalian two hybrid screen in association with site directed mutagenesis and competitive inhibitor peptides, we identified constituent minimal essential domain (152 DEVGEANN 159) through which HIV-1Nef interacts with ASK1 and inhibits its function. Furthermore our study also unravels a novel alternate mechanism underlying HIV-1 Nef mediated ASK1 functional modulation, wherein by potentiating the inhibitory ser(967) phosphorylation of ASK1, HIV-1Nef negatively modulated ASK1 function.


Asunto(s)
VIH-1/fisiología , MAP Quinasa Quinasa Quinasa 5/química , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/química , Secuencia de Aminoácidos , Apoptosis , Sitios de Unión , Unión Competitiva , Células HEK293 , Interacciones Huésped-Patógeno , Humanos , Células Jurkat , MAP Quinasa Quinasa Quinasa 5/metabolismo , Fragmentos de Péptidos/química , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Técnicas del Sistema de Dos Híbridos , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/metabolismo
18.
Free Radic Biol Med ; 53(10): 1929-41, 2012 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-23000059

RESUMEN

The estrogen receptor (ER) plays a cardinal role in estrogen-responsive breast carcinogenesis. It is, however, unclear as to how estrogen-ER interaction potentiates breast cancer progression. Compelling evidence supports estrogen-induced redox alterations, such as augmented reactive oxygen species (ROS) levels, as having a crucial role in breast carcinogenesis. Despite ER being a biological mediator of the majority of estrogen-induced cellular responses; its role in estrogen-induced tissue-specific ROS generation remains largely debatable. We examined a panel of human breast cancer specimens and found that ER-positive breast cancer specimens exhibited a higher incidence of augmented O(2)(•-) levels compared to matched normal tissue. ROS are known to function as signal transducers and ROS-mediated signaling remains a key complementary mechanism that drives carcinogenesis by activating redox-sensitive oncogenic pathways. Additional studies revealed that augmented O(2)(•-) levels in breast cancer specimens coincided with mammalian target of rapamycin complex 2 (mTORC2) hyperactivation. Detailed investigations using in vitro experiments established that 17ß-estradiol (E2)-stimulated breast cancer cells exhibited transiently upregulated O(2)(•-) levels, with the presence of ER being a crucial determinant for the phenomenon to take place. Gene expression, ER transactivation, and confocal studies revealed that the E2-induced transient O(2)(•-) upregulation was effected by ER through a nongenomic pathway possibly involving mitochondria. Furthermore, E2 treatment activated mTORC2 in breast cancer cells in a characteristically ER-dependent manner. Interestingly, altering O(2)(•-) anion levels through chemical/genetic methods caused significant modulation of the mTORC2 signaling cascade. Taken together, our findings unravel a novel nongenomic pathway unique to estrogen-responsive breast cancer cells wherein, upon stimulation by E2, ER may regulate mTORC2 activity in a redox-dependent manner by transiently modulating O(2)(•-) levels particularly within mitochondria. The findings suggest that therapies aimed at counteracting these redox alterations and/or resultant signaling cascades may complement conventional treatments for estrogen-responsive breast cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , Complejos Multiproteicos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptores de Estrógenos/metabolismo , Superóxidos/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Línea Celular Tumoral , Progresión de la Enfermedad , Estradiol/farmacología , Femenino , Humanos , Células MCF-7 , Diana Mecanicista del Complejo 2 de la Rapamicina , Mitocondrias/metabolismo , Oxidación-Reducción , Interferencia de ARN , ARN Interferente Pequeño , Transducción de Señal/fisiología , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...