Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
Acad Med ; 94(5): 708-714, 2019 05.
Article in English | MEDLINE | ID: mdl-30520806

ABSTRACT

PURPOSE: This analysis examined the role of a National Institutes of Health (NIH) individual Mentored Career Development Award (K01, K08, K23) on launching and sustaining independent research careers for early-career scientists, and investigated the effects of these awards during and after the doubling of the NIH budget. METHOD: The authors used grants data from the NIH covering the period 1990 through 2016, and compared success in receipt of R01 equivalent awards (R01 Eq.) and Research Project Grants (RPGs) for K awardees and K applicants who did not receive funding. The analysis combined regression discontinuity design with coarsened exact matching, and regression. RESULTS: Overall, receipt of K award was associated with a 24.1% increase in likelihood of first independent NIH award (P < .01), and a larger number of R01 Eq. and RPG awards. After accounting for first major independent awards, K awards were uncorrelated with receiving second major independent research awards. Comparing different funding periods, K01 awards were predictive of subsequent R01 Eq. and RPG awards after but not during the NIH doubling, K08 awards were predictive only during the NIH doubling, and K23 awards were predictive during both periods. CONCLUSIONS: Receipt of Mentored Career Development Awards was linked to increased likelihood that early-career scientists successfully transitioned to an independent research career. These findings indicate that extending funding to additional K award applicants with meritorious scores could significantly strengthen the pipeline of biomedical researchers. In addition, enhancing K awards may be relevant to sustaining research careers for clinician scientists.


Subject(s)
Biomedical Research/economics , Career Choice , Education, Medical/organization & administration , Financing, Organized/economics , National Institutes of Health (U.S.)/economics , Research Personnel/economics , Research Support as Topic/economics , Adult , Biomedical Research/statistics & numerical data , Education, Medical/statistics & numerical data , Female , Humans , Male , Mentors/statistics & numerical data , National Institutes of Health (U.S.)/statistics & numerical data , Research Personnel/statistics & numerical data , United States
2.
FASEB J ; : fj201800639, 2018 Jun 19.
Article in English | MEDLINE | ID: mdl-29920223

ABSTRACT

Here, we use recent U.S. National Institutes of Health (NIH) data to document trends in the NIH-funded workforce over time. Consistent with previous studies that were initiated by NIH, we find that the number of scientists funded on competing R01-equivalent (R01 Eq.) and research project grants (RPGs) increased 2-5% per year between 2009 and 2016. Primary beneficiaries of this growth were experienced investigators (Exp), whereas the share of funding awarded to early-stage investigators (ESIs) and new investigators (NIs) declined. The decline occurred even after NIH instituted the New and Early-Stage Investigator policy in 2009. When we evaluate the investigator pool, we find that women and racial and ethnic minorities represent a higher percentage of NIs and ESIs relative to Exp. Thus, trends of diminishing support for NIs and ESIs may negatively impact the diversity of the current and future biomedical research workforce. We find some recent gains among women and Hispanics as part of the applicant and awardee pool for both R01 Eq. and RPGs, but significant, large gaps persist among nationally underrepresented racial minorities. Our findings suggest a need to prioritize investments and support of ESIs and NIs, groups in which women and racial and ethnic minorities represent a larger proportion of the applicant pool, to enhance diversity in the NIH-funded workforce.-Nikaj, S., Roychowdhury, D., Lund, P. K., Matthews, M., Pearson, K. Examining trends in the diversity of the U.S. National Institutes of Health participating and funded workforce.

3.
Acad Med ; 92(10): 1382-1389, 2017 10.
Article in English | MEDLINE | ID: mdl-28767499

ABSTRACT

Clinician-investigators, also called physician-scientists, offer critical knowledge and perspectives that benefit research on basic science mechanisms, improved diagnostic and therapeutic approaches, population and outcomes medicine, health policy, and health services, yet few clinically trained health professionals pursue a research career. Sustaining this workforce requires attention to the unique challenges faced by investigators who must achieve clinical and research competence during training and their careers. These challenges include the duration of required clinical training, limited or discontinuous research opportunities, high levels of educational debt, balancing the dual obligations and rewards of clinical care and research, competition for research funding, and the need for leadership development after training. Women and individuals from underrepresented racial and ethnic groups comprise a small percentage of this workforce.The authors summarize the recent literature on training for clinician-investigators, emphasizing approaches with encouraging outcomes that warrant broader implementation. Using this overview as background, they convened three workshops at the National Institutes of Health in 2016 to identify and refine key priorities for potential new pilot programs to recruit and retain the clinician-investigator workforce. From these workshops emerged three priorities for future pilot programs: (1) support for research in residency, (2) new research on-ramps for health professionals at multiple career stages, and (3) national networks to diversify and sustain clinician-investigator faculty. Implementation of any pilot program will require coordinated commitment from academic health centers, medical licensing/certification boards, professional societies, and clinician-investigators themselves, in addition to support from the National Institutes of Health.


Subject(s)
Biomedical Research/education , Health Personnel/education , Minority Groups/education , Personnel Selection/methods , Research Personnel/education , Female , Humans , Pilot Projects , United States
4.
J Clin Invest ; 127(2): 593-607, 2017 Feb 01.
Article in English | MEDLINE | ID: mdl-28094771

ABSTRACT

Orphan GPCRs provide an opportunity to identify potential pharmacological targets, yet their expression patterns and physiological functions remain challenging to elucidate. Here, we have used a genetically engineered knockin reporter mouse to map the expression pattern of the Gpr182 during development and adulthood. We observed that Gpr182 is expressed at the crypt base throughout the small intestine, where it is enriched in crypt base columnar stem cells, one of the most active stem cell populations in the body. Gpr182 knockdown had no effect on homeostatic intestinal proliferation in vivo, but led to marked increases in proliferation during intestinal regeneration following irradiation-induced injury. In the ApcMin mouse model, which forms spontaneous intestinal adenomas, reductions in Gpr182 led to more adenomas and decreased survival. Loss of Gpr182 enhanced organoid growth efficiency ex vivo in an EGF-dependent manner. Gpr182 reduction led to increased activation of ERK1/2 in basal and challenge models, demonstrating a potential role for this orphan GPCR in regulating the proliferative capacity of the intestine. Importantly, GPR182 expression was profoundly reduced in numerous human carcinomas, including colon adenocarcinoma. Together, these results implicate Gpr182 as a negative regulator of intestinal MAPK signaling-induced proliferation, particularly during regeneration and adenoma formation.


Subject(s)
Adenomatous Polyposis Coli/metabolism , Cell Proliferation , Intestine, Small/metabolism , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase 3/metabolism , Neoplasms, Experimental/metabolism , Receptors, G-Protein-Coupled/metabolism , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/pathology , Adenomatous Polyposis Coli Protein/genetics , Adenomatous Polyposis Coli Protein/metabolism , Animals , Gene Knockdown Techniques , Intestine, Small/pathology , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 3/genetics , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Receptors, G-Protein-Coupled/genetics
5.
J Biol Chem ; 292(7): 2586-2600, 2017 02 17.
Article in English | MEDLINE | ID: mdl-28053090

ABSTRACT

Gut microbiota play an important role in regulating the development of the host immune system, metabolic rate, and at times, disease pathogenesis. The factors and mechanisms that mediate interactions between microbiota and the intestinal epithelium are not fully understood. We provide novel evidence that microbiota may control intestinal epithelial stem cell (IESC) proliferation in part through microRNAs (miRNAs). We demonstrate that miRNA profiles differ dramatically across functionally distinct cell types of the mouse jejunal intestinal epithelium and that miRNAs respond to microbiota in a highly cell type-specific manner. Importantly, we also show that miRNAs in IESCs are more prominently regulated by microbiota compared with miRNAs in any other intestinal epithelial cell subtype. We identify miR-375 as one miRNA that is significantly suppressed by the presence of microbiota in IESCs. Using a novel method to knockdown gene and miRNA expression ex vivo enteroids, we demonstrate that we can knock down gene expression in Lgr5+ IESCs. Furthermore, when we knock down miR-375 in IESCs, we observe significantly increased proliferative capacity. Understanding the mechanisms by which microbiota regulate miRNA expression in IESCs and other intestinal epithelial cell subtypes will elucidate a critical molecular network that controls intestinal homeostasis and, given the heightened interest in miRNA-based therapies, may offer novel therapeutic strategies in the treatment of gastrointestinal diseases associated with altered IESC function.


Subject(s)
Intestinal Mucosa/metabolism , Stem Cells/microbiology , Transcriptome , Animals , Female , Germ-Free Life , Green Fluorescent Proteins/genetics , Intestinal Mucosa/cytology , Intestinal Mucosa/microbiology , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , SOX9 Transcription Factor/genetics , Stem Cells/cytology
6.
J Womens Health (Larchmt) ; 26(5): 525-529, 2017 05.
Article in English | MEDLINE | ID: mdl-27509297

ABSTRACT

While women have been well represented in medical school and biomedical doctoral degree programs, they do not comprise half of academic medicine faculty positions. Furthermore, there is a significant paucity of women in academic medicine leadership positions, as evidenced by the fact that only 16% of dean positions at United States Medical schools are filled by women. In this commentary, the authors review the state of women in academic medicine and argue that increased representation of women in the academic workforce will lead to economic gains, increased scientific discovery, and improvements to women's health.


Subject(s)
Biomedical Research , Career Mobility , Physicians, Women , Science , Women's Health , Academic Medical Centers , Faculty, Medical , Female , Humans , Leadership , Schools, Medical , Sexism/psychology , Workforce
7.
CBE Life Sci Educ ; 15(3)2016.
Article in English | MEDLINE | ID: mdl-27587850

ABSTRACT

The National Institutes of Health (NIH) is committed to attracting, developing, and supporting the best scientists from all groups as an integral part of excellence in training. Biomedical research workforce diversity, capitalizing on the full spectrum of skills, talents, and viewpoints, is essential for solving complex human health challenges. Over the past few decades, the biomedical research workforce has benefited from NIH programs aimed at enhancing diversity. However, there is considerable room for improvement, particularly at the level of independent scientists and within scientific leadership. We provide a rationale and specific opportunities to develop and sustain a diverse biomedical research workforce through interventions that promote the successful transitions to different stages on the path toward completion of training and entry into the biomedical workforce.


Subject(s)
Biomedical Research , Cultural Diversity , National Institutes of Health (U.S.) , Female , Humans , Male , Minority Groups , Program Development , United States , Workforce
8.
Gut Microbes ; 7(5): 414-23, 2016 09 02.
Article in English | MEDLINE | ID: mdl-27459363

ABSTRACT

Doxorubicin (DOXO) induces significant, but transient, increases in apoptosis in the stem cell zone of the jejunum, followed by mucosal damage involving a decrease in crypt proliferation, crypt number, and villus height. The gastrointestinal tract is home to a vast population of commensal bacteria and numerous studies have demonstrated a symbiotic relationship between intestinal bacteria and intestinal epithelial cells (IEC) in maintaining homeostatic functions of the intestine. However, whether enteric bacteria play a role in DOXO-induced damage is not well understood. We hypothesized that enteric bacteria are necessary for induction of apoptosis and damage associated with DOXO treatment. Conventionally raised (CONV) and germ free (GF) mice were given a single injection of DOXO, and intestinal tissue was collected at 6, 72, and 120 h after treatment and from no treatment (0 h) controls. Histology and morphometric analyses quantified apoptosis, mitosis, crypt depth, villus height, and crypt density. Immunostaining for muc2 and lysozyme evaluated Paneth cells, goblet cells or dual stained intermediate cells. DOXO administration induced significant increases in apoptosis in jejunal epithelium regardless of the presence of enteric bacteria; however, the resulting injury, as demonstrated by statistically significant changes in crypt depth, crypt number, and proliferative cell number, was dependent upon the presence of enteric bacteria. Furthermore, we observed expansion of Paneth and goblet cells and presence of intermediate cells only in CONV and not GF mice. These findings provide evidence that manipulation and/or depletion of the enteric microbiota may have clinical significance in limiting chemotherapy-induced mucositis.


Subject(s)
Antibiotics, Antineoplastic/adverse effects , Apoptosis/drug effects , Bacteria/drug effects , Doxorubicin/adverse effects , Gastrointestinal Microbiome/drug effects , Intestines/drug effects , Intestines/microbiology , Animals , Antibiotics, Antineoplastic/administration & dosage , Bacteria/genetics , Bacteria/isolation & purification , Doxorubicin/administration & dosage , Female , Germ-Free Life , Goblet Cells/drug effects , Goblet Cells/microbiology , Intestines/cytology , Mice , Mice, Inbred C57BL , Paneth Cells/drug effects , Paneth Cells/microbiology
9.
J Biol Chem ; 291(31): 15975-84, 2016 07 29.
Article in English | MEDLINE | ID: mdl-27261459

ABSTRACT

Proliferation and differentiation of intestinal epithelial cells (IECs) occur in part through precise regulation of key transcription factors, such as SOX9. MicroRNAs (miRNAs) have emerged as prominent fine-tuners of transcription factor expression and activity. We hypothesized that miRNAs, in part through the regulation of SOX9, may mediate IEC homeostasis. Bioinformatic analyses of the SOX9 3'-UTR revealed highly conserved target sites for nine different miRNAs. Of these, only the miR-30 family members were both robustly and variably expressed across functionally distinct cell types of the murine jejunal epithelium. Inhibition of miR-30 using complementary locked nucleic acids (LNA30bcd) in both human IECs and human colorectal adenocarcinoma-derived Caco-2 cells resulted in significant up-regulation of SOX9 mRNA but, interestingly, significant down-regulation of SOX9 protein. To gain mechanistic insight into this non-intuitive finding, we performed RNA sequencing on LNA30bcd-treated human IECs and found 2440 significantly increased genes and 2651 significantly decreased genes across three time points. The up-regulated genes are highly enriched for both predicted miR-30 targets, as well as genes in the ubiquitin-proteasome pathway. Chemical suppression of the proteasome rescued the effect of LNA30bcd on SOX9 protein levels, indicating that the regulation of SOX9 protein by miR-30 is largely indirect through the proteasome pathway. Inhibition of the miR-30 family led to significantly reduced IEC proliferation and a dramatic increase in markers of enterocyte differentiation. This in-depth analysis of a complex miRNA regulatory program in intestinal epithelial cell models provides novel evidence that the miR-30 family likely plays an important role in IEC homeostasis.


Subject(s)
Cell Differentiation/physiology , Cell Proliferation/physiology , Enterocytes/metabolism , Gene Expression Regulation/physiology , MicroRNAs/metabolism , SOX9 Transcription Factor/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Caco-2 Cells , Enterocytes/cytology , Humans , Male , Mice , Mice, Mutant Strains , MicroRNAs/genetics , SOX9 Transcription Factor/genetics , Ubiquitin-Protein Ligases/genetics
10.
Am J Physiol Gastrointest Liver Physiol ; 309(7): G578-89, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26251471

ABSTRACT

Current views suggest that apoptosis eliminates genetically damaged cells that may otherwise form tumors. Prior human studies link elevated insulin and reduced apoptosis to risk of colorectal adenomas. We hypothesized that hyperinsulinemia associated with obesity would lead to reduced colon epithelial cell (CEC) apoptosis after radiation and that this effect would be altered by deletion of the insulin-like growth factor (IGF) 1 receptor (IGF1R) or the insulin receptor (IR). Mice with villin-Cre-mediated IGF1R or IR deletion in CECs and floxed littermates were fed a high-fat diet to induce obesity and hyperinsulinemia or control low-fat chow. Mice were exposed to 5-Gy abdominal radiation to induce DNA damage and euthanized 4 h later for evaluation of apoptosis by localization of cleaved caspase-3. Obese mice exhibited decreased apoptosis of genetically damaged CECs. IGF1R deletion did not affect CEC apoptosis in lean or obese animals. In contrast, IR loss increased CEC apoptosis in both diet groups but did not prevent antiapoptotic effects of obesity. Levels of p53 protein were significantly reduced in CECs of obese mice with intact IR but increased in both lean and obese mice without IR. Levels of mRNAs encoding proapoptotic Perp and the cell cycle inhibitor Cdkn1b/p27 were reduced in CECs of obese mice and increased in lean mice lacking IR. Together, our studies provide novel evidence for antiapoptotic roles of obesity and IR, but not IGF1R, in colonic epithelium after DNA damage. However, neither IR nor IGF1R deletion prevented a reduction in radiation-induced CEC apoptosis during obesity and hyperinsulinemia.


Subject(s)
Apoptosis/radiation effects , Colon/pathology , Intestinal Mucosa/metabolism , Obesity/metabolism , Receptor, IGF Type 1/metabolism , Receptor, Insulin/metabolism , Animals , Blotting, Western , Caspase 3 , Colon/metabolism , Immunohistochemistry , Male , Mice , Radiation Injuries, Experimental , Real-Time Polymerase Chain Reaction , Receptor, IGF Type 1/genetics , Receptor, Insulin/genetics
11.
FASEB J ; 29(7): 2828-42, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25837582

ABSTRACT

Insulin-like growth factor 1 (IGF1) has potent trophic effects on normal or injured intestinal epithelium, but specific effects on intestinal stem cells (ISCs) are undefined. We used Sox9-enhanced green fluorescent protein (EGFP) reporter mice that permit analyses of both actively cycling ISCs (Sox9-EGFP(Low)) and reserve/facultative ISCs (Sox9-EGFP(High)) to study IGF1 action on ISCs in normal intestine or during crypt regeneration after high-dose radiation-induced injury. We hypothesized that IGF1 differentially regulates proliferation and gene expression in actively cycling and reserve/facultative ISCs. IGF1 was delivered for 5 days using subcutaneously implanted mini-pumps in uninjured mice or after 14 Gy abdominal radiation. ISC numbers, proliferation, and transcriptome were assessed. IGF1 increased epithelial growth in nonirradiated mice and enhanced crypt regeneration after radiation. In uninjured and regenerating intestines, IGF1 increased total numbers of Sox9-EGFP(Low) ISCs and percentage of these cells in M-phase. IGF1 increased percentages of Sox9-EGFP(High) ISCs in S-phase but did not expand this population. Microarray revealed that IGF1 activated distinct gene expression signatures in the 2 Sox9-EGFP ISC populations. In vitro IGF1 enhanced enteroid formation by Sox9-EGFP(High) facultative ISCs but not Sox9-EGFP(Low) actively cycling ISCs. Our data provide new evidence that IGF1 activates 2 ISC populations via distinct regulatory pathways to promote growth of normal intestinal epithelium and crypt regeneration after irradiation.


Subject(s)
Adult Stem Cells/classification , Insulin-Like Growth Factor I/physiology , Intestine, Small/cytology , Adult Stem Cells/drug effects , Adult Stem Cells/physiology , Animals , Cell Cycle , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Insulin-Like Growth Factor I/pharmacology , Intestinal Mucosa/cytology , Intestinal Mucosa/drug effects , Intestinal Mucosa/physiology , Intestine, Small/drug effects , Intestine, Small/physiology , Mice , Mice, Transgenic , Multipotent Stem Cells/classification , Multipotent Stem Cells/drug effects , Multipotent Stem Cells/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Radiation Injuries, Experimental/drug therapy , Radiation Injuries, Experimental/pathology , Radiation Injuries, Experimental/physiopathology , Receptor, IGF Type 1/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Regeneration/drug effects , Regeneration/physiology , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism
12.
Am J Physiol Gastrointest Liver Physiol ; 308(6): G562-71, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25573176

ABSTRACT

Myeloid translocation genes (MTGs) are transcriptional corepressors implicated in development, malignancy, differentiation, and stem cell function. While MTG16 loss renders mice sensitive to chemical colitis, the role of MTG16 in the small intestine is unknown. Histological examination revealed that Mtg16(-/-) mice have increased enterocyte proliferation and goblet cell deficiency. After exposure to radiation, Mtg16(-/-) mice exhibited increased crypt viability and decreased apoptosis compared with wild-type (WT) mice. Flow cytometric and immunofluorescence analysis of intestinal epithelial cells for phospho-histone H2A.X also indicated decreased DNA damage and apoptosis in Mtg16(-/-) intestines. To determine if Mtg16 deletion affected epithelial cells in a cell-autonomous fashion, intestinal crypts were isolated from Mtg16(-/-) mice. Mtg16(-/-) and WT intestinal crypts showed similar enterosphere forming efficiencies when cultured in the presence of EGF, Noggin, and R-spondin. However, when Mtg16(-/-) crypts were cultured in the presence of Wnt3a, they demonstrated higher enterosphere forming efficiencies and delayed progression to mature enteroids. Mtg16(-/-) intestinal crypts isolated from irradiated mice exhibited increased survival compared with WT intestinal crypts. Interestingly, Mtg16 expression was reduced in a stem cell-enriched population at the time of crypt regeneration. This is consistent with MTG16 negatively regulating regeneration in vivo. Taken together, our data demonstrate that MTG16 loss promotes radioresistance and impacts intestinal stem cell function, possibly due to shifting cellular response away from DNA damage-induced apoptosis and towards DNA repair after injury.


Subject(s)
Cell Proliferation , Gamma Rays , Intestinal Mucosa/metabolism , Intestine, Small/metabolism , Nuclear Proteins/metabolism , Radiation Injuries, Experimental/metabolism , Regeneration , Transcription Factors/metabolism , Animals , Apoptosis , Cell Proliferation/drug effects , Cell Survival , DNA Damage , Female , Gene Expression Regulation , Goblet Cells/metabolism , Goblet Cells/pathology , Histones/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Intestine, Small/drug effects , Intestine, Small/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Phenotype , Radiation Injuries, Experimental/etiology , Radiation Injuries, Experimental/pathology , Radiation Tolerance , Regeneration/drug effects , Repressor Proteins , Signal Transduction , Stem Cells/metabolism , Stem Cells/pathology , Tissue Culture Techniques , Transcription Factors/deficiency , Transcription Factors/genetics , Wnt3A Protein/pharmacology
13.
Am J Physiol Gastrointest Liver Physiol ; 308(2): G100-11, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25394660

ABSTRACT

The insulin receptor (IR) regulates nutrient uptake and utilization in multiple organs, but its role in the intestinal epithelium is not defined. This study developed a mouse model with villin-Cre (VC) recombinase-mediated intestinal epithelial cell (IEC)-specific IR deletion (VC-IR(Δ/Δ)) and littermate controls with floxed, but intact, IR (IR(fl/fl)) to define in vivo roles of IEC-IR in mice fed chow or high-fat diet (HFD). We hypothesized that loss of IEC-IR would alter intestinal growth, biomarkers of intestinal epithelial stem cells (IESC) or other lineages, body weight, adiposity, and glucose or lipid handling. In lean, chow-fed mice, IEC-IR deletion did not affect body or fat mass, plasma glucose, or IEC proliferation. In chow-fed VC-IR(Δ/Δ) mice, mRNA levels of the Paneth cell marker lysozyme (Lyz) were decreased, but markers of other differentiated lineages were unchanged. During HFD-induced obesity, IR(fl/fl) and VC-IR(Δ/Δ) mice exhibited similar increases in body and fat mass, plasma insulin, mRNAs encoding several lipid-handling proteins, a decrease in Paneth cell number, and impaired glucose tolerance. In IR(fl/fl) mice, HFD-induced obesity increased circulating cholesterol; numbers of chromogranin A (CHGA)-positive enteroendocrine cells (EEC); and mRNAs encoding Chga, glucose-dependent insulinotrophic peptide (Gip), glucagon (Gcg), Lyz, IESC biomarkers, and the enterocyte cholesterol transporter Scarb1. All these effects were attenuated or lost in VC-IR(Δ/Δ) mice. These results demonstrate that IEC-IR is not required for normal growth of the intestinal epithelium in lean adult mice. However, our findings provide novel evidence that, during HFD-induced obesity, IEC-IR contributes to increases in EEC, plasma cholesterol, and increased expression of Scarb1 or IESC-, EEC-, and Paneth cell-derived mRNAs.


Subject(s)
Cholesterol/metabolism , Diet, High-Fat , Enteroendocrine Cells/metabolism , Intestines/pathology , Paneth Cells/metabolism , Receptor, Insulin/metabolism , Stem Cells/metabolism , Animals , Cell Differentiation , Gastric Inhibitory Polypeptide/metabolism , Insulin/blood , Intestinal Mucosa/metabolism , Mice , Mice, Transgenic , Obesity/metabolism , RNA, Messenger/metabolism
14.
Cancer Epidemiol Biomarkers Prev ; 23(10): 2093-100, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25017244

ABSTRACT

BACKGROUND: Hyperinsulinemia resulting from obesity and insulin resistance is associated with increased risk of many cancers, but the biology underlying this risk is unclear. We hypothesized that increased mRNA levels of the insulin-like growth factor I receptor (IGFIR) versus the insulin receptor (IR) or elevated ratio of IR-A:IR-B isoforms in normal rectal mucosa would predict adenoma risk, particularly in individuals with high body mass index (BMI) or plasma insulin. METHODS: Biopsies from normal rectal mucosa were obtained from consenting patients undergoing routine colonoscopy at University of North Carolina Hospitals (Chapel Hill, NC). Subjects with colorectal adenomas were classified as cases (n = 100) and were matched to adenoma-free controls (n = 98) based on age, sex, and BMI. IGFIR and IR mRNA levels were assessed by qRT-PCR, and IR-A:IR-B mRNA ratios by standard PCR. Plasma insulin and crypt apoptosis were measured by ELISA and terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL), respectively. Logistic regression models examined relationships between receptor mRNAs, BMI, plasma insulin, and adenoma risk. RESULTS: Unexpectedly, cases were significantly more likely to have lower IGFIR mRNA levels than controls. No overall differences in total IR mRNA or IR-A:IR-B ratios were observed between cases and controls. Interestingly, in patients with high plasma insulin, increased IR-A:IR-B ratio was associated with increased likelihood of having adenomas. CONCLUSIONS: Our work shows novel findings that reduced IGFIR mRNA and, during high plasma insulin, increased IR-A:IR-B ratios in normal rectal mucosa are associated with colorectal adenoma risk. IMPACT: Our work provides evidence supporting a link between IGFIR and IR isoform expression levels and colorectal adenoma risk.


Subject(s)
Adenoma/metabolism , Colorectal Neoplasms/metabolism , Receptor, IGF Type 1/biosynthesis , Receptor, Insulin/biosynthesis , Adenoma/pathology , Apoptosis , Colorectal Neoplasms/pathology , Enzyme-Linked Immunosorbent Assay , Female , Humans , In Situ Nick-End Labeling , Male , Middle Aged , Polymerase Chain Reaction , Protein Isoforms , RNA, Messenger
15.
Endocrinology ; 155(9): 3302-14, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24914941

ABSTRACT

Nutrient intake regulates intestinal epithelial mass and crypt proliferation. Recent findings in model organisms and rodents indicate nutrient restriction impacts intestinal stem cells (ISC). Little is known about the impact of diet-induced obesity (DIO), a model of excess nutrient intake on ISC. We used a Sox9-EGFP reporter mouse to test the hypothesis that an adaptive response to DIO or associated hyperinsulinemia involves expansion and hyperproliferation of ISC. The Sox9-EGFP reporter mouse allows study and isolation of ISC, progenitors, and differentiated lineages based on different Sox9-EGFP expression levels. Sox9-EGFP mice were fed a high-fat diet for 20 weeks to induce DIO and compared with littermates fed low-fat rodent chow. Histology, fluorescence activated cell sorting, and mRNA analyses measured impact of DIO on jejunal crypt-villus morphometry, numbers, and proliferation of different Sox9-EGFP cell populations and gene expression. An in vitro culture assay directly assessed functional capacity of isolated ISC. DIO mice exhibited significant increases in body weight, plasma glucose, insulin, and insulin-like growth factor 1 (IGF1) levels and intestinal Igf1 mRNA. DIO mice had increased villus height and crypt density but decreased intestinal length and decreased numbers of Paneth and goblet cells. In vivo, DIO resulted in a selective expansion of Sox9-EGFP(Low) ISC and percentage of ISC in S-phase. ISC expansion significantly correlated with plasma insulin levels. In vitro, isolated ISC from DIO mice formed fewer enteroids in standard 3D Matrigel culture compared to controls, indicating impaired ISC function. This decreased enteroid formation in isolated ISC from DIO mice was rescued by exogenous insulin, IGF1, or both. We conclude that DIO induces specific increases in ISC and ISC hyperproliferation in vivo. However, isolated ISC from DIO mice have impaired intrinsic survival and growth in vitro that can be rescued by exogenous insulin or IGF1.


Subject(s)
Cell Proliferation , Diet, High-Fat/adverse effects , Insulin-Like Growth Factor I/metabolism , Insulin/metabolism , Intestines/cytology , Obesity/metabolism , Stem Cells/cytology , Animals , Female , Humans , Intestinal Mucosa/metabolism , Male , Mice , Mice, Transgenic , Obesity/genetics , Obesity/physiopathology , Signal Transduction , Stem Cells/metabolism
16.
Diabetes ; 63(9): 3141-8, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24722248

ABSTRACT

MicroRNAs (miRNAs) have emerged as biomarkers of metabolic status, etiological factors in complex disease, and promising drug targets. Recent reports suggest that miRNAs are critical regulators of pathways underlying the pathophysiology of type 2 diabetes. In this study, we demonstrate by deep sequencing and real-time quantitative PCR that hepatic levels of Foxa2 mRNA and miR-29 are elevated in a mouse model of diet-induced insulin resistance. We also show that Foxa2 and miR-29 are significantly upregulated in the livers of Zucker diabetic fatty (fa/fa) rats and that the levels of both returned to normal upon treatment with the insulin-sensitizing agent pioglitazone. We present evidence that miR-29 expression in human hepatoma cells is controlled in part by FOXA2, which is known to play a critical role in hepatic energy homeostasis. Moreover, we demonstrate that miR-29 fine-tunes FOXA2-mediated activation of key lipid metabolism genes, including PPARGC1A, HMGCS2, and ABHD5. These results suggest that miR-29 is an important regulatory factor in normal metabolism and may represent a novel therapeutic target in type 2 diabetes and related metabolic syndromes.


Subject(s)
Insulin Resistance/genetics , MicroRNAs/physiology , Animals , Cell Line, Tumor , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/genetics , Female , Hepatocyte Nuclear Factor 3-beta/genetics , Hepatocyte Nuclear Factor 3-beta/physiology , Humans , Mice , Models, Animal , Pioglitazone , Rats , Rats, Zucker , Thiazolidinediones/therapeutic use , Up-Regulation
17.
J Cell Sci ; 126(Pt 24): 5645-56, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24127567

ABSTRACT

Despite evidence for the impact of insulin on intestinal epithelial physiology and pathophysiology, the expression patterns, roles, and regulation of insulin receptor (IR) and IR isoforms in the intestinal epithelium are not well characterized. IR-A is thought to mediate the proliferative effects of insulin or insulin growth factors (IGFs) in fetal or cancer cells. IR-B is considered to be the metabolic receptor for insulin in specialized tissues. This study used a novel Sox9-EGFP reporter mouse that permits isolation of intestinal epithelial stem cells (IESCs), progenitors, enteroendocrine cells and differentiated lineages, the Apc(Min/+) mouse model of precancerous adenoma and normal human intestinal and colorectal cancer (CRC) cell lines. We tested the hypothesis that there is differential expression of IR-A or IR-B in stem and tumor cells versus differentiated intestinal epithelial cells (IECs) and that IR-B impacts cell proliferation. Our findings provide evidence that IR-B expression is significantly lower in highly proliferative IESCs and progenitor cells versus post-mitotic, differentiated IECs and in subconfluent and undifferentiated versus differentiated Caco-2 cells. IR-B is also reduced in Apc(Min/+) tumors and highly tumorigenic CRC cells. These differences in IR-B were accompanied by altered levels of mRNAs encoding muscleblind-like 2 (MBNL2), a known regulator of IR alternative splicing. Forced IR-B expression in subconfluent and undifferentiated Caco-2 cells reduced proliferation and increased biomarkers of differentiation. Our findings indicate that the impact of insulin on different cell types in the intestinal epithelium might differ depending on relative IR-B IR-A expression levels and provide new evidence for the roles of IR-B to limit proliferation of CRC cells.


Subject(s)
Cell Proliferation , Colorectal Neoplasms/metabolism , Receptor, Insulin/metabolism , Stem Cells/metabolism , Animals , Caco-2 Cells , Cell Differentiation , DNA Replication , Gene Expression , Humans , Intestinal Mucosa/metabolism , Mice , Phenotype , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptor, Insulin/genetics , Signal Transduction , Zonula Occludens-1 Protein/metabolism , beta Catenin/metabolism
18.
PLoS One ; 8(8): e73140, 2013.
Article in English | MEDLINE | ID: mdl-24015295

ABSTRACT

Ileocecal resection (ICR) is a commonly required surgical intervention in unmanageable Crohn's disease and necrotizing enterocolitis. However, the impact of ICR, and the concomitant doses of antibiotic routinely given with ICR, on the intestinal commensal microbiota has not been determined. In this study, wild-type C57BL6 mice were subjected to ICR and concomitant single intraperitoneal antibiotic injection. Intestinal lumen contents were collected from jejunum and colon at 7, 14, and 28 days after resection and compared to non-ICR controls. Samples were analyzed by 16S rRNA gene pyrosequencing and quantitative PCR. The intestinal microbiota was altered by 7 days after ICR and accompanying antibiotic treatment, with decreased diversity in the colon. Phylogenetic diversity (PD) decreased from 11.8 ± 1.8 in non-ICR controls to 5.9 ± 0.5 in 7-day post-ICR samples. There were also minor effects in the jejunum where PD values decreased from 8.3 ± 0.4 to 7.5 ± 1.4. PCoA analysis indicated that bacterial populations 28 days post-ICR differed significantly from non-ICR controls. Moreover, colon and jejunum bacterial populations were remarkably similar 28 days after resection, whereas the initial communities differed markedly. Firmicutes and Bacteroidetes were the predominant phyla in jejunum and colon before ICR; however, Firmicutes became the vastly predominant phylum in jejunum and colon 28 days after ICR. Although the microbiota returned towards a homeostatic state, with re-establishment of Firmicutes as the predominant phylum, we did not detect Bacteroidetes in the colon 28 days after ICR. In the jejunum Bacteroidetes was detected at a 0.01% abundance after this time period. The changes in jejunal and colonic microbiota induced by ICR and concomitant antibiotic injection may therefore be considered as potential regulators of post-surgical adaptive growth or function, and in a setting of active IBD, potential contributors to post-surgical pathophysiology of disease recurrence.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteroidetes/genetics , Colon , Jejunum , Microbiota/genetics , Phylogeny , RNA, Ribosomal, 16S/genetics , Animals , Bacteroidetes/growth & development , Colon/microbiology , Colon/surgery , Crohn Disease/microbiology , Crohn Disease/surgery , Humans , Jejunum/microbiology , Jejunum/surgery , Mice , Sequence Analysis, RNA
20.
Am J Physiol Gastrointest Liver Physiol ; 302(10): G1111-32, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22361729

ABSTRACT

Recent identification of intestinal epithelial stem cell (ISC) markers and development of ISC reporter mice permit visualization and isolation of regenerating ISCs after radiation to define their functional and molecular phenotypes. Previous studies in uninjured intestine of Sox9-EGFP reporter mice demonstrate that ISCs express low levels of Sox9-EGFP (Sox9-EGFP Low), whereas enteroendocrine cells (EEC) express high levels of Sox9-EGFP (Sox9-EGFP High). We hypothesized that Sox9-EGFP Low ISCs would expand after radiation, exhibit enhanced proliferative capacities, and adopt a distinct gene expression profile associated with rapid proliferation. Sox9-EGFP mice were given 14 Gy abdominal radiation and studied between days 3 and 9 postradiation. Radiation-induced changes in number, growth, and transcriptome of the different Sox9-EGFP cell populations were determined by histology, flow cytometry, in vitro culture assays, and microarray. Microarray confirmed that nonirradiated Sox9-EGFP Low cells are enriched for Lgr5 mRNA and mRNAs enriched in Lgr5-ISCs and identified additional putative ISC markers. Sox9-EGFP High cells were enriched for EEC markers, as well as Bmi1 and Hopx, which are putative markers of quiescent ISCs. Irradiation caused complete crypt loss, followed by expansion and hyperproliferation of Sox9-EGFP Low cells. From nonirradiated intestine, only Sox9-EGFP Low cells exhibited ISC characteristics of forming organoids in culture, whereas during regeneration both Sox9-EGFP Low and High cells formed organoids. Microarray demonstrated that regenerating Sox9-EGFP High cells exhibited transcriptomic changes linked to p53-signaling and ISC-like functions including DNA repair and reduced oxidative metabolism. These findings support a model in which Sox9-EGFP Low cells represent active ISCs, Sox9-EGFP High cells contain radiation-activatable cells with ISC characteristics, and both participate in crypt regeneration.


Subject(s)
Green Fluorescent Proteins/biosynthesis , Jejunum/radiation effects , SOX9 Transcription Factor/biosynthesis , Stem Cells/radiation effects , Animals , Cell Proliferation/radiation effects , Cells, Cultured , Gene Expression/radiation effects , Homeodomain Proteins/biosynthesis , Jejunum/metabolism , Mice , Mice, Transgenic , Nuclear Proteins/biosynthesis , Polycomb Repressive Complex 1 , Proto-Oncogene Proteins/biosynthesis , Receptors, G-Protein-Coupled/biosynthesis , Regeneration/radiation effects , Repressor Proteins/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...