Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros












Base de dados
Intervalo de ano de publicação
1.
Clin Sci (Lond) ; 137(24): 1823-1838, 2023 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-38126208

RESUMO

Preventing kidney graft dysfunction and rejection is a critical step in addressing the nationwide organ shortage and improving patient outcomes. While kidney transplants (KT) are performed more frequently, the overall number of patients on the waitlist consistently exceeds organ availability. Despite improved short-term outcomes in KT, comparable progress in long-term allograft survival has not been achieved. Major cause of graft loss at 5 years post-KT is chronic allograft dysfunction (CAD) characterized by interstitial fibrosis and tubular atrophy (IFTA). Accordingly, proactive prevention of CAD requires a comprehensive understanding of the immune mechanisms associated with either further dysfunction or impaired repair. Allograft rejection is primed by innate immune cells and carried out by adaptive immune cells. The rejection process is primarily facilitated by antibody-mediated rejection (ABMR) and T cell-mediated rejection (TCMR). It is essential to better elucidate the actions of individual immune cell subclasses (e.g. B memory, Tregs, Macrophage type 1 and 2) throughout the rejection process, rather than limiting our understanding to broad classes of immune cells. Embracing multi-omic approaches may be the solution in acknowledging these intricacies and decoding these enigmatic pathways. A transition alongside advancing technology will better allow organ biology to find its place in this era of precision and personalized medicine.


Assuntos
Rejeição de Enxerto , Transplante de Rim , Humanos , Rejeição de Enxerto/etiologia , Rim , Transplante Homólogo , Transplante de Rim/efeitos adversos , Aloenxertos
2.
Am J Transplant ; 23(9): 1434-1445, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37201755

RESUMO

Operational tolerance (OT) after kidney transplantation is defined as stable graft acceptance without the need for immunosuppression therapy. However, it is not clear which cellular and molecular pathways are driving tolerance in these patients. In this first-of-its-kind pilot study, we assessed the immune landscape associated with OT using single-cell analyses. Peripheral mononuclear cells from a kidney transplant recipient with OT (Tol), 2 healthy individuals (HC), and a kidney transplant recipient with normal kidney function on standard-of-care immunosuppression (SOC) were evaluated. The immune landscape of the Tol was drastically different from that of SOC and emerged closer to the profile of HC. TCL1A+ naive B cells and LSGAL1+ regulatory T cells (Tregs) were in higher proportions in Tol. We were unable to identify the Treg subcluster in SOC. The ligand-receptor analysis in HC and Tol identified interactions between B cells, and Tregs that enhance the proliferation and suppressive function of Tregs. SOC reported the highest proportion of activated B cells with more cells in the G2M phase. Our single-cell RNA sequencing study identified the mediators of tolerance; however, it emphasizes the requirement of similar investigations on a larger cohort to reaffirm the role of immune cells in tolerance.


Assuntos
Transplante de Rim , Humanos , Transplante de Rim/efeitos adversos , Leucócitos Mononucleares , Projetos Piloto , Rejeição de Enxerto/etiologia , Tolerância Imunológica , Linfócitos T Reguladores , Análise de Sequência de RNA , Tolerância ao Transplante
3.
J Invest Dermatol ; 143(10): 1964-1972.e4, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37004877

RESUMO

Ligand activation of the aryl hydrocarbon receptor (AHR) accelerates keratinocyte differentiation and the formation of the epidermal permeability barrier. Several classes of lipids, including ceramides, are critical to the epidermal permeability barrier. In normal human epidermal keratinocytes, the AHR ligand, 2,3,7,8-tetrachlorodibenzo-p-dioxin, increased RNA levels of ceramide metabolism and transport genes: uridine diphosphate glucose ceramide glucosyltransferase (UGCG), ABCA12, GBA1, and SMPD1. Levels of abundant skin ceramides were also increased by 2,3,7,8-tetrachlorodibenzo-p-dioxin. These included the metabolites synthesized by UGCG, glucosylceramides, and acyl glucosylceramides. Chromatin immunoprecipitation-sequence analysis and luciferase reporter assays identified UGCG as a direct AHR target. The AHR antagonist, GNF351, inhibited the 2,3,7,8-tetrachlorodibenzo-p-dioxin-mediated RNA and transcriptional increases. Tapinarof, an AHR ligand approved for the treatment of psoriasis, increased UGCG RNA, protein, and its lipid metabolites hexosylceramides as well as increased the RNA expression of ABCA12, GBA1, and SMPD1. In Ahr-null mice, Ugcg RNA and hexosylceramides were lower than those in the wild type. These results indicate that the AHR regulates the expression of UGCG, a ceramide-metabolizing enzyme required for ceramide trafficking, keratinocyte differentiation, and epidermal permeability barrier formation.


Assuntos
Glucosilceramidas , Dibenzodioxinas Policloradas , Animais , Camundongos , Humanos , Glucosilceramidas/metabolismo , Uridina Difosfato Glucose , Receptores de Hidrocarboneto Arílico/genética , Receptores de Hidrocarboneto Arílico/metabolismo , Ligantes , RNA
4.
Transplantation ; 107(10): 2143-2154, 2023 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-36814094

RESUMO

Solid organ transplantation saves thousands of lives suffering from end-stage diseases. Although early transplants experienced acute organ injury, medical breakthroughs, such as tissue typing, and use of immunosuppressive agents have considerably improved graft survival. However, the overall incidence of allograft injury and chronic rejection remains high. Often the clinical manifestations of organ injury or rejection are nonspecific and late. Current requirement for successful organ transplantation is the identification of reliable, accurate, disease-specific, noninvasive methods for the early diagnosis of graft injury or rejection. Development of noninvasive techniques is important to allow routine follow-ups without the discomfort and risks associated with a graft biopsy. Multiple biofluids have been successfully tested for the presence of potential proteomic biomarkers; these include serum, plasma, urine, and whole blood. Kidney transplant research has provided significant evidence to the potential of proteomics-based biomarkers for acute and chronic kidney rejection, delayed graft function, early detection of declining allograft health. Multiple proteins have been implicated as biomarkers; however, recent observations implicate the use of similar canonical pathways and biofunctions associated with graft injury/rejection with altered proteins as potential biomarkers. Unfortunately, the current biomarker studies lack high sensitivity and specificity, adding to the complexity of their utility in the clinical space. In this review, we first describe the high-throughput proteomics technologies and then discuss the outcomes of proteomics profiling studies in the transplantation of several organs. Existing literature provides hope that novel biomarkers will emerge from ongoing efforts and guide physicians in delivering specific therapies to prolong graft survival.


Assuntos
Transplante de Rim , Transplante de Órgãos , Proteômica/métodos , Transplante de Rim/efeitos adversos , Transplante de Órgãos/efeitos adversos , Transplante Homólogo , Rejeição de Enxerto/diagnóstico , Rejeição de Enxerto/patologia , Biomarcadores/metabolismo
5.
EMBO Rep ; 24(3): e55643, 2023 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-36592158

RESUMO

Extensive desmoplasia and poor vasculature renders pancreatic tumors severely hypoxic, contributing to their aggressiveness and therapy resistance. Here, we identify the HuR/MYB/HIF1α axis as a critical regulator of the metabolic plasticity and hypoxic survival of pancreatic cancer cells. HuR undergoes nuclear-to-cytoplasmic translocation under hypoxia and stabilizes MYB transcripts, while MYB transcriptionally upregulates HIF1α. Upon MYB silencing, pancreatic cancer cells fail to survive and adapt metabolically under hypoxia, despite forced overexpression of HIF1α. MYB induces the transcription of several HIF1α-regulated glycolytic genes by directly binding to their promoters, thus enhancing the recruitment of HIF1α to hypoxia-responsive elements through its interaction with p300-dependent histone acetylation. MYB-depleted pancreatic cancer cells exhibit a dramatic reduction in tumorigenic ability, glucose-uptake and metabolism in orthotopic mouse model, even after HIF1α restoration. Together, our findings reveal an essential role of MYB in metabolic reprogramming that supports pancreatic cancer cell survival under hypoxia.


Assuntos
Neoplasias Pancreáticas , Camundongos , Animais , Neoplasias Pancreáticas/genética , Hipóxia , Regiões Promotoras Genéticas , Hipóxia Celular/genética , Linhagem Celular Tumoral , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética
6.
Am J Physiol Gastrointest Liver Physiol ; 324(3): G207-G218, 2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36648139

RESUMO

Treatment of advanced liver disease using surgical modalities is possible due to the liver's innate ability to regenerate following resection. Several key cellular events in the regenerative process converge at the mitochondria, implicating their crucial roles in liver regeneration. Mitochondria enable the regenerating liver to meet massive metabolic demands by coordinating energy production to drive cellular proliferative processes and vital homeostatic functions. Mitochondria are also involved in terminating the regenerative process by mediating apoptosis. Studies have shown that attenuation of mitochondrial activity results in delayed liver regeneration, and liver failure following resection is associated with mitochondrial dysfunction. Emerging mitochondria therapy (i.e., mitotherapy) strategies involve isolating healthy donor mitochondria for transplantation into diseased organs to promote regeneration. This review highlights mitochondria's inherent role in liver regeneration.


Assuntos
Hepatectomia , Regeneração Hepática , Fígado/metabolismo , Mitocôndrias , Proliferação de Células
7.
J Proteome Res ; 19(2): 794-804, 2020 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-31928012

RESUMO

Earlier we have shown important roles of MYB in pancreatic tumor pathobiology. To better understand the role of MYB in the tumor microenvironment and identify MYB-associated secreted biomarker proteins, we conducted mass spectrometry analysis of the secretome from MYB-modulated and control pancreatic cancer cell lines. We also performed in silico analyses to determine MYB-associated biofunctions, gene networks, and altered biological pathways. Our data demonstrated significant modulation (p < 0.05) of 337 secreted proteins in MYB-silenced MiaPaCa cells, whereas 282 proteins were differentially present in MYB-overexpressing BxPC3 cells, compared to their respective control cells. Alteration of several phenotypes such as cellular movement, cell death and survival, inflammatory response, protein synthesis, etc. was associated with MYB-induced differentially expressed proteins (DEPs) in secretomes. DEPs from MYB-silenced MiaPaCa PC cells were suggestive of the downregulation of genes primarily associated with glucose metabolism, PI3K/AKT signaling, and oxidative stress response, among others. DEPs from MYB-overexpressing BxPC3 cells suggested the enhanced release of proteins associated with glucose metabolism and cellular motility. We also observed that MYB positively regulated the expression of four proteins with potential biomarker properties, i.e., FLNB, ENO1, ITGB1, and INHBA. Mining of publicly available databases using Oncomine and UALCAN demonstrated that these genes are overexpressed in pancreatic tumors and associated with reduced patient survival. Altogether, these data provide novel avenues for future investigations on diverse biological functions of MYB, specifically in the tumor microenvironment, and could also be exploited for biomarker development.


Assuntos
Neoplasias Pancreáticas , Proteômica , Biomarcadores , Biomarcadores Tumorais/genética , Humanos , Neoplasias Pancreáticas/genética , Fosfatidilinositol 3-Quinases , Transdução de Sinais , Microambiente Tumoral
8.
Mol Cancer Res ; 16(2): 187-196, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29117940

RESUMO

The ETS family transcription factor ETV4 is aberrantly expressed in a variety of human tumors and plays an important role in carcinogenesis through upregulation of relevant target gene expression. Here, it is demonstrated that ETV4 is overexpressed in pancreatic cancer tissues as compared with the normal pancreas, and is associated with enhanced growth and rapid cell-cycle progression of pancreatic cancer cells. ETV4 expression was silenced through stable expression of a specific short hairpin RNA (shRNA) in two pancreatic cancer cell lines (ASPC1 and Colo357), while it was ectopically expressed in BXPC3 cells. Silencing of ETV4 in ASPC1 and Colo357 cells reduced the growth by 55.3% and 38.9%, respectively, while forced expression of ETV4 in BXPC3 cells increased the growth by 46.8% in comparison with respective control cells. Furthermore, ETV4-induced cell growth was facilitated by rapid transition of cells from G1- to S-phase of the cell cycle. Mechanistic studies revealed that ETV4 directly regulates the expression of Cyclin D1 CCND1, a protein crucial for cell-cycle progression from G1- to S-phase. These effects on the growth and cell cycle were reversed by the forced expression of Cyclin D1 in ETV4-silenced pancreatic cancer cells. Altogether, these data provide the first experimental evidence for a functional role of ETV4 in pancreatic cancer growth and cell-cycle progression.Implications: The functional and mechanistic data presented here regarding ETV4 in pancreatic cancer growth and cell-cycle progression suggest that ETV4 could serve as a potential biomarker and novel target for pancreatic cancer therapy. Mol Cancer Res; 16(2); 187-96. ©2017 AACR.


Assuntos
Proteínas E1A de Adenovirus/genética , Proteínas E1A de Adenovirus/metabolismo , Ciclina D1/genética , Neoplasias Pancreáticas/genética , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Humanos , Neoplasias Pancreáticas/metabolismo , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-ets , Transcrição Gênica , Regulação para Cima
9.
Int J Mol Sci ; 18(4)2017 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-28383487

RESUMO

Pancreatic cancer (PC) continues to rank among the most lethal cancers. The consistent increase in incidence and mortality has made it the seventh leading cause of cancer-associated deaths globally and the third in the United States. The biggest challenge in combating PC is our insufficient understanding of the molecular mechanism(s) underlying its complex biology. Studies during the last several years have helped identify several putative factors and events, both genetic and epigenetic, as well as some deregulated signaling pathways, with implications in PC onset and progression. In this review article, we make an effort to summarize our current understanding of molecular and cellular events involved in the pathogenesis of pancreatic malignancy. Specifically, we provide up-to-date information on the genetic and epigenetic changes that occur during the initiation and progression of PC and their functional involvement in the pathogenic processes. We also discuss the impact of the tumor microenvironment on the molecular landscape of PC and its role in aggressive disease progression. It is envisioned that a better understanding of these molecular factors and the mechanisms of their actions can help unravel novel diagnostic and prognostic biomarkers and can also be exploited for future targeted therapies.


Assuntos
Biomarcadores Tumorais/genética , Epigênese Genética , Mutação , Neoplasias Pancreáticas/patologia , Progressão da Doença , Regulação Neoplásica da Expressão Gênica , Predisposição Genética para Doença , Humanos , Terapia de Alvo Molecular , Neoplasias Pancreáticas/genética , Microambiente Tumoral
10.
Molecules ; 22(3)2017 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-28273819

RESUMO

Phytochemicals are an important part of traditional medicine and have been investigated in detail for possible inclusion in modern medicine as well. These compounds often serve as the backbone for the synthesis of novel therapeutic agents. For many years, phytochemicals have demonstrated encouraging activity against various human cancer models in pre-clinical assays. Here, we discuss select phytochemicals-curcumin, epigallocatechin-3-gallate (EGCG), resveratrol, plumbagin and honokiol-in the context of their reported effects on the processes of inflammation and oxidative stress, which play a key role in tumorigenesis. We also discuss the emerging evidence on modulation of tumor microenvironment by these phytochemicals which can possibly define their cancer-specific action. Finally, we provide recent updates on how low bioavailability, a major concern with phytochemicals, is being circumvented and the general efficacy being improved, by synthesis of novel chemical analogs and nanoformulations.


Assuntos
Anti-Inflamatórios/uso terapêutico , Neoplasias/prevenção & controle , Compostos Fitoquímicos/farmacologia , Anti-Inflamatórios/farmacologia , Humanos , Medicina Tradicional , Estresse Oxidativo/efeitos dos fármacos , Compostos Fitoquímicos/uso terapêutico , Microambiente Tumoral/efeitos dos fármacos
11.
Biochim Biophys Acta Rev Cancer ; 1868(1): 16-28, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28108348

RESUMO

Despite efforts at various levels, racial health disparities still exist in cancer patients. These inequalities in incidence and/or clinical outcome can only be explained by a multitude of factors, with genetic basis being one of them. Several investigations have provided convincing evidence to support epigenetic regulation of cancer-associated genes, which results in the differential transcriptome and proteome, and may be linked to a pre-disposition of individuals of certain race/ethnicity to early or more aggressive cancers. Recent technological advancements and the ability to quickly analyze whole genome have aided in these efforts, and owing to their relatively easy detection, methylation events are much well-characterized, than the acetylation events, across human populations. The early trend of investigating a pre-determined set of genes for differential epigenetic regulation is paving way for more unbiased screening. This review summarizes our current understanding of the epigenetic events that have been tied to the racial differences in cancer incidence and mortality. A better understanding of the epigenetics of racial diversity holds promise for the design and execution of novel strategies targeting the human epigenome for reducing the disparity gaps.


Assuntos
Epigênese Genética/genética , Neoplasias/genética , Acetilação , Animais , Metilação de DNA/genética , Humanos , Proteoma/genética , Transcriptoma/genética
12.
Am J Cancer Res ; 7(1): 1-12, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28123843

RESUMO

Last few decades have witnessed remarkable progress in our understanding of cancer initiation and progression leading to refinement of prevention and treatment approaches. Although these advances have improved the survival of cancer patients in general, certain racial/ethnic groups have benefited only partially. Footprints of cancer-associated racial disparities are very much evident in cancers of the prostate, breast, cervical, colorectal, endometrium, liver and lung. These health inequalities are mostly attributed to socioeconomic differences among races, but there is a growing realization that these may actually be due to inherent biological differences as well. Indeed, significant data now exist to support the biological basis of racial disparities in cancer, warranting basic research investigations, using appropriate tools and model systems. In this article, we have aimed to succinctly review the literature supporting the biological bases of racial disparities in cancer, along with available resources, databases and model systems that will be of interest to researchers. Moreover, we have highlighted the specific areas that need attention in terms of development of resources and/or tools, and discuss the opportunities and challenges in basic biological research in cancer health disparities.

13.
Cancer Res ; 76(24): 7254-7264, 2016 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-27923829

RESUMO

Aberrant expression of the kinase IKKε in pancreatic ductal adenocarcinoma (PDAC) has been associated with poor prognosis. In this study, we define a pathobiologic function for IKKε in reprogramming glucose metabolism and driving progression in PDAC. Silencing IKKε in PDAC cells, which overexpressed it endogenously, was sufficient to reduce malignant cell growth, clonogenic potential, glucose consumption, lactate secretion, and expression of genes involved in glucose metabolism, without impacting the basal oxygen consumption rate. IKKε silencing also attenuated c-Myc in a manner associated with diminished signaling through an AKT/GSK3ß/c-MYC phosphorylation cascade that promoted MYC nuclear accumulation. In an orthotopic mouse model, IKKε-silenced PDAC exhibited a relative reduction in glucose uptake, tumorigenicity, and metastasis. Overall, our findings offer a preclinical mechanistic rationale to target IKKε to improve the therapeutic management of PDAC in patients. Cancer Res; 76(24); 7254-64. ©2016 AACR.


Assuntos
Carcinoma Ductal Pancreático/patologia , Glucose/metabolismo , Quinase I-kappa B/biossíntese , Neoplasias Pancreáticas/patologia , Animais , Carcinoma Ductal Pancreático/metabolismo , Linhagem Celular Tumoral , Técnicas de Silenciamento de Genes , Xenoenxertos , Humanos , Immunoblotting , Imunoprecipitação , Camundongos , Neoplasias Pancreáticas/metabolismo , Reação em Cadeia da Polimerase , Regulação para Cima
14.
Sci Rep ; 6: 28446, 2016 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-27354262

