Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
Am J Physiol Gastrointest Liver Physiol ; 302(12): G1359-63, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22461030

ABSTRACT

Many advances have been reported in the long-term culture of intestinal mucosal cells in recent years. A significant number of publications have described new culture media, cell formations, and growth patterns. Furthermore, it is now possible to study, e.g., the capabilities of isolated stem cells or the interactions between stem cells and mesenchyme. However, at the moment there is significant variation in the way these structures are described and named. A standardized nomenclature would benefit the ability to communicate and compare findings from different laboratories using the different culture systems. To address this issue, members of the NIH Intestinal Stem Cell Consortium herein propose a systematic nomenclature for in vitro cultures of the small and large intestine. We begin by describing the structures that are generated by preparative steps. We then define and describe structures produced in vitro, specifically: enterosphere, enteroid, reconstituted intestinal organoid, induced intestinal organoid, colonosphere, colonoid, and colonic organoid.


Subject(s)
Cell Culture Techniques/classification , Colon/cytology , Intestine, Small/cytology , Humans , Intestinal Mucosa/cytology , Stem Cells/cytology , Terminology as Topic
9.
J Surg Res ; 169(2): 169-78, 2011 Aug.
Article in English | MEDLINE | ID: mdl-20444471

ABSTRACT

Production of tissue engineered small intestine (TESI) has been limited by the relatively large amount of native tissue required to generate neomucosa. The influence of growth factors and three-dimensional (3D) extracellular matrices on TESI has been studied both in vitro and in vivo, and positive growth effects on tissue mass and differentiation were noted. The present study investigates the impact of single doses of glucagon-like peptide-2 (GLP-2), hepatocyte growth factor (HGF), or holo-transferrin adsorbed onto a polyglycolic (PGA) mesh scaffold using a rat small-intestinal organoid transplant model. In Experiment I, intestinal organoids were seeded onto PGA mesh discs, suspended in either Matrigel (n=8) or a vehicle control (n=8), and implanted into syngenic recipients. In Experiment II, GLP-2 (n=8), HGF (n=8), or transferrin (n=8) were adsorbed onto PGA mesh discs. Intestinal organoids were then suspended in Matrigel and seeded onto each growth factor-loaded PGA disc or onto control discs without growth factors (n=12). In addition, organoids were suspended in vehicle and seeded onto control discs (n=12). All discs were implanted into syngenic recipients. After 4 wk, histologic analysis of the samples revealed significantly greater neomucosal surface area (3.62±0.33 mm(2)versus 0.92±0.11 mm(2), P<0.0001) and cyst diameter (2.83±0.14 mm versus 2.06±0.07 mm, P<0.0001) in groups treated with Matrigel compared with vehicle controls. The addition of holo-transferrin to the scaffolds further augmented neomucosal surface area (9.11±0.66 mm(2)versus 3.01±0.22 mm(2), P<0.01), whereas that of GLP-2 stimulated the formation of increased numbers of cysts (8.88±0.46 versus 4.18±0.25, P<0.01). These data suggest that Matrigel and growth factors adsorbed to polymer scaffolds can be used to manipulate the morphology of TESI.


Subject(s)
Bioengineering/methods , Intercellular Signaling Peptides and Proteins/pharmacology , Intestinal Mucosa/drug effects , Polyglycolic Acid , Surgical Mesh , Animals , Animals, Newborn , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Glucagon-Like Peptide 2/pharmacology , Hepatocyte Growth Factor/pharmacology , Intestinal Mucosa/cytology , Male , Models, Animal , Pilot Projects , Rats , Rats, Inbred Lew , Transferrin/pharmacology
10.
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
11.
Langenbecks Arch Surg ; 395(8): 1083-91, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20614132

ABSTRACT

PURPOSE: Detailed understanding of the functional anatomy of the lower esophageal sphincter (LES) is germane to successful surgical treatment of esophageal disorders. However, a comprehensive concept of the structure-function relationship of the LES is currently lacking. METHODS: We reviewed published anatomic evidence, medical imaging, and impedance manometry data sets and developed a novel functional concept of the LES. RESULTS: Morphologic evidence accumulated over the past three decades indicates that the LES is an anatomic structure that differs markedly from typical ring sphincters of the gastrointestinal tract (e.g., upper esophageal sphincter or anal sphincters). Recent impedance manometry investigations shed new light on the functional anatomy of the LES. These data corroborate a concept of this sphincter as a double-layer, twisted stretch sphincter. This sphincter requires tissue tension for optimal function. Retightening of the longitudinal stretch of the esophagus provides an effective therapy if this tension is lost, e.g., for patients with hiatal hernias. Paralysis of the muscle fibers of this sphincter system results in functional obstruction, and this explains the pathophysiology of "achalasia". CONCLUSIONS: Based on available data, we propose a novel concept that better explains the structure-function relationship of the LES. Improved knowledge of the biomechanical factors of esophageal disorders can be expected to advance surgical treatment for these diseases.


