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










Publication year range
1.
J Family Med Prim Care ; 13(4): 1496-1499, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38827669

ABSTRACT

Background: Care-seeking after injury episodes is generally associated with major uncertainties concerning its incidence, care, and cure/disappearance, and the price of care. Though the utilization pattern of the population is shaped by social, economic, cultural and political factors, it varies widely for the rich and the poor. With this background, a community-based epidemiological study was conducted to determine the care-seeking behavior of victims of unintentional injuries. Methods: A cross-sectional study was conducted from May 2023-September 2023 in the Bhatar block of Purba Bardhaman district, West Bengal. Cluster random sampling was applied to select a required sample of 555 individuals from 24 villages. Study tools used were predesigned and pretested schedules developed by the researchers with the help of guidelines for conducting community surveys on injuries provided by the World Health Organization (WHO). The study had approval from the Institutional Ethics Committee. Descriptive statistics were performed using SPSS V16. Results: Out of 49 injury episodes, 51.0% had received first aid. Out of 39 injury episodes seeking medical attention, a traditional practitioner, healer or bone setter was the type of first medical contact in the case of the majority of injury episodes (38.5%), followed by a hospital (35.9%). Conclusion: Low utilization of government healthcare facilities is a cause for concern. Reliance on unqualified informal healthcare providers is also a cause for concern. Our findings will help design community interventions to increase the provision of appropriate first aid in unintentional injuries.

2.
Pharmaceuticals (Basel) ; 17(2)2024 Jan 30.
Article in English | MEDLINE | ID: mdl-38399394

ABSTRACT

Early stage chemical development presents numerous challenges, and achieving a functional balance is a major hurdle, with many early compounds not meeting the clinical requirements for advancement benchmarks due to issues like poor oral bioavailability. There is a need to develop strategies for achieving the desired systemic concentration for these compounds. This will enable further evaluation of the biological response upon a compound-target interaction, providing deeper insight into the postulated biological pathways. Our study elucidates alternative drug delivery paradigms by comparing formulation strategies across oral (PO), intraperitoneal (IP), subcutaneous (SC), and intravenous (IV) routes. While each modality boasts its own set of merits and constraints, it is the drug's formulation that crucially influences its pharmacokinetic (PK) trajectory and the maintenance of its therapeutic levels. Our examination of model compounds G7883 and G6893 highlighted their distinct physio-chemical attributes. By harnessing varied formulation methods, we sought to fine-tune their PK profiles. PK studies showcased G7883's extended half-life using an SC oil formulation, resulting in a 4.5-fold and 2.5-fold enhancement compared with the IP and PO routes, respectively. In contrast, with G6893, we achieved a prolonged systemic coverage time above the desired target concentration through a different approach using an IV infusion pump. These outcomes underscore the need for tailored formulation strategies, which are dictated by the compound's innate properties, to reach the optimal in vivo systemic concentrations. Prioritizing formulation and delivery optimization early on is pivotal for effective systemic uptake, thereby facilitating a deeper understanding of biological pathways and expediting the overall clinical drug development timeline.

3.
ACS Omega ; 8(48): 45653-45667, 2023 Dec 05.
Article in English | MEDLINE | ID: mdl-38075834

ABSTRACT

A dimer of Pd(II), [(bpy)Pd(µ-OH)2Pd(bpy)]2+, (complex 1) (where bpy = 2,2'-bipyridyl) has been synthesized at physiological pH (7.4) and characterized by electronic spectroscopy, electrospray ionization mass spectrometry (ESI-MS) spectroscopy, and Fourier transform infrared (FT-IR) analysis. Reaction kinetics of 1 with glycine (L1H), l-glutamic acid (L2H), and l-arginine (L3H) were investigated in an aqueous medium at pH of 7.4 and constant ionic strength via a spectrophotometer as a function of temperature and different concentrations of substrate-complex and ligand. The interactions were supported by two discrete successive steps, i.e., ligand-dependent and ligand-independent steps. The equilibrium constant of complex formation (outer-sphere association) and the rate constant during complex-substrate-ligand interaction were calculated. The Eyring equation was applied to evaluate activation factors (ΔH‡ and ΔS‡), and associative mechanisms of all reactions were proposed. Thermodynamic parameters (ΔH° and ΔS°) were also estimated from the standard plot of ln KE against 103/T. Spectroscopic titration of 1 at pH 7.4 in Tris-HCl buffer with calf thymus DNA, electronic emission titration with ethidium bromide (EtBr), antimicrobial activities, and an agarose gel electrophoresis run of 1 on pBR322 plasmid DNA have shown strong evidence of anticancer activity. Moreover, it has nontoxic water molecules as leaving groups.

4.
Nat Commun ; 14(1): 5923, 2023 09 22.
Article in English | MEDLINE | ID: mdl-37740028

ABSTRACT

Treatment of osteoporosis commonly diminishes osteoclast number which suppresses bone formation thus compromising fracture prevention. Bone formation is not suppressed, however, when bone degradation is reduced by retarding osteoclast functional resorptive capacity, rather than differentiation. We find deletion of deubiquitinase, BRCA1-associated protein 1 (Bap1), in myeloid cells (Bap1∆LysM), arrests osteoclast function but not formation. Bap1∆LysM osteoclasts fail to organize their cytoskeleton which is essential for bone degradation consequently increasing bone mass in both male and female mice. The deubiquitinase activity of BAP1 modifies osteoclast function by metabolic reprogramming. Bap1 deficient osteoclast upregulate the cystine transporter, Slc7a11, by enhanced H2Aub occupancy of its promoter. SLC7A11 controls cellular reactive oxygen species levels and redirects the mitochondrial metabolites away from the tricarboxylic acid cycle, both being necessary for osteoclast function. Thus, in osteoclasts BAP1 appears to regulate the epigenetic-metabolic axis and is a potential target to reduce bone degradation while maintaining osteogenesis in osteoporotic patients.


Subject(s)
Osteoclasts , Osteogenesis , Animals , Female , Humans , Male , Mice , Bone Density , Citric Acid Cycle , Deubiquitinating Enzymes , Osteogenesis/genetics , Tumor Suppressor Proteins/genetics , Ubiquitin Thiolesterase/genetics
5.
Nat Cancer ; 4(6): 812-828, 2023 06.
Article in English | MEDLINE | ID: mdl-37277530

ABSTRACT

The Hippo pathway is a key growth control pathway that is conserved across species. The downstream effectors of the Hippo pathway, YAP (Yes-associated protein) and TAZ (transcriptional coactivator with PDZ-binding motif), are frequently activated in cancers to drive proliferation and survival. Based on the premise that sustained interactions between YAP/TAZ and TEADs (transcriptional enhanced associate domain) are central to their transcriptional activities, we discovered a potent small-molecule inhibitor (SMI), GNE-7883, that allosterically blocks the interactions between YAP/TAZ and all human TEAD paralogs through binding to the TEAD lipid pocket. GNE-7883 effectively reduces chromatin accessibility specifically at TEAD motifs, suppresses cell proliferation in a variety of cell line models and achieves strong antitumor efficacy in vivo. Furthermore, we uncovered that GNE-7883 effectively overcomes both intrinsic and acquired resistance to KRAS (Kirsten rat sarcoma viral oncogene homolog) G12C inhibitors in diverse preclinical models through the inhibition of YAP/TAZ activation. Taken together, this work demonstrates the activities of TEAD SMIs in YAP/TAZ-dependent cancers and highlights their potential broad applications in precision oncology and therapy resistance.