RESUMO

We have recently demonstrated that the transcription factor MYB can modulate several cancer-associated phenotypes in pancreatic cancer. In order to understand the molecular basis of these MYB-associated changes, we conducted deep-sequencing of transcriptome of MYB-overexpressing and -silenced pancreatic cancer cells, followed by in silico pathway analysis. We identified significant modulation of 774 genes upon MYB-silencing (p < 0.05) that were assigned to 25 gene networks by in silico analysis. Further analyses placed genes in our RNA sequencing-generated dataset to several canonical signalling pathways, such as cell-cycle control, DNA-damage and -repair responses, p53 and HIF1α. Importantly, we observed downregulation of the pancreatic adenocarcinoma signaling pathway in MYB-silenced pancreatic cancer cells exhibiting suppression of EGFR and NF-κB. Decreased expression of EGFR and RELA was validated by both qPCR and immunoblotting and they were both shown to be under direct transcriptional control of MYB. These observations were further confirmed in a converse approach wherein MYB was overexpressed ectopically in a MYB-null pancreatic cancer cell line. Our findings thus suggest that MYB potentially regulates growth and genomic stability of pancreatic cancer cells via targeting complex gene networks and signaling pathways. Further in-depth functional studies are warranted to fully understand MYB signaling in pancreatic cancer.


Assuntos
Redes Reguladoras de Genes/genética , Proteínas Proto-Oncogênicas c-myb/metabolismo , Transdução de Sinais/genética , Linhagem Celular Tumoral , Regulação para Baixo , Receptores ErbB/genética , Receptores ErbB/metabolismo , Instabilidade Genômica , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Regiões Promotoras Genéticas , Ligação Proteica , Proteínas Proto-Oncogênicas c-myb/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-myb/genética , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Análise de Sequência de DNA , Fator de Transcrição RelA/genética , Fator de Transcrição RelA/metabolismo , Regulação para Cima
15.
Int J Mol Sci ; 17(5)2016 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-27331811

RESUMO

There is compelling evidence that serum, tissue and intracellular levels of copper are elevated in all types of cancer. Copper has been suggested as an important co-factor for angiogenesis. It is also a major metal ion present inside the nucleus, bound to DNA bases, particularly guanine. We have earlier proposed that the interaction of phenolic-antioxidants with intracellular copper leads to the generation of reactive oxygen species (ROS) that ultimately serve as DNA cleaving agents. To further validate our hypothesis we show here that the antioxidant gossypol and its semi-synthetic derivative apogossypolone induce copper-mediated apoptosis in breast MDA-MB-231, prostate PC3 and pancreatic BxPC-3 cancer cells, through the generation of ROS. MCF10A breast epithelial cells refractory to the cytotoxic property of these compounds become sensitized to treatment against gossypol, as well as apogossypolone, when pre-incubated with copper. Our present results confirm our earlier findings and strengthen our hypothesis that plant-derived antioxidants mobilize intracellular copper instigating ROS-mediated cellular DNA breakage. As cancer cells exist under significant oxidative stress, this increase in ROS-stress to cytotoxic levels could be a successful anticancer approach.


Assuntos
Antineoplásicos/farmacologia , Apoptose , Cobre/metabolismo , Gossipol/análogos & derivados , Espécies Reativas de Oxigênio/metabolismo , Linhagem Celular Tumoral , Dano ao DNA , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Gossipol/farmacologia , Humanos , Estresse Oxidativo
16.
Br J Cancer ; 113(12): 1694-703, 2015 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-26657649

