Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Am Chem Soc ; 143(12): 4714-4724, 2021 03 31.
Article in English | MEDLINE | ID: mdl-33739832

ABSTRACT

Prodrugs engineered for preferential activation in diseased versus normal tissues offer immense potential to improve the therapeutic indexes (TIs) of preclinical and clinical-stage active pharmaceutical ingredients that either cannot be developed otherwise or whose efficacy or tolerability it is highly desirable to improve. Such approaches, however, often suffer from trial-and-error design, precluding predictive synthesis and optimization. Here, using bromodomain and extra-terminal (BET) protein inhibitors (BETi)-a class of epigenetic regulators with proven anticancer potential but clinical development hindered in large part by narrow TIs-we introduce a macromolecular prodrug platform that overcomes these challenges. Through tuning of traceless linkers appended to a "bottlebrush prodrug" scaffold, we demonstrate correlation of in vitro prodrug activation kinetics with in vivo tumor pharmacokinetics, enabling the predictive design of novel BETi prodrugs with enhanced antitumor efficacies and devoid of dose-limiting toxicities in a syngeneic triple-negative breast cancer murine model. This work may have immediate clinical implications, introducing a platform for predictive prodrug design and potentially overcoming hurdles in drug development.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Design , Prodrugs/pharmacology , Proteins/antagonists & inhibitors , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Humans , Macromolecular Substances/chemical synthesis , Macromolecular Substances/chemistry , Macromolecular Substances/pharmacology , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Molecular Structure , Prodrugs/chemical synthesis , Prodrugs/chemistry , Proteins/metabolism
2.
Elife ; 82019 11 01.
Article in English | MEDLINE | ID: mdl-31663852

ABSTRACT

Even though pancreatic ductal adenocarcinoma (PDAC) is associated with fibrotic stroma, the molecular pathways regulating the formation of cancer associated fibroblasts (CAFs) are not well elucidated. An epigenomic analysis of patient-derived and de-novo generated CAFs demonstrated widespread loss of cytosine methylation that was associated with overexpression of various inflammatory transcripts including CXCR4. Co-culture of neoplastic cells with CAFs led to increased invasiveness that was abrogated by inhibition of CXCR4. Metabolite tracing revealed that lactate produced by neoplastic cells leads to increased production of alpha-ketoglutarate (aKG) within mesenchymal stem cells (MSCs). In turn, aKG mediated activation of the demethylase TET enzyme led to decreased cytosine methylation and increased hydroxymethylation during de novo differentiation of MSCs to CAF. Co-injection of neoplastic cells with TET-deficient MSCs inhibited tumor growth in vivo. Thus, in PDAC, a tumor-mediated lactate flux is associated with widespread epigenomic reprogramming that is seen during CAF formation.


Subject(s)
Cancer-Associated Fibroblasts/pathology , Cellular Reprogramming/drug effects , Epigenesis, Genetic/drug effects , Lactic Acid/pharmacology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , 5-Methylcytosine/analogs & derivatives , 5-Methylcytosine/metabolism , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/drug effects , Cellular Reprogramming/genetics , DNA Methylation/drug effects , Humans , Ketoglutaric Acids/metabolism , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Neoplasm Invasiveness , Receptors, CXCR4/metabolism , Stromal Cells/drug effects , Stromal Cells/metabolism , Transcriptome/genetics , Pancreatic Neoplasms
3.
Nat Biomed Eng ; 2(11): 822-830, 2018 11.
Article in English | MEDLINE | ID: mdl-30918745

ABSTRACT

At present there are no drugs for the treatment of chronic liver fibrosis that have been approved by the Food and Drug administration of the United States. Telmisartan, a small-molecule antihypertensive drug, displays antifibrotic activity, but its clinical use is limited because it causes systemic hypotension. Here, we report the scalable and convergent synthesis of macromolecular telmisartan prodrugs optimized for preferential release in diseased liver tissue. We optimized the release of active telmisartan in fibrotic liver to be depot-like (that is, a constant therapeutic concentration) through the molecular design of telmisartan brush-arm star polymers, and show that these lead to improved efficacy and to the avoidance of dose-limiting hypotension in both metabolically and chemically induced mouse models of hepatic fibrosis, as determined by histopathology, enzyme levels in the liver, intact-tissue protein markers, hepatocyte necrosis protection, and gene-expression analyses. In rats and dogs, the prodrugs are retained long-term in liver tissue and have a well-tolerated safety profile. Our findings support the further development of telmisartan prodrugs that enable infrequent dosing in the treatment of liver fibrosis.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/therapeutic use , Drug Design , Liver Cirrhosis/drug therapy , Prodrugs/therapeutic use , Telmisartan/therapeutic use , Angiotensin II Type 1 Receptor Blockers/chemistry , Angiotensin II Type 1 Receptor Blockers/pharmacokinetics , Animals , Carbon Tetrachloride/toxicity , Disease Models, Animal , Female , Gene Expression Regulation/drug effects , Half-Life , Liver/metabolism , Liver Cirrhosis/chemically induced , Liver Cirrhosis/pathology , Male , Mice , Mice, Inbred BALB C , Polymers/chemistry , Prodrugs/chemistry , Prodrugs/pharmacokinetics , Rats , Rats, Sprague-Dawley , Telmisartan/chemistry
4.
Nat Biomed Eng ; 2(9): 707, 2018 Sep.
Article in English | MEDLINE | ID: mdl-31015683

ABSTRACT

In the version of this Article originally published, the author Peter Blume-Jensen was not denoted as a corresponding author; this has now been amended and the author's email address has been added. The 'Correspondence and requests for materials' statement was similarly affected and has now been updated with the author's initials 'P.B-J.'

5.
Nat Commun ; 8(1): 783, 2017 10 04.
Article in English | MEDLINE | ID: mdl-28978906

ABSTRACT

The roles of long non-coding RNAs in cancer metabolism remain largely unexplored. Here we identify FILNC1 (FoxO-induced long non-coding RNA 1) as an energy stress-induced long non-coding RNA by FoxO transcription factors. FILNC1 deficiency in renal cancer cells alleviates energy stress-induced apoptosis and markedly promotes renal tumor development. We show that FILNC1 deficiency leads to enhanced glucose uptake and lactate production through upregulation of c-Myc. Upon energy stress, FILNC1 interacts with AUF1, a c-Myc mRNA-binding protein, and sequesters AUF1 from binding c-Myc mRNA, leading to downregulation of c-Myc protein. FILNC1 is specifically expressed in kidney, and is downregulated in renal cell carcinoma; also, its low expression correlates with poor clinical outcomes in renal cell carcinoma. Together, our study not only identifies FILNC1 as a negative regulator of renal cancer with potential clinical value, but also reveals a regulatory mechanism by long non-coding RNAs to control energy metabolism and tumor development.FoxO are commonly down-regulated transcription factors and tumor suppressors in renal cell cancer (RCC). Here, the authors show that upon energy stress FoxOs induce the expression of the long non-coding RNA FILNC1, which inhibits survival of RCC by downregulating c-Myc and c-Myc-dependent metabolic rewiring.


Subject(s)
Carcinoma, Renal Cell/genetics , Energy Metabolism/genetics , Genes, myc/physiology , Kidney Neoplasms/genetics , RNA, Long Noncoding/physiology , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Down-Regulation , Glucose/metabolism , HEK293 Cells , Heterogeneous Nuclear Ribonucleoprotein D0 , Heterogeneous-Nuclear Ribonucleoprotein D/genetics , Heterogeneous-Nuclear Ribonucleoprotein D/metabolism , Heterogeneous-Nuclear Ribonucleoprotein D/physiology , Humans , Kidney/metabolism , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Lactic Acid/biosynthesis , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Stress, Physiological , Up-Regulation
6.
Nature ; 542(7639): 119-123, 2017 02 02.
Article in English | MEDLINE | ID: mdl-28099419

ABSTRACT

The genome of pancreatic ductal adenocarcinoma (PDAC) frequently contains deletions of tumour suppressor gene loci, most notably SMAD4, which is homozygously deleted in nearly one-third of cases. As loss of neighbouring housekeeping genes can confer collateral lethality, we sought to determine whether loss of the metabolic gene malic enzyme 2 (ME2) in the SMAD4 locus would create cancer-specific metabolic vulnerability upon targeting of its paralogous isoform ME3. The mitochondrial malic enzymes (ME2 and ME3) are oxidative decarboxylases that catalyse the conversion of malate to pyruvate and are essential for NADPH regeneration and reactive oxygen species homeostasis. Here we show that ME3 depletion selectively kills ME2-null PDAC cells in a manner consistent with an essential function for ME3 in ME2-null cancer cells. Mechanistically, integrated metabolomic and molecular investigation of cells deficient in mitochondrial malic enzymes revealed diminished NADPH production and consequent high levels of reactive oxygen species. These changes activate AMP activated protein kinase (AMPK), which in turn directly suppresses sterol regulatory element-binding protein 1 (SREBP1)-directed transcription of its direct targets including the BCAT2 branched-chain amino acid transaminase 2) gene. BCAT2 catalyses the transfer of the amino group from branched-chain amino acids to α-ketoglutarate (α-KG) thereby regenerating glutamate, which functions in part to support de novo nucleotide synthesis. Thus, mitochondrial malic enzyme deficiency, which results in impaired NADPH production, provides a prime 'collateral lethality' therapeutic strategy for the treatment of a substantial fraction of patients diagnosed with this intractable disease.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Gene Deletion , Malate Dehydrogenase/deficiency , Pancreatic Neoplasms/genetics , AMP-Activated Protein Kinases/metabolism , Amino Acids, Branched-Chain/metabolism , Animals , Biocatalysis , Carcinoma, Pancreatic Ductal/enzymology , Carcinoma, Pancreatic Ductal/psychology , Carcinoma, Pancreatic Ductal/therapy , Humans , Ketoglutaric Acids/metabolism , Malate Dehydrogenase/genetics , Male , Mice , Minor Histocompatibility Antigens/biosynthesis , Minor Histocompatibility Antigens/genetics , Mitochondria/enzymology , Mitochondria/pathology , NADP/biosynthesis , NADP/metabolism , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/therapy , Pregnancy Proteins/biosynthesis , Pregnancy Proteins/genetics , Reactive Oxygen Species/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Transaminases/biosynthesis , Transaminases/genetics
7.
Nat Commun ; 7: 13525, 2016 11 28.
Article in English | MEDLINE | ID: mdl-27892457

ABSTRACT

Dysregulated energetic metabolism has been recently identified as a hallmark of cancer. Although mutations in metabolic enzymes hardwire metabolism to tumourigenesis, they are relatively infrequent in ovarian cancer. More often, cancer metabolism is re-engineered by altered abundance and activity of the metabolic enzymes. Here we identify ubiquitin-specific peptidase 13 (USP13) as a master regulator that drives ovarian cancer metabolism. USP13 specifically deubiquitinates and thus upregulates ATP citrate lyase and oxoglutarate dehydrogenase, two key enzymes that determine mitochondrial respiration, glutaminolysis and fatty acid synthesis. The USP13 gene is co-amplified with PIK3CA in 29.3% of high-grade serous ovarian cancers and its overexpression is significantly associated with poor clinical outcome. Inhibiting USP13 remarkably suppresses ovarian tumour progression and sensitizes tumour cells to the treatment of PI3K/AKT inhibitor. Our results reveal an important metabolism-centric role of USP13, which may lead to potential therapeutics targeting USP13 in ovarian cancers.


Subject(s)
Endopeptidases/genetics , Gene Amplification , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Carcinogenesis/drug effects , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Progression , Endopeptidases/metabolism , Female , Gene Knockdown Techniques , Genome, Human , Glutamine/metabolism , HEK293 Cells , Humans , Lipids/biosynthesis , Mitochondria/drug effects , Mitochondria/metabolism , Neoplasm Metastasis , Ovarian Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Protein Stability/drug effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Substrate Specificity/drug effects , Ubiquitin-Specific Proteases , Ubiquitination/drug effects
8.
Cell Metab ; 24(5): 685-700, 2016 11 08.
Article in English | MEDLINE | ID: mdl-27829138

ABSTRACT

Reactive stromal cells are an integral part of tumor microenvironment (TME) and interact with cancer cells to regulate their growth. Although targeting stromal cells could be a viable therapy to regulate the communication between TME and cancer cells, identification of stromal targets that make cancer cells vulnerable has remained challenging and elusive. Here, we identify a previously unrecognized mechanism whereby metabolism of reactive stromal cells is reprogrammed through an upregulated glutamine anabolic pathway. This dysfunctional stromal metabolism confers atypical metabolic flexibility and adaptive mechanisms in stromal cells, allowing them to harness carbon and nitrogen from noncanonical sources to synthesize glutamine in nutrient-deprived conditions existing in TME. Using an orthotopic mouse model for ovarian carcinoma, we find that co-targeting glutamine synthetase in stroma and glutaminase in cancer cells reduces tumor weight, nodules, and metastasis. We present a synthetic lethal approach to target tumor stroma and cancer cells simultaneously for desirable therapeutic outcomes.


Subject(s)
Glutamate-Ammonia Ligase/metabolism , Neoplasms/enzymology , Neoplasms/pathology , Tumor Microenvironment , Amino Acids/metabolism , Animals , Aspartate Aminotransferases/metabolism , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Carbon/metabolism , Cell Line, Tumor , Cell Proliferation , Citric Acid Cycle , Disease Models, Animal , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , Metabolome , Mice, Nude , Nitrogen/metabolism , Nucleotides/metabolism , Stromal Cells/enzymology , Up-Regulation
9.
Elife ; 5: e10250, 2016 Feb 27.
Article in English | MEDLINE | ID: mdl-26920219

ABSTRACT

Cancer-associated fibroblasts (CAFs) are a major cellular component of tumor microenvironment in most solid cancers. Altered cellular metabolism is a hallmark of cancer, and much of the published literature has focused on neoplastic cell-autonomous processes for these adaptations. We demonstrate that exosomes secreted by patient-derived CAFs can strikingly reprogram the metabolic machinery following their uptake by cancer cells. We find that CAF-derived exosomes (CDEs) inhibit mitochondrial oxidative phosphorylation, thereby increasing glycolysis and glutamine-dependent reductive carboxylation in cancer cells. Through 13C-labeled isotope labeling experiments we elucidate that exosomes supply amino acids to nutrient-deprived cancer cells in a mechanism similar to macropinocytosis, albeit without the previously described dependence on oncogenic-Kras signaling. Using intra-exosomal metabolomics, we provide compelling evidence that CDEs contain intact metabolites, including amino acids, lipids, and TCA-cycle intermediates that are avidly utilized by cancer cells for central carbon metabolism and promoting tumor growth under nutrient deprivation or nutrient stressed conditions.


Subject(s)
Exosomes , Fibroblasts/metabolism , Glucose/metabolism , Neoplasms/physiopathology , Tumor Microenvironment , Exosomes/metabolism , Fermentation , Glycolysis , Lactic Acid/metabolism , Oxidative Phosphorylation
10.
J Natl Cancer Inst ; 108(6): djv426, 2016 Jan 26.
Article in English | MEDLINE | ID: mdl-26819345

ABSTRACT

BACKGROUND: The clinical and biological effects of metabolic alterations in cancer are not fully understood. METHODS: In high-grade serous ovarian cancer (HGSOC) samples (n = 101), over 170 metabolites were profiled and compared with normal ovarian tissues (n = 15). To determine NAT8L gene expression across different cancer types, we analyzed the RNA expression of cancer types using RNASeqV2 data available from the open access The Cancer Genome Atlas (TCGA) website (http://www.cbioportal.org/public-portal/). Using NAT8L siRNA, molecular techniques and histological analysis, we determined cancer cell viability, proliferation, apoptosis, and tumor growth in in vitro and in vivo (n = 6-10 mice/group) settings. Data were analyzed with the Student's t test and Kaplan-Meier analysis. Statistical tests were two-sided. RESULTS: Patients with high levels of tumoral NAA and its biosynthetic enzyme, aspartate N-acetyltransferase (NAT8L), had worse overall survival than patients with low levels of NAA and NAT8L. The overall survival duration of patients with higher-than-median NAA levels (3.6 years) was lower than that of patients with lower-than-median NAA levels (5.1 years, P = .03). High NAT8L gene expression in other cancers (melanoma, renal cell, breast, colon, and uterine cancers) was associated with worse overall survival. NAT8L silencing reduced cancer cell viability (HEYA8: control siRNA 90.61% ± 2.53, NAT8L siRNA 39.43% ± 3.00, P < .001; A2780: control siRNA 90.59% ± 2.53, NAT8L siRNA 7.44% ± 1.71, P < .001) and proliferation (HEYA8: control siRNA 74.83% ± 0.92, NAT8L siRNA 55.70% ± 1.54, P < .001; A2780: control siRNA 50.17% ± 4.13, NAT8L siRNA 26.52% ± 3.70, P < .001), which was rescued by addition of NAA. In orthotopic mouse models (ovarian cancer and melanoma), NAT8L silencing reduced tumor growth statistically significantly (A2780: control siRNA 0.52 g ± 0.15, NAT8L siRNA 0.08 g ± 0.17, P < .001; HEYA8: control siRNA 0.79 g ± 0.42, NAT8L siRNA 0.24 g ± 0.18, P = .008, A375-SM: control siRNA 0.55 g ± 0.22, NAT8L siRNA 0.21 g ± 0.17 g, P = .001). NAT8L silencing downregulated the anti-apoptotic pathway, which was mediated through FOXM1. CONCLUSION: These findings indicate that the NAA pathway has a prominent role in promoting tumor growth and represents a valuable target for anticancer therapy.Altered energy metabolism is a hallmark of cancer (1). Proliferating cancer cells have much greater metabolic requirements than nonproliferating differentiated cells (2,3). Moreover, altered cancer metabolism elevates unique metabolic intermediates, which can promote cancer survival and progression (4,5). Furthermore, emerging evidence suggests that proliferating cancer cells exploit alternative metabolic pathways to meet their high demand for energy and to accumulate biomass (6-8).


Subject(s)
Acetyltransferases/metabolism , Aspartic Acid/analogs & derivatives , Biomarkers, Tumor/metabolism , Cystadenocarcinoma, Serous/metabolism , Ovarian Neoplasms/metabolism , Ovary/metabolism , Animals , Apoptosis , Aspartic Acid/metabolism , Cell Line, Tumor , Cell Proliferation , Cell Survival , Chromatography, High Pressure Liquid , Cystadenocarcinoma, Serous/pathology , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Mice , Neoplasm Grading , Ovarian Neoplasms/pathology , Tandem Mass Spectrometry
11.
Oncotarget ; 6(32): 33705-19, 2015 Oct 20.
Article in English | MEDLINE | ID: mdl-26378042

ABSTRACT

Warburg effect has emerged as a potential hallmark of many cancers. However, the molecular mechanisms that led to this metabolic state of aerobic glycolysis, particularly in ovarian cancer (OVCA) have not been completely elucidated. HSulf-1 predominantly functions by limiting the bioavailability of heparan binding growth factors and hence their downstream signaling. Here we report that HSulf-1, a known putative tumor suppressor, is a negative regulator of glycolysis. Silencing of HSulf-1 expression in OV202 cell line increased glucose uptake and lactate production by upregulating glycolytic genes such as Glut1, HKII, LDHA, as well as metabolites. Conversely, HSulf-1 overexpression in TOV21G cells resulted in the down regulation of glycolytic enzymes and reduced glycolytic phenotype, supporting the role of HSulf-1 loss in enhanced aerobic glycolysis. HSulf-1 deficiency mediated glycolytic enhancement also resulted in increased inhibitory phosphorylation of pyruvate dehydrogenase (PDH) thus blocking the entry of glucose flux into TCA cycle. Consistent with this, metabolomic and isotope tracer analysis showed reduced glucose flux into TCA cycle. Moreover, HSulf-1 loss is associated with lower oxygen consumption rate (OCR) and impaired mitochondrial function. Mechanistically, lack of HSulf-1 promotes c-Myc induction through HB-EGF-mediated p-ERK activation. Pharmacological inhibition of c-Myc reduced HB-EGF induced glycolytic enzymes implicating a major role of c-Myc in loss of HSulf-1 mediated altered glycolytic pathway in OVCA. Similarly, PG545 treatment, an agent that binds to heparan binding growth factors and sequesters growth factors away from their ligand also blocked HB-EGF signaling and reduced glucose uptake in vivo in HSulf-1 deficient cells.


Subject(s)
Ovarian Neoplasms/metabolism , Sulfotransferases/deficiency , Animals , Cell Line, Tumor , Cell Proliferation/physiology , Citric Acid Cycle , Female , Glycolysis , Humans , Mice , Mice, Knockout , Microarray Analysis , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Signal Transduction , Sulfotransferases/metabolism
12.
Curr Top Dev Biol ; 108: 217-46, 2014.
Article in English | MEDLINE | ID: mdl-24512711

ABSTRACT

Aging is marked by changes that affect organs and resident stem cell function. Shorting of telomeres, DNA damage, oxidative stress, deregulation of genes and proteins, impaired cell-cell communication, and an altered systemic environment cause the eventual demise of cells. At the same time, reparative activities also decline. It is intriguing to correlate aging with the decline of regenerative abilities. Animal models with strong regenerative capabilities imply that aging processes might not be affecting regeneration. In this review, we selectively present age-dependent changes in stem/progenitor cells that are vital for tissue homeostasis and repair. In addition, the aging effect on regeneration following injury in organs such as lung, skeletal muscle, heart, nervous system, cochlear hair, lens, and liver are discussed. These tissues are also known for diseases such as heart attack, stroke, cognitive impairment, cataract, and hearing loss that occur mostly during aging in humans. Conclusively, vertebrate regeneration declines with age with the loss of stem/progenitor cell function. Future studies on improving the function of stem cells, along with studies in fish and amphibians where regeneration does not decline with age, will undoubtedly provide insights into both processes.


Subject(s)
Aging , Regeneration/physiology , Vertebrates/physiology , Animals , DNA Repair , Endothelial Cells/physiology , Hair Cells, Auditory/physiology , Hematopoietic Stem Cells/physiology , Humans , Mesenchymal Stem Cells/cytology , Muscles/cytology , Muscles/pathology , Myocardium/pathology , Neurons/metabolism , Stem Cells/cytology , Stem Cells/physiology
13.
Birth Defects Res C Embryo Today ; 96(1): 1-29, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22457174

ABSTRACT

A number of organs have the intrinsic ability to regenerate, a distinctive feature that varies among organisms. Organ regeneration is a process not fully yet understood. However, when its underlying mechanisms are unraveled, it holds tremendous therapeutic potential for humans. In this review, we chose to summarize the repair and regenerative potential of the following organs and organ systems: thymus, adrenal gland, thyroid gland, intestine, lungs, heart, liver, blood vessels, germ cells, nervous system, eye tissues, hair cells, kidney and bladder, skin, hair follicles, pancreas, bone, and cartilage. For each organ, a review of the following is presented: (a) factors, pathways, and cells that are involved in the organ's intrinsic regenerative ability, (b) contribution of exogenous cells - such as progenitor cells, embryonic stem cells, induced pluripotent stem cells, and bone marrow-, adipose- and umbilical cord blood-derived stem cells - in repairing and regenerating organs in the absence of an innate intrinsic regenerative capability, (c) and the progress made in engineering bio-artificial scaffolds, tissues, and organs. Organ regeneration is a promising therapy that can alleviate humans from diseases that have not been yet cured. It is also superior to already existing treatments that utilize exogenous sources to substitute for the organ's lost structure and/or function(s).


Subject(s)
Regeneration/physiology , Regenerative Medicine/methods , Animals , Humans , Models, Animal , Organ Specificity/physiology , Stem Cell Transplantation , Stem Cells/cytology , Stem Cells/physiology , Tissue Engineering
SELECTION OF CITATIONS
SEARCH DETAIL