Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
J Pediatr Surg ; 56(2): 346-351, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32709529

ABSTRACT

BACKGROUND/PURPOSE: Intraluminal springs have recently been shown to lengthen segments of intestine in a process known as distraction enterogenesis. We hypothesized that biocompatible springs could be used to lengthen defunctionalized murine small intestine and would lead to identifiable intestinal adaptations at the molecular level. METHODS: Age and weight matched C57BL/6 mice underwent surgical insertion of nitinol spring-loaded capsules into a Roux limb of jejunum. Segment lengths were measured at initial spring placement and at euthanasia after 14 and 21 days. Histology and gene expression of the Roux limb were evaluated at scarification and compared to untreated control segments. RESULTS: Intestinal segments loaded with compressed springs lengthened an average of 240%, which was significantly longer than control segments loaded with either empty capsules or uncompressed springs. Muscularis thickening was greater in spring-treated mice compared to controls without springs. Crypt depth and Lgr5+ expression was greater in mice that received compressed spring treatments when compared to control groups. CONCLUSIONS: Insertion of a compressed nitinol spring into a Roux limb results in significant intestinal lengthening, smooth muscle thickening, and Lgr5+ expression in a mouse model. The ability to increase small bowel length in a defunctionalized murine model may be used to understand the mechanism of distraction enterogenesis.


Subject(s)
Intestines/surgery , Short Bowel Syndrome , Tissue Expansion Devices , Animals , Jejunum/surgery , Mice , Mice, Inbred C57BL , Short Bowel Syndrome/surgery , Tissue Expansion
2.
Cell Transplant ; 29: 963689720903709, 2020.
Article in English | MEDLINE | ID: mdl-32907378

ABSTRACT

Human small intestinal crypts are the source of intestinal stem cells (ISCs) that are capable of undergoing self-renewal and differentiation to an epithelial layer. The development of methods to expand the ISCs has provided opportunities to model human intestinal epithelial disorders. Human crypt samples are usually obtained from either endoscopic or discarded surgical samples, and are thereby exposed to warm ischemia, which may impair their in vitro growth as three-dimensional culture as spheroids or enteroids. In this study we compared duodenal samples obtained from discarded surgical samples to those isolated from whole-body preserved cadaveric donors to generate in vitro cultures. We also examined the effect of storage solution (phosphate-buffered saline or University of Wisconsin [UW] solution) as well as multiple storage times on crypt isolation and growth in culture. We found that intestinal crypts were successfully isolated from cadaveric tissue stored for up to 144 h post-procurement and also were able to generate enteroids and spheroids in certain media conditions. Surgical samples stored in UW after procurement were sufficiently viable up to 24 h and also allowed the generation of enteroids and spheroids. We conclude that surgical samples stored for up to 24 h post-procurement in UW solution allowed for delayed crypt isolation and viable in vitro cultures. Furthermore, in situ, hypothermic preservation in cadaveric duodenal samples permitted crypt/ISC isolation, and successful culture of spheroids and enteroids from tissues held for up to 6 days post-procurement.


Subject(s)
Cell Culture Techniques/methods , Intestines/physiopathology , Cadaver , Cell Differentiation , Humans
3.
J Biol Chem ; 294(41): 15182-15192, 2019 10 11.
Article in English | MEDLINE | ID: mdl-31341016

ABSTRACT

Neurogenin-3 (NEUROG3) is a helix-loop-helix (HLH) transcription factor involved in the production of endocrine cells in the intestine and pancreas of humans and mice. However, the human NEUROG3 loss-of-function phenotype differs subtly from that in mice, but the reason for this difference remains poorly understood. Because NEUROG3 expression precedes exit of the cell cycle and the expression of endocrine cell markers during differentiation, we investigated the effect of lentivirus-mediated overexpression of the human NEUROG3 gene on the cell cycle of BON4 cells and various human nonendocrine cell lines. NEUROG3 overexpression induced a reversible cell cycle exit, whereas expression of a neuronal lineage homolog, NEUROG1, had no such effect. In endocrine lineage cells, the cellular quiescence induced by short-term NEUROG3 expression required cyclin-dependent kinase inhibitor 1A (CDKN1A)/p21CIP1 expression. Expression of endocrine differentiation markers required sustained NEUROG3 expression in the quiescent, but not in the senescent, state. Inhibition of the phosphatase and tensin homolog (PTEN) pathway reversed quiescence by inducing cyclin-dependent kinase 2 (CDK2) and reducing p21CIP1 and NEUROG3 protein levels in BON4 cells and human enteroids. We discovered that NEUROG3 expression stimulates expression of CDKN2a/p16INK4a and BMI1 proto-oncogene polycomb ring finger (BMI1), with the latter limiting expression of the former, delaying the onset of CDKN2a/p16INK4a -driven cellular senescence. Furthermore, NEUROG3 bound to the promoters of both CDKN1a/p21CIP1 and BMI1 genes, and BMI1 attenuated NEUROG3 binding to the CDKN1a/p21CIP1 promoter. Our findings reveal how human NEUROG3 integrates inputs from multiple signaling pathways and thereby mediates cell cycle exit at the onset of differentiation.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Cycle Checkpoints , Mitogen-Activated Protein Kinase 7/metabolism , Nerve Tissue Proteins/metabolism , PTEN Phosphohydrolase/metabolism , Cell Line , Cellular Senescence , Gene Expression Regulation , Genes, p16 , Humans , Proto-Oncogene Mas
4.
PLoS One ; 14(5): e0216326, 2019.
Article in English | MEDLINE | ID: mdl-31150401

ABSTRACT

Adult intestinal epithelial stem cells are a promising resource for treatment of intestinal epithelial disorders that cause intestinal failure and for intestinal tissue engineering. We developed two different animal models to study the implantation of cultured murine and human intestinal epithelial cells in the less differentiated "spheroid" state and the more differentiated "enteroid" state into the denuded small intestine of mice. Engraftment of donor cells could not be achieved while the recipient intestine remained in continuity. However, we were able to demonstrate successful implantation of murine and human epithelial cells when the graft segment was in a bypassed loop of jejunum. Implantation of donor cells occurred in a random fashion in villus and crypt areas. Engraftment was observed in 75% of recipients for murine and 36% of recipients for human cells. Engrafted spheroid cells differentiated into the full complement of intestinal epithelial cells. These findings demonstrate for the first time successful engraftment into the small bowel which is optimized in a bypassed loop surgical model.


Subject(s)
Epithelial Cells/transplantation , Intestine, Small/cytology , Animals , Cell Differentiation , Cells, Cultured , Disease Models, Animal , Graft Survival , Humans , Jejunum , Mice , Spheroids, Cellular/transplantation
5.
J Surg Res ; 215: 219-224, 2017 07.
Article in English | MEDLINE | ID: mdl-28688651

ABSTRACT

BACKGROUND: Distraction enterogenesis has been investigated as a novel treatment for short bowel syndrome (SBS). With variable intestinal sizes, it is critical to determine safe, translatable spring characteristics in differently sized animal models before clinical use. Nitinol springs have been shown to lengthen intestines in rats and pigs. Here, we show spring-mediated intestinal lengthening is scalable and feasible in a murine model. MATERIALS AND METHODS: A 10-mm nitinol spring was compressed to 3 mm and placed in a 5-mm intestinal segment isolated from continuity in mice. A noncompressed spring placed in a similar fashion served as a control. Spring parameters were proportionally extrapolated from previous spring parameters to accommodate the smaller size of murine intestines. After 2-3 wk, the intestinal segments were examined for size and histology. RESULTS: Experimental group with spring constants, k = 0.2-1.4 N/m, showed intestinal lengthening from 5.0 ± 0.6 mm to 9.5 ± 0.8 mm (P < 0.0001), whereas control segments lengthened from 5.3 ± 0.5 mm to 6.4 ± 1.0 mm (P < 0.02). Diameter increased similarly in both groups. Isolated segment perforation was noted when k ≥ 0.8 N/m. Histologically, lengthened segments had increased muscularis thickness and crypt depth in comparison to normal intestine. CONCLUSIONS: Nitinol springs with k ≤ 0.4 N/m can safely yield nearly 2-fold distraction enterogenesis in length and diameter in a scalable mouse model. Not only does this study derive the safe ranges and translatable spring characteristics in a scalable murine model for patients with short bowel syndrome, it also demonstrates the feasibility of spring-mediated intestinal lengthening in a mouse, which can be used to study underlying mechanisms in the future.


Subject(s)
Short Bowel Syndrome/surgery , Tissue Expansion Devices , Tissue Expansion/instrumentation , Alloys , Animals , Feasibility Studies , Mice , Mice, Inbred C57BL , Tissue Expansion/methods , Treatment Outcome
6.
Stem Cells Transl Med ; 6(2): 666-676, 2017 02.
Article in English | MEDLINE | ID: mdl-28191783

ABSTRACT

Intestinal failure is a rare life-threatening condition that results in the inability to maintain normal growth and hydration status by enteral nutrition alone. Although parenteral nutrition and whole organ allogeneic transplantation have improved the survival of these patients, current therapies are associated with a high risk for morbidity and mortality. Development of methods to propagate adult human intestinal stem cells (ISCs) and pluripotent stem cells raises the possibility of using stem cell-based therapy for patients with monogenic and polygenic forms of intestinal failure. Organoids have demonstrated the capacity to proliferate indefinitely and differentiate into the various cellular lineages of the gut. Genome-editing techniques, including the overexpression of the corrected form of the defective gene, or the use of CRISPR (clustered regularly interspaced short palindromic repeats)/Cas9 to selectively correct the monogenic disease-causing variant within the stem cell, make autologous ISC transplantation a feasible approach. However, numerous techniques still need to be further optimized, including more robust ex vivo ISC expansion, native ISC ablation, and engraftment protocols. Large-animal models can to be used to develop such techniques and protocols and to establish the safety of autologous ISC transplantation because outcomes in such models can be extrapolated more readily to humans. Stem Cells Translational Medicine 2017;6:666-676.


Subject(s)
Gene Editing/methods , Intestines/transplantation , Regeneration , Short Bowel Syndrome/surgery , Stem Cell Transplantation/methods , Stem Cells , Animals , CRISPR-Cas Systems , Cell Differentiation , Cell Lineage , Cell Proliferation , Gene Expression Regulation, Developmental , Humans , Intestines/pathology , Intestines/physiopathology , Recovery of Function , Risk Factors , Short Bowel Syndrome/genetics , Short Bowel Syndrome/pathology , Short Bowel Syndrome/physiopathology , Signal Transduction , Stem Cell Transplantation/adverse effects , Stem Cells/metabolism , Stem Cells/pathology , Treatment Outcome
7.
J Pediatr Surg ; 51(6): 995-1000, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26995514

ABSTRACT

PURPOSE: Current culture schema for human intestinal stem cells (hISCs) frequently rely on a 3D culture system using Matrigel™, a laminin-rich matrix derived from murine sarcoma that is not suitable for clinical use. We have developed a novel 2D culture system for the in vitro expansion of hISCs as an intestinal epithelial monolayer without the use of Matrigel. METHODS: Cadaveric duodenal samples were processed to isolate intestinal crypts from the mucosa. Crypts were cultured on a thin coat of type I collagen or laminin. Intestinal epithelial monolayers were supported with growth factors to promote self-renewal or differentiation of the hISCs. Proliferating monolayers were sub-cultured every 4-5days. RESULTS: Intestinal epithelial monolayers were capable of long-term cell renewal. Less differentiated monolayers expressed high levels of gene marker LGR5, while more differentiated monolayers had higher expressions of CDX2, MUC2, LYZ, DEF5, and CHGA. Furthermore, monolayers were capable of passaging into a 3D culture system to generate spheroids and enteroids. CONCLUSION: This 2D system is an important step to expand hISCs for further experimental studies and for clinical cell transplantation. LEVEL OF EVIDENCE: 1 Experimental.


Subject(s)
Epithelial Cells/cytology , Intestinal Mucosa/cytology , Stem Cells/cytology , Biocompatible Materials , Cadaver , Cell Culture Techniques , Collagen Type I , Humans , In Vitro Techniques , Laminin
8.
Cell Tissue Res ; 365(1): 123-34, 2016 07.
Article in English | MEDLINE | ID: mdl-26928041

ABSTRACT

Porcine models are useful for investigating therapeutic approaches to short bowel syndrome and potentially to intestinal stem cell (ISC) transplantation. Whereas techniques for the culture and genetic manipulation of ISCs from mice and humans are well established, similar methods for porcine stem cells have not been reported. Jejunal crypts were isolated from murine, human, and juvenile and adult porcine small intestine, suspended in Matrigel, and co-cultured with syngeneic intestinal subepithelial myofibroblasts (ISEMFs) or cultured without feeder cells in various culture media. Media containing epidermal growth factor, noggin, and R-spondin 1 (ENR medium) were supplemented with various combinations of Wnt3a- or ISEMF-conditioned medium (CM) and with glycogen synthase kinase 3 inhibitor (GSK3i), and their effects were studied on cultured crypts. Cell lineage differentiation was assessed by immunohistochemistry and quantitative polymerase chain reaction. Cultured porcine cells were serially passaged and transduced with a lentiviral vector. Whereas ENR medium supported murine enteroid growth, it did not sustain porcine crypts beyond 5 days. Supplementation of Wnt3a-CM and GSK3i resulted in the formation of complex porcine enteroids with budding extensions. These enteroids contained a mixture of stem and differentiated cells and were successfully passaged in the presence of GSK3i. Crypts grown in media supplemented with porcine ISEMF-CM formed spheroids that were less well differentiated than enteroids. Enteroids and spheroids were transfected with a lentivirus with high efficiency. Thus, our method maintains juvenile and adult porcine crypt cells long-term in culture. Porcine enteroids and spheroids can be successfully passaged and transduced by using lentiviral vectors.


Subject(s)
Aging/physiology , Intestines/cytology , Tissue Culture Techniques/methods , Animals , Cryopreservation , Culture Media, Conditioned/pharmacology , Immunohistochemistry , Intestinal Mucosa/metabolism , Mice , Myofibroblasts/cytology , Myofibroblasts/drug effects , Sus scrofa , Temperature , Transduction, Genetic
9.
PLoS One ; 11(1): e0148216, 2016.
Article in English | MEDLINE | ID: mdl-26820624

ABSTRACT

BACKGROUND & AIMS: Intestinal microfold (M) cells are specialized epithelial cells that act as gatekeepers of luminal antigens in the intestinal tract. They play a critical role in the intestinal mucosal immune response through transport of viruses, bacteria and other particles and antigens across the epithelium to immune cells within Peyer's patch regions and other mucosal sites. Recent studies in mice have demonstrated that M cells are generated from Lgr5+ intestinal stem cells (ISCs), and that infection with Salmonella enterica serovar Typhimurium increases M cell formation. However, it is not known whether and how these findings apply to primary human small intestinal epithelium propagated in an in vitro setting. METHODS: Human intestinal crypts were grown as monolayers with growth factors and treated with recombinant RANKL, and assessed for mRNA transcripts, immunofluorescence and uptake of microparticles and S. Typhimurium. RESULTS: Functional M cells were generated by short-term culture of freshly isolated human intestinal crypts in a dose- and time-dependent fashion. RANKL stimulation of the monolayer cultures caused dramatic induction of the M cell-specific markers, SPIB, and Glycoprotein-2 (GP2) in a process primed by canonical WNT signaling. Confocal microscopy demonstrated a pseudopod phenotype of GP2-positive M cells that preferentially take up microparticles. Furthermore, infection of the M cell-enriched cultures with the M cell-tropic enteric pathogen, S. Typhimurium, led to preferential association of the bacteria with M cells, particularly at lower inoculum sizes. Larger inocula caused rapid induction of M cells. CONCLUSIONS: Human intestinal crypts containing ISCs can be cultured and differentiate into an epithelial layer with functional M cells with characteristic morphological and functional properties. This study is the first to demonstrate that M cells can be induced to form from primary human intestinal epithelium, and that S. Typhimurium preferentially infect these cells in an in vitro setting. We anticipate that this model can be used to generate large numbers of M cells for further functional studies of these key cells of intestinal immune induction and their impact on controlling enteric pathogens and the intestinal microbiome.


Subject(s)
Intestinal Mucosa/cytology , Intestine, Small/cytology , Peyer's Patches/cytology , Stem Cells/cytology , Cell Culture Techniques/methods , Cell Differentiation , Cells, Cultured , Humans , Immunity, Mucosal , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestine, Small/immunology , Intestine, Small/microbiology , Peyer's Patches/immunology , Peyer's Patches/microbiology , RANK Ligand/immunology , Salmonella typhimurium/immunology , Stem Cells/immunology
10.
A A Case Rep ; 6(5): 121-3, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26462163

ABSTRACT

We report an unfortunate case of accidental administration of intrathecal gadolinium through an external ventricular drain in a postcraniotomy patient during magnetic resonance imaging of the brain. The incident occurred after the venous contrast line was connected mistakenly to the ventricular drainage catheter. The patient subsequently developed confusion, aphasia, and right facial droop with new computed tomography evidence of diffuse cerebral edema and stroke. Review of the magnetic resonance image revealed the inappropriate presence of subarachnoid gadolinium. Despite all interventions, the patient developed irreversible neurologic disability. We address the clinical sequelae, management strategies, and factors contributing to the catheter misconnection that led to this event.


Subject(s)
Brain Edema/etiology , Gadolinium/administration & dosage , Gadolinium/adverse effects , Medication Errors/adverse effects , Stroke/etiology , Critical Illness , Drainage , Humans , Magnetic Resonance Imaging/methods , Male , Middle Aged
11.
PLoS One ; 10(5): e0127258, 2015.
Article in English | MEDLINE | ID: mdl-26016485

ABSTRACT

The myofibroblast is an important stromal cell of the gastrointestinal tract. Current in vitro and in vivo models either do not accurately recreate stromal-epithelial interactions or are not specific to myofibroblasts. We sought to create an animal model that would allow the study of myofibroblast-epithelial interactions. We isolated and cultured colonic myofibroblasts from FVB mice. Cells were α-SMA and vimentin positive but desmin negative on immunoblot analysis. We injected the myofibroblasts into the colonic submucosa of syngeneic adult mice (n = 8) via a miniendoscopic system. We then isolated green fluorescent protein (GFP) positive colonic myofibroblasts from C57BL/6-Tg(CAG-EGFP)1Osb/J mice and injected them into the colonic lamina propria of C57BL/6J mice at 1x10(5) (n = 14), 1x10(6) (n = 9), or 5x10(6) cells/mL (n = 4). A subset of mice were injected with serum-free media and ink without cells (n = 3). Mice underwent repeat endoscopy and euthanasia one or 7 days after injection. Colons were isolated and either fixed in 10% formalin or the inked sites were individually excised and lysed for DNA. We assessed the injection sites via histology and immunohistochemical stains for α-SMA and GFP. We used qPCR to quantify GFP DNA transcripts at the lamina propria injection sites. Submucosal injection of myofibroblasts resulted in the formation of a subepithelial wheal on endoscopy, which persisted to day 7. Myofibroblasts injected either into the submucosa or lamina propria maintained viability on post-injection day 7 as evidenced by positive α-SMA staining. qPCR of lamina propria injections showed a dose-dependent increase in GFP DNA transcripts on post-injection day 1, whereas the number of transcripts on day 7 was equivalent for the concentrations injected. We demonstrate short-term survival of primary cultured colonic myofibroblasts in syngeneic mice. This may prove to be a valuable model for studying the role of myofibroblasts in states of health and disease.


Subject(s)
Colonoscopy , Immunocompetence , Injections , Myofibroblasts/cytology , Animals , Cell Survival , Colon/cytology , Female , Intestinal Mucosa/cytology , Male , Mice , Mice, Inbred C57BL , Time Factors
12.
Sci Rep ; 5: 8566, 2015 Apr 10.
Article in English | MEDLINE | ID: mdl-25858503

ABSTRACT

Exposure to high levels of ionizing radiation (IR) leads to debilitating and dose-limiting gastrointestinal (GI) toxicity. Using three-dimensional mouse crypt culture, we demonstrated that p53 target PUMA mediates radiation-induced apoptosis via a cell-intrinsic mechanism, and identified the GSK-3 inhibitor CHIR99021 as a potent radioprotector. CHIR99021 treatment improved Lgr5+ cell survival and crypt regeneration after radiation in culture and mice. CHIR99021 treatment specifically blocked apoptosis and PUMA induction and K120 acetylation of p53 mediated by acetyl-transferase Tip60, while it had no effect on p53 stabilization, phosphorylation or p21 induction. CHIR99021 also protected human intestinal cultures from radiation by PUMA but not p21 suppression. These results demonstrate that p53 posttranslational modifications play a key role in the pathological and apoptotic response of the intestinal stem cells to radiation and can be targeted pharmacologically.


Subject(s)
Apoptosis/drug effects , Apoptosis/genetics , Intestines/cytology , Radiation, Ionizing , Radiation-Protective Agents/pharmacology , Stem Cells/drug effects , Stem Cells/metabolism , Tumor Suppressor Protein p53/metabolism , Acetylation , Animals , Apoptosis Regulatory Proteins/deficiency , Apoptosis Regulatory Proteins/genetics , Cells, Cultured , Disease Models, Animal , Gastrointestinal Diseases/etiology , Gene Expression Regulation/drug effects , Gene Expression Regulation/radiation effects , Glycogen Synthase Kinase 3/antagonists & inhibitors , Humans , Mice , Mice, Knockout , Pyridines/pharmacology , Pyrimidines/pharmacology , Radiation Injuries, Experimental , Radiation Tolerance/drug effects , Radiation Tolerance/genetics , Radiotherapy/adverse effects , Receptors, G-Protein-Coupled/metabolism , Stem Cells/radiation effects , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics
13.
BMC Oral Health ; 15: 21, 2015 Feb 11.
Article in English | MEDLINE | ID: mdl-25888138

ABSTRACT

BACKGROUND: Anecdotal reports assert a relationship between weather and lunar activity and the odontogenic abscess (OA) incidence, but this relationship has not been validated. Therefore, the present study investigated the relationship between oral pain caused by OA and a variety of meteorological parameters and cyclic lunar activity. METHODS: The records of all dental emergency patients treated at the AllDent Zahnzentrum Emergency Unit in Munich, Germany during 2012 were retrospectively reviewed. Patients with oral pain who were diagnosed with OA and treated surgically (n = 1211) were included in the analysis. The OA incidence was correlated to daily meteorological data, biosynoptic weather analysis, and cyclic lunar activity. RESULTS: There was no seasonal variation in the OA incidence. None of the meteorological parameters, lunar phase, or biosynoptic weather class were significantly correlated with the OA incidence, except the mean barometric pressure, which was weakly correlated (rho = -0.204). The OA incidence showed a decreasing trend as barometric pressure increased (p < 0.001). On multiple linear regression, the barometric pressure accounted for approximately 4% of the OA incidence. CONCLUSION: There is no evidence supporting a correlation between the incidence of odontogenic abscess and the weather and lunar activities.


Subject(s)
Abscess/epidemiology , Moon , Tooth Diseases/epidemiology , Weather , Adolescent , Adult , Aged , Atmospheric Pressure , Ecological and Environmental Phenomena , Female , Germany/epidemiology , Humans , Humidity , Incidence , Male , Middle Aged , Mythology , Rain , Retrospective Studies , Seasons , Sunlight , Temperature , Toothache/epidemiology , Young Adult
14.
PLoS One ; 9(9): e107814, 2014.
Article in English | MEDLINE | ID: mdl-25222024

ABSTRACT

BACKGROUND: We previously reported in vitro maintenance and proliferation of human small intestinal epithelium using Matrigel, a proprietary basement membrane product. There are concerns over the applicability of Matrigel-based methods for future human therapies. We investigated type I collagen as an alternative for the culture of human intestinal epithelial cells. METHODS: Human small intestine was procured from fresh surgical pathology specimens. Small intestinal crypts were isolated using EDTA chelation. Intestinal subepithelial myofibroblasts were isolated from a pediatric sample and expanded in vitro. After suspension in Matrigel or type I collagen gel, crypts were co-cultured above a confluent layer of myofibroblasts. Crypts were also grown in monoculture with exposure to myofibroblast conditioned media; these were subsequently sub-cultured in vitro and expanded with a 1∶2 split ratio. Cultures were assessed with light microscopy, RT-PCR, histology, and immunohistochemistry. RESULTS: Collagen supported viable human epithelium in vitro for at least one month in primary culture. Sub-cultured epithelium expanded through 12 passages over 60 days. Histologic sections revealed polarized columnar cells, with apical brush borders and basolaterally located nuclei. Collagen-based cultures gave rise to monolayer epithelial sheets at the gel-liquid interface, which were not observed with Matrigel. Immunohistochemical staining identified markers of differentiated intestinal epithelium and myofibroblasts. RT-PCR demonstrated expression of α-smooth muscle actin and vimentin in myofibroblasts and E-Cadherin, CDX2, villin 1, intestinal alkaline phosphatase, chromogranin A, lysozyme, and Lgr5 in epithelial cells. These markers were maintained through several passages. CONCLUSION: Type I collagen gel supports long-term in vitro maintenance and expansion of fully elaborated human intestinal epithelium. Collagen-based methods yield familiar enteroid structures as well as a new pattern of sheet-like growth, and they eliminate the need for Matrigel for in vitro human intestinal epithelial growth. Future research is required to further develop this cell culture system for tissue engineering applications.


Subject(s)
Collagen Type I/metabolism , In Vitro Techniques , Intestinal Mucosa/cytology , Intestine, Small/cytology , Basement Membrane/cytology , Basement Membrane/metabolism , Coculture Techniques , Collagen/chemistry , Collagen Type I/chemistry , Drug Combinations , Extracellular Matrix/chemistry , Humans , Intestinal Mucosa/growth & development , Intestinal Mucosa/metabolism , Intestine, Small/growth & development , Intestine, Small/metabolism , Laminin/chemistry , Myofibroblasts/cytology , Proteoglycans/chemistry
15.
PLoS One ; 9(1): e84651, 2014.
Article in English | MEDLINE | ID: mdl-24400106

ABSTRACT

Intestinal epithelial stem cells (ISCs) are the focus of recent intense study. Current in vitro models rely on supplementation with the Wnt agonist R-spondin1 to support robust growth, ISC self-renewal, and differentiation. Intestinal subepithelial myofibroblasts (ISEMFs) are important supportive cells within the ISC niche. We hypothesized that co-culture with ISEMF enhances the growth of ISCs in vitro and allows for their successful in vivo implantation and engraftment. ISC-containing small intestinal crypts, FACS-sorted single ISCs, and ISEMFs were procured from C57BL/6 mice. Crypts and single ISCs were grown in vitro into enteroids, in the presence or absence of ISEMFs. ISEMFs enhanced the growth of intestinal epithelium in vitro in a proximity-dependent fashion, with co-cultures giving rise to larger enteroids than monocultures. Co-culture of ISCs with supportive ISEMFs relinquished the requirement of exogenous R-spondin1 to sustain long-term growth and differentiation of ISCs. Mono- and co-cultures were implanted subcutaneously in syngeneic mice. Co-culture with ISEMFs proved necessary for successful in vivo engraftment and proliferation of enteroids; implants without ISEMFs did not survive. ISEMF whole transcriptome sequencing and qPCR demonstrated high expression of specific R-spondins, well-described Wnt agonists that supports ISC growth. Specific non-supportive ISEMF populations had reduced expression of R-spondins. The addition of ISEMFs in intestinal epithelial culture therefore recapitulates a critical element of the intestinal stem cell niche and allows for its experimental interrogation and biodesign-driven manipulation.


Subject(s)
Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Myofibroblasts/metabolism , Stem Cells/metabolism , Animals , Cell Culture Techniques , Cell Proliferation , Cells, Cultured , Cluster Analysis , Gene Expression Profiling , Mice , Mice, Transgenic , Stem Cells/cytology , Transcriptome
16.
Am J Physiol Gastrointest Liver Physiol ; 305(8): G542-51, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23928185

ABSTRACT

Fluorescence-activated cell sorting (FACS) is an essential tool for studies requiring isolation of distinct intestinal epithelial cell populations. Inconsistent or lack of reporting of the critical parameters associated with FACS methodologies has complicated interpretation, comparison, and reproduction of important findings. To address this problem a comprehensive multicenter study was designed to develop guidelines that limit experimental and data reporting variability and provide a foundation for accurate comparison of data between studies. Common methodologies and data reporting protocols for tissue dissociation, cell yield, cell viability, FACS, and postsort purity were established. Seven centers tested the standardized methods by FACS-isolating a specific crypt-based epithelial population (EpCAM+/CD44+) from murine small intestine. Genetic biomarkers for stem/progenitor (Lgr5 and Atoh 1) and differentiated cell lineages (lysozyme, mucin2, chromogranin A, and sucrase isomaltase) were interrogated in target and control populations to assess intra- and intercenter variability. Wilcoxon's rank sum test on gene expression levels showed limited intracenter variability between biological replicates. Principal component analysis demonstrated significant intercenter reproducibility among four centers. Analysis of data collected by standardized cell isolation methods and data reporting requirements readily identified methodological problems, indicating that standard reporting parameters facilitate post hoc error identification. These results indicate that the complexity of FACS isolation of target intestinal epithelial populations can be highly reproducible between biological replicates and different institutions by adherence to common cell isolation methods and FACS gating strategies. This study can be considered a foundation for continued method development and a starting point for investigators that are developing cell isolation expertise to study physiology and pathophysiology of the intestinal epithelium.


Subject(s)
Epithelial Cells/physiology , Flow Cytometry/standards , Intestinal Mucosa/cytology , Animals , Cell Culture Techniques , Cell Survival , Gene Expression Regulation , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Male , Mice , Mice, Inbred C57BL , Observer Variation , Staining and Labeling
17.
Gastroenterology ; 145(2): 383-95.e1-21, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23644405

ABSTRACT

BACKGROUND & AIMS: Identification of intestinal stem cells (ISCs) has relied heavily on the use of transgenic reporters in mice, but this approach is limited by mosaic expression patterns and difficult to directly apply to human tissues. We sought to identify reliable surface markers of ISCs and establish a robust functional assay to characterize ISCs from mouse and human tissues. METHODS: We used immunohistochemistry, real-time reverse-transcription polymerase chain reaction, and fluorescence-activated cell sorting (FACS) to analyze intestinal epithelial cells isolated from mouse and human intestinal tissues. We compared different combinations of surface markers among ISCs isolated based on expression of Lgr5-green fluorescent protein. We developed a culture protocol to facilitate the identification of functional ISCs from mice and then tested the assay with human intestinal crypts and putative ISCs. RESULTS: CD44(+)CD24(lo)CD166(+) cells, isolated by FACS from mouse small intestine and colon, expressed high levels of stem cell-associated genes. Transit-amplifying cells and progenitor cells were then excluded based on expression of GRP78 or c-Kit. CD44(+)CD24(lo)CD166(+) GRP78(lo/-) putative stem cells from mouse small intestine included Lgr5-GFP(hi) and Lgr5-GFP(med/lo) cells. Incubation of these cells with the GSK inhibitor CHIR99021 and the E-cadherin stabilizer Thiazovivin resulted in colony formation by 25% to 30% of single-sorted ISCs. CONCLUSIONS: We developed a culture protocol to identify putative ISCs from mouse and human tissues based on cell surface markers. CD44(+)CD24(lo)CD166(+), GRP78(lo/-), and c-Kit(-) facilitated identification of putative stem cells from the mouse small intestine and colon, respectively. CD44(+)CD24(-/lo)CD166(+) also identified putative human ISCs. These findings will facilitate functional studies of mouse and human ISCs.


Subject(s)
Adult Stem Cells/metabolism , Antigens, Surface/metabolism , Intestinal Mucosa/cytology , Activated-Leukocyte Cell Adhesion Molecule/metabolism , Animals , CD24 Antigen/metabolism , Cell Culture Techniques , Colon/cytology , Colony-Forming Units Assay , Endoplasmic Reticulum Chaperone BiP , Flow Cytometry , Heat-Shock Proteins/genetics , Humans , Hyaluronan Receptors/metabolism , Intestine, Small/cytology , Mice , Proto-Oncogene Proteins c-kit/metabolism
18.
Stem Cells ; 31(9): 2024-30, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23553902

ABSTRACT

Recent seminal studies have rapidly advanced the understanding of intestinal epithelial stem cell (IESC) biology in murine models. However, the lack of techniques suitable for isolation and subsequent downstream analysis of IESCs from human tissue has hindered the application of these findings toward the development of novel diagnostics and therapies with direct clinical relevance. This study demonstrates that the cluster of differentiation genes CD24 and CD44 are differentially expressed across LGR5 positive "active" stem cells as well as HOPX positive "facultative" stem cells. Fluorescence-activated cell sorting enables differential enrichment of LGR5 (CD24-/CD44+) and HOPX (CD24+/CD44+) cells for gene expression analysis and culture. These findings provide the fundamental methodology and basic cell surface signature necessary for isolating and studying intestinal stem cell populations in human physiology and disease.


Subject(s)
CD24 Antigen/metabolism , Epithelial Cells/cytology , Hyaluronan Receptors/metabolism , Intestines/cytology , Stem Cells/cytology , Stem Cells/metabolism , Adult , Biomarkers/metabolism , Female , Humans , Middle Aged
19.
Tissue Eng Part C Methods ; 19(12): 961-9, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23566043

ABSTRACT

Methods for the in vitro culture of primary small intestinal epithelium have improved greatly in recent years. A critical barrier for the translation of this methodology to the patient's bedside is the ability to grow intestinal stem cells using a well-defined extracellular matrix. Current methods rely on the use of Matrigel(™), a proprietary basement membrane-enriched extracellular matrix gel produced in mice that is not approved for clinical use. We demonstrate for the first time the capacity to support the long-term in vitro growth of murine intestinal epithelium in monoculture, using type I collagen. We further demonstrate successful in vivo engraftment of enteroids co-cultured with intestinal subepithelial myofibroblasts in collagen gel. Small intestinal crypts were isolated from 6 to 10 week old transgenic enhanced green fluorescent protein (eGFP+) mice and suspended within either Matrigel or collagen gel; cultures were supported using previously reported media and growth factors. After 1 week, cultures were either lysed for DNA or RNA extraction or were implanted subcutaneously in syngeneic host mice. Quantitative real-time polymerase chain reaction (qPCR) was performed to determine expansion of the transgenic eGFP-DNA and to determine the mRNA gene expression profile. Immunohistochemistry was performed on in vitro cultures and recovered in vivo explants. Small intestinal crypts reliably expanded to form enteroids in either Matrigel or collagen in both mono- and co-cultures as confirmed by microscopy and eGFP-DNA qPCR quantification. Collagen-based cultures yielded a distinct morphology with smooth enteroids and epithelial monolayer growth at the gel surface; both enteroid and monolayer cells demonstrated reactivity to Cdx2, E-cadherin, CD10, Periodic Acid-Schiff, and lysozyme. Collagen-based enteroids were successfully subcultured in vitro, whereas pure monolayer epithelial sheets did not survive passaging. Reverse transcriptase-polymerase chain reaction demonstrated evidence of Cdx2, villin 1, mucin 2, chromogranin A, lysozyme 1, and Lgr5 expression, suggesting a fully elaborated intestinal epithelium. Additionally, collagen-based enteroids co-cultured with myofibroblasts were successfully recovered after 5 weeks of in vivo implantation, with a preserved immunophenotype. These results indicate that collagen-based techniques have the capacity to eliminate the need for Matrigel in intestinal stem cell culture. This is a critical step towards producing neo-mucosa using good manufacturing practices for clinical applications in the future.


Subject(s)
Collagen/chemistry , Extracellular Matrix/chemistry , Intestinal Mucosa/cytology , Intestine, Small/cytology , Animals , Antigens, Differentiation/metabolism , Cell Culture Techniques , Cells, Cultured , Gels/chemistry , Intestinal Mucosa/metabolism , Intestine, Small/metabolism , Mice , Mice, Transgenic
20.
J Hosp Med ; 7(9): 697-701, 2012.
Article in English | MEDLINE | ID: mdl-22961756

ABSTRACT

BACKGROUND: A structured, medical preoperative evaluation may positively impact the perioperative course of medically complex patients. Hospitalists are in a unique position to assist in preoperative evaluations, given their expertise with inpatient medicine and postoperative surgical consultation. OBJECTIVE: To evaluate specific outcomes after addition of a Hospitalist-run, medical Preoperative clinic to the standard Anesthesia preoperative evaluation. DESIGN, SETTING, PATIENTS: A pre/post retrospective, comparative review of outcomes of 5223 noncardiac surgical patients at a tertiary care Veterans Administration (VA) medical center. RESULTS: Length of stay was reduced for inpatients with an American Society of Anesthesia (ASA) score of 3 or higher (P < 0.0001). There was a trend towards a reduction in same-day, medically avoidable surgical cancellations (8.5% vs 4.9%, P = 0.065). More perioperative beta blockers were used (P < 0.0001) and more stress tests were ordered (P = 0.012). Inpatient mortality rates were reduced (1.27% vs 0.36%, P = 0.0158). CONCLUSION: A structured medical preoperative evaluation may benefit medically complex patients and improve perioperative processes and outcomes.


Subject(s)
Anesthesia/methods , Hospitalists/organization & administration , Outpatient Clinics, Hospital/organization & administration , Preoperative Care/methods , Tertiary Care Centers/organization & administration , Aged , Anesthesia/statistics & numerical data , Female , Humans , Male , Middle Aged , Outcome and Process Assessment, Health Care , Outpatient Clinics, Hospital/statistics & numerical data , Perioperative Care/statistics & numerical data , Quality Indicators, Health Care , Retrospective Studies , Tertiary Care Centers/statistics & numerical data , United States , United States Department of Veterans Affairs
SELECTION OF CITATIONS
SEARCH DETAIL
...