RESUMO

BACKGROUND: MYB encodes for a transcription factor regulating the expression of a wide array of genes involved in cellular functions. It is reported to be amplified in a sub-set of pancreatic cancer (PC) cases; however, its pathobiological association has remained unclear thus far. METHODS: Expression of MYB and other cellular proteins was analysed by immunoblot or qRT-PCR analyses. MYB was stably overexpressed in non-expressing (BxPC3) and silenced in highly expressing (MiaPaCa and Panc1) PC cells. Effect on growth was analysed by automated cell counting at 24-h interval. Cell-cycle progression and apoptotic indices of PC cells with altered MYB expression were measured through flow cytometry upon staining with respective biomarkers. Cell motility/invasion was examined in a Boyden's chamber assay using non-coated or Matrigel-coated membranes. Effect on tumorigenicity and metastatic potential was examined by non-invasive imaging and through end-point measurements of luciferase-tagged MYB-altered PC implanted in the pancreas of nude mice. RESULTS: MYB was aberrantly expressed in all malignant cases of pancreas, whereas remained undetectable in normal pancreas. All the tested established PC cell lines except BxPC3 also exhibited MYB expression. Forced expression of MYB in BxPC3 cells promoted their growth, cell-cycle progression, survival and malignant behaviour, whereas its silencing in MiaPaCa and Panc1 cells produced converse effects. More importantly, ectopic MYB expression was sufficient to confer tumorigenic and metastatic capabilities to non-tumorigenic BxPC3 cells, while its silencing resulted in significant loss of the same in MYB-overexpressing cells as demonstrated in orthotopic mouse model. We also identified several MYB-regulated genes in PC cells that might potentially mediate its effect on tumour growth and metastasis. CONCLUSIONS: MYB is aberrantly overexpressed in PC cells and acts as a key determinant of pancreatic tumour growth and metastasis.


Assuntos
Divisão Celular/genética , Genes myb , Metástase Neoplásica/genética , Neoplasias Pancreáticas/patologia , Animais , Ciclo Celular , Xenoenxertos , Humanos , Camundongos , Neoplasias Pancreáticas/genética
17.
Cell Biol Int ; 37(7): 687-93, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23456795

RESUMO

Prkaca gene of mouse encodes for a cAMP dependent protein kinase catalytic alpha subunit. PKA occurs naturally as a 4-membered structure having two regulatory (R) and two catalytic (C) subunits each encoded by separate gene. Alternatively spliced two transcript variants are known for the Prkaca gene, which encode for two isoforms of PKA C-subunits, namely Cα1 and Cα2. These isoforms arise as a result of alternative splicing of the first coding exon with the internal exons. We have identified a new transcript variant using combinatorial approach of bioinformatics and molecular biology techniques involving RT-PCR, semi-nested PCR and sequencing. The new transcript variant encoding Cα3 isoform has N-terminus that differs from Cα1 and Cα2 isoforms. Cα3 isoform also arise as a result of alternative splicing of first coding exon with the internal exon. Newly identified transcript is expressed ubiquitously in different tissues examined.


Assuntos
Subunidades Catalíticas da Proteína Quinase Dependente de AMP Cíclico/genética , Processamento Alternativo , Sequência de Aminoácidos , Animais , Sequência de Bases , Domínio Catalítico , Biologia Computacional , Subunidades Catalíticas da Proteína Quinase Dependente de AMP Cíclico/metabolismo , Bases de Dados Genéticas , Éxons , Camundongos , Dados de Sequência Molecular , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
18.
Cell Mol Neurobiol ; 32(6): 957-63, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22488527

RESUMO

Gamma-subunit of nicotinic acetylcholine receptor is encoded by chrng gene of mouse. This gene is located on chromosome 1, spans 6.5 kb, and contains 12 exons and 11 introns. Previous studies have reported three transcript variants (C1-3) produced by alternative splicing; C1 contains all the 12 reported exons, C2 uses an in-frame alternate splice site in exon-2, and C3 produced by exon-5 skipping. These variants differ in their channel kinetics and opening times. In our study, we report the presence of two new transcript variants (T1 and T2) of chrng expressed in mouse postnatal day 3 and adult skeletal muscles. These transcripts contain novel first coding exon either N1 or N2. N1 is located in the 5' UTR, while N2 is an extended exon-2. 5' extension of exon-2 contains an initiation codon which produces a novel transcript variant. Either of the two exons can splice with the internal exons to produce mature transcripts making different 5' ends of the transcripts. Consequently, the proteins encoded by these two transcripts differ at N-termini. The presence of N2 exon containing transcript was further supported by the availability of EST from the database. These new variants display heterogeneous properties. They differ in the presence of signal peptide, phosphorylation, and acetylation of their amino acid residues of the new N-termini of the gamma subunit.


Assuntos
Envelhecimento/metabolismo , Processamento Alternativo/genética , Feto/metabolismo , Músculo Esquelético/crescimento & desenvolvimento , Músculo Esquelético/metabolismo , Receptores Nicotínicos/genética , Sequência de Aminoácidos , Animais , Biologia Computacional , Eletroforese em Gel de Ágar , Éxons/genética , Feminino , Íntrons/genética , Masculino , Camundongos , Dados de Sequência Molecular , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Nicotínicos/química , Receptores Nicotínicos/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
19.
Gene ; 500(1): 73-9, 2012 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-22446042

RESUMO

The Prkar1b gene encodes regulatory type 1 beta subunit of protein kinase A. Here we report that mouse R1ß gene produces three alternative splice variants (designated as mR1ß1, mR1ß2 and mR1ß3) that have different N-terminal protein structures. New splice variants were identified using combinatorial approach of bioinformatics pipeline involving online available tools and databases, and molecular biology techniques involving RT-PCR, semi-nested PCR and sequencing. Except mR1ß3, which was not detected by RT-PCR in brain and muscle tissues of 3day old mice, all three spliced isoforms were found to be ubiquitously expressed in tissues and postnatal developmental stages examined. The presence of different N-termini in isoforms may be important for unique docking interactions with A kinase anchoring proteins.


Assuntos
Processamento Alternativo , Subunidade RIbeta da Proteína Quinase Dependente de AMP Cíclico/química , Subunidade RIbeta da Proteína Quinase Dependente de AMP Cíclico/genética , Biologia Molecular/métodos , Sequência de Aminoácidos , Animais , Sequência de Bases , Éxons , Camundongos , Dados de Sequência Molecular , Especificidade de Órgãos , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Alinhamento de Sequência
20.
Mol Biol Rep ; 39(4): 3375-83, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21706350

RESUMO

Prkar1b gene encodes regulatory type I, beta subunit (RIß) of cAMP dependent protein kinase A in mouse. Among the various isoforms of regulatory and catalytic subunits that comprise mammalian PKA, RIß subunit is considered to be one of the important subunits for neuronal functions. This is involved in multiple forms of synaptic plasticity, and influences memory and learning by maintaining hippocampal long-term potentiation (LTP). Deficient expression of this gene has been implicated in autoimmune disease systemic lupus erythematosus (SLE). We have identified two novel non-coding exons of the Prkar1b gene (designated as exon 1A and exon 1B), which are spliced to the canonical exon 2 and constitute the 5' untranslated region giving rise to three alternative transcript isoforms. We have also confirmed the expression of the previously known first exon (designated as exon 1C) with known transcript published earlier. The transcripts containing exons 1A, 1B and 1C are differentially regulated during the development and tissue types. In silico study of more than 20 kb nucleotide sequence upstream of known translational initiation codon revealed three distinct promoter regions named as PA, PB, and PC upstream of the exon 1A, exon 1B and exon 1C respectively. PB is non-CpG related promoter but PA and PC are CpG related promoters, however all three promoters are TATA less. Further analysis showed that these promoters possess potential signature sequences for common as well as different transcription factors suggesting complex regulation of Prkar1b gene.


Assuntos
Regiões 5' não Traduzidas/genética , Processamento Alternativo/genética , Subunidade RIbeta da Proteína Quinase Dependente de AMP Cíclico/genética , DNA Intergênico/genética , Éxons/genética , Perfilação da Expressão Gênica , Animais , Sequência de Bases , Ilhas de CpG/genética , Subunidade RIbeta da Proteína Quinase Dependente de AMP Cíclico/metabolismo , DNA Complementar/genética , Regulação Enzimológica da Expressão Gênica , Variação Genética , Genoma/genética , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Especificidade de Órgãos/genética , Regiões Promotoras Genéticas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...