Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Daniel E. Levin is active.

Publication


Featured researches published by Daniel E. Levin.


Cell | 2015

Gremlin 1 Identifies a Skeletal Stem Cell with Bone, Cartilage, and Reticular Stromal Potential

Daniel L. Worthley; Michael Churchill; Jocelyn T. Compton; Yagnesh Tailor; Meenakshi Rao; Yiling Si; Daniel E. Levin; Matthew G. Schwartz; Aysu Uygur; Yoku Hayakawa; Stefanie Gross; Bernhard W. Renz; Wanda Setlik; Ashley N. Martinez; Xiaowei Chen; Saqib Nizami; Heon Goo Lee; H. Paco Kang; Jon-Michael Caldwell; Samuel Asfaha; C. Benedikt Westphalen; Trevor A. Graham; Guangchun Jin; Karan Nagar; Hongshan Wang; Mazen A. Kheirbek; Alka Kolhe; Jared Carpenter; Mark A. Glaire; Abhinav Nair

The stem cells that maintain and repair the postnatal skeleton remain undefined. One model suggests that perisinusoidal mesenchymal stem cells (MSCs) give rise to osteoblasts, chondrocytes, marrow stromal cells, and adipocytes, although the existence of these cells has not been proven through fate-mapping experiments. We demonstrate here that expression of the bone morphogenetic protein (BMP) antagonist gremlin 1 defines a population of osteochondroreticular (OCR) stem cells in the bone marrow. OCR stem cells self-renew and generate osteoblasts, chondrocytes, and reticular marrow stromal cells, but not adipocytes. OCR stem cells are concentrated within the metaphysis of long bones not in the perisinusoidal space and are needed for bone development, bone remodeling, and fracture repair. Grem1 expression also identifies intestinal reticular stem cells (iRSCs) that are cells of origin for the periepithelial intestinal mesenchymal sheath. Grem1 expression identifies distinct connective tissue stem cells in both the bone (OCR stem cells) and the intestine (iRSCs).


American Journal of Physiology-gastrointestinal and Liver Physiology | 2015

Human and mouse tissue-engineered small intestine both demonstrate digestive and absorptive function

Christa N. Grant; Salvador Garcia Mojica; Frederic G. Sala; J. Ryan Hill; Daniel E. Levin; Allison L. Speer; Erik R. Barthel; Hiroyuki Shimada; Nicholas C. Zachos; Tracy C. Grikscheit

Short bowel syndrome (SBS) is a devastating condition in which insufficient small intestinal surface area results in malnutrition and dependence on intravenous parenteral nutrition. There is an increasing incidence of SBS, particularly in premature babies and newborns with congenital intestinal anomalies. Tissue-engineered small intestine (TESI) offers a therapeutic alternative to the current standard treatment, intestinal transplantation, and has the potential to solve its biggest challenges, namely donor shortage and life-long immunosuppression. We have previously demonstrated that TESI can be generated from mouse and human small intestine and histologically replicates key components of native intestine. We hypothesized that TESI also recapitulates native small intestine function. Organoid units were generated from mouse or human donor intestine and implanted into genetically identical or immunodeficient host mice. After 4 wk, TESI was harvested and either fixed and paraffin embedded or immediately subjected to assays to illustrate function. We demonstrated that both mouse and human tissue-engineered small intestine grew into an appropriately polarized sphere of intact epithelium facing a lumen, contiguous with supporting mesenchyme, muscle, and stem/progenitor cells. The epithelium demonstrated major ultrastructural components, including tight junctions and microvilli, transporters, and functional brush-border and digestive enzymes. This study demonstrates that tissue-engineered small intestine possesses a well-differentiated epithelium with intact ion transporters/channels, functional brush-border enzymes, and similar ultrastructural components to native tissue, including progenitor cells, whether derived from mouse or human cells.


Journal of Pediatric Surgery | 2013

Human tissue-engineered small intestine forms from postnatal progenitor cells.

Daniel E. Levin; Erik R. Barthel; Allison L. Speer; Frederic G. Sala; Xiaogang Hou; Yasuhiro Torashima; Tracy C. Grikscheit

PURPOSE Tissue-engineered small intestine (TESI) represents a potential cure for short bowel syndrome (SBS). We previously reported full-thickness intestine formation using an organoid units-on-scaffold approach in rodent and swine models. Transplanted intestinal xenografts have been documented to survive from human fetal tissue but not from postnatal tissue. We now present the first report of human TESI from postnatal tissue. METHODS Organoid units (OU) were prepared from human small bowel resection specimens, loaded onto biodegradable scaffolds and implanted into NOD/SCID gamma chain-deficient mice. After 4 weeks, TESI was harvested and immunostained for β2-microglobulin to identify human tissue, villin for enterocytes, lysozyme for Paneth cells, chromogranin-A for enteroendocrine cells, mucin-2 for goblet cells, smooth muscle actin and desmin to demonstrate muscularis, and S-100 for nerves. RESULTS All TESI was of human origin. Immunofluorescence staining of human TESI reveals the presence of all four differentiated cell types of mature human small intestine, in addition to the muscularis and the supporting intestinal subepithelial myofibroblasts. Nerve tissue is also present. CONCLUSIONS Our technique demonstrates survival, growth, and differentiation of postnatally derived human small intestinal OU into full thickness TESI in murine hosts. This regenerative medicine strategy may eventually assist in the treatment of SBS.


Journal of Visualized Experiments | 2012

Tissue engineering of the intestine in a murine model.

Erik R. Barthel; Allison L. Speer; Daniel E. Levin; Frederic G. Sala; Xiaogang Hou; Yasuhiro Torashima; Clarence M. Wigfall; Tracy C. Grikscheit

Tissue-engineered small intestine (TESI) has successfully been used to rescue Lewis rats after massive small bowel resection, resulting in return to preoperative weights within 40 days.(1) In humans, massive small bowel resection can result in short bowel syndrome, a functional malabsorptive state that confers significant morbidity, mortality, and healthcare costs including parenteral nutrition dependence, liver failure and cirrhosis, and the need for multivisceral organ transplantation.(2) In this paper, we describe and document our protocol for creating tissue-engineered intestine in a mouse model with a multicellular organoid units-on-scaffold approach. Organoid units are multicellular aggregates derived from the intestine that contain both mucosal and mesenchymal elements,(3) the relationship between which preserves the intestinal stem cell niche.(4) In ongoing and future research, the transition of our technique into the mouse will allow for investigation of the processes involved during TESI formation by utilizing the transgenic tools available in this species.(5)The availability of immunocompromised mouse strains will also permit us to apply the technique to human intestinal tissue and optimize the formation of human TESI as a mouse xenograft before its transition into humans. Our method employs good manufacturing practice (GMP) reagents and materials that have already been approved for use in human patients, and therefore offers a significant advantage over approaches that rely upon decellularized animal tissues. The ultimate goal of this method is its translation to humans as a regenerative medicine therapeutic strategy for short bowel syndrome.


Current Opinion in Pediatrics | 2012

Tissue-engineering of the gastrointestinal tract.

Daniel E. Levin; Tracy C. Grikscheit

Purpose of review The purpose of this review is to describe recent advancements in tissue-engineering of the gastrointestinal system. For some patients, a congenital or acquired defect in the alimentary system results in digestive or nutritional deficiencies requiring intervention. Unfortunately, these treatments are associated with morbid complications. Advances in the growth of tissue-engineered esophagus, stomach, small intestine, colon and anus have been made in recent years. The progress reviewed here hopefully will someday benefit patients with gastrointestinal organ loss by providing a tissue replacement with morphology and function similar to native tissue. Recent findings In native gastrointestinal tissue, epithelial homeostasis is governed largely by the interaction of the stem cell and its surrounding cellular niche. In particular, the small intestinal stem cell populations identified as the crypt base columnar cell (CBCC) and at cell position 4 (cp4) are responsible for mucosal maintenance and response to injury. This work influences efforts to generate bioengineered tissues for both in-vitro mucosal models and full-thickness in-vivo tissue-engineered esophagus, stomach, intestine and colon. Summary Gastrointestinal organ loss is a challenge to manage. Current therapy can be life-saving, but is associated with morbid complications. Tissue-engineering will someday restore normal gastrointestinal function and eliminate the need for nutritional supplementation or transplant.


Regenerative Medicine | 2012

Human tissue-engineered colon forms from postnatal progenitor cells: an in vivo murine model

Erik R. Barthel; Daniel E. Levin; Allison L. Speer; Frederic G. Sala; Yasuhiro Torashima; Xiaogang Hou; Tracy C. Grikscheit

AIM Loss of colon reservoir function after colectomy can adversely affect patient outcomes. In previous work, human fetal intestinal cells developed epithelium without mesenchyme following implantation in mice. However, for humans, postnatal tissue would be the preferred donor source. We generated tissue-engineered colon (TEC) from postnatal human organoid units. MATERIALS & METHODS Organoid units were prepared from human colon waste specimens, loaded onto biodegradable scaffolds and implanted into immunocompromised mice. After 4 weeks, human TEC was harvested. Immunofluorescence staining confirmed human origin, identified differentiated epithelial cell types and verified the presence of supporting mesenchyme. RESULTS Human TEC demonstrated a simple columnar epithelium. Immunofluorescence staining demonstrated human origin and the three differentiated cell types of mature colon epithelium. Key mesenchymal components (smooth muscle, intestinal subepithelial myofibroblasts and ganglion cells) were seen. CONCLUSION Colon can form from human progenitor cells on a scaffold in a mouse host. This proof-of-concept experiment is an important step in transitioning TEC to human therapy.


American Journal of Physiology-gastrointestinal and Liver Physiology | 2015

Adult zebrafish intestine resection: a novel model of short bowel syndrome, adaptation, and intestinal stem cell regeneration

Kathy A. Schall; K. A. Holoyda; Christa N. Grant; Daniel E. Levin; E. R. Torres; A. Maxwell; H. A. Pollack; Rex Moats; Mark R. Frey; A. Darehzereshki; D. Al Alam; Ching-Ling Lien; Tracy C. Grikscheit

Loss of significant intestinal length from congenital anomaly or disease may lead to short bowel syndrome (SBS); intestinal failure may be partially offset by a gain in epithelial surface area, termed adaptation. Current in vivo models of SBS are costly and technically challenging. Operative times and survival rates have slowed extension to transgenic models. We created a new reproducible in vivo model of SBS in zebrafish, a tractable vertebrate model, to facilitate investigation of the mechanisms of intestinal adaptation. Proximal intestinal diversion at segment 1 (S1, equivalent to jejunum) was performed in adult male zebrafish. SBS fish emptied distal intestinal contents via stoma as in the human disease. After 2 wk, S1 was dilated compared with controls and villus ridges had increased complexity, contributing to greater villus epithelial perimeter. The number of intervillus pockets, the intestinal stem cell zone of the zebrafish increased and contained a higher number of bromodeoxyuridine (BrdU)-labeled cells after 2 wk of SBS. Egf receptor and a subset of its ligands, also drivers of adaptation, were upregulated in SBS fish. Igf has been reported as a driver of intestinal adaptation in other animal models, and SBS fish exposed to a pharmacological inhibitor of the Igf receptor failed to demonstrate signs of intestinal adaptation, such as increased inner epithelial perimeter and BrdU incorporation. We describe a technically feasible model of human SBS in the zebrafish, a faster and less expensive tool to investigate intestinal stem cell plasticity as well as the mechanisms that drive intestinal adaptation.


Methods of Molecular Biology | 2013

A “Living Bioreactor” for the Production of Tissue-Engineered Small Intestine

Daniel E. Levin; Frederic G. Sala; Erik R. Barthel; Allison L. Speer; Xiaogang Hou; Yasuhiro Torashima; Tracy C. Grikscheit

Here, we describe the use of a mouse model as a living bioreactor for the generation of tissue-engineered small intestine. Small intestine is harvested from donor mice with subsequent isolation of organoid units (a cluster of mesenchymal and epithelial cells). Some of these organoid units contain pluripotent stem cells with a preserved relationship with the mesenchymal stem cell niche. A preparation of organoid units is seeded onto a biodegradable scaffold and implanted intraperitoneally within the omentum of the host animal. The cells are nourished initially via imbibition until neovascularization occurs. This technique allows the growth of fully differentiated epithelium (composed of Paneth cells, goblet cells, enterocytes and enteroendocrine cells), muscle, nerve, and blood vessels of donor origin. Variations of this technique have been used to generate tissue-engineered stomach, large intestine, and esophagus. The variations include harvest technique, length of digestion, and harvest times.


Journal of Tissue Engineering and Regenerative Medicine | 2016

Fgf10 overexpression enhances the formation of tissue-engineered small intestine.

Yasuhiro Torashima; Daniel E. Levin; Erik R. Barthel; Allison L. Speer; Frederic G. Sala; Xiaogang Hou; Tracy C. Grikscheit

Short bowel syndrome (SBS) is a morbid and mortal condition characterized in most patients by insufficient intestinal surface area. Current management strategies are inadequate, but tissue‐engineered small intestine (TESI) offers a potential therapy. A barrier to translation of TESI is the generation of scalable mucosal surface area to significantly increase nutritional absorption. Fibroblast growth factor 10 (Fgf10) is a critical growth factor essential for the development of the gastrointestinal tract. We hypothesized that overexpression of Fgf10 would improve the generation of TESI. Organoid units, the multicellular donor tissue that forms TESI, were derived from Rosa26rtTA/+, tet(o)Fgf10/– or Fgf10Mlc‐nlacZ‐v24 (hereafter called Fgf10lacZ ) mice. These were implanted into the omentum of NOD/SCID γ‐chain‐deficient mice and induced with doxycycline in the case of tet(o)Fgf10/–. Resulting TESI were explanted at 4 weeks and studied by histology, quantitative RT–PCR and immunofluorescence. Four weeks after implantation, Fgf10 overexpressing TESI was larger and weighed more than the control tissues. Within the mucosa, the villus height was significantly longer and crypts contained a greater percentage of proliferating epithelial cells. A fully differentiated intestinal epithelium with enterocytes, goblet cells, enteroendocrine cells and Paneth cells was identified in the Fgf10‐overexpressing TESI, comparable to native small intestine. β‐Galactosidase expression was found in both the epithelium and the mesenchyme of the TESI derived from the Fgf10LacZ duodenum. However, this was not the case with TESI generated from jejunum and ileum. We conclude that Fgf10 enhances the formation of TESI. Copyright


Expert Review of Medical Devices | 2011

Tissue-engineered small intestine

Daniel E. Levin; Justin M.L. Dreyfuss; Tracy C. Grikscheit

“An ideal intestinal replacement would be derived from immunocompatible cells, would grow with the recipient, and be fully capable of performing all metabolic and physiologic functions of native small bowel.” Short bowel syndrome (SBS) is defined by both the metabolic and physiologic disturbances observed with significant truncation of normal bowel length. With a 70–75% reduction in small intestine length, alterations in fluid and nutrient bal ance occur with potentially life threatening consequences. These include profound decrease in the absorption of carbohydrates and protein as well as severe mineral and vitamin deficiencies. Additional sequalae include formation of renal and biliary calculi, low body weight, failure to thrive and death [1–3]. Both adults and children may be affected. In the adult population, bowel resection secondary to mesenteric ischemia, trauma, inflammatory bowel disease and malignancy are common etiologies. Children are susceptible to a wide range of congenital or acquired conditions such as intestinal atresia, volvulus and necrotizing enterocolitis. The incidence of SBS is estimated to be 24.5 per 100,000 live births and is associated with 30% 5-year mortality [2]. Current treatment options are inadequate and are focused on nutritional supplementation via total parenteral nutrition or intestinal replacement with transplanted allograft. The risks of hepatic failure and central line sepsis associated with prolonged total parenteral nutrition have been described [4]. Although outcomes for small intestine transplantation are improving, this therapy has disadvantages of supply shortages, rejection and lifelong immunosuppression. An ideal intestinal replacement would

Collaboration


Dive into the Daniel E. Levin's collaboration.

Top Co-Authors

Avatar

Tracy C. Grikscheit

Children's Hospital Los Angeles

View shared research outputs
Top Co-Authors

Avatar

Allison L. Speer

Children's Hospital Los Angeles

View shared research outputs
Top Co-Authors

Avatar

Erik R. Barthel

Children's Hospital Los Angeles

View shared research outputs
Top Co-Authors

Avatar

Xiaogang Hou

Children's Hospital Los Angeles

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Frederic G. Sala

Children's Hospital Los Angeles

View shared research outputs
Top Co-Authors

Avatar

Christa N. Grant

Children's Hospital Los Angeles

View shared research outputs
Top Co-Authors

Avatar

Ryan G. Spurrier

Children's Hospital Los Angeles

View shared research outputs
Top Co-Authors

Avatar

Justin M.L. Dreyfuss

Children's Hospital Los Angeles

View shared research outputs
Top Co-Authors

Avatar

Andre Panossian

Children's Hospital Los Angeles

View shared research outputs
Researchain Logo
Decentralizing Knowledge