Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 98
Filtrar
1.
EMBO J ; 39(4): e103315, 2020 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-31930531

RESUMO

Somatic cells acclimate to changes in the environment by temporary reprogramming. Much has been learned about transcription factors that induce these cell-state switches in both plants and animals, but how cells rapidly modulate their proteome remains elusive. Here, we show rapid induction of autophagy during temporary reprogramming in plants triggered by phytohormones, immune, and danger signals. Quantitative proteomics following sequential reprogramming revealed that autophagy is required for timely decay of previous cellular states and for tweaking the proteome to acclimate to the new conditions. Signatures of previous cellular programs thus persist in autophagy-deficient cells, affecting cellular decision-making. Concordantly, autophagy-deficient cells fail to acclimatize to dynamic climate changes. Similarly, they have defects in dedifferentiating into pluripotent stem cells, and redifferentiation during organogenesis. These observations indicate that autophagy mediates cell-state switches that underlie somatic cell reprogramming in plants and possibly other organisms, and thereby promotes phenotypic plasticity.


Assuntos
Arabidopsis/fisiologia , Autofagia , Reprogramação Celular , Proteoma , Transdução de Sinais , Aclimatação , Arabidopsis/citologia , Arabidopsis/imunologia , Fenótipo , Reguladores de Crescimento de Plantas/metabolismo , Proteômica
2.
Mol Med ; 29(1): 59, 2023 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-37098464

RESUMO

BACKGROUND: Myofibroblasts (MFB), one of the major effectors of pathologic fibrosis, mainly derived from the activation of fibroblast to myofibroblast transition (FMT). Although MFBs were historically considered terminally differentiated cells, their potential for de-differentiation was recently recognized and implied with therapeutic value in treating fibrotic diseases, for instance, idiopathic pulmonary fibrosis (IPF) and post allogeneic hematopoietic stem cell transplantation bronchiolitis obliterans (BO). During the past decade, several methods were reported to block or reverse MFB differentiation, among which mesenchymal stem cells (MSC) have demonstrated potential but undetermined therapeutic values. However, the MSC-mediated regulation of FMT and underlying mechanisms remained largely undefined. METHOD: By identifying TGF-ß1 hypertension as the pivotal landmark during the pro-fibrotic FMT, TGF-ß1-induced MFB and MSC co-culture models were established and utilized to investigate regulations by MSC on FMT in vitro. Methods including RNA sequencing (RNA-seq), Western blot, qPCR and flow cytometry were used. RESULT: Our data revealed that TGF-ß1 readily induced invasive signatures identified in fibrotic tissues and initiated MFB differentiation in normal FB. MSC reversibly de-differentiated MFB into a group of FB-like cells by selectively inhibiting the TGF-ß-SMAD2/3 signaling. Importantly, these proliferation-boosted FB-like cells remained sensitive to TGF-ß1 and could be re-induced into MFB. CONCLUSION: Our findings highlighted the reversibility of MSC-mediated de-differentiation of MFB through TGF-ß-SMAD2/3 signaling, which may explain MSC's inconsistent clinical efficacies in treating BO and other fibrotic diseases. These de-differentiated FB-like cells are still sensitive to TGF-ß1 and may further deteriorate MFB phenotypes unless the pro-fibrotic microenvironment is corrected.


Assuntos
Células-Tronco Mesenquimais , Miofibroblastos , Humanos , Diferenciação Celular , Fibroblastos/metabolismo , Fibrose , Células-Tronco Mesenquimais/metabolismo , Miofibroblastos/metabolismo , Proteína Smad2/metabolismo , Fator de Crescimento Transformador beta1/farmacologia , Fator de Crescimento Transformador beta1/metabolismo , Proteína Smad3/metabolismo
3.
Semin Cell Dev Biol ; 103: 83-93, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32417220

RESUMO

ß cells uniquely produce and secrete insulin under the control of several, integrated signals, to maintain blood glucose concentrations within a narrow physiological interval. ß cell failure is key to the onset and progression of type 2 diabetes, due to impaired function and reduced mass. In this review we focus on several features of human ß cell dysfunction and pathology in type 2 diabetes, as revealed by direct assessment of isolated islet traits and examination of pancreatic tissue from organ donors, surgical samples or autoptic specimens. Insulin secretion defects and pathology findings are discussed in relation to some of the major underlying mechanisms, to also provide clues for conceiving better prevention and treatment of type 2 diabetes by targeting the pancreatic ß cells.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Células Secretoras de Insulina/metabolismo , Humanos
4.
J Cell Physiol ; 237(3): 1639-1647, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34796916

RESUMO

Adipocytes are the key constituents of adipose tissue, and their de-differentiation process has been widely observed in physiological and pathological conditions. For obese people, the promotion of adipocyte de-differentiation or maintenance of an undifferentiated state of adipocytes may help to improve their metabolic condition. Thus, understanding the regulatory mechanisms of adipocyte de-differentiation is necessary for treating metabolic diseases. Attractively, in addition to intracellular signals regulating adipocyte de-differentiation, external factors such as temperature and pressure also affect adipocyte de-differentiation. In this review, we summarize the recent progress in the field and discuss the regulatory roles and mechanisms of involved endogenous and exogenous factors during the process of de-differentiation.


Assuntos
Adipócitos , Doenças Metabólicas , Adipócitos/metabolismo , Tecido Adiposo/metabolismo , Diferenciação Celular , Humanos , Doenças Metabólicas/metabolismo , Obesidade/genética , Obesidade/metabolismo
5.
Int J Neurosci ; 132(8): 843-849, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-33148105

RESUMO

BACKGROUND: Central nervous system is a rare occurring location of solitary fibrous tumors (SFTs). SFTs have a potential for recurrence, which is the leading cause of death in patients with these disease entities. De-differentiation phenomenon combined with cerebrospinal fluid (CSF) dissemination through drop metastasis of STFs from intracranial to intraspinal has only been reported in extremely limited cases. CASE DESCRIPTION: Herein, we present a case of SFT in a 54-year old male. MRI showed characteristic of mixed high and low signal with 6.3 cm × 6.5 cm × 5.9 cm. After radical surgical resection, the pathology indicated benign SFT. However, MRI re-examination of 22 months later detected local recurrence, concomitant with spreading of intracranial and intraspinal through CSF dissemination. And interestingly, the second pathology found de-differentiation phenomenon and malignance of SFT, in which some areas transformed to rhabdomyosarcoma. CONCLUSION: This is the first case report of recurrent intracranial SFT de-differentiating to rhabdomyosarcoma concurrent with CSF pathway drop metastasis. Benign intracranial SFTs have the potential of de-differentiation, which may play an important role in its distant metastasis. The underlying molecular biological and pathological mechanisms of benign SFT malignance transformation still warrant further exploration.


Assuntos
Neoplasias Encefálicas , Rabdomiossarcoma , Febre Grave com Síndrome de Trombocitopenia , Tumores Fibrosos Solitários , Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/cirurgia , Humanos , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Tumores Fibrosos Solitários/diagnóstico por imagem , Tumores Fibrosos Solitários/cirurgia
6.
Int J Mol Sci ; 23(7)2022 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-35409189

RESUMO

Adipocytes from the superficial layer of subcutaneous adipose tissue undergo cyclic de- and re-differentiation, which can significantly influence the development of skin inflammation under different cutaneous conditions. This inflammation can be connected with local loading of the reticular dermis with lipids released due to de-differentiation of adipocytes during the catagen phase of the hair follicle cycle. Alternatively, the inflammation parallels a widespread release of cathelicidin, which typically takes place in the anagen phase (especially in the presence of pathogens). Additionally, trans-differentiation of dermal adipocytes into myofibroblasts, which can occur under some pathological conditions, can be responsible for the development of collateral scarring in acne. Here, we provide an overview of such cellular conversions in the skin and discuss their possible involvement in the pathophysiology of inflammatory skin conditions, such as acne and psoriasis.


Assuntos
Acne Vulgar , Dermatopatias , Adipócitos , Folículo Piloso/fisiologia , Humanos , Inflamação , Pele
7.
Int J Mol Sci ; 23(19)2022 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-36232796

RESUMO

Pancreatic ß cell expansion and functional maturation during the birth-to-weaning period is driven by epigenetic programs primarily triggered by growth factors, hormones, and nutrients provided by human milk. As shown recently, exosomes derived from various origins interact with ß cells. This review elucidates the potential role of milk-derived exosomes (MEX) and their microRNAs (miRs) on pancreatic ß cell programming during the postnatal period of lactation as well as during continuous cow milk exposure of adult humans to bovine MEX. Mechanistic evidence suggests that MEX miRs stimulate mTORC1/c-MYC-dependent postnatal ß cell proliferation and glycolysis, but attenuate ß cell differentiation, mitochondrial function, and insulin synthesis and secretion. MEX miR content is negatively affected by maternal obesity, gestational diabetes, psychological stress, caesarean delivery, and is completely absent in infant formula. Weaning-related disappearance of MEX miRs may be the critical event switching ß cells from proliferation to TGF-ß/AMPK-mediated cell differentiation, whereas continued exposure of adult humans to bovine MEX miRs via intake of pasteurized cow milk may reverse ß cell differentiation, promoting ß cell de-differentiation. Whereas MEX miR signaling supports postnatal ß cell proliferation (diabetes prevention), persistent bovine MEX exposure after the lactation period may de-differentiate ß cells back to the postnatal phenotype (diabetes induction).


Assuntos
Exossomos , MicroRNAs , Proteínas Quinases Ativadas por AMP/metabolismo , Adulto , Animais , Bovinos , Diferenciação Celular , Proliferação de Células , Exossomos/metabolismo , Feminino , Humanos , Lactente , Insulina/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Leite/metabolismo , Leite Humano/metabolismo , Gravidez , Fator de Crescimento Transformador beta/metabolismo
8.
J Biol Chem ; 295(30): 10245-10254, 2020 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-32444498

RESUMO

Telomeres comprise specialized nucleic acid-protein complexes that help protect chromosome ends from DNA damage. Moreover, telomeres associate with subtelomeric regions through looping. This results in altered expression of subtelomeric genes. Recent observations further reveal telomere length-dependent gene regulation and epigenetic modifications at sites spread across the genome and distant from telomeres. This regulation is mediated through the telomere-binding protein telomeric repeat-binding factor 2 (TRF2). These observations suggest a role of telomeres in extra-telomeric functions. Most notably, telomeres have a broad impact on pluripotency and differentiation. For example, cardiomyocytes differentiate with higher efficacy from induced pluripotent stem cells having long telomeres, and differentiated cells obtained from human embryonic stem cells with relatively long telomeres have a longer lifespan. Here, we first highlight reports on these two seemingly distinct research areas: the extra-telomeric role of telomere-binding factors and the role of telomeres in pluripotency/stemness. On the basis of the observations reported in these studies, we draw attention to potential molecular connections between extra-telomeric biology and pluripotency. Finally, in the context of the nonlocal influence of telomeres on pluripotency and stemness, we discuss major opportunities for progress in molecular understanding of aging-related disorders and neurodegenerative diseases.


Assuntos
Envelhecimento/metabolismo , Diferenciação Celular , Doenças Neurodegenerativas/metabolismo , Células-Tronco Pluripotentes/metabolismo , Telômero/metabolismo , Envelhecimento/genética , Envelhecimento/patologia , Dano ao DNA , Humanos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/patologia , Células-Tronco Pluripotentes/patologia , Telômero/genética , Telômero/patologia , Homeostase do Telômero , Proteína 2 de Ligação a Repetições Teloméricas/metabolismo
9.
FASEB J ; 34(9): 12663-12676, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32761888

RESUMO

Phospholipase Cß1 is activated by Gαq to generate calcium signals in response to hormones and neurotransmitters. Besides carrying out this plasma membrane function, PLCß1 has a cytosolic population that helps to drive the differentiation of PC12 cells by inhibiting a nuclease that promotes RNA-induced silencing (C3PO). Here, we show that down-regulating PLCß1 or reducing its cytosolic population by activating Gαq to localize it to the plasma membrane returns differentiated PC12 and SK-N-SH cells to an undifferentiated state. In this state, PC12 cells have a spherical morphology, resume proliferation, and express the stem cell transcription factors nanog and Oct4. Similar changes are seen when C3PO is down-regulated. This return to a stem-like state is accompanied by shifts in multiple miR populations. Surprisingly, de-differentiation can be induced by extended stimulation of Gαq where cells return to a spherical morphology and levels of specific miRs return to their undifferentiated values. In complementary studies, we followed the real-time hydrolysis of a fluorescent-tagged miR in cells where PLCß1 or C3PO were down-regulated in PC12 cells and find substantial differences in miR processing in the undifferentiated and differentiated states. Taken together, our studies suggest that PLCß1, through its ability to regulate C3PO and endogenous miR populations, mediates the differentiation of two types of cultured neuronal cells.


Assuntos
Desdiferenciação Celular , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Fosfolipase C beta/metabolismo , Complexo de Inativação Induzido por RNA/metabolismo , Animais , Linhagem Celular Tumoral , Humanos , MicroRNAs/metabolismo , Células PC12 , Interferência de RNA , Ratos , Transdução de Sinais
10.
Biotechnol Bioeng ; 118(2): 979-991, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33205831

RESUMO

Scalable processes are requisite for the robust biomanufacturing of human pluripotent stem cell (hPSC)-derived therapeutics. Toward this end, we demonstrate the xeno-free expansion and directed differentiation of human embryonic and induced pluripotent stem cells to definitive endoderm (DE) in a controlled stirred suspension bioreactor (SSB). Based on previous work on converting hPSCs to insulin-producing progeny, differentiation of two hPSC lines was optimized in planar cultures yielding up to 87% FOXA2+ /SOX17+ cells. Next, hPSCs were propagated in an SSB with controlled pH and dissolved oxygen. Cultures displayed a 10- to 12-fold increase in cell number over 5-6 days with the maintenance of pluripotency (>85% OCT4+ ) and viability (>85%). For differentiation, SSB cultures yielded up to 89% FOXA2+ /SOX17+ cells or ~ 8 DE cells per seeded hPSC. Specification to DE cell fate was consistently more efficient in the bioreactor compared to planar cultures. Hence, a tunable strategy is established that is suitable for the xeno-free manufacturing of DE cells from different hPSC lines in scalable SSBs. This study advances bioprocess development for producing a wide gamut of human DE cell-derived therapeutics.


Assuntos
Reatores Biológicos , Endoderma/metabolismo , Células-Tronco Embrionárias Humanas/metabolismo , Linhagem Celular , Endoderma/citologia , Células-Tronco Embrionárias Humanas/citologia , Humanos , Células-Tronco Pluripotentes Induzidas/citologia
11.
J Theor Biol ; 503: 110384, 2020 10 21.
Artigo em Inglês | MEDLINE | ID: mdl-32603669

RESUMO

The cancer stem cell hypothesis claims that tumor growth and progression are driven by a (typically) small niche of the total cancer cell population called cancer stem cells (CSCs). These CSCs can go through symmetric or asymmetric divisions to differentiate into specialised, progenitor cells or reproduce new CSCs. While it was once held that this differentiation pathway was unidirectional, recent research has demonstrated that differentiated cells are more plastic than initially considered. In particular, differentiated cells can de-differentiate and recover their stem-like capacity. Two recent papers have considered how this rate of plasticity affects the evolutionary dynamic of an invasive, malignant population of stem cells and differentiated cells into existing tissue (Mahdipour-Shirayeh et al., 2017; Wodarz, 2018). These papers arrive at seemingly opposing conclusions, one claiming that increased plasticity results in increased invasive potential, and the other that increased plasticity decreases invasive potential. Here, we show that what is most important, when determining the effect on invasive potential, is how one distributes this increased plasticity between the compartments of resident and mutant-type cells. We also demonstrate how these results vary, producing non-monotone fixation probability curves, as inter-compartmental plasticity changes when differentiated cell compartments are allowed to continue proliferating, highlighting a fundamental difference between the two models. We conclude by demonstrating the stability of these qualitative results over various parameter ranges. Keywords: cancer stem cells, plasticity, de-differentiation, fixation probability.


Assuntos
Neoplasias , Células-Tronco Neoplásicas , Adaptação Fisiológica , Diferenciação Celular , Humanos , Neoplasias/genética , Probabilidade
12.
Exp Cell Res ; 385(2): 111683, 2019 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-31634482

RESUMO

Transitioning from a differentiated state to a higher-order of plasticity, by partial rather than full reactivation of pluripotency genes, might be a better approach in regenerative medicine. Hydrogen sulfide plays a crucial role in the maintenance and differentiation of mesenchymal stem cells (MSC) that have the potential to differentiate to a diverse group of mesenchymally derived cells. It was shown that these cells show a heavy reliance on cystathionine-ß-synthase (CBS)-derived hydrogen sulfide (H2S) during differentiation. We have found that expression and activity of 3-mercaptopyruvate sulfurtransferase (MPST), one of three enzymes that hat regulates H2S biosynthesis, is significantly lower in MSC as compared with lineage-restricted dermal fibroblasts. Here, we tested the hypothesis that suppression of MPST in dermal fibroblasts might induce plasticity-related changes and broaden the transdifferentiation potency. Inactivation of MPST with phenylpyruvate (PP) or by siRNA silencing led to diminished H2S production associated with increased production of reactive oxygen species (ROS) and lactic acid. Accumulation of α-ketoglutarate (α-KG), a key metabolite required for the expression of ten-eleven translocation hydroxylase (TET), was associated with stimulated transcription of pluripotency related genes including OCT4, KLF4, SOX2, and NANOG. Suppression of TET1 gene and inhibition of glycolysis with glucose analog, 2-desoxy-d-glucose, or hexokinase II inhibitor significantly reduced expression of pluripotency genes following MPST inactivation or knockdown. MPST disruption promoted the conversion of fibroblasts into adipocytes as evidenced by a significant increase in expression of adipocyte-specific genes, PPARγ, and UCP1, and intracellular accumulation of oil Red-O positive fat droplets. Inhibition of glycolysis inhibited these changes. Under induced differentiation conditions, fibroblasts with disrupted MPST show the potency to differentiate to white adipogenic lineage. Thus, MPST inactivation or silencing enhances the plasticity of dermal fibroblasts in a TET1 and glycolysis dependent manner and promotes adipogenic transdifferentiation.


Assuntos
Adipócitos/citologia , Transdiferenciação Celular , Fibroblastos/metabolismo , Sulfurtransferases/genética , Adipócitos/metabolismo , Adulto , Células Cultivadas , Fibroblastos/citologia , Glicólise , Humanos , Sulfeto de Hidrogênio/metabolismo , Fator 4 Semelhante a Kruppel , Ácido Láctico/metabolismo , Masculino , Oxigenases de Função Mista/metabolismo , PPAR gama/genética , PPAR gama/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Sulfurtransferases/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteína Desacopladora 1/genética , Proteína Desacopladora 1/metabolismo
13.
Cell Mol Life Sci ; 76(21): 4179-4201, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31563998

RESUMO

Uncontrolled scarring, or fibrosis, can interfere with the normal function of virtually all tissues of the body, ultimately leading to organ failure and death. Fibrotic diseases represent a major cause of death in industrialized countries. Unfortunately, no curative treatments for these conditions are yet available, highlighting the critical need for a better fundamental understanding of molecular mechanisms that may be therapeutically tractable. The ultimate indispensable effector cells responsible for deposition of extracellular matrix proteins that comprise scars are mesenchymal cells, namely fibroblasts and myofibroblasts. In this review, we focus on the biology of these cells and the molecular mechanisms that regulate their pertinent functions. We discuss key pro-fibrotic mediators, signaling pathways, and transcription factors that dictate their activation and persistence. Because of their possible clinical and therapeutic relevance, we also consider potential brakes on mesenchymal cell activation and cellular processes that may facilitate myofibroblast clearance from fibrotic tissue-topics that have in general been understudied.


Assuntos
Células-Tronco Mesenquimais/metabolismo , Diferenciação Celular , Matriz Extracelular/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Fibrose , Humanos , Células-Tronco Mesenquimais/citologia , Miofibroblastos/citologia , Miofibroblastos/metabolismo , RNA não Traduzido/metabolismo , Transdução de Sinais
14.
Int J Mol Sci ; 21(21)2020 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-33171818

RESUMO

Immune evasion and altered metabolism, where glucose utilization is diverted to increased lactic acid production, are two fundamental hallmarks of cancer. Although lactic acid has long been considered a waste product of this alteration, it is now well accepted that increased lactic acid production and the resultant acidification of the tumor microenvironment (TME) promotes multiple critical oncogenic processes including angiogenesis, tissue invasion/metastasis, and drug resistance. We and others have hypothesized that excess lactic acid in the TME is responsible for suppressing anticancer immunity. Recent studies support this hypothesis and provide mechanistic evidence explaining how lactic acid and the acidic TME impede immune cell functions. In this review, we consider lactic acid's role as a critical immunoregulatory molecule involved in suppressing immune effector cell proliferation and inducing immune cell de-differentiation. This results in the inhibition of antitumor immune responses and the activation of potent, negative regulators of innate and adaptive immune cells. We also consider the role of an acidic TME in suppressing anticancer immunity. Finally, we provide insights to help translate this new knowledge into impactful anticancer immune therapies.


Assuntos
Ácido Láctico/metabolismo , Neoplasias/imunologia , Microambiente Tumoral/fisiologia , Humanos , Concentração de Íons de Hidrogênio , Imunidade/imunologia , Terapia de Imunossupressão , Imunossupressores/farmacologia , Imunoterapia/métodos , Neoplasias/metabolismo
15.
Int J Mol Sci ; 21(15)2020 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-32751354

RESUMO

Since material stiffness controls many cell functions, we reviewed the currently available knowledge on stiffness sensing and elucidated what is known in the context of clinical and experimental articular cartilage (AC) repair. Remarkably, no stiffness information on the various biomaterials for clinical AC repair was accessible. Using mRNA expression profiles and morphology as surrogate markers of stiffness-related effects, we deduced that the various clinically available biomaterials control chondrocyte (CH) phenotype well, but not to equal extents, and only in non-degenerative settings. Ample evidence demonstrates that multiple molecular aspects of CH and mesenchymal stromal cell (MSC) phenotype are susceptible to material stiffness, because proliferation, migration, lineage determination, shape, cytoskeletal properties, expression profiles, cell surface receptor composition, integrin subunit expression, and nuclear shape and composition of CHs and/or MSCs are stiffness-regulated. Moreover, material stiffness modulates MSC immuno-modulatory and angiogenic properties, transforming growth factor beta 1 (TGF-ß1)-induced lineage determination, and CH re-differentiation/de-differentiation, collagen type II fragment production, and TGF-ß1- and interleukin 1 beta (IL-1ß)-induced changes in cell stiffness and traction force. We then integrated the available molecular signaling data into a stiffness-regulated CH phenotype model. Overall, we recommend using material stiffness for controlling cell phenotype, as this would be a promising design cornerstone for novel future-oriented, cell-instructive biomaterials for clinical high-quality AC repair tissue.


Assuntos
Materiais Biocompatíveis/química , Cartilagem Articular/efeitos dos fármacos , Condrócitos/efeitos dos fármacos , Mecanotransdução Celular/genética , Osteoartrite/terapia , Regeneração/efeitos dos fármacos , Materiais Biocompatíveis/uso terapêutico , Biomarcadores/metabolismo , Cartilagem Articular/imunologia , Cartilagem Articular/patologia , Cartilagem Articular/cirurgia , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Condrócitos/citologia , Condrócitos/metabolismo , Condrogênese/efeitos dos fármacos , Condrogênese/genética , Colágeno Tipo II/genética , Colágeno Tipo II/metabolismo , Regulação da Expressão Gênica , Dureza/fisiologia , Humanos , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Osteoartrite/genética , Osteoartrite/imunologia , Osteoartrite/cirurgia , Fenótipo , Regeneração/genética , Fatores de Transcrição SOX9/genética , Fatores de Transcrição SOX9/metabolismo , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
16.
Diabetologia ; 62(2): 223-232, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30465066

RESUMO

Adipose tissue remains a cryptic organ. The ubiquitous presence of adipocytes, the different fat pads in distinct anatomical locations, the many different types of fat, in each case with their distinct precursor populations, and the ability to interchange into other types of fat cells or even de-differentiate altogether, offers a staggering amount of complexity to the adipose tissue organ as a whole. Adipose tissue holds the key to improving our understanding of systemic metabolic homeostasis. As such, understanding adipose tissue physiology offers the basis for a mechanistic understanding of the pathophysiology of diabetes. This review presents some of the lesser known aspects of this fascinating tissue, which consistently still offers much opportunity for the discovery of novel targets for pharmacological intervention.


Assuntos
Adipócitos/metabolismo , Tecido Adiposo/metabolismo , Diabetes Mellitus/metabolismo , Obesidade/metabolismo , Adipocinas/metabolismo , Animais , Metabolismo Energético/fisiologia , Humanos
17.
EMBO J ; 34(11): 1572-88, 2015 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-25908840

RESUMO

Transcriptional networks defining stemness in adult neural stem cells (NSCs) are largely unknown. We used the proximal cis-regulatory element (pCRE) of the retina-specific homeobox gene 2 (rx2) to address such a network. Lineage analysis in the fish retina identified rx2 as marker for multipotent NSCs. rx2-positive cells located in the peripheral ciliary marginal zone behave as stem cells for the neuroretina, or the retinal pigmented epithelium. We identified upstream regulators of rx2 interrogating the rx2 pCRE in a trans-regulation screen and focused on four TFs (Sox2, Tlx, Gli3, and Her9) activating or repressing rx2 expression. We demonstrated direct interaction of the rx2 pCRE with the four factors in vitro and in vivo. By conditional mosaic gain- and loss-of-function analyses, we validated the activity of those factors on regulating rx2 transcription and consequently modulating neuroretinal and RPE stem cell features. This becomes obvious by the rx2-mutant phenotypes that together with the data presented above identify rx2 as a transcriptional hub balancing stemness of neuroretinal and RPE stem cells in the adult fish retina.


Assuntos
Células-Tronco Adultas/metabolismo , Proteínas de Peixes/metabolismo , Proteínas de Homeodomínio/metabolismo , Células-Tronco Neurais/metabolismo , Oryzias/metabolismo , Retina/metabolismo , Fatores de Transcrição/metabolismo , Células-Tronco Adultas/citologia , Animais , Proteínas de Peixes/genética , Regulação da Expressão Gênica/fisiologia , Proteínas de Homeodomínio/genética , Células-Tronco Neurais/citologia , Oryzias/genética , Retina/citologia , Fatores de Transcrição/genética
18.
Biochem Biophys Res Commun ; 508(2): 440-444, 2019 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-30502082

RESUMO

Hyperuricemia contributes to vascular injury and dysfunction, yet the potential mechanisms are not well understood. Uric acid (UA) has been found to stimulate macrophage migration inhibitory factor (MIF) up-regulation in renal tubules from rats subjected to UA-induced nephropathy. Given that MIF is able to induce vascular smooth muscle cell (VSMC) de-differentiation (from contractile state to a secretory state), we thus hypothesized that UA-induced vascular injury is via up-regulating of MIF in VSMCs, which enhancing vascular inflammation and VSMC transition. Within a mouse model of UA injection (500 mg/kg, twice/day, 14 days), we measured circulating and vascular MIF levels under UA stimulation at 6 h, day 1, and 14. We tested the efficacy of MIF inhibitor (10 mg/kg, twice/day, 14 days) on UA-induced vascular inflammation and remodeling. High plasma level of UA induced vascular MIF release into the plasma at acute phase. In the chronic phase, the protein level of MIF is up-regulated in the vessels. MIF inhibitor suppressed vascular inflammatory responses, repressed VSMC de-differentiation, and attenuated vascular remodeling and dysfunction following UA stimulation. Knockdown of MIF in cultured VSMCs repressed UA-induced de-differentiation. Our results provided a novel mechanism for MIF-mediated vascular injury in response to UA stimulation, and suggested that anti-MIF interventions may be of therapeutic value in hyperuricemic patients.


Assuntos
Fatores Inibidores da Migração de Macrófagos/antagonistas & inibidores , Remodelação Vascular/fisiologia , Animais , Desdiferenciação Celular/efeitos dos fármacos , Desdiferenciação Celular/fisiologia , Células Cultivadas , Modelos Animais de Doenças , Técnicas de Silenciamento de Genes , Humanos , Hiperuricemia/patologia , Hiperuricemia/fisiopatologia , Oxirredutases Intramoleculares/antagonistas & inibidores , Oxirredutases Intramoleculares/genética , Oxirredutases Intramoleculares/fisiologia , Fatores Inibidores da Migração de Macrófagos/genética , Fatores Inibidores da Migração de Macrófagos/fisiologia , Masculino , Camundongos , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , RNA Interferente Pequeno/genética , Ratos , Ratos Sprague-Dawley , Ácido Úrico/toxicidade , Remodelação Vascular/efeitos dos fármacos , Vasculite/induzido quimicamente , Vasculite/prevenção & controle
19.
BMC Cancer ; 19(1): 259, 2019 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-30902086

RESUMO

BACKGROUND: Though the management of malignancies has improved vastly in recent years, many treatment options lack the desired efficacy and fail to adequately augment patient morbidity and mortality. It is increasingly clear that patient response to therapy is unique to each individual, necessitating personalised, or 'precision' medical care. This demand extends to thyroid cancer; ~ 10% patients fail to respond to radioiodine treatment due to loss of phenotypic differentiation, exposing the patient to unnecessary ionising radiation, as well as delaying treatment with alternative therapies. METHODS: Human thyroid tissue (n = 23, malignant and benign) was live-sliced (5 mm diameter × 350-500 µm thickness) then analysed or incorporated into a microfluidic culture device for 96 h (37 °C). Successful maintenance of tissue was verified by histological (H&E), flow cytometric propidium iodide or trypan blue uptake, immunohistochemical (Ki67 detection/ BrdU incorporation) and functional analysis (thyroxine [T4] output) in addition to analysis of culture effluent for the cell death markers lactate dehydrogenase (LDH) and dead-cell protease (DCP). Apoptosis was investigated by Terminal deoxynucleotidyl transferase dUTP nick end labelling (TUNEL). Differentiation was assessed by evaluation of thyroid transcription factor (TTF1) and sodium iodide symporter (NIS) expression (western blotting). RESULTS: Maintenance of gross tissue architecture was observed. Analysis of dissociated primary thyroid cells using flow cytometry both prior to and post culture demonstrated no significant change in the proportion of viable cells. LDH and DCP release from on-chip thyroid tissue indicated that after an initial raised level of release, signifying cellular damage, detectable levels dropped markedly. A significant increase in apoptosis (p < 0.01) was observed after tissue was perfused with etoposide and JNK inhibitor, but not in control tissue incubated for the same time period. No significant difference in Ki-67 positivity or TTF1/NIS expression was detected between fresh and post-culture thyroid tissue samples, moreover BrdU positive nuclei indicated on-chip cellular proliferation. Cultured thyroid explants were functionally viable as determined by production of T4 throughout the culture period. CONCLUSIONS: The described microfluidic platform can maintain the viability of thyroid tissue slices ex vivo for a minimum of four days, providing a platform for the assessment of thyroid tissue radioiodine sensitivity/adjuvant therapies in real time.


Assuntos
Antineoplásicos/farmacologia , Ensaios de Seleção de Medicamentos Antitumorais/instrumentação , Dispositivos Lab-On-A-Chip , Técnicas de Cultura de Tecidos/instrumentação , Antineoplásicos/uso terapêutico , Resistencia a Medicamentos Antineoplásicos , Humanos , Glândula Tireoide/efeitos dos fármacos , Glândula Tireoide/patologia , Neoplasias da Glândula Tireoide/tratamento farmacológico , Neoplasias da Glândula Tireoide/patologia
20.
BMC Cancer ; 19(1): 518, 2019 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-31146720

RESUMO

BACKGROUND: Prostate cancer displays different morphologies which, in turn, affect patient outcome. This fact prompted questions about the lineage relationship between differentiated, more treatable prostate adenocarcinoma and poorly differentiated, less treatable non-adenocarcinoma including small cell carcinoma, and the molecular mechanism underlying prostate cancer differentiation. METHODS: Newly available non-adenocarcinoma/small cell carcinoma PDX LuCaP lines were analyzed for expression of stem cell transcription factors (scTF) LIN28A, NANOG, POU5F1, SOX2, which are responsible for reprogramming or de-differentiation. cDNA of these genes were cloned from small cell carcinoma LuCaP 145.1 into expression vectors to determine if they could function in reprogramming. RESULTS: Expression of scTF was detected in small cell carcinoma LuCaP 93, 145.1, 145.2, and non-adenocarcinoma LuCaP 173.1, 173.2A. Transfection of scTF from LuCaP 145.1 altered the gene expression of prostate non-small cell carcinoma cells, as well as fibroblasts. The resultant cells grew in stem-like colonies. Of note was a 10-fold lower expression of B2M in the transfected cells. Low B2M was also characteristic of LuCaP 145.1. Conversely, B2M was increased when stem cells were induced to differentiate. CONCLUSIONS: This work suggested a pathway in the emergence of non-adenocarcinoma/small cell carcinoma from adenocarcinoma through activation of scTF genes that produced cancer de-differentiation.


Assuntos
Adenocarcinoma/patologia , Carcinoma de Células Pequenas/patologia , Linhagem da Célula/genética , Neoplasias da Próstata/patologia , Adenocarcinoma/genética , Carcinoma de Células Pequenas/genética , Desdiferenciação Celular/genética , Linhagem Celular , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Proteína Homeobox Nanog/genética , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Fator 3 de Transcrição de Octâmero/genética , Neoplasias da Próstata/genética , Proteínas de Ligação a RNA/genética , Fatores de Transcrição SOXB1/genética , Microglobulina beta-2/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa