Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 19(6): e0303577, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38843233

RESUMO

Malic Enzyme 1 (ME1) plays an integral role in fatty acid synthesis and cellular energetics through its production of NADPH and pyruvate. As such, it has been identified as a gene of interest in obesity, type 2 diabetes, and an array of epithelial cancers, with most work being performed in vitro. The current standard model for ME1 loss in vivo is the spontaneous Mod-1 null allele, which produces a canonically inactive form of ME1. Herein, we describe two new genetically engineered mouse models exhibiting ME1 loss at dynamic timepoints. Using murine embryonic stem cells and Flp/FRT and Cre/loxP class switch recombination, we established a germline Me1 knockout model (Me1 KO) and an inducible conditional knockout model (Me1 cKO), activated upon tamoxifen treatment in adulthood. Collectively, neither the Me1 KO nor Me1 cKO models exhibited deleterious phenotype under standard laboratory conditions. Knockout of ME1 was validated by immunohistochemistry and genotype confirmed by PCR. Transmission patterns favor Me1 loss in Me1 KO mice when maternally transmitted to male progeny. Hematological examination of these models through complete blood count and serum chemistry panels revealed no discrepancy with their wild-type counterparts. Orthotopic pancreatic tumors in Me1 cKO mice grow similarly to Me1 expressing mice. Similarly, no behavioral phenotype was observed in Me1 cKO mice when aged for 52 weeks. Histological analysis of several tissues revealed no pathological phenotype. These models provide a more modern approach to ME1 knockout in vivo while opening the door for further study into the role of ME1 loss under more biologically relevant, stressful conditions.


Assuntos
Malato Desidrogenase , Camundongos Knockout , Fenótipo , Animais , Malato Desidrogenase/metabolismo , Malato Desidrogenase/genética , Masculino , Camundongos , Feminino , Células Germinativas/metabolismo , Camundongos Endogâmicos C57BL
2.
bioRxiv ; 2024 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-38915707

RESUMO

Injury can cause differentiated cells to undergo massive reprogramming to become proliferative to repair tissue via a cellular program called paligenosis. Gastric digestive-enzyme-secreting chief cells use paligenosis to reprogram into progenitor-like Spasmolytic-Polypeptide Expressing Metaplasia (SPEM) cells. Stage 1 of paligenosis is to downscale mature cell architecture via a process involving lysosomes. Here, we noticed that sulfated glycoproteins (which are metaplasia and cancer markers in mice and humans) were not digested during paligenosis but excreted into the gland lumen. Various genetic and pharmacological approaches showed that endoplasmic reticulum membranes and secretory granule cargo were also excreted and that the process proceeded in parallel with, but was independent lysosomal activity. 3-dimensional light and electron-microscopy demonstrated that excretion occurred via unique, complex, multi-chambered invaginations of the apical plasma membrane. As this lysosome-independent cell cleansing process does not seem to have been priorly described, we termed it "cathartocytosis". Cathartocytosis allows a cell to rapidly eject excess material (likely in times of extreme stress such as are induced by paligenosis) without waiting for autophagic and lysosomal digestion. We speculate the ejection of sulfated glycoproteins (likely mucins) would aid in downscaling and might also help bind and flush pathogens (like H pylori which causes SPEM) away from tissue.

3.
J Clin Invest ; 2024 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-38869951

RESUMO

Neutrophil hyperactivity and neutrophil extracellular trap release (NETosis) appear to play important roles in the pathogenesis of the thromboinflammatory autoimmune disease known as antiphospholipid syndrome (APS). The understanding of neutrophil metabolism has advanced tremendously in the past decade, and accumulating evidence suggests that a variety of metabolic pathways guide neutrophil activities in health and disease. Our previous work characterizing the transcriptome of APS neutrophils revealed that genes related to glycolysis, glycogenolysis, and the pentose phosphate pathway (PPP) were significantly upregulated. Here, we found that APS patient neutrophils used glycolysis more avidly than healthy control neutrophils, especially when the neutrophils were from APS patients with a history of microvascular disease. In vitro, inhibiting either glycolysis or the PPP tempered phorbol myristate acetate- and APS IgG-induced NETosis, but not NETosis triggered by a calcium ionophore. In mice, inhibiting either glycolysis or the PPP reduced neutrophil reactive oxygen species production and suppressed APS IgG-induced NETosis ex vivo. When APS-associated thrombosis was evaluated in mice, inhibiting either glycolysis or the PPP markedly suppressed thrombosis and circulating NET remnants. In summary, these data identify a potential role for restraining neutrophil glucose flux in the treatment of APS.

4.
Cancer Discov ; 14(4): 683-689, 2024 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-38571435

RESUMO

Research on precancers, as defined as at-risk tissues and early lesions, is of high significance given the effectiveness of early intervention. We discuss the need for risk stratification to prevent overtreatment, an emphasis on the role of genetic and epigenetic aging when considering risk, and the importance of integrating macroenvironmental risk factors with molecules and cells in lesions and at-risk normal tissues for developing effective intervention and health policy strategies.


Assuntos
Lesões Pré-Cancerosas , Humanos , Lesões Pré-Cancerosas/genética , Lesões Pré-Cancerosas/patologia , Fatores de Risco
5.
J Nutr ; 154(4): 1153-1164, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38246358

RESUMO

BACKGROUND: Tissue repair and regeneration in the gastrointestinal system are crucial for maintaining homeostasis, with the process relying on intricate cellular interactions and affected by micro- and macro-nutrients. Iron, essential for various biological functions, plays a dual role in tissue healing by potentially causing oxidative damage and participating in anti-inflammatory mechanisms, underscoring its complex relationship with inflammation and tissue repair. OBJECTIVE: The study aimed to elucidate the role of low dietary iron in gastrointestinal tissue repair. METHODS: We utilized quantitative iron measurements to assess iron levels in inflamed regions of patients with ulcerative colitis and Crohn's disease. In addition, 3 mouse models of gastrointestinal injury/repair (dextran sulfate sodium-induced colitis, radiation injury, and wound biopsy) were used to assess the effects of low dietary iron on tissue repair. RESULTS: We found that levels of iron in inflamed regions of both patients with ulcerative colitis and Crohn's disease are elevated. Similarly, during gastrointestinal repair, iron levels were found to be heightened, specifically in intestinal epithelial cells across the 3 injury/repair models. Mice on a low-iron diet showed compromised tissue repair with reduced proliferation. In standard diet, epithelial cells and the stem cell compartment maintain adequate iron stores. However, during a period of iron deficiency, epithelial cells exhaust their iron reserves, whereas the stem cell compartments maintain their iron pools. During injury, when the stem compartment is disrupted, low iron levels impair proliferation and compromise repair mechanisms. CONCLUSIONS: Low dietary iron impairs intestinal repair through compromising the ability of epithelial cells to aid in intestinal proliferation.


Assuntos
Colite Ulcerativa , Colite , Doença de Crohn , Humanos , Camundongos , Animais , Doença de Crohn/patologia , Ferro da Dieta/efeitos adversos , Colite/induzido quimicamente , Cicatrização , Modelos Animais de Doenças , Ferro/farmacologia , Mucosa Intestinal , Sulfato de Dextrana/farmacologia , Camundongos Endogâmicos C57BL
6.
bioRxiv ; 2023 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-37986898

RESUMO

Activating mutations in KRAS extensively reprogram cellular metabolism to support the continuous growth, proliferation, and survival of pancreatic tumors. Targeting these metabolic dependencies are promising approaches for the treatment of established tumors. However, metabolic reprogramming is required early during tumorigenesis to provide transformed cells selective advantage towards malignancy. Acinar cells can give rise to pancreatic tumors through acinar-to-ductal metaplasia (ADM). Dysregulation of pathways that maintain acinar homeostasis accelerate tumorigenesis. During ADM, acinar cells transdifferentiate to duct-like cells, a process driven by oncogenic KRAS. The metabolic reprogramming that is required for the transdifferentiation in ADM is unclear. We performed transcriptomic analysis on mouse acinar cells undergoing ADM and found metabolic programs are globally enhanced, consistent with the transition of a specialized cell to a less differentiated phenotype with proliferative potential. Indeed, we and others have demonstrated how inhibiting metabolic pathways necessary for ADM can prevent transdifferentiation and tumorigenesis. Here, we also find NRF2-target genes are differentially expressed during ADM. Among these, we focused on the increase in the gene coding for NADPH-producing enzyme, Glucose-6-phosphate dehydrogenase (G6PD). Using established mouse models of KrasG12D-driven pancreatic tumorigenesis and G6PD-deficiency, we find that mutant G6pd accelerates ADM and pancreatic intraepithelial neoplasia. Acceleration of cancer initiation with G6PD-deficiency is dependent on its NADPH-generating function in reactive oxygen species (ROS) management, as opposed to other outputs of the pentose phosphate pathway. Together, this work provides new insights into the function of metabolic pathways during early tumorigenesis.

7.
Adv Physiol Educ ; 47(4): 910-918, 2023 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-37769043

RESUMO

The development of science writing and presentation skills is necessary for a successful science career. Too often these skills are not included in pre- or postsecondary science, technology, engineering, and mathematics (STEM) education, leading to a disconnect between high schoolers' expectations for college preparedness and the skills needed to succeed in college. The Young Scientist Program Summer Focus recruits high school students from historically marginalized backgrounds to participate in 8-week summer internships at Washington University in St. Louis. Students conduct hands-on biomedical research projects under the mentorship of Washington University scientists (graduate students, postdoctorates, lab staff). Here, we present the curriculum for a science communication course that accompanies this early research experience. The course is designed to strengthen students' communication skills (critical reading, writing, presenting, and peer review) through a combination of weekly lectures and active learning methods. It prepares students for the capstone of their summer internship: writing a scientific paper and presenting their results at a closing symposium. We administered pre- and postprogram surveys to four Summer Focus cohorts to determine whether the course met its learning objectives. We found significant improvements in students' self-confidence in reading, interpreting, and communicating scientific data. Thus, this course provides a successful model for introducing science literacy and communication skills that are necessary for any career in STEM. We provide a detailed outline of the course structure and content so that this training can be incorporated into any undergraduate and graduate research programs.NEW & NOTEWORTHY Strong communication skills are necessary for a successful scientific career. Here, we describe the curriculum for a science communication course designed to accompany high school students participating in a summer biomedical research program. The course aims to improve their scientific literacy and communication skills. Students learn to read and understand scientific literature, write a paper about their summer research project, present their results, and provide feedback to peers. We found significant improvements in students' self-confidence in reading, interpreting, and communicating scientific data after completing the course. This successful model serves as a guide for students participating in their first research experience and provides the skills for success in future science, technology, engineering, and mathematics education and careers. The curriculum presented here can be easily adapted for any research program, including undergraduate summer research experiences and graduate student laboratory rotations.


Assuntos
Currículo , Instituições Acadêmicas , Humanos , Estudantes , Comunicação , Redação
8.
Med ; 3(2): 87-89, 2022 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-35590211

RESUMO

Dietary interventions hold promise in cancer treatments. However, clinical application has been limited by a lack of mechanistic understanding of the metabolic effects. In this issue, Yang et al. use mouse models and isotope tracing to demonstrate that the ketogenic diet induces reductive stress and primes pancreatic tumors for chemotherapy.1.


Assuntos
Dieta Cetogênica , Neoplasias Pancreáticas , Animais , Carboidratos , Modelos Animais de Doenças , Camundongos , Neoplasias Pancreáticas/tratamento farmacológico
9.
EMBO Rep ; 22(9): e51806, 2021 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-34309175

RESUMO

Differentiated cells across multiple species and organs can re-enter the cell cycle to aid in injury-induced tissue regeneration by a cellular program called paligenosis. Here, we show that activating transcription factor 3 (ATF3) is induced early during paligenosis in multiple cellular contexts, transcriptionally activating the lysosomal trafficking gene Rab7b. ATF3 and RAB7B are upregulated in gastric and pancreatic digestive-enzyme-secreting cells at the onset of paligenosis Stage 1, when cells massively induce autophagic and lysosomal machinery to dismantle differentiated cell morphological features. Their expression later ebbs before cells enter mitosis during Stage 3. Atf3-/- mice fail to induce RAB7-positive autophagic and lysosomal vesicles, eventually causing increased death of cells en route to Stage 3. Finally, we observe that ATF3 is expressed in human gastric metaplasia and during paligenotic injury across multiple other organs and species. Thus, our findings indicate ATF3 is an evolutionarily conserved gene orchestrating the early paligenotic autodegradative events that must occur before cells are poised to proliferate and contribute to tissue repair.


Assuntos
Fator 3 Ativador da Transcrição , Plasticidade Celular , Fator 3 Ativador da Transcrição/genética , Animais , Ciclo Celular , Diferenciação Celular , Metaplasia/genética , Camundongos
10.
JCI Insight ; 6(14)2021 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-34138755

RESUMO

Cancer cells reprogram cellular metabolism to maintain adequate nutrient pools to sustain proliferation. Moreover, autophagy is a regulated mechanism to break down dysfunctional cellular components and recycle cellular nutrients. However, the requirement for autophagy and the integration in cancer cell metabolism is not clear in colon cancer. Here, we show a cell-autonomous dependency of autophagy for cell growth in colorectal cancer. Loss of epithelial autophagy inhibits tumor growth in both sporadic and colitis-associated cancer models. Genetic and pharmacological inhibition of autophagy inhibits cell growth in colon cancer-derived cell lines and patient-derived enteroid models. Importantly, normal colon epithelium and patient-derived normal enteroid growth were not decreased following autophagy inhibition. To couple the role of autophagy to cellular metabolism, a cell culture screen in conjunction with metabolomic analysis was performed. We identified a critical role of autophagy to maintain mitochondrial metabolites for growth. Loss of mitochondrial recycling through inhibition of mitophagy hinders colon cancer cell growth. These findings have revealed a cell-autonomous role of autophagy that plays a critical role in regulating nutrient pools in vivo and in cell models, and it provides therapeutic targets for colon cancer.


Assuntos
Neoplasias Associadas a Colite/imunologia , Mitocôndrias/metabolismo , Mitofagia/imunologia , Nutrientes/deficiência , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Colite/induzido quimicamente , Colite/complicações , Colite/imunologia , Colite/patologia , Neoplasias Associadas a Colite/tratamento farmacológico , Neoplasias Associadas a Colite/genética , Neoplasias Associadas a Colite/patologia , Colo/citologia , Colo/imunologia , Colo/patologia , Sulfato de Dextrana/administração & dosagem , Sulfato de Dextrana/toxicidade , Modelos Animais de Doenças , Feminino , Humanos , Mucosa Intestinal/citologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/patologia , Masculino , Metabolômica , Camundongos , Camundongos Transgênicos , Mitocôndrias/imunologia , Mitofagia/efeitos dos fármacos
11.
EMBO J ; 37(7)2018 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-29467218

RESUMO

In 1900, Adami speculated that a sequence of context-independent energetic and structural changes governed the reversion of differentiated cells to a proliferative, regenerative state. Accordingly, we show here that differentiated cells in diverse organs become proliferative via a shared program. Metaplasia-inducing injury caused both gastric chief and pancreatic acinar cells to decrease mTORC1 activity and massively upregulate lysosomes/autophagosomes; then increase damage associated metaplastic genes such as Sox9; and finally reactivate mTORC1 and re-enter the cell cycle. Blocking mTORC1 permitted autophagy and metaplastic gene induction but blocked cell cycle re-entry at S-phase. In kidney and liver regeneration and in human gastric metaplasia, mTORC1 also correlated with proliferation. In lysosome-defective Gnptab-/- mice, both metaplasia-associated gene expression changes and mTORC1-mediated proliferation were deficient in pancreas and stomach. Our findings indicate differentiated cells become proliferative using a sequential program with intervening checkpoints: (i) differentiated cell structure degradation; (ii) metaplasia- or progenitor-associated gene induction; (iii) cell cycle re-entry. We propose this program, which we term "paligenosis", is a fundamental process, like apoptosis, available to differentiated cells to fuel regeneration following injury.


Assuntos
Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Regeneração/fisiologia , Células Acinares , Animais , Autofagossomos/fisiologia , Ciclo Celular/fisiologia , Transdiferenciação Celular/fisiologia , Reprogramação Celular/fisiologia , Celulas Principais Gástricas/patologia , Trato Gastrointestinal/patologia , Expressão Gênica , Humanos , Lisossomos , Metaplasia/genética , Camundongos , Camundongos Endogâmicos C57BL , Fase S/fisiologia , Fatores de Transcrição SOX9/metabolismo , Estômago/lesões , Estômago/patologia , Transferases (Outros Grupos de Fosfato Substituídos)/genética
12.
Gastroenterology ; 154(4): 839-843.e2, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29248442

RESUMO

Spasmolytic polypeptide-expressing metaplasia (SPEM) develops in patients with chronic atrophic gastritis due to infection with Helicobacter pylori; it might be a precursor to intestinal metaplasia and gastric adenocarcinoma. Lineage tracing experiments of the gastric corpus in mice have not established whether SPEM derives from proliferating stem cells or differentiated, post-mitotic zymogenic chief cells in the gland base. We investigated whether differentiated cells can give rise to SPEM using a nongenetic approach in mice. Mice were given intraperitoneal injections of 5-fluorouracil, which blocked gastric cell proliferation, plus tamoxifen to induce SPEM. Based on analyses of molecular and histologic markers, we found SPEM developed even in the absence of cell proliferation. SPEM therefore did not arise from stem cells. In histologic analyses of gastric resection specimens from 10 patients with adenocarcinoma, we found normal zymogenic chief cells that were transitioning into SPEM cells only in gland bases, rather than the proliferative stem cell zone. Our findings indicate that SPEM can arise by direct reprogramming of existing cells-mainly of chief cells.


Assuntos
Adenocarcinoma/patologia , Transdiferenciação Celular , Celulas Principais Gástricas/patologia , Lesões Pré-Cancerosas/patologia , Neoplasias Gástricas/patologia , Estômago/patologia , Adenocarcinoma/metabolismo , Adenocarcinoma/cirurgia , Animais , Biomarcadores Tumorais/metabolismo , Linhagem da Célula , Proliferação de Células , Transdiferenciação Celular/efeitos dos fármacos , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Celulas Principais Gástricas/efeitos dos fármacos , Celulas Principais Gástricas/metabolismo , Fluoruracila/farmacologia , Gastrectomia , Mucosa Gástrica/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular , Metaplasia , Camundongos , Peptídeos/metabolismo , Fenótipo , Lesões Pré-Cancerosas/metabolismo , Lesões Pré-Cancerosas/cirurgia , Estômago/efeitos dos fármacos , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/cirurgia , Fatores de Tempo
13.
EMBO J ; 36(16): 2318-2320, 2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28666994
14.
J Biol Chem ; 290(2): 706-15, 2015 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-25381248

RESUMO

The dominant paradigm for spectrin function is that (αß)2-spectrin tetramers or higher order oligomers form membrane-associated two-dimensional networks in association with F-actin to reinforce the plasma membrane. Tetramerization is an essential event in such structures. We characterize the tetramerization interaction between α-spectrin and ß-spectrins in Drosophila. Wild-type α-spectrin binds to both ß- and ßH-chains with high affinity, resembling other non-erythroid spectrins. However, α-spec(R22S), a tetramerization site mutant homologous to the pathological α-spec(R28S) allele in humans, eliminates detectable binding to ß-spectrin and reduces binding to ßH-spectrin ∼1000-fold. Even though spectrins are essential proteins, α-spectrin(R22S) rescues α-spectrin mutants to adulthood with only minor phenotypes indicating that tetramerization, and thus conventional network formation, is not the essential function of non-erythroid spectrin. Our data provide the first rigorous test for the general requirement for tetramer-based non-erythroid spectrin networks throughout an organism and find that they have very limited roles, in direct contrast to the current paradigm.


Assuntos
Membrana Celular/genética , Drosophila melanogaster/genética , Espectrina/genética , Citoesqueleto de Actina/genética , Citoesqueleto de Actina/metabolismo , Actinas/genética , Actinas/metabolismo , Animais , Membrana Celular/metabolismo , Drosophila melanogaster/crescimento & desenvolvimento , Drosophila melanogaster/metabolismo , Humanos , Mutação , Multimerização Proteica , Espectrina/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...