Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 98
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 181(4): 848-864.e18, 2020 05 14.
Article in English | MEDLINE | ID: mdl-32298651

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is a progressive condition of chronic bronchitis, small airway obstruction, and emphysema that represents a leading cause of death worldwide. While inflammation, fibrosis, mucus hypersecretion, and metaplastic epithelial lesions are hallmarks of this disease, their origins and dependent relationships remain unclear. Here we apply single-cell cloning technologies to lung tissue of patients with and without COPD. Unlike control lungs, which were dominated by normal distal airway progenitor cells, COPD lungs were inundated by three variant progenitors epigenetically committed to distinct metaplastic lesions. When transplanted to immunodeficient mice, these variant clones induced pathology akin to the mucous and squamous metaplasia, neutrophilic inflammation, and fibrosis seen in COPD. Remarkably, similar variants pre-exist as minor constituents of control and fetal lung and conceivably act in normal processes of immune surveillance. However, these same variants likely catalyze the pathologic and progressive features of COPD when expanded to high numbers.


Subject(s)
Lung/pathology , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/metabolism , Adult , Aged , Animals , Female , Fibrosis/physiopathology , Humans , Inflammation/pathology , Lung/metabolism , Male , Metaplasia/physiopathology , Mice , Middle Aged , Neutrophils/immunology , Pneumonia/pathology , Pulmonary Disease, Chronic Obstructive/physiopathology , Single-Cell Analysis/methods , Stem Cells/metabolism
3.
Pharmacol Rev ; 76(3): 414-453, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38697854

ABSTRACT

Since its discovery over 35 years ago, MDM2 has emerged as an attractive target for the development of cancer therapy. MDM2's activities extend from carcinogenesis to immunity to the response to various cancer therapies. Since the report of the first MDM2 inhibitor more than 30 years ago, various approaches to inhibit MDM2 have been attempted, with hundreds of small-molecule inhibitors evaluated in preclinical studies and numerous molecules tested in clinical trials. Although many MDM2 inhibitors and degraders have been evaluated in clinical trials, there is currently no Food and Drug Administration (FDA)-approved MDM2 inhibitor on the market. Nevertheless, there are several current clinical trials of promising agents that may overcome the past failures, including agents granted FDA orphan drug or fast-track status. We herein summarize the research efforts to discover and develop MDM2 inhibitors, focusing on those that induce MDM2 degradation and exert anticancer activity, regardless of the p53 status of the cancer. We also describe how preclinical and clinical investigations have moved toward combining MDM2 inhibitors with other agents, including immune checkpoint inhibitors. Finally, we discuss the current challenges and future directions to accelerate the clinical application of MDM2 inhibitors. In conclusion, targeting MDM2 remains a promising treatment approach, and targeting MDM2 for protein degradation represents a novel strategy to downregulate MDM2 without the side effects of the existing agents blocking p53-MDM2 binding. Additional preclinical and clinical investigations are needed to finally realize the full potential of MDM2 inhibition in treating cancer and other chronic diseases where MDM2 has been implicated. SIGNIFICANCE STATEMENT: Overexpression/amplification of the MDM2 oncogene has been detected in various human cancers and is associated with disease progression, treatment resistance, and poor patient outcomes. This article reviews the previous, current, and emerging MDM2-targeted therapies and summarizes the preclinical and clinical studies combining MDM2 inhibitors with chemotherapy and immunotherapy regimens. The findings of these contemporary studies may lead to safer and more effective treatments for patients with cancers overexpressing MDM2.


Subject(s)
Antineoplastic Agents , Neoplasms , Proto-Oncogene Proteins c-mdm2 , Humans , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Proto-Oncogene Proteins c-mdm2/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Animals , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology , Molecular Targeted Therapy
4.
Cell ; 145(7): 1023-35, 2011 Jun 24.
Article in English | MEDLINE | ID: mdl-21703447

ABSTRACT

Barrett's esophagus is an intestine-like metaplasia and precursor of esophageal adenocarcinoma. Triggered by gastroesophageal reflux disease, the origin of this metaplasia remains unknown. p63-deficient mice, which lack squamous epithelia, may model acid-reflux damage. We show here that p63 null embryos rapidly develop intestine-like metaplasia with gene expression profiles similar to Barrett's metaplasia. We track its source to a unique embryonic epithelium that is normally undermined and replaced by p63-expressing cells. Significantly, we show that a discrete population of these embryonic cells persists in adult mice and humans at the squamocolumnar junction, the source of Barrett's metaplasia. We show that upon programmed damage to the squamous epithelium, these embryonic cells migrate toward adjacent, specialized squamous cells in a process that may recapitulate early Barrett's. Our findings suggest that certain precancerous lesions, such as Barrett's, initiate not from genetic alterations but from competitive interactions between cell lineages driven by opportunity.


Subject(s)
Barrett Esophagus/pathology , Esophagus/pathology , Animals , Barrett Esophagus/embryology , Gene Expression Profiling , Humans , Intestine, Small/cytology , Metaplasia , Mice , Phosphoproteins/genetics , Phosphoproteins/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism
5.
Cell ; 144(4): 566-76, 2011 Feb 18.
Article in English | MEDLINE | ID: mdl-21335238

ABSTRACT

TAp63α, a homolog of the p53 tumor suppressor, is a quality control factor in the female germline. Remarkably, already undamaged oocytes express high levels of the protein, suggesting that TAp63α's activity is under tight control of an inhibitory mechanism. Biochemical studies have proposed that inhibition requires the C-terminal transactivation inhibitory domain. However, the structural mechanism of TAp63α inhibition remains unknown. Here, we show that TAp63α is kept in an inactive dimeric state. We reveal that relief of inhibition leads to tetramer formation with ∼20-fold higher DNA affinity. In vivo, phosphorylation-triggered tetramerization of TAp63α is not reversible by dephosphorylation. Furthermore, we show that a helix in the oligomerization domain of p63 is crucial for tetramer stabilization and competes with the transactivation domain for the same binding site. Our results demonstrate how TAp63α is inhibited by complex domain-domain interactions that provide the basis for regulating quality control in oocytes.


Subject(s)
Oocytes/metabolism , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Trans-Activators/chemistry , Trans-Activators/metabolism , Animals , DNA/metabolism , Dimerization , Female , Gamma Rays , Mice , Models, Molecular , Phosphorylation , Protein Multimerization , Tumor Suppressor Protein p53/metabolism
6.
Cell ; 147(3): 525-38, 2011 Oct 28.
Article in English | MEDLINE | ID: mdl-22036562

ABSTRACT

The extent of lung regeneration following catastrophic damage and the potential role of adult stem cells in such a process remains obscure. Sublethal infection of mice with an H1N1 influenza virus related to that of the 1918 pandemic triggers massive airway damage followed by apparent regeneration. We show here that p63-expressing stem cells in the bronchiolar epithelium undergo rapid proliferation after infection and radiate to interbronchiolar regions of alveolar ablation. Once there, these cells assemble into discrete, Krt5+ pods and initiate expression of markers typical of alveoli. Gene expression profiles of these pods suggest that they are intermediates in the reconstitution of the alveolar-capillary network eradicated by viral infection. The dynamics of this p63-expressing stem cell in lung regeneration mirrors our parallel finding that defined pedigrees of human distal airway stem cells assemble alveoli-like structures in vitro and suggests new therapeutic avenues to acute and chronic airway disease.


Subject(s)
Bronchi/cytology , Influenza A Virus, H1N1 Subtype , Influenza, Human/pathology , Lung/physiology , Pulmonary Alveoli/cytology , Respiratory Distress Syndrome/pathology , Stem Cells/cytology , Animals , Disease Models, Animal , Gene Expression Profiling , Humans , Lung/cytology , Lung/virology , Mice , Mice, Inbred C57BL , Pulmonary Alveoli/virology , Rats , Transcription Factors/genetics , Wound Healing
8.
Am J Respir Crit Care Med ; 208(9): 930-943, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37695863

ABSTRACT

Rationale: CFTR (cystic fibrosis transmembrane conductance regulator) modulator drugs restore function to mutant channels in patients with cystic fibrosis (CF) and lead to improvements in body mass index and lung function. Although it is anticipated that early childhood treatment with CFTR modulators will significantly delay or even prevent the onset of advanced lung disease, lung neutrophils and inflammatory cytokines remain high in patients with CF with established lung disease despite modulator therapy, underscoring the need to identify and ultimately target the sources of this inflammation in CF lungs. Objectives: To determine whether CF lungs, like chronic obstructive pulmonary disease (COPD) lungs, harbor potentially pathogenic stem cell "variants" distinct from the normal p63/Krt5 lung stem cells devoted to alveolar fates, to identify specific variants that might contribute to the inflammatory state of CF lungs, and to assess the impact of CFTR genetic complementation or CFTR modulators on the inflammatory variants identified herein. Methods: Stem cell cloning technology developed to resolve pathogenic stem cell heterogeneity in COPD and idiopathic pulmonary fibrosis lungs was applied to end-stage lungs of patients with CF (three homozygous CFTR:F508D, one CFTR F508D/L1254X; FEV1, 14-30%) undergoing therapeutic lung transplantation. Single-cell-derived clones corresponding to the six stem cell clusters resolved by single-cell RNA sequencing of these libraries were assessed by RNA sequencing and xenografting to monitor inflammation, fibrosis, and mucin secretion. The impact of CFTR activity on these variants after CFTR gene complementation or exposure to CFTR modulators was assessed by molecular and functional studies. Measurements and Main Results: End-stage CF lungs display a stem cell heterogeneity marked by five predominant variants in addition to the normal lung stem cell, of which three are proinflammatory both at the level of gene expression and their ability to drive neutrophilic inflammation in xenografts in immunodeficient mice. The proinflammatory functions of these three variants were unallayed by genetic or pharmacological restoration of CFTR activity. Conclusions: The emergence of three proinflammatory stem cell variants in CF lungs may contribute to the persistence of lung inflammation in patients with CF with advanced disease undergoing CFTR modulator therapy.


Subject(s)
Cystic Fibrosis , Pulmonary Disease, Chronic Obstructive , Humans , Child, Preschool , Animals , Mice , Cystic Fibrosis/drug therapy , Cystic Fibrosis/genetics , Cystic Fibrosis/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Lung/pathology , Pulmonary Disease, Chronic Obstructive/pathology , Inflammation/metabolism
9.
Gut ; 72(4): 624-637, 2023 04.
Article in English | MEDLINE | ID: mdl-36002248

ABSTRACT

OBJECTIVE: Many cancers engage embryonic genes for rapid growth and evading the immune system. SOX9 has been upregulated in many tumours, yet the role of SOX9 in mediating immunosuppressive tumour microenvironment is unclear. Here, we aim to dissect the role of SOX9-mediated cancer stemness attributes and immunosuppressive microenvironment in advanced gastric adenocarcinoma (GAC) for novel therapeutic discoveries. METHODS: Bulk RNAseq/scRNA-seq, patient-derived cells/models and extensive functional studies were used to identify the expression and functions of SOX9 and its target genes in vitro and in vivo. Immune responses were studied in PBMCs or CD45+ immune cells cocultured with tumour cells with SOX9high or knockout and the KP-Luc2 syngeneic models were used for efficacy of combinations. RESULTS: SOX9 is one of the most upregulated SOX genes in GAC and highly expressed in primary and metastatic tissues and associated with poor prognosis. Depletion of SOX9 in patient-derived GAC cells significantly decreased cancer stemness attributes, tumour formation and metastases and consistently increased CD8+ T cell responses when cocultured with PBMCs/CD45+ cells from GAC patients. RNA sequencing identified the leukaemia inhibitory factor (LIF) as the top secreted molecule regulated by SOX9 in tumour cells and was enriched in malignant ascites and mediated SOX9-induced M2 macrophage repolarisation and inhibited T cell function. CONCLUSION: Epithelial SOX9 is critical in suppressing CD8+ T cell responses and modified macrophage function in GAC through the paracrine LIF factor. Cotargeting LIF/LIFR and CSF1R has great potential in targeting SOX9-mediated cancer stemness, T cell immunosuppression and metastases suggesting the novel combination therapy against advanced GAC.


Subject(s)
Adenocarcinoma , Stomach Neoplasms , Humans , Tumor Microenvironment , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Immunosuppressive Agents , Immunosuppression Therapy , SOX9 Transcription Factor/genetics
10.
Nat Rev Mol Cell Biol ; 12(4): 259-65, 2011 04.
Article in English | MEDLINE | ID: mdl-21427767

ABSTRACT

The p53 family of proteins consists of p53, p63 and p73, which are transcription factors that affect both cancer and development. It is now emerging that these proteins also regulate maternal reproduction. Whereas p63 is important for maturation of the egg, p73 ensures normal mitosis in the developing blastocyst. p53 subsequently regulates implantation of the embryo through transcriptional control of leukaemia inhibitory factor. Elucidating the cell biological basis of how these factors regulate female fertility may lead to new approaches to the control of human maternal reproduction.


Subject(s)
DNA-Binding Proteins/physiology , Nuclear Proteins/physiology , Trans-Activators/physiology , Tumor Suppressor Protein p53/physiology , Tumor Suppressor Proteins/physiology , Animals , Blastocyst/cytology , Blastocyst/physiology , DNA-Binding Proteins/genetics , Female , Fertility/genetics , Fertility/physiology , Humans , Male , Mice , Mice, Knockout , Models, Biological , Nuclear Proteins/genetics , Oocytes/cytology , Oocytes/physiology , Phosphoproteins/genetics , Phosphoproteins/physiology , Reproduction/genetics , Reproduction/physiology , Trans-Activators/genetics , Transcription Factors , Tumor Protein p73 , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/genetics
11.
Med Res Rev ; 41(2): 961-1021, 2021 03.
Article in English | MEDLINE | ID: mdl-33155698

ABSTRACT

There is an urgent need to identify novel therapies for childhood cancers. Neuroblastoma is the most common pediatric solid tumor, and accounts for ~15% of childhood cancer-related mortality. Neuroblastomas exhibit genetic, morphological and clinical heterogeneity, which limits the efficacy of existing treatment modalities. Gaining detailed knowledge of the molecular signatures and genetic variations involved in the pathogenesis of neuroblastoma is necessary to develop safer and more effective treatments for this devastating disease. Recent studies with advanced high-throughput "omics" techniques have revealed numerous genetic/genomic alterations and dysfunctional pathways that drive the onset, growth, progression, and resistance of neuroblastoma to therapy. A variety of molecular signatures are being evaluated to better understand the disease, with many of them being used as targets to develop new treatments for neuroblastoma patients. In this review, we have summarized the contemporary understanding of the molecular pathways and genetic aberrations, such as those in MYCN, BIRC5, PHOX2B, and LIN28B, involved in the pathogenesis of neuroblastoma, and provide a comprehensive overview of the molecular targeted therapies under preclinical and clinical investigations, particularly those targeting ALK signaling, MDM2, PI3K/Akt/mTOR and RAS-MAPK pathways, as well as epigenetic regulators. We also give insights on the use of combination therapies involving novel agents that target various pathways. Further, we discuss the future directions that would help identify novel targets and therapeutics and improve the currently available therapies, enhancing the treatment outcomes and survival of patients with neuroblastoma.


Subject(s)
Molecular Targeted Therapy , Neuroblastoma , Child , Humans , Neuroblastoma/drug therapy , Neuroblastoma/genetics , Signal Transduction
12.
Nature ; 517(7536): 616-20, 2015 Jan 29.
Article in English | MEDLINE | ID: mdl-25383540

ABSTRACT

Lung diseases such as chronic obstructive pulmonary disease and pulmonary fibrosis involve the progressive and inexorable destruction of oxygen exchange surfaces and airways, and have emerged as a leading cause of death worldwide. Mitigating therapies, aside from impractical organ transplantation, remain limited and the possibility of regenerative medicine has lacked empirical support. However, it is clinically known that patients who survive sudden, massive loss of lung tissue from necrotizing pneumonia or acute respiratory distress syndrome often recover full pulmonary function within six months. Correspondingly, we recently demonstrated lung regeneration in mice following H1N1 influenza virus infection, and linked distal airway stem cells expressing Trp63 (p63) and keratin 5, called DASC(p63/Krt5), to this process. Here we show that pre-existing, intrinsically committed DASC(p63/Krt5) undergo a proliferative expansion in response to influenza-induced lung damage, and assemble into nascent alveoli at sites of interstitial lung inflammation. We also show that the selective ablation of DASC(p63/Krt5) in vivo prevents this regeneration, leading to pre-fibrotic lesions and deficient oxygen exchange. Finally, we demonstrate that single DASC(p63/Krt5)-derived pedigrees differentiate to type I and type II pneumocytes as well as bronchiolar secretory cells following transplantation to infected lung and also minimize the structural consequences of endogenous stem cell loss on this process. The ability to propagate these cells in culture while maintaining their intrinsic lineage commitment suggests their potential in stem cell-based therapies for acute and chronic lung diseases.


Subject(s)
Keratin-5/metabolism , Lung/cytology , Lung/physiology , Phosphoproteins/metabolism , Regeneration , Stem Cells/cytology , Stem Cells/metabolism , Trans-Activators/metabolism , Animals , Bronchioles/cytology , Bronchioles/virology , Cell Differentiation , Cell Lineage , Cell Proliferation , Dogs , Humans , Influenza A Virus, H1N1 Subtype/pathogenicity , Lung/pathology , Lung/virology , Madin Darby Canine Kidney Cells , Mice , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology , Oxygen/metabolism , Pedigree , Pneumonia/metabolism , Pneumonia/pathology , Pneumonia/virology , Pulmonary Alveoli/cytology , Pulmonary Alveoli/pathology , Pulmonary Alveoli/virology , Re-Epithelialization , Stem Cell Transplantation
13.
Nature ; 522(7555): 173-8, 2015 Jun 11.
Article in English | MEDLINE | ID: mdl-26040716

ABSTRACT

Stem cells of the gastrointestinal tract, pancreas, liver and other columnar epithelia collectively resist cloning in their elemental states. Here we demonstrate the cloning and propagation of highly clonogenic, 'ground state' stem cells of the human intestine and colon. We show that derived stem-cell pedigrees sustain limited copy number and sequence variation despite extensive serial passaging and display exquisitely precise, cell-autonomous commitment to epithelial differentiation consistent with their origins along the intestinal tract. This developmentally patterned and epigenetically maintained commitment of stem cells is likely to enforce the functional specificity of the adult intestinal tract. Using clonally derived colonic epithelia, we show that toxins A or B of the enteric pathogen Clostridium difficile recapitulate the salient features of pseudomembranous colitis. The stability of the epigenetic commitment programs of these stem cells, coupled with their unlimited replicative expansion and maintained clonogenicity, suggests certain advantages for their use in disease modelling and regenerative medicine.


Subject(s)
Intestines/cytology , Stem Cells/cytology , Stem Cells/metabolism , Bacterial Toxins/pharmacology , Cell Differentiation/drug effects , Cell Lineage , Cells, Cultured , Clone Cells/cytology , Clone Cells/metabolism , Clostridioides difficile/physiology , Colon/cytology , Colon/drug effects , Enterocolitis, Pseudomembranous/microbiology , Enterocolitis, Pseudomembranous/pathology , Epigenesis, Genetic/genetics , Epithelium/drug effects , Epithelium/metabolism , Fetus/cytology , Genomic Instability/genetics , Humans , Intestine, Small/cytology , Intestines/drug effects , Organoids/cytology , Organoids/growth & development
14.
FASEB J ; 33(9): 10528-10537, 2019 09.
Article in English | MEDLINE | ID: mdl-31260634

ABSTRACT

The circadian clock is important for cellular and organ function. However, its function in sickle cell disease (SCD), a life-threatening hemolytic disorder, remains unknown. Here, we performed an unbiased microarray screen, which revealed significantly altered expression of circadian rhythmic genes, inflammatory response genes, and iron metabolic genes in SCD Berkeley transgenic mouse lungs compared with controls. Given the vital role of period 2 (Per2) in the core clock and the unrecognized role of Per2 in SCD, we transplanted the bone marrow (BM) of SCD mice to Per2Luciferase mice, which revealed that Per2 expression was up-regulated in SCD mouse lung. Next, we transplanted the BM of SCD mice to period 1 (Per1)/Per2 double deficient [Per1/Per2 double knockout (dKO)] and wild-type mice, respectively. We discovered that Per1/Per2 dKO mice transplanted with SCD BM (SCD → Per1/Per2 dKO) displayed severe irradiation sensitivity and were more susceptible to an early death. Although we observed an increase of peripheral inflammatory cells, we did not detect differences in erythrocyte sickling. However, there was further lung damage due to elevated pulmonary congestion, inflammatory cell infiltration, iron overload, and secretion of IL-6 in lavage fluid. Overall, we demonstrate that Per1/Per2 is beneficial to counteract elevated systemic inflammation, lung tissue inflammation, and iron overload in SCD.-Adebiyi, M. G., Zhao, Z., Ye, Y., Manalo, J., Hong, Y., Lee, C. C., Xian, W., McKeon, F., Culp-Hill, R., D' Alessandro, A., Kellems, R. E., Yoo, S.-H., Han, L., Xia, Y. Circadian period 2: a missing beneficial factor in sickle cell disease by lowering pulmonary inflammation, iron overload, and mortality.


Subject(s)
Anemia, Sickle Cell/mortality , Circadian Clocks , Circadian Rhythm/genetics , Iron Overload/mortality , Period Circadian Proteins/physiology , Pneumonia/mortality , Anemia, Sickle Cell/genetics , Anemia, Sickle Cell/therapy , Animals , Bone Marrow Transplantation , Gene Expression Profiling , Iron Overload/genetics , Iron Overload/therapy , Mice , Mice, Knockout , Pneumonia/genetics , Pneumonia/therapy
15.
Adv Exp Med Biol ; 1123: 55-69, 2019.
Article in English | MEDLINE | ID: mdl-31016595

ABSTRACT

The incidence of esophageal adenocarcinoma is rapidly increasing in Western countries. This is despite the introduction of sophisticated endoscopic techniques and our ability to readily monitor the presumed precursor lesion known as Barrett's esophagus. Preemptive approaches, including radiofrequency ablation (RFA), and photodynamic therapy (PDT) for Barrett's esophagus and dysplasia are achieving dramatic initial results. Although the long-term efficacy of these nonspecific ablative therapies is awaiting longitudinal studies, reports of recurrences are increasing. More targeted therapies, particularly directed at the stem cells of Barrett's esophagus, demand knowing the origin of this intestinal metaplasia (IM). The prevailing concept holds that Barrett's esophagus arises from the "transcommitment" of esophageal stem cells to produce an intestine-like epithelium. An alternative explanation derives from the discovery of a discrete population of residual embryonic cells (RECs) existing at the gastroesophageal junction in normal individuals that expands and colonizes regions of the esophagus denuded by chronic reflux. These RECs form IM within days of esophageal injury, suggesting a novel mechanism of tumorigenesis.A corollary of this work is that the Barrett's stem cell is distinct from that of the squamous epithelium and, once identified, will form the basis of new preemptive strategies for addressing Barrett's and its related neoplasia.


Subject(s)
Barrett Esophagus/pathology , Esophageal Neoplasms/pathology , Esophagus/cytology , Stem Cells/cytology , Humans , Metaplasia , Neoplasm Recurrence, Local
16.
J Pathol ; 238(4): 519-530, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26415052

ABSTRACT

High-grade serous cancer (HGSC) progresses to advanced stages without symptoms and the 5-year survival rate is a dismal 30%. Recent studies of ovaries and Fallopian tubes in patients with BRCA1 or BRCA2 mutations have documented a pre-metastatic intramucosal neoplasm that is found almost exclusively in the Fallopian tube, termed 'serous tubal intraepithelial carcinoma' or STIC. Moreover, other proliferations, termed p53 signatures, secretory cell outgrowths (SCOUTs), and lower-grade serous tubal intraepithelial neoplasms (STINs) fall short of STIC but share similar alterations in expression, in keeping with an underpinning of genomic disturbances involved in, or occurring in parallel with, serous carcinogenesis. To gain insight into the cellular origins of this unique tubal pathway to high-grade serous cancer, we cloned and both immortalized and transformed Fallopian tube stem cells (FTSCs). We demonstrated that pedigrees of FTSCs were capable of multipotent differentiation and that the tumours derived from transformed FTSCs shared the histological and molecular features of HGSC. We also demonstrated that altered expression of some biomarkers seen in transformed FTSCs and HGSCs (stathmin, EZH2, CXCR4, CXCL12, and FOXM1) could be seen as well in immortalized cells and their in vivo counterparts SCOUTs and STINs. Thus, a whole-genome transcriptome analysis comparing FTSCs, immortalized FTSCs, and transformed FTSCs showed a clear molecular progression sequence that is recapitulated by the spectrum of accumulated perturbations characterizing the range of proliferations seen in vivo. Biomarkers unique to STIC relative to normal tubal epithelium provide a basis for novel detection approaches to early HGSC, but must be viewed critically given their potential expression in lesser proliferations. Perturbations shared by both immortalized and transformed FTSCs may provide unique early targets for prevention strategies. Central to these efforts has been the ability to clone and perpetuate multipotent FTSCs.


Subject(s)
Carcinoma in Situ/pathology , Cystadenocarcinoma, Serous/pathology , Fallopian Tube Neoplasms/pathology , Neoplasms, Cystic, Mucinous, and Serous/pathology , Neoplastic Stem Cells/pathology , Biomarkers, Tumor/metabolism , Cystadenocarcinoma, Serous/genetics , Fallopian Tube Neoplasms/genetics , Female , Humans , Neoplasms, Cystic, Mucinous, and Serous/diagnosis , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology
17.
Gastroenterology ; 149(6): 1599-606, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26296479

ABSTRACT

The development of and adherence to quality indicators in gastroenterology, as in all of medicine, is increasing in importance to ensure that patients receive consistent high-quality care. In addition, government-based and private insurers will be expecting documentation of the parameters by which we measure quality, which will likely affect reimbursements. Barrett's esophagus remains a particularly important disease entity for which we should maintain up-to-date guidelines, given its commonality, potentially lethal outcomes, and controversies regarding screening and surveillance. To achieve this goal, a relatively large group of international experts was assembled and, using the modified Delphi method, evaluated the validity of multiple candidate quality indicators for the diagnosis and management of Barrett's esophagus. Several candidate quality indicators achieved >80% agreement. These statements are intended to serve as a consensus on candidate quality indicators for those who treat patients with Barrett's esophagus.


Subject(s)
Adenocarcinoma/diagnosis , Barrett Esophagus/diagnosis , Consensus Development Conferences as Topic , Disease Management , Esophageal Neoplasms/diagnosis , Esophagus/pathology , Gastroenterology/organization & administration , Adenocarcinoma/pathology , Adenocarcinoma/therapy , Barrett Esophagus/pathology , Barrett Esophagus/therapy , Consensus , Disease Progression , Esophageal Neoplasms/pathology , Esophageal Neoplasms/therapy , Esophagoscopy , Humans , United States
18.
J Pathol ; 236(3): 265-71, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25782708

ABSTRACT

Recent studies have suggested the involvement of a unique population of cells at the cervical squamo-columnar junction (SCJ) in the pathogenesis of early (squamous intraepithelial lesion or SIL) and advanced (squamous cell and adeno-carcinomas) cervical neoplasia. However, there is little evidence to date showing that SCJ cells harbour carcinogenic HPV or are instrumental in the initial phases of neoplasia. This study was designed to (1) determine if normal-appearing SCJ cells contained evidence of carcinogenic HPV infection and (2) trace their transition to early SIL. Sections of cervix from high-risk reproductive age women were selected and SCJ cells were analysed by using several techniques which increasingly implicated HPV infection: HPV DNA (genotyping and in situ hybridization)/RNA (PCR), immunostaining for HPV16 E2 (an early marker of HPV infection), p16(ink4), Ki67, and HPV L1 protein. In 22 cases with a history of SIL and no evidence of preneoplastic lesion in the excision specimen, HPV DNA was isolated from eight of ten with visible SCJ cells, six of which were HPV16/18 DNA-positive. In five of these latter cases, the SCJ cells were positive for p16(ink4) and/or HPV E2. Transcriptionally active HPV infection (E6/E7 mRNAs) was also detected in microdissected SCJ cells. Early squamous atypia associated with the SCJ cells demonstrated in addition diffuse p16(ink4) immunoreactivity, elevated proliferative index, and rare L1 antigen positivity. We present for the first time direct evidence that normal-appearing SCJ cells can be infected by carcinogenic HPV. They initially express HPV E2 and their progression to SIL is heralded by an expanding metaplastic progeny with increased proliferation and p16(ink4) expression. Whether certain SCJs are more vulnerable than others to carcinogenic HPV genotypes and what variables determine transition to high-grade SIL remain unresolved, but the common event appears to be a vulnerable cell at the SCJ.


Subject(s)
Cell Transformation, Viral , Epithelial Cells/pathology , Papillomaviridae/physiology , Papillomavirus Infections/pathology , Uterine Cervical Neoplasms/pathology , Capsid Proteins/metabolism , Cervix Uteri/pathology , Cyclin-Dependent Kinase Inhibitor p16 , DNA, Viral/genetics , DNA-Binding Proteins/metabolism , Epithelial Cells/virology , Female , Genotype , Humans , In Situ Hybridization , Ki-67 Antigen/metabolism , Neoplasm Proteins/metabolism , Oncogene Proteins, Viral/metabolism , Papillomaviridae/genetics , Papillomavirus Infections/virology , RNA, Viral/genetics , Uterine Cervical Neoplasms/virology
19.
Proc Natl Acad Sci U S A ; 110(44): 17969-74, 2013 Oct 29.
Article in English | MEDLINE | ID: mdl-24127607

ABSTRACT

Squamous cell carcinoma (SCC) is highly malignant and refractory to therapy. The majority of existing mouse SCC models involve multiple gene mutations. Very few mouse models of spontaneous SCC have been generated by a single gene deletion. Here we report a haploinsufficient SCC mouse model in which exon 3 of the Tp53BP2 gene (a p53 binding protein) was deleted in one allele in a BALB/c genetic background. Tp53BP2 encodes ASPP2 (ankyrin repeats, SH3 domain and protein rich region containing protein 2). Keratinocyte differentiation induces ASPP2 and its expression is inversely correlated with p63 protein in vitro and in vivo. Up-regulation of p63 expression is required for ASPP2(Δexon3/+) BALB/c mice to develop SCC, as heterozygosity of p63 but not p53 prevents them from developing it. Mechanistically, ASPP2 inhibits ΔNp63 expression through its ability to bind IκB and enhance nuclear Rel/A p65, a component of the NF-κB transcription complex, which mediates the repression of p63. Reduced ASPP2 expression associates with tumor metastasis and increased p63 expression in human head and neck SCCs. This study identifies ASPP2 as a tumor suppressor that suppresses SCC via inflammatory signaling through NF-κB-mediated repression of p63.


Subject(s)
Carcinoma, Squamous Cell/immunology , Disease Models, Animal , Phosphoproteins/metabolism , Signal Transduction/immunology , Trans-Activators/metabolism , Transcription Factor RelA/metabolism , Tumor Suppressor Proteins/immunology , Tumor Suppressor Proteins/metabolism , Animals , Carcinoma, Squamous Cell/genetics , Cell Line , Crosses, Genetic , DNA Primers/genetics , Haploinsufficiency , Humans , Immunoblotting , Immunoprecipitation , Mice , Mice, Inbred BALB C , Microarray Analysis , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Tumor Suppressor Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL