Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 19 de 19
1.
J Clin Invest ; 132(5)2022 03 01.
Article En | MEDLINE | ID: mdl-35229723

Cancer stem-like cells (CSLCs) acquire enhanced immune checkpoint responses to evade immune cell killing and promote tumor progression. Here we showed that signal regulatory protein γ (SIRPγ) determined CSLC properties and immune evasiveness in a small population of lung adenocarcinoma (LUAD) cancer cells. A SIRPγhi population displayed CSLC properties and transmitted the immune escape signal through sustaining CD47 expression in both SIRPγhi and SIRPγlo/- tumor cells. SIRPγ bridged MST1 and PP2A to facilitate MST1 dephosphorylation, resulting in Hippo/YAP activation and leading to cytokine release by CSLCs, which stimulated CD47 expression in LUAD cells and consequently inhibited tumor cell phagocytosis. SIRPγ promoted tumor growth and metastasis in vivo through YAP signaling. Notably, SIRPγ targeting with genetic SIRPγ knockdown or a SIRPγ-neutralizing antibody inhibited CSLC phenotypes and elicited phagocytosis that suppressed tumor growth in vivo. SIRPG was upregulated in human LUAD and its overexpression predicted poor survival outcome. Thus, SIRPγhi cells serve as CSLCs and tumor immune checkpoint-initiating cells, propagating the immune escape signal to the entire cancer cell population. Our study identifies Hippo/YAP signaling as the first mechanism by which SIRPγ is engaged and reveals that targeting SIRPγ represents an immune- and CSLC-targeting strategy for lung cancer therapy.


Adenocarcinoma of Lung , Lung Neoplasms , Adenocarcinoma of Lung/metabolism , CD47 Antigen/genetics , CD47 Antigen/metabolism , Cell Line, Tumor , Hippo Signaling Pathway , Humans , Lung Neoplasms/genetics , Signal Transduction
2.
Nat Rev Gastroenterol Hepatol ; 19(5): 328-342, 2022 05.
Article En | MEDLINE | ID: mdl-35190728

Fibrolamellar carcinoma (FLC), a rare, lethal hepatic cancer, occurs primarily in adolescents and young adults. Unlike hepatocellular carcinoma, FLC has no known association with viral, metabolic or chemical agents that cause cirrhosis. Currently, surgical resection is the only treatment demonstrated to achieve cure, and no standard of care exists for systemic therapy. Progress in FLC research illuminates a transition from an obscure cancer to one for which an interactive community seems poised to uncover fundamental mechanisms and initiate translation towards novel therapies. In this Roadmap, we review advances since the seminal discovery in 2014 that nearly all FLC tumours express a signature oncogene (DNAJB1-PRKACA) encoding a fusion protein (DNAJ-PKAc) in which the J-domain of a heat shock protein 40 (HSP40) co-chaperone replaces an amino-terminal segment of the catalytic subunit of the cyclic AMP-dependent protein kinase (PKA). Important gains include increased understanding of oncogenic pathways driven by DNAJ-PKAc; identification of potential therapeutic targets; development of research models; elucidation of immune mechanisms with potential for the development of immunotherapies; and completion of the first multicentre clinical trials of targeted therapy for FLC. In each of these key areas we propose a Roadmap for future progress.


Carcinoma, Hepatocellular , Liver Neoplasms , Adolescent , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/therapy , Clinical Trials as Topic , HSP40 Heat-Shock Proteins , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/therapy , Multicenter Studies as Topic , Young Adult
3.
Nat Immunol ; 19(1): 29-40, 2018 Jan.
Article En | MEDLINE | ID: mdl-29242539

Although deletion of certain autophagy-related genes has been associated with defects in hematopoiesis, it remains unclear whether hyperactivated mitophagy affects the maintenance and differentiation of hematopoietic stem cells (HSCs) and committed progenitor cells. Here we report that targeted deletion of the gene encoding the AAA+-ATPase Atad3a hyperactivated mitophagy in mouse hematopoietic cells. Affected mice showed reduced survival, severely decreased bone-marrow cellularity, erythroid anemia and B cell lymphopenia. Those phenotypes were associated with skewed differentiation of stem and progenitor cells and an enlarged HSC pool. Mechanistically, Atad3a interacted with the mitochondrial channel components Tom40 and Tim23 and served as a bridging factor to facilitate appropriate transportation and processing of the mitophagy protein Pink1. Loss of Atad3a caused accumulation of Pink1 and activated mitophagy. Notably, deletion of Pink1 in Atad3a-deficient mice significantly 'rescued' the mitophagy defect, which resulted in restoration of the progenitor and HSC pools. Our data indicate that Atad3a suppresses Pink1-dependent mitophagy and thereby serves a key role in hematopoietic homeostasis.


ATPases Associated with Diverse Cellular Activities/metabolism , Hematopoietic Stem Cells/metabolism , Homeostasis , Mitochondrial Proteins/metabolism , Mitophagy , Protein Kinases/metabolism , ATPases Associated with Diverse Cellular Activities/genetics , Animals , Apoptosis/genetics , Cell Differentiation/genetics , Cell Proliferation/genetics , HEK293 Cells , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mice, Knockout , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins , Mitochondrial Precursor Protein Import Complex Proteins , Mitochondrial Proteins/genetics , Protein Kinases/genetics
4.
Sci Transl Med ; 6(220): 220ra10, 2014 Jan 22.
Article En | MEDLINE | ID: mdl-24452262

Loss-of-function mutations in the myotubularin gene (MTM1) cause X-linked myotubular myopathy (XLMTM), a fatal, congenital pediatric disease that affects the entire skeletal musculature. Systemic administration of a single dose of a recombinant serotype 8 adeno-associated virus (AAV8) vector expressing murine myotubularin to Mtm1-deficient knockout mice at the onset or at late stages of the disease resulted in robust improvement in motor activity and contractile force, corrected muscle pathology, and prolonged survival throughout a 6-month study. Similarly, single-dose intravascular delivery of a canine AAV8-MTM1 vector in XLMTM dogs markedly improved severe muscle weakness and respiratory impairment, and prolonged life span to more than 1 year in the absence of toxicity or a humoral or cell-mediated immune response. These results demonstrate the therapeutic efficacy of AAV-mediated gene therapy for myotubular myopathy in small- and large-animal models, and provide proof of concept for future clinical trials in XLMTM patients.


Disease Models, Animal , Genetic Therapy/methods , Myopathies, Structural, Congenital/genetics , Myopathies, Structural, Congenital/therapy , Animals , Dependovirus/genetics , Diaphragm , Dogs , Genetic Vectors , Genotype , HEK293 Cells , Humans , Male , Mice , Mice, Knockout , Muscle Contraction , Muscle Weakness , Mutation , Myopathies, Structural, Congenital/mortality , Protein Tyrosine Phosphatases, Non-Receptor/genetics
5.
Stem Cell Res ; 12(2): 467-80, 2014 Mar.
Article En | MEDLINE | ID: mdl-24434629

The ability to extract somatic cells from a patient and reprogram them to pluripotency opens up new possibilities for personalized medicine. Induced pluripotent stem cells (iPSCs) have been employed to generate beating cardiomyocytes from a patient's skin or blood cells. Here, iPSC methods were used to generate cardiomyocytes starting from the urine of a patient with Duchenne muscular dystrophy (DMD). Urine was chosen as a starting material because it contains adult stem cells called urine-derived stem cells (USCs). USCs express the canonical reprogramming factors c-myc and klf4, and possess high telomerase activity. Pluripotency of urine-derived iPSC clones was confirmed by immunocytochemistry, RT-PCR and teratoma formation. Urine-derived iPSC clones generated from healthy volunteers and a DMD patient were differentiated into beating cardiomyocytes using a series of small molecules in monolayer culture. Results indicate that cardiomyocytes retain the DMD patient's dystrophin mutation. Physiological assays suggest that dystrophin-deficient cardiomyocytes possess phenotypic differences from normal cardiomyocytes. These results demonstrate the feasibility of generating cardiomyocytes from a urine sample and that urine-derived cardiomyocytes retain characteristic features that might be further exploited for mechanistic studies and drug discovery.


Dystrophin/deficiency , Induced Pluripotent Stem Cells/pathology , Muscular Dystrophy, Duchenne/pathology , Muscular Dystrophy, Duchenne/urine , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Adult , Animals , Case-Control Studies , Cell Differentiation/physiology , Cells, Cultured , Drug Discovery , Female , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/biosynthesis , Kruppel-Like Transcription Factors/urine , Male , Mice , Mice, Inbred NOD , Mice, SCID , Myocytes, Cardiac/cytology , Proto-Oncogene Proteins c-myc/biosynthesis , Proto-Oncogene Proteins c-myc/urine , Telomerase/urine , Young Adult
6.
Pharmacol Rev ; 65(3): 1091-133, 2013 Jul.
Article En | MEDLINE | ID: mdl-23818131

Regenerative medicine is a rapidly evolving multidisciplinary, translational research enterprise whose explicit purpose is to advance technologies for the repair and replacement of damaged cells, tissues, and organs. Scientific progress in the field has been steady and expectations for its robust clinical application continue to rise. The major thesis of this review is that the pharmacological sciences will contribute critically to the accelerated translational progress and clinical utility of regenerative medicine technologies. In 2007, we coined the phrase "regenerative pharmacology" to describe the enormous possibilities that could occur at the interface between pharmacology, regenerative medicine, and tissue engineering. The operational definition of regenerative pharmacology is "the application of pharmacological sciences to accelerate, optimize, and characterize (either in vitro or in vivo) the development, maturation, and function of bioengineered and regenerating tissues." As such, regenerative pharmacology seeks to cure disease through restoration of tissue/organ function. This strategy is distinct from standard pharmacotherapy, which is often limited to the amelioration of symptoms. Our goal here is to get pharmacologists more involved in this field of research by exposing them to the tools, opportunities, challenges, and interdisciplinary expertise that will be required to ensure awareness and galvanize involvement. To this end, we illustrate ways in which the pharmacological sciences can drive future innovations in regenerative medicine and tissue engineering and thus help to revolutionize the discovery of curative therapeutics. Hopefully, the broad foundational knowledge provided herein will spark sustained conversations among experts in diverse fields of scientific research to the benefit of all.


Pharmacology , Regenerative Medicine , Animals , Biocompatible Materials , Humans , Stem Cell Transplantation , Tissue Engineering
7.
Stem Cells ; 31(10): 2047-60, 2013 Oct.
Article En | MEDLINE | ID: mdl-23873634

Regenerative medicine is transitioning into clinical programs using stem/progenitor cell therapies for repair of damaged organs. We summarize those for liver and pancreas, organs that share endodermal stem cell populations, biliary tree stem cells (hBTSCs), located in peribiliary glands. They are precursors to hepatic stem/progenitors in canals of Hering and to committed progenitors in pancreatic duct glands. They give rise to maturational lineages along a radial axis within bile duct walls and a proximal-to-distal axis starting at the duodenum and ending with mature cells in the liver or pancreas. Clinical trials have been ongoing for years assessing effects of determined stem cells (fetal-liver-derived hepatic stem/progenitors) transplanted into the hepatic artery of patients with various liver diseases. Immunosuppression was not required. Control subjects, those given standard of care for a given condition, all died within a year or deteriorated in their liver functions. Subjects transplanted with 100-150 million hepatic stem/progenitor cells had improved liver functions and survival extending for several years. Full evaluations of safety and efficacy of transplants are still in progress. Determined stem cell therapies for diabetes using hBTSCs remain to be explored but are likely to occur following ongoing preclinical studies. In addition, mesenchymal stem cells (MSCs) and hematopoietic stem cells (HSCs) are being used for patients with chronic liver conditions or with diabetes. MSCs have demonstrated significant effects through paracrine signaling of trophic and immunomodulatory factors, and there is limited evidence for inefficient lineage restriction into mature parenchymal or islet cells. HSCs' effects are primarily via modulation of immune mechanisms.


Hepatitis/therapy , Mesenchymal Stem Cell Transplantation , Pancreatitis/therapy , Cell Differentiation , Cell Lineage , Hepatitis/immunology , Humans , Liver/embryology , Liver/immunology , Liver/pathology , Mesenchymal Stem Cells/physiology , Pancreas/embryology , Pancreas/immunology , Pancreas/pathology , Pancreatitis/immunology , Stem Cell Niche
8.
Stem Cells ; 31(9): 1966-79, 2013 Sep.
Article En | MEDLINE | ID: mdl-23847135

Peribiliary glands (PBGs) in bile duct walls, and pancreatic duct glands (PDGs) associated with pancreatic ducts, in humans of all ages, contain a continuous, ramifying network of cells in overlapping maturational lineages. We show that proximal (PBGs)-to-distal (PDGs) maturational lineages start near the duodenum with cells expressing markers of pluripotency (NANOG, OCT4, and SOX2), proliferation (Ki67), self-replication (SALL4), and early hepato-pancreatic commitment (SOX9, SOX17, PDX1, and LGR5), transitioning to PDG cells with no expression of pluripotency or self-replication markers, maintenance of pancreatic genes (PDX1), and expression of markers of pancreatic endocrine maturation (NGN3, MUC6, and insulin). Radial-axis lineages start in PBGs near the ducts' fibromuscular layers with stem cells and end at the ducts' lumens with cells devoid of stem cell traits and positive for pancreatic endocrine genes. Biliary tree-derived cells behaved as stem cells in culture under expansion conditions, culture plastic and serum-free Kubota's Medium, proliferating for months as undifferentiated cells, whereas pancreas-derived cells underwent only approximately 8-10 divisions, then partially differentiated towards an islet fate. Biliary tree-derived cells proved precursors of pancreas' committed progenitors. Both could be driven by three-dimensional conditions, islet-derived matrix components and a serum-free, hormonally defined medium for an islet fate (HDM-P), to form spheroids with ultrastructural, electrophysiological and functional characteristics of neoislets, including glucose regulatability. Implantation of these neoislets into epididymal fat pads of immunocompromised mice, chemically rendered diabetic, resulted in secretion of human C-peptide, regulatable by glucose, and able to alleviate hyperglycemia in hosts. The biliary tree-derived stem cells and their connections to pancreatic committed progenitors constitute a biological framework for life-long pancreatic organogenesis.


Biliary Tract/cytology , Cell Lineage , Organogenesis , Pancreas/cytology , Pancreas/growth & development , Stem Cells/cytology , Adult , Animals , Antigens, Neoplasm/metabolism , Biomarkers/metabolism , Cell Adhesion Molecules/metabolism , Cell Differentiation/genetics , Cell Lineage/genetics , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Experimental/therapy , Electrophysiological Phenomena , Epithelial Cell Adhesion Molecule , Gene Expression Regulation , Humans , Hyperglycemia/therapy , Islets of Langerhans/cytology , Islets of Langerhans/physiology , Islets of Langerhans/ultrastructure , Islets of Langerhans Transplantation , Mice , Organogenesis/genetics , Pancreatic Ducts/cytology , Phenotype , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Spheroids, Cellular/cytology , Spheroids, Cellular/metabolism , Spheroids, Cellular/ultrastructure , Stem Cell Niche/genetics , Stem Cells/metabolism
9.
PLoS One ; 6(10): e26535, 2011.
Article En | MEDLINE | ID: mdl-22046303

Amniotic fluid stem (AFS) cells are broadly multipotent, can be expanded extensively in culture, are not tumorigenic and can be readily cryopreserved for cell banking. Mesenchymal stem cells (MSC) show immunomodulatory activity and secrete a wide spectrum of cytokines and chemokines that suppress inflammatory responses, block mixed lymphocyte reactions (MLR) and other immune reactions, and have proven therapeutic against conditions such as graft-versus-host disease. AFS cells resemble MSCs in many respects including surface marker expression and differentiation potential. We therefore hypothesized that AFS cells may exhibit similar immunomodulatory capabilities. We present data to demonstrate that direct contact with AFS cells inhibits lymphocyte activation. In addition, we show that cell-free supernatants derived from AFS cells primed with total blood monocytes or IL-1ß, a cytokine released by monocytes and essential in mediation of the inflammatory response, also inhibited lymphocyte activation. Further investigation of AFS cell-free supernatants by protein array revealed secretion of multiple factors in common with MSCs that are known to be involved in immune regulation including growth related oncogene (GRO) and monocyte chemotactic protein (MCP) family members as well as interleukin-6 (IL-6). AFS cells activated by PBMCs released several additional cytokines as compared to BM-MSCs, including macrophage inflammatory protein-3α (MIP-3α), MIP-1α and Activin. AFS cells also released higher levels of MCP-1 and lower levels of MCP-2 compared to BM-MSCs in response to IL-1ß activation. This suggests that there may be some AFS-specific mechanisms of inhibition of lymphocyte activation. Our results indicate that AFS cells are able to suppress inflammatory responses in vitro and that soluble factors are an essential component in the communication between lymphocytes and AFS cells. Their extensive self-renewal capacity, possibility for banking and absence of tumorigenicity may make AFS cells a superior source of stable, well characterized "off the shelf" immunomodulatory cells for a variety of immunotherapies.


Amniotic Fluid/cytology , Immunity , Proto-Oncogene Proteins c-kit , Stem Cells/immunology , Clone Cells/immunology , Culture Media, Conditioned , Cytokines/metabolism , Humans , Immunotherapy , Inflammation/prevention & control , Tissue Banks
10.
Hepatology ; 54(6): 2159-72, 2011 Dec.
Article En | MEDLINE | ID: mdl-21809358

UNLABELLED: Multipotent stem/progenitors are present in peribiliary glands of extrahepatic biliary trees from humans of all ages and in high numbers in hepato-pancreatic common duct, cystic duct, and hilum. They express endodermal transcription factors (e.g., Sox9, SOX17, FOXA2, PDX1, HES1, NGN3, PROX1) intranuclearly, stem/progenitor surface markers (EpCAM, NCAM, CD133, CXCR4), and sometimes weakly adult liver, bile duct, and pancreatic genes (albumin, cystic fibrosis transmembrane conductance regulator [CFTR], and insulin). They clonogenically expand on plastic and in serum-free medium, tailored for endodermal progenitors, remaining phenotypically stable as undifferentiated cells for months with a cell division initially every ≈36 hours and slowing to one every 2-3 days. Transfer into distinct culture conditions, each comprised of a specific mix of hormones and matrix components, yields either cords of hepatocytes (express albumin, CYP3A4, and transferrin), branching ducts of cholangiocytes (expressing anion exchanger-2-AE2 and CFTR), or regulatable C-peptide secreting neoislet-like clusters (expressing glucagon, insulin) and accompanied by changes in gene expression correlating with the adult fate. Transplantation into quiescent livers of immunocompromised mice results in functional human hepatocytes and cholangiocytes, whereas if into fat pads of streptozocin-induced diabetic mice, results in functional islets secreting glucose-regulatable human C-peptide. CONCLUSION: The phenotypes and availability from all age donors suggest that these stem/progenitors have considerable potential for regenerative therapies of liver, bile duct, and pancreatic diseases including diabetes.


Biliary Tract/cytology , Cell Differentiation/physiology , Hepatocytes/cytology , Islets of Langerhans/cytology , Multipotent Stem Cells/cytology , Animals , Cell Lineage , Cells, Cultured , Colony-Forming Units Assay , Humans , Mice , Mice, SCID , Regeneration/physiology , Transcription Factors/biosynthesis , Transplantation, Heterologous
11.
PM R ; 3(6 Suppl 1): S95-9, 2011 Jun.
Article En | MEDLINE | ID: mdl-21703588

Human stem cells derived from bone marrow are currently used in clinical medicine for bone and cartilage repair for injuries such as meniscal tears. New clinical stem cell studies underway include the treatment of patients with spinal cord injuries. Rapid advances in stem cell science are opening new avenues for drug discovery and may lead to new uses of stem cells for other musculoskeletal disorders.


Musculoskeletal Diseases/surgery , Stem Cell Transplantation/methods , Bone Marrow Cells/cytology , Humans , Musculoskeletal Diseases/rehabilitation , Treatment Outcome
12.
J Immunol Methods ; 352(1-2): 174-7, 2010 Jan 31.
Article En | MEDLINE | ID: mdl-19925805

The ability of an experimental treatment to induce primitive, undifferentiated stem cells towards an epidermal fate may be tested by comparing the treated stem cells with a positive control, such as primary keratinocytes. In an effort to perfect methods used for this comparison, we tested two commercially available antibodies and three fixation methods to determine which antibody/fixation interaction produced the best immunofluorescent images of the nuclear localization of p63, a canonical marker of epidermal fate, in keratinocytes. Here, we report the methods used, and the experimental outcome.


Keratinocytes/metabolism , Membrane Proteins/biosynthesis , Microscopy, Fluorescence/methods , Nuclear Proteins/biosynthesis , Pluripotent Stem Cells/metabolism , Antibodies/metabolism , Cell Culture Techniques , Cell Differentiation , Cells, Cultured , Foreskin/pathology , Humans , Infant, Newborn , Keratinocytes/immunology , Male , Pluripotent Stem Cells/immunology , Reference Standards , Tissue Fixation
14.
J Cell Biochem ; 106(4): 507-11, 2009 Mar 01.
Article En | MEDLINE | ID: mdl-19130494

We review progress towards the goal of utilizing stem cells as a source of engineered pancreatic beta-cells for therapy of diabetes. Protocols for the in vitro differentiation of embryonic stem (ES) cells based on normal developmental cues have generated beta-like cells that produce high levels of insulin, albeit at low efficiency and without full responsiveness to extracellular levels of glucose. Induced pluripotent stem (iPS) cells also can yield insulin-producing cells following similar approaches. An important recent report shows that when transplanted into mice, human ES-derived cells with a phenotype corresponding to pancreatic endoderm matured to yield cells capable of maintaining near-normal regulation of blood sugar [Kroon et al., 2008]. Major hurdles that must be overcome to enable the broad clinical translation of these advances include teratoma formation by ES and iPS cells, and the need for immunosuppressive drugs. Classes of stem cells that can be expanded extensively in culture but do not form teratomas, such as amniotic fluid-derived stem cells and hepatic stem cells, offer possible alternatives for the production of beta-like cells, but further evidence is required to document this potential. Generation of autologous iPS cells should prevent transplant rejection, but may prove prohibitively expensive. Banking strategies to identify small numbers of stem cell lines homozygous for major histocompatibility loci have been proposed to enable beneficial genetic matching that would decrease the need for immunosuppression.


Diabetes Mellitus/therapy , Embryonic Stem Cells/cytology , Insulin-Secreting Cells/cytology , Animals , Cell Culture Techniques , Cell Differentiation , Cell Transplantation/methods , Humans , Insulin-Secreting Cells/transplantation , Tissue Engineering/methods
16.
J Exp Med ; 204(8): 1973-87, 2007 Aug 06.
Article En | MEDLINE | ID: mdl-17664288

Human hepatic stem cells (hHpSCs), which are pluripotent precursors of hepatoblasts and thence of hepatocytic and biliary epithelia, are located in ductal plates in fetal livers and in Canals of Hering in adult livers. They can be isolated by immunoselection for epithelial cell adhesion molecule-positive (EpCAM+) cells, and they constitute approximately 0.5-2.5% of liver parenchyma of all donor ages. The self-renewal capacity of hHpSCs is indicated by phenotypic stability after expansion for >150 population doublings in a serum-free, defined medium and with a doubling time of approximately 36 h. Survival and proliferation of hHpSCs require paracrine signaling by hepatic stellate cells and/or angioblasts that coisolate with them. The hHpSCs are approximately 9 microm in diameter, express cytokeratins 8, 18, and 19, CD133/1, telomerase, CD44H, claudin 3, and albumin (weakly). They are negative for alpha-fetoprotein (AFP), intercellular adhesion molecule (ICAM) 1, and for markers of adult liver cells (cytochrome P450s), hemopoietic cells (CD45), and mesenchymal cells (vascular endothelial growth factor receptor and desmin). If transferred to STO feeders, hHpSCs give rise to hepatoblasts, which are recognizable by cordlike colony morphology and up-regulation of AFP, P4503A7, and ICAM1. Transplantation of freshly isolated EpCAM+ cells or of hHpSCs expanded in culture into NOD/SCID mice results in mature liver tissue expressing human-specific proteins. The hHpSCs are candidates for liver cell therapies.


Cell Culture Techniques/methods , Liver/cytology , Liver/embryology , Stem Cells/cytology , Cell Adhesion , Cell Membrane/metabolism , Culture Media, Serum-Free/metabolism , Epithelial Cells/cytology , Hematopoietic Stem Cells/metabolism , Hepatocytes/metabolism , Humans , Intercellular Adhesion Molecule-1/metabolism , Leukocyte Common Antigens/biosynthesis , Liver/metabolism , Mesoderm/metabolism , Signal Transduction , alpha-Fetoproteins/metabolism
17.
Biomaterials ; 28(34): 5068-73, 2007 Dec.
Article En | MEDLINE | ID: mdl-17706763

As a prominent tool in regenerative medicine, tissue engineering (TE) has been an active field of scientific research for nearly three decades. Clinical application of TE technologies has been relatively restricted, however, owing in part to the limited number of biomaterials that are approved for human use. While many excellent biomaterials have been developed in recent years, their translation into clinical practice has been slow. As a consequence, many investigators still employ biodegradable polymers that were first approved for use in humans over 30 years ago. During normal development tissue morphogenesis is heavily influenced by the interaction of cells with the extracellular matrix (ECM). Yet simple polymers, while providing architectural support for neo-tissue development, do not adequately mimic the complex interactions between adult stem and progenitor cells and the ECM that promote functional tissue regeneration. Future advances in TE and regenerative medicine will depend on the development of "smart" biomaterials that actively participate in the formation of functional tissue. Clinical translation of these new classes of biomaterials will be supported by many of the same evaluation tools as those developed and described by Professor David F. Williams and colleagues over the past 30 years.


Biocompatible Materials/chemistry , Regenerative Medicine/instrumentation , Regenerative Medicine/methods , Tissue Engineering/instrumentation , Tissue Engineering/methods , Animals , Bioartificial Organs , Biopolymers/chemistry , Cell Culture Techniques/instrumentation , Cell Culture Techniques/methods , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Humans , Hydrogel, Polyethylene Glycol Dimethacrylate/chemistry , Keratins/chemistry , Polymers/chemistry , Proteins/chemistry , Stem Cells/metabolism
18.
Nat Biotechnol ; 25(1): 100-6, 2007 Jan.
Article En | MEDLINE | ID: mdl-17206138

Stem cells capable of differentiating to multiple lineages may be valuable for therapy. We report the isolation of human and rodent amniotic fluid-derived stem (AFS) cells that express embryonic and adult stem cell markers. Undifferentiated AFS cells expand extensively without feeders, double in 36 h and are not tumorigenic. Lines maintained for over 250 population doublings retained long telomeres and a normal karyotype. AFS cells are broadly multipotent. Clonal human lines verified by retroviral marking were induced to differentiate into cell types representing each embryonic germ layer, including cells of adipogenic, osteogenic, myogenic, endothelial, neuronal and hepatic lineages. Examples of differentiated cells derived from human AFS cells and displaying specialized functions include neuronal lineage cells secreting the neurotransmitter L-glutamate or expressing G-protein-gated inwardly rectifying potassium channels, hepatic lineage cells producing urea, and osteogenic lineage cells forming tissue-engineered bone.


Amniotic Fluid/cytology , Cell Culture Techniques/methods , Cell Separation/methods , Pluripotent Stem Cells/cytology , Stem Cell Transplantation/methods , Tissue Engineering/methods , Amniotic Fluid/metabolism , Animals , Biomarkers/analysis , Cell Differentiation , Cells, Cultured , Humans , Pluripotent Stem Cells/metabolism , Proto-Oncogene Proteins c-kit/analysis
19.
Liver Int ; 24(4): 361-70, 2004 Aug.
Article En | MEDLINE | ID: mdl-15287860

BACKGROUND: The availability of well-characterized human liver cell populations that can be frozen and thawed will be critical for cell therapy. We addressed whether human hepatocytes can recover after cryopreservation and engraft in immunodeficient mice. METHODS: We isolated cells from discarded human livers and studied the properties of cryopreserved cells. The viability of thawed cells was established with multiple in vitro assays, including analysis of liver gene expression, ureagenesis, cytochrome P450 activity, and growth factor-induced cell proliferation. The fate of transplanted cells was analysed in immunodeficient NOD-SCID mice. RESULTS: After thawing, the viability of human hepatocytes exceeded 60%. Cells attached to culture dishes, proliferated following growth factor stimulation and exhibited liver-specific functions. After transplantation in NOD-SCID mice, cells engrafted in the peritoneal cavity, a heterologous site, as well as the liver itself, retained hepatic function and proliferated in response to liver injury. Transplanted hepatocytes were integrated in the liver parenchyma. Occasionally, transplanted cells were integrated in bile ducts. CONCLUSIONS: Cryopreserved human liver cell showed the ability to retain functional integrity and to reconstitute both hepatic and biliary lineages in mice. These studies offer suitable paradigms aimed at characterizing liver cells prior to transplantation in people.


Cryopreservation , Graft Survival , Hepatocytes/cytology , Hepatocytes/transplantation , Animals , Biological Assay/methods , Cell Division , Cell Movement , Cell Survival , Humans , Liver/cytology , Mice , Mice, Inbred NOD , Mice, SCID , Transplantation, Heterologous
...