Subject(s)
Esophageal Sphincter, Lower/physiopathology , Manometry/methods , Esophageal Achalasia/pathology , Esophageal Achalasia/physiopathology , Esophageal Achalasia/surgery , Esophageal Sphincter, Lower/pathology , Gastroesophageal Reflux/pathology , Gastroesophageal Reflux/physiopathology , Gastroesophageal Reflux/surgery , Hernia, Hiatal/pathology , Hernia, Hiatal/physiopathology , Hernia, Hiatal/surgery , Humans , Muscle, Smooth/pathology , Muscle, Smooth/physiopathology , Muscle, Smooth/surgery , Reference Values
12.
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
13.
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
14.
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
15.
Surgery ; 140(3): 423-34, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16934605

ABSTRACT

BACKGROUND: Orthotopic transplantation of intestinal mucosal organoids that contain putative mucosal stem cells serves as an important step toward implementing intestinal gene therapy and treatment for malabsorption syndromes in animals and humans. We hypothesized that intestinal mucosal organoids can be transplanted along the axis of the small bowel giving rise to a neomucosa expressing proteins of its donor origin. METHODS: Epithelial organoids were harvested from neonatal mice or rat small intestine with the use of a combination of enzymatic digestion with dispase and collagenase, and gravity sedimentation. In adult syngeneic recipients, a 7-cm segment of midjejunum was isolated, leaving its vascular pedicle intact. The remaining proximal and distal segments were anastomosed to restore intestinal continuity. The isolated segments were randomly subjected to surgical or chemical mucosectomy with a chelator solution for 30, 45, or 60 minutes and then compared. Histologic examination was used to confirm the presence of enterocytes, goblet cells, enteroendocrine cells, and Paneth cells in the neomucosal segments. To confirm the presence of ileal bile acid transporter (IBAT) gene message and function, we measured sodium-dependent bile acid uptake and IBAT-messenger RNA. Immunohistochemical examination using anti-IBAT antibodies was performed to demonstrate the expression of IBAT in the neomucosal segments. Experiments were repeated in a murine model transgenic for the green fluorescent protein to verify donor origin of the engrafted mucosa expressing IBAT. RESULTS: The area of peak IBAT function was found to be located in the terminal ileum. Organoid units harvested from this region were capable of generating a small-bowel neoileal mucosa after being seeded into the jejunum. This mucosa was histologically confirmed to differentiate into all 4 intestinal lineages and to express IBAT signal, confirming its donor-derived origin. Optimal engraftment of mucosa expressing the IBAT protein was found in isolated jejunal segments debrided for 45 minutes. Sodium-dependent bile acid uptake was 5-fold higher in the neoileum, compared with the jejunum. IBAT-mRNA levels in the neoileum were 18-100-fold higher than those in the jejunum. Areas of green fluorescent protein-positive mucosa stained positively with anti-IBAT antibody in adjacent sections, suggesting that the regenerated mucosa is from transplanted ileal stem cells. CONCLUSIONS: Orthotopic transplantation of epithelial organoids containing ileal stem cells was used to generate a neoileal mucosa that expressed all 4 intestinal lineages along with a new zone of active bile acid uptake and IBAT expression in a recipient jejunal segment.


Subject(s)
Intestinal Mucosa/transplantation , Malabsorption Syndromes/surgery , Organoids/transplantation , Stem Cell Transplantation/methods , Animals , Bile Acids and Salts/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Lineage , Female , Gene Expression Regulation , Genetic Therapy/methods , Ileum/cytology , Ileum/metabolism , Ileum/transplantation , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Malabsorption Syndromes/metabolism , Malabsorption Syndromes/pathology , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Transgenic , Organoids/cytology , Organoids/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Inbred Lew
16.
Rejuvenation Res ; 9(1): 20-5, 2006.
Article in English | MEDLINE | ID: mdl-16608391

ABSTRACT

A number of clinical conditions are caused by disorders affecting the mucosal lining of the gastrointestinal tract. Some patients suffer from a loss of mucosal surface area due to congenital defects or due to surgical resections ("short bowel syndrome"). Other patients have inborn or acquired defects of certain mucosal functions (e.g., glucose-galactose malabsorption, bile acid malabsorption). Many patients with these mucosal disorders could be more effectively treated if healthy mucosa were available in larger quantities as a replacement or functional supplement. We therefore developed methods to transplant mucosal stem cells from one part of the intestine to another and to make bioengineered intestinal mucosa. We generated an animal model of bile acid malabsorption using rats that underwent resection of the distal 25% of their small intestine (ileum). This resulted in significant losses of bile acids with the fecal excretions in these animals. We subsequently harvested ileal stem cell clusters from neonatal donors, removed the mucosa from a segment of proximal intestine (jejunum), and implanted the stem cell clusters into the debrided segment of jejunum. After four weeks, the animals had developed a functional "neomucosa." We inserted the "neo-ileal" segment into continuity as a substitute ileum. Postoperative measurements of fecal bile acid excretion showed that we were able to reverse the malabsorption syndrome in this model. This was the first reported neo-mucosa-based treatment of a malabsorption syndrome in vivo. We subsequently studied different biodegradable PGA and PLLA scaffoldings to generate bioengineered intestinal mucosa. We implanted these materials into omentum of rats and were able to identify a PGA/PLLA hybrid material on which engraftment rates of 36% of the available surface area could be achieved. Most recently, we developed a novel technique that permits direct observation of cell-biomaterial interactions after implantation into omentum or intestine in vivo. This method will help to optimize engraftment conditions for stem cell clusters on biomaterials.


Subject(s)
Intestinal Mucosa/cytology , Intestinal Mucosa/transplantation , Tissue Engineering , Animals , Ileum/cytology , Ileum/surgery , Jejunum/cytology , Jejunum/surgery , Malabsorption Syndromes/surgery , Rats , Stem Cells/cytology
17.
Cells Tissues Organs ; 184(3-4): 154-65, 2006.
Article in English | MEDLINE | ID: mdl-17409741

ABSTRACT

INTRODUCTION: Biodegradable polyester scaffolds have proven useful for growing neointestinal tissue equivalents both in vitro and in vivo. These scaffolds allow cells to attach and grow in a 3-dimensional space while nutrient flow is maintained throughout the matrix. The purpose of this study was to evaluate different biopolymer constructs and to determine mucosal engraftment rates and mucosal morphology. HYPOTHESIS: We hypothesized that different biopolymer constructs may vary in their ability to provide a good scaffolding onto which intestinal stem cell organoids may be engrafted. STUDY DESIGN: Eight different microporous biodegradable polymer tubes composed of polyglycolic acid (PGA), polylactic acid, or a combination of both, using different fabrication techniques were seeded with intestinal stem cell clusters obtained from neonatal rats. Three different seeded polymer constructs were subsequently placed into the omentum of syngeneic adult recipient rats (n = 8). Neointestinal grafts were harvested 4 weeks after implantation. Polymers were microscopically evaluated for the presence of mucosal growth, morphology, scar formation and residual polymer. RESULTS: Mucosal engraftment was observed in 7 out of 8 of the polymer constructs. A maximal surface area engraftment of 36% (range 5-36%) was seen on nonwoven, randomly entangled, small fiber PGA mesh coated with aerosolized 5% poly-L-lactic acid. Villous and crypt development, morphology and created surface area were best on PGA nonwoven mesh constructs treated with poly-L-lactic acid. Electrospun microfiber PGA had poor overall engraftment with little or no crypt or villous formation. CONCLUSION: Intestinal organoids can be engrafted onto biodegradable polyester scaffoldings with restitution of an intestinal mucosal layer. Variability in polymer composition, processing techniques and material properties (fiber size, luminal dimensions and pore size) affect engraftment success. Future material refinements should lead to improvements in the development of a tissue-engineered intestine.


Subject(s)
Absorbable Implants , Intestinal Mucosa/cytology , Intestinal Mucosa/physiology , Polyesters , Stem Cell Transplantation/methods , Tissue Engineering/methods , Animals , Animals, Newborn , Male , Polyglactin 910 , Rats , Rats, Inbred Lew , Transplantation, Isogeneic
18.
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
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL