Review Open Access
Copyright ©The Author(s) 2016. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Transplant. Mar 24, 2016; 6(1): 1-9
Published online Mar 24, 2016. doi: 10.5500/wjt.v6.i1.1
New approaches to increase intestinal length: Methods used for intestinal regeneration and bioengineering
Ali Shirafkan, Mauro Montalbano, Joshua McGuire, Cristiana Rastellini, Luca Cicalese, Texas Transplant Center, Department of Surgery, University of Texas Medical Branch, Galveston, TX 77555, United States
Author contributions: All authors equally contributed to this paper and design of the study, literature review, editing and final approval of the final version.
Conflict-of-interest statement: No conflicts of interest to disclose.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Luca Cicalese, MD, FACS, Professor, Texas Transplant Center, Department of Surgery, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555, United States. lucicale@utmb.edu
Telephone: +1-409-7722405 Fax: +1-409-7477364
Received: October 1, 2015
Peer-review started: October 9, 2015
First decision: November 4, 2015
Revised: December 27, 2015
Accepted: January 8, 2016
Article in press: January 11, 2016
Published online: March 24, 2016

Abstract

Inadequate absorptive surface area poses a great challenge to the patients suffering a variety of intestinal diseases causing short bowel syndrome. To date, these patients are managed with total parenteral nutrition or intestinal transplantation. However, these carry significant morbidity and mortality. Currently, by emergence of tissue engineering, anticipations to utilize an alternative method to increase the intestinal absorptive surface area are increasing. In this paper, we will review the improvements made over time in attempting elongating the intestine with surgical techniques as well as using intestinal bioengineering. Performing sequential intestinal lengthening was the preliminary method applied in humans. However, these methods did not reach widespread use and has limited outcome. Subsequent experimental methods were developed utilizing scaffolds to regenerate intestinal tissue and organoids unit from the intestinal epithelium. Stem cells also have been studied and applied in all types of tissue engineering. Biomaterials were utilized as a structural support for naive cells to produce bio-engineered tissue that can achieve a near-normal anatomical structure. A promising novel approach is the elongation of the intestine with an acellular biologic scaffold to generate a neo-formed intestinal tissue that showed, for the first time, evidence of absorption in vivo. In the large intestine, studies are more focused on regeneration and engineering of sphincters and will be briefly reviewed. From the review of the existing literature, it can be concluded that significant progress has been achieved in these experimental methods but that these now need to be fully translated into a pre-clinical and clinical experimentation to become a future viable therapeutic option.

Key Words: Bioengineered intestine, Tissue engineered, Scaffolds, Organoids, Stem cells, Intestinal elongation techniques

Core tip: Several methods were used to elongate the short and insufficient segment of intestine in patients suffering short bowel syndrome. These methods include transplantation of an intestinal graft, intestinal elongation, and techniques to create a bioengineered segment of intestine. Innovations in using stem cells, organoid units of intestine and bio-scaffolds allow the modern medicine to engineer segments of functional intestinal tissue in animal models. However, to reach the goal of implanting a fully functional bioengineered intestine in human improvements are still required. This article will review various methods to approach this condition from surgical techniques to elongate the intestine to the application of stem cells and bio scaffolds for creating three dimensional intestinal structure.



INTRODUCTION

Intestinal absorptive function is the result of fine regulation between different cell types and signaling, cooperating within this organ. Intestinal failure is the consequence of various diseases that limit intestinal length or function. These include, but are not limited to: Intestinal atresia, gastroschisis, pseudo-obstruction, motility disorders, Crohn’s disease, mesenteric thrombosis, intestinal necrosis, trauma and lead to short bowel syndrome. When the remaining portion of the intestine is functionally insufficient, intestinal failure results and this is characterized by fluid imbalance, electrolyte loss and altered nutrients absorption[1].Total parenteral nutrition (TPN) has been used as a treatment option, however, hepatic insufficiency, catheter related thrombosis and sepsis are the most significant limiting factors[2-5].

Intestinal transplantation offers a physiologic cure in the treatment of these patients as an alternative treatment[6]. Limitations of intestinal transplantation include sepsis and infections, chronic immunosuppression to avoid rejection and shortage in optimal organ donors[7]. Various techniques have been proposed to develop a safe and functional method to take advantages of bioengineering in the field of intestinal elongation. In this article, we will review the current knowledge on this subject, explain the limitation and benefits of each method and finally elaborate on the future direction and goals.

In general, the methods in intestinal tissue engineering can be classified into the following groups: Surgical techniques that can physically elongate the patient’s intestinal length; development of intestinal tissue using stem cells (SCs) in culture; development of organoid units from intestinal cells implanted on biologic materials in vivo and then incorporated in continuity with the intestine; utilization of biologic scaffold in vivo to obtain a neo-formed intestinal segment.

SURGICAL TECHNIQUES

Early surgical procedures to address short bowel syndrome attempted to increase nutrient absorption prolonging food transit time. Those procedures included vagotomy and pyloroplasty procedures, reversing small intestine segment, pouch formation, and prejejunal or preileal colon transposition[8-14]. In the early 1980s, Bianchi[15] described a reproducible technique to increase the length of the small intestine. Briefly, the procedure consisted in dividing an intestinal loop longitudinally in the midline where the vessels alternately go to one or other side of the loop from the mesentery. Then each side would be sutured to form a hemiloop. The final step was to anastomose the newly formed loops iso-peristaltically. As a result, the length of that bowel loop would be doubled, however, the diameter was halved. The advantage of this procedure was preservation of all available mucosa while tailoring the intestine length[15,16].

An alternative approach, called serial transverse enteroplasty (STEP), was introduced in early 2000. Following intentional dilatation of the small bowel, surgical stapling would be performed in an alternating direction from side to side in a “zig-zag fashion” perpendicular to the long axis of the bowel to elongate the existing small intestine. This procedure would be basically equivalent of the Bianchi procedure, however STEP had several theoretic advantages. The procedure was easier to perform and there was no need for anastomoses. Additionally, the intestine would never be opened, and the mesentery would never be jeopardized. In contrast, the over-all theoretical increase in length would depend on the amount of bowel dilatation and the size of the created intestinal lumen[17].

However, the patients who had undergone the Bianchi procedure would wean off TPN more than those with STEP, and they eventually would require intestinal transplants more than those with STEP. In addition, STEP was shown to be associated with higher rates of complication[18]. A study describes results from 38 patients who underwent STEP procedure for different diagnosis including intestinal atresia, gastroschisis with or without volvulus and necrotizing enterocolitis. Overall, the mean intestinal length increased considerably. The percentage of total calories tolerated enterally also increased. The most common complication was: Staple line leak, obstruction and abscess. It should be acknowledged that both these procedures have an acceptable short-term outcome while bridging the patients to intestinal transplants and do not seem to constitute a permanent treatment for intestinal failure[19].

SCS

SCs application in regenerative medicine is relatively new. The peculiarity of SCs differentiation is based on their plasticity and mainly on the microenvironment in which they are placed. Recently, it was shown that bone marrow derived hematopoietic stem cells (HSCs) after transplantation in mice, lethally irradiated with 60Cobalt, induce regeneration of gastrointestinal tissues[20]. Bone marrow mesenchymal stromal cells (BMMSCs) are able to mitigate lethal intestinal injury and their intravenous injection will increase the level of intestinal growth factors in the blood and induce regeneration of the intestinal SCs niche of the irradiated host[21].

Utilizing soluble growth factors, like epidermal growth factor (EGF) and hepatic growth factor (HGF), in the culture medium of intestinal SCs improves results obtained by increasing the homing of transplanted cells[22]. Supporting stem cell application, Qu et al[23] reported that transplantation of BMMSCs and soluble stem cell factors cooperate in regeneration of GI mucosa in a rat model in which indomethacin-induced GI injury was performed.

Hori et al[24] in 2002 seeded autologous mesenchymal stem cells (MSCs) on a collagen sponge graft to evaluate intestinal regeneration. Despite a complete mucosa was developed, they did not induce regeneration of the muscle layers. To develop smooth muscle cells with peristaltic features, Yoshida et al[25] employed induced pluripotent stem cells (iPSCs) from mice to induce differentiation of the muscularis into active and functional intestinal smooth muscle cells. However, they were not able to control the produced differentiated cells, since they include cardiac-like cells, mucosal cells and smooth muscle cells.

The intestine is a complex organ composed by many cell types. Today, no SC sources permit the generation of all cell types. During the last years, many studies analyzed stem-cell differentiation mechanisms. Studies on population of muscle-derivedstem cells confirmed that they are capable of self-renewal and multi-lineage differentiation including the ability to differentiate into intestinal smooth muscle cells[15,16].

Neuronal progenitor cells are present both in the central nervous system as well as enteric nervous system (ENS). Advances in cell culture techniques allowed isolation of enteric stem/progenitor cells and glial precursor cells. Several groups were able to isolate the neuronal crest-derived cells by sorting according to the markers for Sox10, p75 and Nestin. Following transplantation of these cells in the aganglionic bowel of mice Ret (-/-), the ENS was rebuilt[26].

Interestingly, it has been shown that inducing the CNS-neuronal progenitor cells with gut-derived soluble growth factors, will cause these cells to acquire enteric neuronal phenotype[27]. Likewise, transfected BMMSCs with glial cell-derived neurotrophic factor (GDNF) and Neurotrophin-3 (NT-3) genes, resulted in differentiation of BMMSCs into neuron-like cells with expression of neuronal markers as MAP-2 and GFAP[28,29].

In 2011, Spence et al[20] mimicked embryonic intestinal development in an in vitro model by using a series of specific growth factors at different time points and they successfully induced human pluripotent stem cells (PSCs) to differentiate into the new intestinal epithelium tissue and crypt-villus units. In order to mimic the natural intestinal peristalsis and physiology in vitro, Kim et al[30] developed a microfluidic “Gut-on-a-Chip” technology that exposed established epithelial cell lines to physiological peristalsis motions and liquid flow. This particular condition spontaneously induced morphogenesis of three-dimensional intestinal villi. However, these studies supported SCs applications, these in vitro models can only partially reiterate the whole in vivo intestinal complexity including absorptive or enteric barrier functions, and are far from offering a complete intestinal tissue that could be utilized in an in vivo model.

SCS AND BIO-SCAFFOLDS

SCs use has been improved by the attempt to create a three-dimensional (3-D) gel supporting structure system in vitro but this remains a major challenge for translational studies. McCracken et al[29] enhanced the 3-D tissue culture model. They transformed the PSCs implanted on a matrigel layer for a period of one to three months into intestinal mesenchyme and epithelium.

Generation of 3-D milieu provides a microenvironment with superior cell-cell interaction and communication that mimic an in vivo condition. For this aim, tissue engineering has used biocompatible scaffolds. Polymeric materials have two main characteristics; they are bio inert and easily biodegradable while they support all cell functions including adhesion, proliferation and differentiation.

Many studies supported that, these scaffolds provide a matrix for the seeding of cells in high density, which promotes reorganization of a functional tissue in a shorter time-frame. Biodegradable materials must give a perfect mechanical support until cells become able to produce extracellular matrix and other cellular factors. Then they are obligated to be wiped out gradually while being replaced by cellular and extracellular components. Persistence of these materials in the body and prolonged exposition to them can trigger an inflammatory response in the implantation site. Kim et al[31] used biodegradable matrices of polyglycolic acid (PGA) fibers, and seeded smooth muscle cells in tissue culture dishes (static seeding) and a cell suspension in spinner flasks (stirred seeding). They observed that seeding with dynamic model produced more uniform distribution and resulted in a neo-formed tissue with higher cellularity and greater elastin deposition. In the course of optimization of the tissue engineering methods, Qin et al[32] isolated intestinal smooth muscle cells from rats and seeded them in small intestinal submucosa (SIS) that is an acellular porcine-derived collagen-based matrix. SIS were implanted in an adult rat jejunal interposition model. Cell-seeded SIS displayed significantly improvement in contracting ability in respect to the SIS when no cells are seeded. However, there were no organized smooth muscle cell layers. Totonelli et al[33] and Maghsoudlou et al[34] used a detergent enzymatic treatment (DET) procedure to wash the cellular components of the rat’s intestine and to construct a natural acellular intestinal scaffold for regeneration of new intestinal tissue. The yielded scaffolds preserved the native architecture and connective tissue components.

Nakase et al[35] used a mixture of autologous smooth muscle cells from the stomach wall of a canine model with collagen solution, which was poured into a sponge to develop a collagen scaffold. Then, these structures have been implanted into the isolated defects of ileum as a patch graft. After 12 wk, the patch turned into relatively well-developed regenerated epithelium, villi and a smooth muscle layer in the lamina propria, however, the lack of contraction of these grafts presented as a significant problem.

Autologous MSCs from bone marrow were used by Hori et al[24] and seeded onto collagen scaffolds to induce the regeneration of a muscular layer. One month after implantation, they observed regeneration of the intestine with a muscular layer at the reconstructed site by - smooth muscle actin positive cells; however, this layer was thin and disappeared by 16 wk.

To stimulate proliferation of smooth muscle layer and angiogenesis, Lee et al[36] used basic fibroblast growth factors (bFGF). They compared two different concentrations of local administration of bFGF with the control. They found that incorporation of bFGF into the collagen coating layer of scaffolds would result in a significantly higher density of cells and blood vessels. They also found that when the bFGF is incorporated in encapsulated poly D, L-lactic-co-glycolic acid microsphere, it is more effective than its simple employment in collagen scaffolds suggesting that the addition of specific growth factors improves scaffold performance.

Previously, Zakhem et al[37] utilized a composite chitosan/collagen scaffold three-dimensional matrix to support the smooth muscle cells to restore lost innervation. They grew the rabbit colonic circular smooth muscle cells (RCSMCs) on chitosan-coated plates with a ratio of 1:1 and observed that cells maintained their morphology and physiologic functionality over time. The muscle constructs contracted in response to acetylcholine and potassium chloride and they relaxed in response to vasoactive intestinal peptide. Furthermore, they showed that this scaffold supports neo-innervation of non-innervated smooth muscle cells[38].

In 2015, Zakhem et al[38] showed that neural progenitor cells derived from the appendix and small intestine, will differentiate into mature functional enteric neurons, should they be incorporated in bioengineered internal anal sphincters. Raghavan et al[39,40] found that according to the extracellular matrix microenvironment of culture medium, enteric neuronal progenitor cells, will generate excitatory or inhibitory neuronal subtypes. Microenvironment enriched with collagen I and laminin resulted in contraction pattern, collagen IV induced a nitrergic neuronal population (neurons where transmission is mediated by nitric oxide) and laminin and/or heparin sulfate resulted in a balanced expression of relaxant and contractile motor neurons.

ORGANOID UNITS ON BIO-SCAFFOLDS

Another approach to regenerate intestinal tissue employs the use of organoids. Haffen et al[41] in the 1980s, demonstrated that intestinal crypt cells require interacting with mesenchymal cells for survival, proliferation and differentiation. Then Organ et al[42] isolated progenitor cells from the intestinal crypt and seeded them onto sheets of polyglycolic acid. They observed generation of stratified epithelium suggestive of fetal intestinal development. Of the limitations of this technique was the absence of epithelial-mesenchymal cell-cell interaction, which is thought to be of importance in organogenesis. Subsequently, Tait et al[43] demonstrated that dissociated post-natal small intestinal epithelium of rats, will generate small intestine-like structures when transplanted in the subcutaneous plane of adult rats. They confirmed that those small aggregates of intestinal epithelium and stroma are able to generate the required signals for 3-D regeneration of intestinal tissue. Then Choi and Vacanti[44], developed a villus structure with a core of mesenchymal stromal cells overlaid with epithelium called “Organoid Unit”. They believed that these units possess the epithelial-mesenchymal interaction required for mucosal regeneration. They seeded the organoid units isolated from neonatal rat intestine, and seeded them on poly glycolic acid scaffolds. They implanted them into the rats’ omentum and observed that cysts were generated after 8 wk, composed of columnar epithelium, Paneth’s cells, goblet cells, and crypt-villus-like structures.

To improve their previous work, Choi et al[45] later demonstrated that by collagen coating the scaffolds, the cells engraftment will enhance significantly and cyst sizes will be larger. Since it was known that the small intestine is a dynamic organ and responds differently to various factors, Vacanti’s lab, also investigated the effect of massive small bowel resections, partial hepatectomy and portocaval shunt on the development of organoid units. These interventions would increase the serum level of the epithelial growth factor (EGF) and hepatocyte growth factor (HGF). Interestingly, they observed that the length and diameter are larger and the villus numbers, height, area and mucosal surface are significantly greater in the group with resected small bowel[46]. As the next step, to evaluate the effect of incorporation of these organoid units in the intestine, they anastomosed the units side-to-side to the jejunum after three wk of implantation. They demonstrated that anastomosis had no complication. It also had trophic effects on the villus number, height, and surface length[47]. However, they also described a patchy distribution of the obtained neo mucosa[48].

Later, Grikscheit et al[49,50] adapted the organoid unit transplantation technique to develop tissue engineered colon. They produced organoid units from the rats’ sigmoid colon and implanted them into the omentum. Then, these organoids were anastomosed to the ileum of the rats that previously underwent ileostomies. After 41 d, they found the rats had less stool transit time and moisture content. Histology also showed a normal large intestine architecture including epithelium, vasculature, ganglion cells, and muscularis propria.

To evaluate the function of the tissue engineered small intestine (TESI), Grikscheit et al[51] replaced small intestine with these TESIs. After development of TESIs, they anastomosed them side-to-side to the duodenum, when the rats had 95% of their small bowel resected. Forty days post operation, they found an appropriate architecture and a well formed muscularis mucosa with appropriately distributed Aurbach and Meissner’s plexus and increased blood levels of B-12.

Following the successful results of TESI in rat model, Sala et al[52] transitioned this model in mice to take advantage of transgenic tools available in this species for studying the processes involved in formation of tissue engineered intestine. They found that TESI contains all four differentiated epithelial cell types present in the native small intestine including Goblet, Paneth, Enteroendocrine, and microvilli. They also confirmed that TESI contains innervated muscularis as well as presence of intact stem cell niche.

These investigators, also studied as a preclinical model an autologous-derived organoid unit transplantation in a large animal model. They generated organoid units from a short segment of jejunum of a swine model and implanted them onto omentum to the autologous host. They found that the TESIs replicated the native intestine with all epithelium, muscularis mucosa and stem cell niche[53].

Levin et al[54] investigated the possibility of development of organoid units from the postnatal human small intestine. They implanted organoid units, loaded on polyglycolic acid scaffolds in mice omentum. After 4 wk, they found all TESIs were of human origin with all differentiated cell types of mature human small intestine as well as muscularis and nerve tissue. This study was critical since the majority of the patients acquire the pathology after birth and the tissue engineering should be able to develop the tissues from post-natal stem cells. Then, recently they confirmed that both TESIs derived from human and mice developed intact epithelium with ultrastructural components of tight junctions, microvilli, ion transporter/channels, brush border enzymes similar to native tissue[55].

SCAFFOLDS

Observing the development of a neomucosa after patching the intestinal defects with abdominal wall or serosa of the adjacent colon, brought hope in using these methods for expanding the small bowel absorptive area[56-59]. Due to the limited availability of the tissues as well as anatomical restrictions, Thompson et al[60] investigated the outcome of the patching with prosthetic materials at 8 wk. They studied the outcome of patching the ileal defects of antimesenteric borders of rabbits’ intestine by using a variety of prosthetic materials including knitted Dacron, PGA mesh and polytetrafluoroethylene (PTFE). They also performed an interposition in the distal ileum with a Dacron tube in another group of animals. They only observed development of thin neomucosa covering 15% of the defect with the patches and no neomucosa formation in interposition tubes. They concluded that the use of prosthetic material was not useful for clinical management of short bowel syndrome[60].

Biological Scaffolds derived from extracellular matrixes of different types of tissues are being applied in tissue engineering to replicate the organs both structurally and functionally. In intestinal tissue engineering, these biocompatible materials are thought to increase the intestinal mucosal surface area and absorption.

Chen et al[61] used scaffolds derived from submucosal extracellular matrix of porcine small intestine “small intestine submucosa” (SIS) to evaluate the regeneration of small bowel in dogs. SIS has been previously used to create vascular grafts, abdominal wall, bladder, tendons, and dura mater in animals[62-66]. They applied the SIS as a patch to repair a partial defect created in the small bowel wall. They observed development of mucosal epithelium, smooth muscles and serosa, however, the layers were not architecturally well organized. They also tried to interpose SIS as a tubular segment in the small intestine, which was unsuccessful and all animals died postoperatively due to obstruction or leakage[61].

Then, Wang et al[67] interposed rat derived SIS between an isolated ileal loop in a rat model. They found development of a well-organized three-layer small intestine including mucosa, smooth muscle and serosa after 24 wk, however, there were no signs of innervation.

Another type of scaffolds applied is a collagen-rich membrane derived from submucosal layer of the pig’s small intestine called “Surgisis”. Since it is biocompatible, resistant to infection and contains growth factors, it seemed prudent to use it as a bioscaffold for small intestine regeneration[68-74].

Cicalese et al[75] utilized an acellularized matrix of connective tissue obtained from the dermis of cadaveric donors to develop “acellular dermal matrix” (ADM) with preserved proteins of basement membrane, elastin and collagen fibers. We hypothesized that this matrix will be vascularized by host capillaries and stem cells either circulating or derived from the adjacent crypts would induce tissue regeneration. We implanted these ADMs into the rats’ intestine either in continuity of the functioning bowel loops or as a blind-ended pouch in a defunctionalized jejunal limb. The blind-ended pouch group immediately showed full thickness ingrowth of capillaries, myofibroblasts and a fully regenerated mucosa at 6 mo. Despite the first group developing peritonitis in the first week without any signs of mucosa or muscular development, in subsequent studies, and using a ticker ADM placed immediately in continuity with the resected intestine, we were able to obtain successful generation of a neo-normal intestinal segment without obstructions or abscesses similar in morphology to the blind-end pouch group.

Similarly, Ansolani et al[74] utilized a three-centimeter long tubular Surgisis graft to interpose it in an isolated ileal loop in a rat model. After 24 wk, they found a neovascularized, well-developed layers of serosa, smooth muscle and mucosa. This biomaterial showed to offer a promising alternative in small intestine regeneration, however, the fact that it was not placed in continuity with the functional intestinal tract and there was no confirmation of absorption were the limiting factors.

Recently, we studied the function of such obtained bioengineered intestinal segment transplanting on the rats’ proximal jejunum a Surgisis scaffold. Besides performing a detailed anatomic and functional evaluation, we measured the absorptive function of this neo intestine in vivo. The structural characteristics of the bio artificial intestinal segment was comparable to normal intestine while we also observed brush border development with preserved microvilli as well as the presence of water and ion transporter/channels. In order to unequivocally demonstrate absorption, the animals underwent to a laparotomy after 12 wk from the primary surgery. Upon isolated of the newly formed intestinal segment and its vascular pedicle, we evaluated the absorption of D-Xylose from that specific surface area alone, which confirmed comparable absorption with normal intestine[75]. These promising results providing absorptive functional evidence for the first time in vivo, offer the basis for investigation of this method in a large animal model and its possible rapid translation into the clinical settings.

FUTURE DIRECTIONS

Through the years, significant improvements have been made in the development of new methods to create neo-formed bioengineered intestinal tissue. In the last few years, we have assisted an increment of interest in the field. At this time, most of the proposed models described in the literature present several limitations to translate into human. The main limitations are due to the complexity of some models. For example, the need to perform multiple surgeries to re-implant in continuity with the intestine preformed omental organoids. Moreover, many of the methods described are still rudimental and do not offer a complete structure that can be used in a clinical application. Even more limiting, most methods do not offer evidence of in vivo absorptive function. We believe that constitute a minimum and fundamental requirement to embark in using any neo-formed bioengineered intestinal structure in a clinical setting to treat intestinal failure. On these bases, we believe that the simpler model that we have described and proven functional in vivo utilizing an acellular biologic scaffold placed immediately in continuity with the short intestinal segment appears to be more promising to translate into clinical application for patients with intestinal failure. With these new approaches, if proven successful in a preclinical model, a breakthrough could take place in development of bio-artificial organs.

Footnotes

P- Reviewer: Fulop T, Salvadori M S- Editor: Qiu S L- Editor: A E- Editor: Wang CH

References
1.  Seguy D, Vahedi K, Kapel N, Souberbielle JC, Messing B. Low-dose growth hormone in adult home parenteral nutrition-dependent short bowel syndrome patients: a positive study. Gastroenterology. 2003;124:293-302.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 176]  [Cited by in F6Publishing: 183]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
2.  Ukleja A, Romano MM. Complications of parenteral nutrition. Gastroenterol Clin North Am. 2007;36:23-46, v.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 35]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
3.  Galea MH, Holliday H, Carachi R, Kapila L. Short-bowel syndrome: a collective review. J Pediatr Surg. 1992;27:592-596.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in F6Publishing: 79]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
4.  Georgeson KE, Breaux CW. Outcome and intestinal adaptation in neonatal short-bowel syndrome. J Pediatr Surg. 1992;27:344-348; discussion 348-350.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 137]  [Cited by in F6Publishing: 122]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
5.  Howard L, Heaphey L, Fleming CR, Lininger L, Steiger E. Four years of North American registry home parenteral nutrition outcome data and their implications for patient management. JPEN J Parenter Enteral Nutr. 1991;15:384-393.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 165]  [Cited by in F6Publishing: 166]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
6.  Lalan S, Pomerantseva I, Vacanti JP. Tissue engineering and its potential impact on surgery. World J Surg. 2001;25:1458-1466.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 88]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
7.  Grant D, Abu-Elmagd K, Reyes J, Tzakis A, Langnas A, Fishbein T, Goulet O, Farmer D. 2003 report of the intestine transplant registry: a new era has dawned. Ann Surg. 2005;241:607-613.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 379]  [Cited by in F6Publishing: 394]  [Article Influence: 20.7]  [Reference Citation Analysis (0)]
8.  Budding J, Smith CC. Role of recirculating loops in the management of massive resection of the small intestine. Surg Gynecol Obstet. 1967;125:243-249.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Frederick PL, Craig TV. The effect of vagotomy and pyloroplasty on weight loss and survival of dogs after massive intestinal resection. Surgery. 1964;56:135-143.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Keller JW, Stewart WC, Westerheide R, Pace WG. Prolonged survival with paired reversed segment after massive intestinal resection. Arch Surg. 1965;91:174-179.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 31]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
11.  Venables CW, Ellis H, Smith AD. Antiperistaltic segments after massive intestinal resections. Lancet. 1966;2:1390-1394.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 41]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
12.  Hutcher NE, Salzberg AM. Pre-ileal transposition of colon to prevent the development of short bowel syndrome in puppied with 90 percent small intestinal resection. Surgery. 1971;70:189-197.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Hutcher NE, Mendez-Picon G, Salzberg AM. Prejejunal transposition of colon to prevent the development of short bowel syndrome in puppies with 90 per cent small intestine resection. J Pediatr Surg. 1973;8:771-777.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 32]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
14.  Garcia VF, Templeton JM, Eichelberger MR, Koop CE, Vinograd I. Colon interposition for the short bowel syndrome. J Pediatr Surg. 1981;16:994-995.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Bianchi A. Intestinal loop lengthening--a technique for increasing small intestinal length. J Pediatr Surg. 1980;15:145-151.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Yildiz BD. Where are we at with short bowel syndrome and small bowel transplant. World J Transplant. 2012;2:95-103.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 16]  [Cited by in F6Publishing: 12]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
17.  Kim HB, Fauza D, Garza J, Oh JT, Nurko S, Jaksic T. Serial transverse enteroplasty (STEP): a novel bowel lengthening procedure. J Pediatr Surg. 2003;38:425-429.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 306]  [Cited by in F6Publishing: 253]  [Article Influence: 12.0]  [Reference Citation Analysis (0)]
18.  Sudan D, DiBaise J, Torres C, Thompson J, Raynor S, Gilroy R, Horslen S, Grant W, Botha J, Langnas A. A multidisciplinary approach to the treatment of intestinal failure. J Gastrointest Surg. 2005;9:165-176; discussion 176-177.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Modi BP, Javid PJ, Jaksic T, Piper H, Langer M, Duggan C, Kamin D, Kim HB. First report of the international serial transverse enteroplasty data registry: indications, efficacy, and complications. J Am Coll Surg. 2007;204:365-371.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 130]  [Cited by in F6Publishing: 131]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
20.  Spence JR, Mayhew CN, Rankin SA, Kuhar MF, Vallance JE, Tolle K, Hoskins EE, Kalinichenko VV, Wells SI, Zorn AM. Directed differentiation of human pluripotent stem cells into intestinal tissue in vitro. Nature. 2011;470:105-109.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1455]  [Cited by in F6Publishing: 1296]  [Article Influence: 99.7]  [Reference Citation Analysis (0)]
21.  Saha S, Bhanja P, Kabarriti R, Liu L, Alfieri AA, Guha C. Bone marrow stromal cell transplantation mitigates radiation-induced gastrointestinal syndrome in mice. PLoS One. 2011;6:e24072.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 98]  [Cited by in F6Publishing: 99]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
22.  Kim SS, Kaihara S, Benvenuto M, Choi RS, Kim BS, Mooney DJ, Taylor GA, Vacanti JP. Regenerative signals for tissue-engineered small intestine. Transplant Proc. 1999;31:657-660.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 26]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
23.  Qu B, Xin GR, Zhao LX, Xing H, Lian LY, Jiang HY, Tong JZ, Wang BB, Jin SZ. Testing stem cell therapy in a rat model of inflammatory bowel disease: role of bone marrow stem cells and stem cell factor in mucosal regeneration. PLoS One. 2014;9:e107891.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
24.  Hori Y, Nakamura T, Kimura D, Kaino K, Kurokawa Y, Satomi S, Shimizu Y. Experimental study on tissue engineering of the small intestine by mesenchymal stem cell seeding. J Surg Res. 2002;102:156-160.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in F6Publishing: 77]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
25.  Yoshida A, Chitcholtan K, Evans JJ, Nock V, Beasley SW. In vitro tissue engineering of smooth muscle sheets with peristalsis using a murine induced pluripotent stem cell line. J Pediatr Surg. 2012;47:329-335.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 17]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
26.  Nishikawa R, Hotta R, Shimojima N, Shibata S, Nagoshi N, Nakamura M, Matsuzaki Y, Okano HJ, Kuroda T, Okano H. Migration and differentiation of transplanted enteric neural crest-derived cells in murine model of Hirschsprung’s disease. Cytotechnology. 2015;67:661-670.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 15]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
27.  Kulkarni S, Zou B, Hanson J, Micci MA, Tiwari G, Becker L, Kaiser M, Xie XS, Pasricha PJ. Gut-derived factors promote neurogenesis of CNS-neural stem cells and nudge their differentiation to an enteric-like neuronal phenotype. Am J Physiol Gastrointest Liver Physiol. 2011;301:G644-G655.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 18]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
28.  Zhu T, Yu D, Feng J, Wu X, Xiang L, Gao H, Zhang X, Wei M. GDNF and NT-3 induce progenitor bone mesenchymal stem cell differentiation into neurons in fetal gut culture medium. Cell Mol Neurobiol. 2015;35:255-264.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 15]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
29.  McCracken KW, Howell JC, Wells JM, Spence JR. Generating human intestinal tissue from pluripotent stem cells in vitro. Nat Protoc. 2011;6:1920-1928.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 305]  [Cited by in F6Publishing: 302]  [Article Influence: 23.2]  [Reference Citation Analysis (0)]
30.  Kim HJ, Ingber DE. Gut-on-a-Chip microenvironment induces human intestinal cells to undergo villus differentiation. Integr Biol (Camb). 2013;5:1130-1140.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 434]  [Cited by in F6Publishing: 467]  [Article Influence: 46.7]  [Reference Citation Analysis (0)]
31.  Kim BS, Putnam AJ, Kulik TJ, Mooney DJ. Optimizing seeding and culture methods to engineer smooth muscle tissue on biodegradable polymer matrices. Biotechnol Bioeng. 1998;57:46-54.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Qin HH, Dunn JC. Small intestinal submucosa seeded with intestinal smooth muscle cells in a rodent jejunal interposition model. J Surg Res. 2011;171:e21-e26.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 38]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
33.  Totonelli G, Maghsoudlou P, Garriboli M, Riegler J, Orlando G, Burns AJ, Sebire NJ, Smith VV, Fishman JM, Ghionzoli M. A rat decellularized small bowel scaffold that preserves villus-crypt architecture for intestinal regeneration. Biomaterials. 2012;33:3401-3410.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 164]  [Cited by in F6Publishing: 146]  [Article Influence: 12.2]  [Reference Citation Analysis (0)]
34.  Maghsoudlou P, Totonelli G, Loukogeorgakis SP, Eaton S, De Coppi P. A decellularization methodology for the production of a natural acellular intestinal matrix. J Vis Exp. 2013;Epub ahead of print.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 30]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
35.  Nakase Y, Hagiwara A, Nakamura T, Kin S, Nakashima S, Yoshikawa T, Fukuda K, Kuriu Y, Miyagawa K, Sakakura C. Tissue engineering of small intestinal tissue using collagen sponge scaffolds seeded with smooth muscle cells. Tissue Eng. 2006;12:403-412.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 80]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
36.  Lee M, Wu BM, Stelzner M, Reichardt HM, Dunn JC. Intestinal smooth muscle cell maintenance by basic fibroblast growth factor. Tissue Eng Part A. 2008;14:1395-1402.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 40]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
37.  Zakhem E, Raghavan S, Bitar KN. Neo-innervation of a bioengineered intestinal smooth muscle construct around chitosan scaffold. Biomaterials. 2014;35:1882-1889.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 21]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
38.  Zakhem E, Rego SL, Raghavan S, Bitar KN. The appendix as a viable source of neural progenitor cells to functionally innervate bioengineered gastrointestinal smooth muscle tissues. Stem Cells Transl Med. 2015;4:548-554.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 8]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
39.  Raghavan S, Bitar KN. The influence of extracellular matrix composition on the differentiation of neuronal subtypes in tissue engineered innervated intestinal smooth muscle sheets. Biomaterials. 2014;35:7429-7440.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 22]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
40.  Raghavan S, Gilmont RR, Bitar KN. Neuroglial differentiation of adult enteric neuronal progenitor cells as a function of extracellular matrix composition. Biomaterials. 2013;34:6649-6658.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 38]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
41.  Haffen K, Kedinger M, Simon-Assmann P. Mesenchyme-dependent differentiation of epithelial progenitor cells in the gut. J Pediatr Gastroenterol Nutr. 1987;6:14-23.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Organ GM, Mooney DJ, Hansen LK, Schloo B, Vacanti JP. Transplantation of enterocytes utilizing polymer-cell constructs to produce a neointestine. Transplant Proc. 1992;24:3009-3011.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Tait IS, Flint N, Campbell FC, Evans GS. Generation of neomucosa in vivo by transplantation of dissociated rat postnatal small intestinal epithelium. Differentiation. 1994;56:91-100.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Choi RS, Vacanti JP. Preliminary studies of tissue-engineered intestine using isolated epithelial organoid units on tubular synthetic biodegradable scaffolds. Transplant Proc. 1997;29:848-851.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 124]  [Cited by in F6Publishing: 126]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
45.  Choi RS, Riegler M, Pothoulakis C, Kim BS, Mooney D, Vacanti M, Vacanti JP. Studies of brush border enzymes, basement membrane components, and electrophysiology of tissue-engineered neointestine. J Pediatr Surg. 1998;33:991-996; discussion 996-997.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 82]  [Cited by in F6Publishing: 80]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
46.  Kim SS, Kaihara S, Benvenuto MS, Choi RS, Kim BS, Mooney DJ, Taylor GA, Vacanti JP. Regenerative signals for intestinal epithelial organoid units transplanted on biodegradable polymer scaffolds for tissue engineering of small intestine. Transplantation. 1999;67:227-233.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Kaihara S, Kim SS, Benvenuto M, Choi R, Kim BS, Mooney D, Tanaka K, Vacanti JP. Successful anastomosis between tissue-engineered intestine and native small bowel. Transplantation. 1999;67:241-245.  [PubMed]  [DOI]  [Cited in This Article: ]
48.  Kaihara S, Kim SS, Kim BS, Mooney D, Tanaka K, Vacanti JP. Long-term follow-up of tissue-engineered intestine after anastomosis to native small bowel. Transplantation. 2000;69:1927-1932.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  Grikscheit TC, Ogilvie JB, Ochoa ER, Alsberg E, Mooney D, Vacanti JP. Tissue-engineered colon exhibits function in vivo. Surgery. 2002;132:200-204.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 55]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
50.  Grikscheit TC, Ochoa ER, Ramsanahie A, Alsberg E, Mooney D, Whang EE, Vacanti JP. Tissue-engineered large intestine resembles native colon with appropriate in vitro physiology and architecture. Ann Surg. 2003;238:35-41.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 82]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
51.  Grikscheit TC, Siddique A, Ochoa ER, Srinivasan A, Alsberg E, Hodin RA, Vacanti JP. Tissue-engineered small intestine improves recovery after massive small bowel resection. Ann Surg. 2004;240:748-754.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 177]  [Cited by in F6Publishing: 185]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
52.  Sala FG, Matthews JA, Speer AL, Torashima Y, Barthel ER, Grikscheit TC. A multicellular approach forms a significant amount of tissue-engineered small intestine in the mouse. Tissue Eng Part A. 2011;17:1841-1850.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 66]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
53.  Sala FG, Kunisaki SM, Ochoa ER, Vacanti J, Grikscheit TC. Tissue-engineered small intestine and stomach form from autologous tissue in a preclinical large animal model. J Surg Res. 2009;156:205-212.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 129]  [Cited by in F6Publishing: 107]  [Article Influence: 7.1]  [Reference Citation Analysis (0)]
54.  Levin DE, Barthel ER, Speer AL, Sala FG, Hou X, Torashima Y, Grikscheit TC. Human tissue-engineered small intestine forms from postnatal progenitor cells. J Pediatr Surg. 2013;48:129-137.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 59]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
55.  Grant CN, Mojica SG, Sala FG, Hill JR, Levin DE, Speer AL, Barthel ER, Shimada H, Zachos NC, Grikscheit TC. Human and mouse tissue-engineered small intestine both demonstrate digestive and absorptive function. Am J Physiol Gastrointest Liver Physiol. 2015;308:G664-G677.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 73]  [Cited by in F6Publishing: 70]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
56.  Binnington HB, Siegel BA, Kissane JM, Ternberg JL. A technique to increase jejunal mucosa surface area. J Pediatr Surg. 1973;8:765-769.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 40]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
57.  Lillemoe KD, Berry WR, Harmon JW, Tai YH, Weichbrod RH, Cogen MA. Use of vascularized abdominal wall pedicle flaps to grow small bowel neomucosa. Surgery. 1982;91:293-300.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  Thompson JS, Vanderhoof JA, Antonson DL, Newland JR, Hodgson PE. Comparison of techniques for growing small bowel neomucosa. J Surg Res. 1984;36:401-406.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 38]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
59.  Watson LC, Friedman HI, Griffin DG, Norton LW, Mellick PW. Small bowel neomucosa. J Surg Res. 1980;28:280-291.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 31]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
60.  Thompson JS, Kampfe PW, Newland JR, Vanderhoof JA. Growth of intestinal neomucosa on prosthetic materials. J Surg Res. 1986;41:484-492.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 32]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
61.  Chen MK, Badylak SF. Small bowel tissue engineering using small intestinal submucosa as a scaffold. J Surg Res. 2001;99:352-358.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 146]  [Cited by in F6Publishing: 155]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
62.  Cobb MA, Badylak SF, Janas W, Boop FA. Histology after dural grafting with small intestinal submucosa. Surg Neurol. 1996;46:389-393; discussion 393-394.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 98]  [Cited by in F6Publishing: 99]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
63.  Kropp BP, Eppley BL, Prevel CD, Rippy MK, Harruff RC, Badylak SF, Adams MC, Rink RC, Keating MA. Experimental assessment of small intestinal submucosa as a bladder wall substitute. Urology. 1995;46:396-400.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 268]  [Cited by in F6Publishing: 275]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
64.  Badylak SF, Lantz GC, Coffey A, Geddes LA. Small intestinal submucosa as a large diameter vascular graft in the dog. J Surg Res. 1989;47:74-80.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 464]  [Cited by in F6Publishing: 479]  [Article Influence: 13.7]  [Reference Citation Analysis (0)]
65.  Clarke KM, Lantz GC, Salisbury SK, Badylak SF, Hiles MC, Voytik SL. Intestine submucosa and polypropylene mesh for abdominal wall repair in dogs. J Surg Res. 1996;60:107-114.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 207]  [Cited by in F6Publishing: 210]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
66.  Badylak SF, Tullius R, Kokini K, Shelbourne KD, Klootwyk T, Voytik SL, Kraine MR, Simmons C. The use of xenogeneic small intestinal submucosa as a biomaterial for Achilles tendon repair in a dog model. J Biomed Mater Res. 1995;29:977-985.  [PubMed]  [DOI]  [Cited in This Article: ]
67.  Wang ZQ, Watanabe Y, Toki A. Experimental assessment of small intestinal submucosa as a small bowel graft in a rat model. J Pediatr Surg. 2003;38:1596-1601.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 51]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
68.  Hodde J. Naturally occurring scaffolds for soft tissue repair and regeneration. Tissue Eng. 2002;8:295-308.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 248]  [Cited by in F6Publishing: 250]  [Article Influence: 11.4]  [Reference Citation Analysis (0)]
69.  Hodde JP, Record RD, Liang HA, Badylak SF. Vascular endothelial growth factor in porcine-derived extracellular matrix. Endothelium. 2001;8:11-24.  [PubMed]  [DOI]  [Cited in This Article: ]
70.  Sarikaya A, Record R, Wu CC, Tullius B, Badylak S, Ladisch M. Antimicrobial activity associated with extracellular matrices. Tissue Eng. 2002;8:63-71.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 136]  [Cited by in F6Publishing: 138]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
71.  Hodde J, Record R, Tullius R, Badylak S. Fibronectin peptides mediate HMEC adhesion to porcine-derived extracellular matrix. Biomaterials. 2002;23:1841-1848.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 91]  [Cited by in F6Publishing: 92]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
72.  Pahari MP, Raman A, Bloomenthal A, Costa MA, Bradley SP, Banner B, Rastellini C, Cicalese L. A novel approach for intestinal elongation using acellular dermal matrix: an experimental study in rats. Transplant Proc. 2006;38:1849-1850.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 20]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
73.  Pahari MP, Brown ML, Elias G, Nseir H, Banner B, Rastellini C, Cicalese L. Development of a bioartificial new intestinal segment using an acellular matrix scaffold. Gut. 2007;56:885-886.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 8]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
74.  Ansaloni L, Bonasoni P, Cambrini P, Catena F, De Cataldis A, Gagliardi S, Gazzotti F, Peruzzi S, Santini D, Taffurelli M. Experimental evaluation of Surgisis as scaffold for neointestine regeneration in a rat model. Transplant Proc. 2006;38:1844-1848.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 22]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
75.  Cicalese L, Corsello T, Stevenson HL, Damiano G, Tuveri M, Zorzi D, Montalbano M, Shirafkan A, Rastellini C. Evidence of Absorptive Function in vivo in a Neo-Formed Bio-Artificial Intestinal Segment Using a Rodent Model. J Gastrointest Surg. 2015;Epub ahead of print.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 7]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]