Subject(s)
Neoplasms , Proto-Oncogene Proteins p21(ras) , Humans , Proto-Oncogene Proteins p21(ras)/genetics , Precision Medicine , Transcription Factors/metabolism , Signal Transduction
6.
Mol Cell Proteomics ; 22(2): 100496, 2023 02.
Article in English | MEDLINE | ID: mdl-36640924

ABSTRACT

Transcriptional enhanced associate domain family members 1 to 4 (TEADs) are a family of four transcription factors and the major transcriptional effectors of the Hippo pathway. In order to activate transcription, TEADs rely on interactions with other proteins, such as the transcriptional effectors Yes-associated protein and transcriptional co-activator with PDZ-binding motif. Nuclear protein interactions involving TEADs influence the transcriptional regulation of genes involved in cell growth, tissue homeostasis, and tumorigenesis. Clearly, protein interactions for TEADs are functionally important, but the full repertoire of TEAD interaction partners remains unknown. Here, we employed an affinity purification mass spectrometry approach to identify nuclear interacting partners of TEADs. We performed affinity purification mass spectrometry experiment in parallel in two different cell types and compared a wildtype TEAD bait protein to a nuclear localization sequence mutant that does not localize to the nucleus. We quantified the results using SAINT analysis and found a significant enrichment of proteins linked to DNA damage including X-ray repair cross-complementing protein 5 (XRCC5), X-ray repair cross-complementing protein 6 (XRCC6), poly(ADP-ribose) polymerase 1 (PARP1), and Rap1-interacting factor 1 (RIF1). In cellular assays, we found that TEADs co-localize with DNA damage-induced nuclear foci marked by histone H2AX phosphorylated on S139 (γH2AX) and Rap1-interacting factor 1. We also found that depletion of TEAD proteins makes cells more susceptible to DNA damage by various agents and that depletion of TEADs promotes genomic instability. Additionally, depleting TEADs dampens the efficiency of DNA double-stranded break repair in reporter assays. Our results connect TEADs to DNA damage response processes, positioning DNA damage as an important avenue for further research of TEAD proteins.


Subject(s)
DNA Damage , DNA Repair , TEA Domain Transcription Factors , Humans , Carcinogenesis/metabolism , DNA Repair/physiology , DNA-Binding Proteins/metabolism , Transcription Factors/metabolism , TEA Domain Transcription Factors/metabolism
7.
Cells ; 11(14)2022 07 17.
Article in English | MEDLINE | ID: mdl-35883668

ABSTRACT

The Hippo pathway regulates tissue homeostasis in normal development and drives oncogenic processes. In this review, we extensively discuss how YAP/TAZ/TEAD cooperate with other master transcription factors and epigenetic cofactors to orchestrate a broad spectrum of transcriptional responses. Even though these responses are often context- and lineage-specific, we do not have a good understanding of how such precise and specific transcriptional control is achieved-whether they are driven by differences in TEAD paralogs, or recruitment of cofactors to tissue-specific enhancers. We believe that emerging single-cell technologies would enable a granular understanding of how the Hippo pathway influences cell fate and drives oncogenic processes, ultimately allowing us to design better pharmacological agents against TEADs and identify robust pharmacodynamics markers of Hippo pathway inhibition.


Subject(s)
Hippo Signaling Pathway , Protein Serine-Threonine Kinases , Single-Cell Analysis , Carcinogenesis , Gene Expression Regulation , Hippo Signaling Pathway/genetics , Humans , Protein Serine-Threonine Kinases/genetics , Signal Transduction , Transcription Factors/metabolism
8.
Adv Exp Med Biol ; 1358: 141-163, 2022.
Article in English | MEDLINE | ID: mdl-35641869

ABSTRACT

Male infertility is one of the major challenging and prevalent diseases having diverse etiologies of which bacteriospermia play a significant role. It has been estimated that approximately 15% of all infertility cases are due to infections caused by uropathogens and in most of the cases bacteria are involved in infection and inflammation leading to the development of bacteriospermia. In response to bacterial load, excess infiltration of leukocytes in the urogenital tract occurs and concomitantly generates oxidative stress (OS). Bacteria may induce infertility either by directly interacting with sperm or by generating reactive oxygen species (ROS) and impair sperm parameters such as motility, volume, capacitation, hyperactivation. They may also induce apoptosis leading to sperm death. Acute bacteriospermia is related with another clinical condition called leukocytospermia and both compromise male fertility potential by OS-mediated damage to sperm leading to male infertility. However, bacteriospermia as a clinical condition as well as the mechanism of action remains poorly understood, necessitating further research in order to understand the role of individual bacterial species and their impact in male infertility.


Subject(s)
Infertility, Male , Antioxidants/pharmacology , Humans , Infertility, Male/etiology , Infertility, Male/metabolism , Male , Oxidative Stress , Reactive Oxygen Species/metabolism , Spermatozoa/physiology
9.
Cancer Discov ; 12(1): 204-219, 2022 01.
Article in English | MEDLINE | ID: mdl-34544753

ABSTRACT

PIK3CA is one of the most frequently mutated oncogenes; the p110a protein it encodes plays a central role in tumor cell proliferation. Small-molecule inhibitors targeting the PI3K p110a catalytic subunit have entered clinical trials, with early-phase GDC-0077 studies showing antitumor activity and a manageable safety profile in patients with PIK3CA-mutant breast cancer. However, preclinical studies have shown that PI3K pathway inhibition releases negative feedback and activates receptor tyrosine kinase signaling, reengaging the pathway and attenuating drug activity. Here we discover that GDC-0077 and taselisib more potently inhibit mutant PI3K pathway signaling and cell viability through unique HER2-dependent mutant p110a degradation. Both are more effective than other PI3K inhibitors at maintaining prolonged pathway suppression. This study establishes a new strategy for identifying inhibitors that specifically target mutant tumors by selective degradation of the mutant oncoprotein and provide a strong rationale for pursuing PI3Kα degraders in patients with HER2-positive breast cancer. SIGNIFICANCE: The PI3K inhibitors GDC-0077 and taselisib have a unique mechanism of action; both inhibitors lead to degradation of mutant p110a protein. The inhibitors that have the ability to trigger specific degradation of mutant p110a without significant change in wild-type p110a protein may result in improved therapeutic index in PIK3CA-mutant tumors.See related commentary by Vanhaesebroeck et al., p. 20.This article is highlighted in the In This Issue feature, p. 1.


Subject(s)
Antineoplastic Agents , Breast Neoplasms , Class I Phosphatidylinositol 3-Kinases , Imidazoles , Oxazepines , Phosphoinositide-3 Kinase Inhibitors , Receptor, ErbB-2 , Female , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Line, Tumor/drug effects , Class I Phosphatidylinositol 3-Kinases/genetics , Imidazoles/pharmacology , Imidazoles/therapeutic use , Oxazepines/pharmacology , Oxazepines/therapeutic use , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Phosphoinositide-3 Kinase Inhibitors/therapeutic use , Receptor, ErbB-2/genetics
10.
Cancer Discov ; 11(3): 778-793, 2021 03.
Article in English | MEDLINE | ID: mdl-33208393

ABSTRACT

Hippo pathway dysregulation occurs in multiple cancers through genetic and nongenetic alterations, resulting in translocation of YAP to the nucleus and activation of the TEAD family of transcription factors. Unlike other oncogenic pathways such as RAS, defining tumors that are Hippo pathway-dependent is far more complex due to the lack of hotspot genetic alterations. Here, we developed a machine-learning framework to identify a robust, cancer type-agnostic gene expression signature to quantitate Hippo pathway activity and cross-talk as well as predict YAP/TEAD dependency across cancers. Further, through chemical genetic interaction screens and multiomics analyses, we discover a direct interaction between MAPK signaling and TEAD stability such that knockdown of YAP combined with MEK inhibition results in robust inhibition of tumor cell growth in Hippo dysregulated tumors. This multifaceted approach underscores how computational models combined with experimental studies can inform precision medicine approaches including predictive diagnostics and combination strategies. SIGNIFICANCE: An integrated chemicogenomics strategy was developed to identify a lineage-independent signature for the Hippo pathway in cancers. Evaluating transcriptional profiles using a machine-learning method led to identification of a relationship between YAP/TAZ dependency and MAPK pathway activity. The results help to nominate potential combination therapies with Hippo pathway inhibition.This article is highlighted in the In This Issue feature, p. 521.


Subject(s)
Cheminformatics/methods , Computational Biology/methods , Genomics/methods , Hippo Signaling Pathway , MAP Kinase Signaling System , Machine Learning , Signal Transduction , Humans
11.
J Am Soc Nephrol ; 31(10): 2341-2354, 2020 10.
Article in English | MEDLINE | ID: mdl-32651223

ABSTRACT

BACKGROUND: The glomerulus is a specialized capillary bed that is involved in urine production and BP control. Glomerular injury is a major cause of CKD, which is epidemic and without therapeutic options. Single-cell transcriptomics has radically improved our ability to characterize complex organs, such as the kidney. Cells of the glomerulus, however, have been largely underrepresented in previous single-cell kidney studies due to their paucity and intractability. METHODS: Single-cell RNA sequencing comprehensively characterized the types of cells in the glomerulus from healthy mice and from four different disease models (nephrotoxic serum nephritis, diabetes, doxorubicin toxicity, and CD2AP deficiency). RESULTS: All cell types in the glomerulus were identified using unsupervised clustering analysis. Novel marker genes and gene signatures of mesangial cells, vascular smooth muscle cells of the afferent and efferent arterioles, parietal epithelial cells, and three types of endothelial cells were identified. Analysis of the disease models revealed cell type-specific and injury type-specific responses in the glomerulus, including acute activation of the Hippo pathway in podocytes after nephrotoxic immune injury. Conditional deletion of YAP or TAZ resulted in more severe and prolonged proteinuria in response to injury, as well as worse glomerulosclerosis. CONCLUSIONS: Generation of comprehensive high-resolution, single-cell transcriptomic profiles of the glomerulus from healthy and injured mice provides resources to identify novel disease-related genes and pathways.


Subject(s)
Kidney Diseases/etiology , Kidney Glomerulus/pathology , Animals , Disease Models, Animal , Gene Expression Profiling , Kidney Diseases/metabolism , Kidney Diseases/pathology , Kidney Glomerulus/metabolism , Mesangial Cells/pathology , Mice , Mice, Inbred C57BL , Podocytes/pathology
12.
Cancer Discov ; 10(8): 1103-1120, 2020 08.
Article in English | MEDLINE | ID: mdl-32690542

ABSTRACT

Among more than 200 BAP1-mutant families affected by the "BAP1 cancer syndrome," nearly all individuals inheriting a BAP1 mutant allele developed one or more malignancies during their lifetime, mostly uveal and cutaneous melanoma, mesothelioma, and clear-cell renal cell carcinoma. These cancer types are also those that, when they occur sporadically, are more likely to carry somatic biallelic BAP1 mutations. Mechanistic studies revealed that the tumor suppressor function of BAP1 is linked to its dual activity in the nucleus, where it is implicated in a variety of processes including DNA repair and transcription, and in the cytoplasm, where it regulates cell death and mitochondrial metabolism. BAP1 activity in tumor suppression is cell type- and context-dependent. BAP1 has emerged as a critical tumor suppressor across multiple cancer types, predisposing to tumor development when mutated in the germline as well as somatically. Moreover, BAP1 has emerged as a key regulator of gene-environment interaction.This article is highlighted in the In This Issue feature, p. 1079.


Subject(s)
Kidney Neoplasms , Melanoma , Skin Neoplasms , Uveal Neoplasms , Humans , Mutation , Tumor Suppressor Proteins/genetics , Ubiquitin Thiolesterase/genetics
13.
Nat Rev Drug Discov ; 19(7): 480-494, 2020 07.
Article in English | MEDLINE | ID: mdl-32555376

ABSTRACT

The Hippo pathway is an evolutionarily conserved signalling pathway with key roles in organ development, epithelial homeostasis, tissue regeneration, wound healing and immune modulation. Many of these roles are mediated by the transcriptional effectors YAP and TAZ, which direct gene expression via control of the TEAD family of transcription factors. Dysregulated Hippo pathway and YAP/TAZ-TEAD activity is associated with various diseases, most notably cancer, making this pathway an attractive target for therapeutic intervention. This Review highlights the key findings from studies of Hippo pathway signalling across biological processes and diseases, and discusses new strategies and therapeutic implications of targeting this pathway.


Subject(s)
Neoplasms/therapy , Protein Serine-Threonine Kinases/metabolism , Regenerative Medicine/methods , Acyltransferases , Adaptor Proteins, Signal Transducing/genetics , Animals , DNA-Binding Proteins/genetics , Fibrosis/genetics , Fibrosis/pathology , Fibrosis/therapy , Gene Expression Regulation , Hippo Signaling Pathway , Humans , Neoplasms/genetics , Neoplasms/pathology , Nuclear Proteins/genetics , Signal Transduction , TEA Domain Transcription Factors , Transcription Factors/genetics , Wound Healing/physiology , YAP-Signaling Proteins
14.
Trends Cancer ; 6(9): 781-796, 2020 09.
Article in English | MEDLINE | ID: mdl-32446746

ABSTRACT

The Hippo pathway regulates myriad biological processes in diverse species and is a key cancer signaling network in humans. Although Hippo has been linked to multiple aspects of cancer, its role in this disease is incompletely understood. Large-scale pan-cancer analyses of core Hippo pathway genes reveal that the pathway is mutated at a high frequency only in select human cancers, including malignant mesothelioma and meningioma. Hippo pathway deregulation is also enriched in squamous epithelial cancers. We discuss cancer-related functions of the Hippo pathway and potential explanations for the cancer-restricted mutation profile of core Hippo pathway genes. Greater understanding of Hippo pathway deregulation in cancers will be essential to guide the imminent use of Hippo-targeted therapies.


Subject(s)
Antineoplastic Agents/pharmacology , Biomarkers, Tumor/genetics , Neoplasms/genetics , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/genetics , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/metabolism , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Competition/genetics , Cell Differentiation/genetics , Cell Proliferation/genetics , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Hippo Signaling Pathway , Humans , Molecular Targeted Therapy/methods , Mutation , Neoplasms/drug therapy , Neoplasms/pathology , Precision Medicine/methods , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Signal Transduction/drug effects , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/antagonists & inhibitors , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/genetics , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/metabolism , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Ubiquitin Thiolesterase/antagonists & inhibitors , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism
15.
J Clin Invest ; 130(5): 2644-2656, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32310225

ABSTRACT

We previously established that global deletion of the enhancer of trithorax and polycomb (ETP) gene, Asxl2, prevents weight gain. Because proinflammatory macrophages recruited to adipose tissue are central to the metabolic complications of obesity, we explored the role of ASXL2 in myeloid lineage cells. Unexpectedly, mice without Asxl2 only in myeloid cells (Asxl2ΔLysM) were completely resistant to diet-induced weight gain and metabolically normal despite increased food intake, comparable activity, and equivalent fecal fat. Asxl2ΔLysM mice resisted HFD-induced adipose tissue macrophage infiltration and inflammatory cytokine gene expression. Energy expenditure and brown adipose tissue metabolism in Asxl2ΔLysM mice were protected from the suppressive effects of HFD, a phenomenon associated with relatively increased catecholamines likely due to their suppressed degradation by macrophages. White adipose tissue of HFD-fed Asxl2ΔLysM mice also exhibited none of the pathological remodeling extant in their control counterparts. Suppression of macrophage Asxl2 expression, via nanoparticle-based siRNA delivery, prevented HFD-induced obesity. Thus, ASXL2 controlled the response of macrophages to dietary factors to regulate metabolic homeostasis, suggesting modulation of the cells' inflammatory phenotype may impact obesity and its complications.


Subject(s)
Energy Metabolism , Myeloid Cells/metabolism , Obesity/prevention & control , Repressor Proteins/deficiency , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/pathology , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Animals , Diet, High-Fat/adverse effects , Female , Gene Knockdown Techniques , Inflammation/metabolism , Inflammation/pathology , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/pathology , Obesity/metabolism , Obesity/pathology , Organ Specificity , RNA, Small Interfering/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Weight Gain/genetics , Weight Gain/physiology
16.
Cancer Res ; 80(8): 1656-1668, 2020 04 15.
Article in English | MEDLINE | ID: mdl-31988076

ABSTRACT

The deubiquitinating enzyme BAP1 is mutated in a hereditary cancer syndrome with a high risk for mesothelioma and melanocytic tumors. Here, we show that pancreatic intraepithelial neoplasia driven by oncogenic mutant KrasG12D progressed to pancreatic adenocarcinoma in the absence of BAP1. The Hippo pathway was deregulated in BAP1-deficient pancreatic tumors, with the tumor suppressor LATS exhibiting enhanced ubiquitin-dependent proteasomal degradation. Therefore, BAP1 may limit tumor progression by stabilizing LATS and thereby promoting activity of the Hippo tumor suppressor pathway. SIGNIFICANCE: BAP1 is mutated in a broad spectrum of tumors. Pancreatic Bap1 deficiency causes acinar atrophy but combines with oncogenic Ras to produce pancreatic tumors. BAP1-deficient tumors exhibit deregulation of the Hippo pathway.See related commentary by Brekken, p. 1624.


Subject(s)
Adenocarcinoma , Pancreatic Neoplasms , Hippo Signaling Pathway , Humans , Protein Serine-Threonine Kinases , Signal Transduction , Tumor Suppressor Proteins , Ubiquitin Thiolesterase
17.
Front Cell Dev Biol ; 7: 156, 2019.
Article in English | MEDLINE | ID: mdl-31475147

ABSTRACT

The Hippo pathway is a critical regulator of cell and organ growth and has emerged as a target for therapeutic intervention in cancers. Its signaling is thought to play an important role in various physiological processes including homeostasis and tissue regeneration. To date there has been limited information about potential pharmacology-related (on-target) safety liabilities of Hippo pathway inhibitors in the context of cancer indications. Herein, we review data from human genetic disorders and genetically engineered rodent models to gain insight into safety liabilities that may emerge from the inhibition of Hippo pathway. Germline systemic deletion of murine Hippo pathway effectors (Yap, Taz, and Teads) resulted in embryonic lethality or developmental phenotypes. Mouse models with tissue-specific deletion (or mutant overexpression) of the key effectors in Hippo pathways have indicated that, at least in some tissues, Hippo signaling may be dispensable for physiological homeostasis; and appears to be critical for regeneration upon tissue damage, indicating that patients with underlying comorbidities and/or insults caused by therapeutic agents and/or comedications may have a higher risk. Caution should be taken in interpreting phenotypes from tissue-specific transgenic animal models since some tissue-specific promoters are turned on during development. In addition, therapeutic agents may result in systemic effects not well-predicted by animal models with tissue-specific gene deletion. Therefore, the development of models that allows for systemic deletion of Yap and/or Taz in adult animals will be key in evaluating the potential safety liabilities of Hippo pathway modulation. In this review, we focus on potential challenges and strategies for targeting the Hippo pathway in cancers.

18.
JCI Insight ; 52019 06 18.
Article in English | MEDLINE | ID: mdl-31211697

ABSTRACT

The lung is a relatively quiescent organ during homeostasis, but has a remarkable capacity for repair after injury. Alveolar epithelial type I cells (AEC1s) line airspaces and mediate gas exchange. After injury, they are regenerated by differentiation from their progenitors - alveolar epithelial type II cells (AEC2s) - which also secrete surfactant to maintain surface tension and alveolar patency. While recent studies showed that the maintenance of AEC2 stemness is Wnt dependent, the molecular mechanisms underlying AEC2-AEC1 differentiation in adult lung repair are still incompletely understood. Here we show that WWTR1 (TAZ) plays a crucial role in AEC differentiation. Using an in vitro organoid culture system, we found that tankyrase inhibition can efficiently block AEC2-AEC1 differentiation, and this effect was due to the inhibition of TAZ. In a bleomycin induced lung injury model, conditional deletion of TAZ in AEC2s dramatically reduced AEC1 regeneration during recovery, leading to exacerbated alveolar lesions and fibrosis. In patients with idiopathic pulmonary fibrosis (IPF), decreased blood levels of RAGE, a biomarker of AEC1 health, were associated with more rapid disease progression. Our findings implicate TAZ as a critical factor involved in AEC2 to AEC1 differentiation, and hence the maintenance of alveolar integrity after injury.


Subject(s)
Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/metabolism , Cell Differentiation/drug effects , Cell Differentiation/physiology , Lung Injury/metabolism , Trans-Activators/metabolism , Trans-Activators/pharmacology , Adaptor Proteins, Signal Transducing , Animals , Bleomycin/adverse effects , Disease Models, Animal , Humans , Idiopathic Pulmonary Fibrosis/pathology , Lung/pathology , Lung Injury/chemically induced , Lung Injury/pathology , Mice, Inbred C57BL , Mice, Knockout , Organogenesis/drug effects , Organogenesis/physiology , Organoids/metabolism , Regeneration/physiology , Stem Cells/metabolism , Trans-Activators/genetics , Transcriptome , beta Catenin/genetics , beta Catenin/metabolism
19.
Trends Cancer ; 5(5): 297-307, 2019 05.
Article in English | MEDLINE | ID: mdl-31174842

ABSTRACT

The Hippo pathway remains a central focus in both basic and translational research and is a key modulator of developmental biology. Dysregulation of the pathway is associated with a plethora of human cancers and there are multiple efforts to target key components of the pathway for disease intervention. In this review, we briefly highlight the latest research advances around the core components of the Hippo pathway in cancer. More specifically, we discuss several genetic aberrations of these factors as mechanisms for the development of cancers, including genetic amplification, deletion, and gene fusions. Additionally, we highlight the role of the Hippo pathway in cancer therapy resistance and tumor immunogenicity. Last, we summarize the ongoing efforts to target the pathway in cancers.


Subject(s)
Neoplasms/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Biomarkers , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Drug Discovery , Gene Expression Regulation, Neoplastic , Hippo Signaling Pathway , Humans , Neoplasms/etiology , Neoplasms/pathology , Neoplasms/therapy , Protein Serine-Threonine Kinases/genetics , Signal Transduction/drug effects
20.
Science ; 364(6437): 283-285, 2019 Apr 19.
Article in English | MEDLINE | ID: mdl-31000662

ABSTRACT

Malignancies arising from mutation of tumor suppressors have unexplained tissue proclivity. For example, BAP1 encodes a widely expressed deubiquitinase for histone H2A, but germline mutations are predominantly associated with uveal melanomas and mesotheliomas. We show that BAP1 inactivation causes apoptosis in mouse embryonic stem cells, fibroblasts, liver, and pancreatic tissue but not in melanocytes and mesothelial cells. Ubiquitin ligase RNF2, which silences genes by monoubiquitinating H2A, promoted apoptosis in BAP1-deficient cells by suppressing expression of the prosurvival genes Bcl2 and Mcl1. In contrast, BAP1 loss in melanocytes had little impact on expression of prosurvival genes, instead inducing Mitf Thus, BAP1 appears to modulate gene expression by countering H2A ubiquitination, but its loss only promotes tumorigenesis in cells that do not engage an RNF2-dependent apoptotic program.


Subject(s)
Apoptosis/genetics , Carcinogenesis/genetics , Gene Expression Regulation, Neoplastic , Melanoma/genetics , Polycomb Repressive Complex 1/metabolism , Tumor Suppressor Proteins/genetics , Ubiquitin Thiolesterase/genetics , Ubiquitin-Protein Ligases/metabolism , Uveal Neoplasms/genetics , Animals , Gene Knock-In Techniques , Germ-Line Mutation , Histones , Humans , Melanocytes/metabolism , Melanocytes/pathology , Melanoma/pathology , Mesothelioma/genetics , Mesothelioma/pathology , Mice , Mice, Mutant Strains , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Ubiquitination , Uveal Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...