Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53.465
Filtrar
Más filtros

Intervalo de año de publicación
1.
Annu Rev Immunol ; 42(1): 179-206, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38166256

RESUMEN

T cell responses must be balanced to ensure adequate protection against malignant transformation and an array of pathogens while also limiting damage to healthy cells and preventing autoimmunity. T cell exhaustion serves as a regulatory mechanism to limit the activity and effector function of T cells undergoing chronic antigen stimulation. Exhausted T cells exhibit poor proliferative potential; high inhibitory receptor expression; altered transcriptome, epigenome, and metabolism; and, most importantly, reduced effector function. While exhaustion helps to restrain damage caused by aberrant T cells in settings of autoimmune disease, it also limits the ability of cells to respond against persistent infection and cancer, leading to disease progression. Here we review the process of T cell exhaustion, detailing the key characteristics and drivers as well as highlighting our current understanding of the underlying transcriptional and epigenetic programming. We also discuss how exhaustion can be targeted to enhance T cell functionality in cancer.


Asunto(s)
Neoplasias , Linfocitos T , Humanos , Animales , Neoplasias/inmunología , Neoplasias/etiología , Neoplasias/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Epigénesis Genética , Activación de Linfocitos/inmunología , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/metabolismo , Agotamiento de Células T
2.
Annu Rev Immunol ; 42(1): 521-550, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38382538

RESUMEN

Immune checkpoint blockade (ICB) induces a remarkable and durable response in a subset of cancer patients. However, most patients exhibit either primary or acquired resistance to ICB. This resistance arises from a complex interplay of diverse dynamic mechanisms within the tumor microenvironment (TME). These mechanisms include genetic, epigenetic, and metabolic alterations that prevent T cell trafficking to the tumor site, induce immune cell dysfunction, interfere with antigen presentation, drive heightened expression of coinhibitory molecules, and promote tumor survival after immune attack. The TME worsens ICB resistance through the formation of immunosuppressive networks via immune inhibition, regulatory metabolites, and abnormal resource consumption. Finally, patient lifestyle factors, including obesity and microbiome composition, influence ICB resistance. Understanding the heterogeneity of cellular, molecular, and environmental factors contributing to ICB resistance is crucial to develop targeted therapeutic interventions that enhance the clinical response. This comprehensive overview highlights key mechanisms of ICB resistance that may be clinically translatable.


Asunto(s)
Resistencia a Antineoplásicos , Inhibidores de Puntos de Control Inmunológico , Inmunoterapia , Neoplasias , Microambiente Tumoral , Humanos , Microambiente Tumoral/inmunología , Neoplasias/inmunología , Neoplasias/terapia , Neoplasias/metabolismo , Neoplasias/etiología , Resistencia a Antineoplásicos/inmunología , Animales , Inmunoterapia/métodos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inhibidores de Puntos de Control Inmunológico/farmacología , Epigénesis Genética
3.
Annu Rev Immunol ; 42(1): 455-488, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38360546

RESUMEN

Ten-eleven translocation (TET) proteins are iron-dependent and α-ketoglutarate-dependent dioxygenases that sequentially oxidize the methyl group of 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC), 5-formylcytosine (5fC) and 5-carboxylcytosine (5caC). All three epigenetic modifications are intermediates in DNA demethylation. TET proteins are recruited by transcription factors and by RNA polymerase II to modify 5mC at enhancers and gene bodies, thereby regulating gene expression during development, cell lineage specification, and cell activation. It is not yet clear, however, how the established biochemical activities of TET enzymes in oxidizing 5mC and mediating DNA demethylation relate to the known association of TET deficiency with inflammation, clonal hematopoiesis, and cancer. There are hints that the ability of TET deficiency to promote cell proliferation in a signal-dependent manner may be harnessed for cancer immunotherapy. In this review, we draw upon recent findings in cells of the immune system to illustrate established as well as emerging ideas of how TET proteins influence cellular function.


Asunto(s)
Desmetilación del ADN , Dioxigenasas , Inmunoterapia , Inflamación , Neoplasias , Humanos , Neoplasias/terapia , Neoplasias/inmunología , Neoplasias/etiología , Neoplasias/metabolismo , Animales , Inflamación/metabolismo , Inflamación/inmunología , Inmunoterapia/métodos , Dioxigenasas/metabolismo , Sistema Inmunológico/metabolismo , Sistema Inmunológico/inmunología , Epigénesis Genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas/genética , Metilación de ADN , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , Oxigenasas de Función Mixta/metabolismo , Oxigenasas de Función Mixta/genética
4.
Annu Rev Immunol ; 40: 45-74, 2022 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-35471840

RESUMEN

The transformative success of antibodies targeting the PD-1 (programmed death 1)/B7-H1 (B7 homolog 1) pathway (anti-PD therapy) has revolutionized cancer treatment. However, only a fraction of patients with solid tumors and some hematopoietic malignancies respond to anti-PD therapy, and the reason for failure in other patients is less known. By dissecting the mechanisms underlying this resistance, current studies reveal that the tumor microenvironment is a major location for resistance to occur. Furthermore, the resistance mechanisms appear to be highly heterogeneous. Here, we discuss recent human cancer data identifying mechanisms of resistance to anti-PD therapy. We review evidence for immune-based resistance mechanisms such as loss of neoantigens, defects in antigen presentation and interferon signaling, immune inhibitory molecules, and exclusion of T cells. We also review the clinical evidence for emerging mechanisms of resistance to anti-PD therapy, such as alterations in metabolism, microbiota, and epigenetics. Finally, we discuss strategies to overcome anti-PD therapy resistance and emphasize the need to develop additional immunotherapies based on the concept of normalization cancer immunotherapy.


Asunto(s)
Neoplasias , Receptor de Muerte Celular Programada 1 , Animales , Antígeno B7-H1 , Humanos , Inmunoterapia , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Linfocitos T , Microambiente Tumoral
5.
Annu Rev Immunol ; 36: 221-246, 2018 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-29328786

RESUMEN

Researchers are intensifying efforts to understand the mechanisms by which changes in metabolic states influence differentiation programs. An emerging objective is to define how fluctuations in metabolites influence the epigenetic states that contribute to differentiation programs. This is because metabolites such as S-adenosylmethionine, acetyl-CoA, α-ketoglutarate, 2-hydroxyglutarate, and butyrate are donors, substrates, cofactors, and antagonists for the activities of epigenetic-modifying complexes and for epigenetic modifications. We discuss this topic from the perspective of specialized CD4+ T cells as well as effector and memory T cell differentiation programs. We also highlight findings from embryonic stem cells that give mechanistic insight into how nutrients processed through pathways such as glycolysis, glutaminolysis, and one-carbon metabolism regulate metabolite levels to influence epigenetic events and discuss similar mechanistic principles in T cells. Finally, we highlight how dysregulated environments, such as the tumor microenvironment, might alter programming events.


Asunto(s)
Diferenciación Celular/genética , Diferenciación Celular/inmunología , Metabolismo Energético , Epigénesis Genética , Animales , Biomarcadores , Regulación del Desarrollo de la Expresión Génica , Humanos , Neoplasias/etiología , Neoplasias/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología
6.
Annu Rev Immunol ; 36: 247-277, 2018 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-29328785

RESUMEN

The liver is a key, frontline immune tissue. Ideally positioned to detect pathogens entering the body via the gut, the liver appears designed to detect, capture, and clear bacteria, viruses, and macromolecules. Containing the largest collection of phagocytic cells in the body, this organ is an important barrier between us and the outside world. Importantly, as portal blood also transports a large number of foreign but harmless molecules (e.g., food antigens), the liver's default immune status is anti-inflammatory or immunotolerant; however, under appropriate conditions, the liver is able to mount a rapid and robust immune response. This balance between immunity and tolerance is essential to liver function. Excessive inflammation in the absence of infection leads to sterile liver injury, tissue damage, and remodeling; insufficient immunity allows for chronic infection and cancer. Dynamic interactions between the numerous populations of immune cells in the liver are key to maintaining this balance and overall tissue health.


Asunto(s)
Fenómenos del Sistema Inmunológico , Hígado/inmunología , Hígado/metabolismo , Inmunidad Adaptativa , Animales , Hepatitis Viral Humana/inmunología , Hepatitis Viral Humana/metabolismo , Hepatitis Viral Humana/virología , Humanos , Tolerancia Inmunológica , Inmunidad Innata , Hígado/irrigación sanguínea , Hígado/citología , Neoplasias/etiología , Neoplasias/metabolismo , Neoplasias/patología
7.
Annu Rev Biochem ; 93(1): 367-387, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38594929

RESUMEN

Lysosomes are the degradative endpoints of material delivered by endocytosis and autophagy and are therefore particularly prone to damage. Membrane permeabilization or full rupture of lysosomal or late endosomal compartments is highly deleterious because it threatens cellular homeostasis and can elicit cell death and inflammatory signaling. Cells have developed a complex response to endo-lysosomal damage that largely consists of three branches. Initially, a number of repair pathways are activated to restore the integrity of the lysosomal membrane. If repair fails or if damage is too extensive, lysosomes are isolated and degraded by a form of selective autophagy termed lysophagy. Meanwhile, an mTORC1-governed signaling cascade drives biogenesis and regeneration of new lysosomal components to reestablish the full lysosomal capacity of the cell. This damage response is vital to counteract the effects of various conditions, including neurodegeneration and infection, and can constitute a critical vulnerability in cancer cells.


Asunto(s)
Autofagia , Endosomas , Lisosomas , Diana Mecanicista del Complejo 1 de la Rapamicina , Transducción de Señal , Lisosomas/metabolismo , Humanos , Animales , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Endosomas/metabolismo , Endocitosis , Neoplasias/metabolismo , Neoplasias/patología , Neoplasias/genética
8.
Annu Rev Biochem ; 93(1): 289-316, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38316136

RESUMEN

RAF family protein kinases are a key node in the RAS/RAF/MAP kinase pathway, the signaling cascade that controls cellular proliferation, differentiation, and survival in response to engagement of growth factor receptors on the cell surface. Over the past few years, structural and biochemical studies have provided new understanding of RAF autoregulation, RAF activation by RAS and the SHOC2 phosphatase complex, and RAF engagement with HSP90-CDC37 chaperone complexes. These studies have important implications for pharmacologic targeting of the pathway. They reveal RAF in distinct regulatory states and show that the functional RAF switch is an integrated complex of RAF with its substrate (MEK) and a 14-3-3 dimer. Here we review these advances, placing them in the context of decades of investigation of RAF regulation. We explore the insights they provide into aberrant activation of the pathway in cancer and RASopathies (developmental syndromes caused by germline mutations in components of the pathway).


Asunto(s)
Transducción de Señal , Quinasas raf , Proteínas ras , Humanos , Proteínas ras/metabolismo , Proteínas ras/genética , Proteínas ras/química , Quinasas raf/metabolismo , Quinasas raf/genética , Animales , Neoplasias/metabolismo , Neoplasias/genética , Neoplasias/patología , Proteínas 14-3-3/metabolismo , Proteínas 14-3-3/genética
9.
Annu Rev Biochem ; 93(1): 447-469, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38603559

RESUMEN

Lysosomes catabolize and recycle lipids and other biological molecules to maintain cellular homeostasis in diverse nutrient environments. Lysosomal lipid catabolism relies on the stimulatory activity of bis(monoacylglycero)phosphate (BMP), an enigmatic lipid whose levels are altered across myriad lysosome-associated diseases. Here, we review the discovery of BMP over half a century ago and its structural properties that facilitate the activation of lipid hydrolases and recruitment of their coactivators. We further discuss the current, yet incomplete, understanding of BMP catabolism and anabolism. To conclude, we discuss its role in lysosome-associated diseases and the potential for modulating its levels by pharmacologically activating and inhibiting the BMP synthase to therapeutically target lysosomal storage disorders, drug-induced phospholipidosis, Alzheimer's disease, Parkinson's disease, frontotemporal dementia, cancer, and viral infection.


Asunto(s)
Lisofosfolípidos , Enfermedades por Almacenamiento Lisosomal , Lisosomas , Monoglicéridos , Humanos , Lisosomas/metabolismo , Lisofosfolípidos/metabolismo , Monoglicéridos/metabolismo , Monoglicéridos/química , Animales , Enfermedades por Almacenamiento Lisosomal/metabolismo , Enfermedades por Almacenamiento Lisosomal/patología , Enfermedades por Almacenamiento Lisosomal/genética , Enfermedades por Almacenamiento Lisosomal/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/genética , Metabolismo de los Lípidos
10.
Cell ; 187(15): 3824-3828, 2024 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-39059359

RESUMEN

If you are a scientist and you only know one thing about tumor metabolism, it's likely the Warburg effect. But who was Otto Warburg, and how did his discoveries regarding the metabolism of tumors shape our current thinking about the metabolic needs of cancer cells?


Asunto(s)
Neoplasias , Efecto Warburg en Oncología , Humanos , Neoplasias/metabolismo , Neoplasias/patología , Historia del Siglo XX , Glucólisis , Historia del Siglo XXI , Animales
11.
Cell ; 187(14): 3602-3618.e20, 2024 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-38823389

RESUMEN

Purine nucleotides are vital for RNA and DNA synthesis, signaling, metabolism, and energy homeostasis. To synthesize purines, cells use two principal routes: the de novo and salvage pathways. Traditionally, it is believed that proliferating cells predominantly rely on de novo synthesis, whereas differentiated tissues favor the salvage pathway. Unexpectedly, we find that adenine and inosine are the most effective circulating precursors for supplying purine nucleotides to tissues and tumors, while hypoxanthine is rapidly catabolized and poorly salvaged in vivo. Quantitative metabolic analysis demonstrates comparative contribution from de novo synthesis and salvage pathways in maintaining purine nucleotide pools in tumors. Notably, feeding mice nucleotides accelerates tumor growth, while inhibiting purine salvage slows down tumor progression, revealing a crucial role of the salvage pathway in tumor metabolism. These findings provide fundamental insights into how normal tissues and tumors maintain purine nucleotides and highlight the significance of purine salvage in cancer.


Asunto(s)
Neoplasias , Nucleótidos de Purina , Purinas , Animales , Ratones , Purinas/metabolismo , Purinas/biosíntesis , Neoplasias/metabolismo , Neoplasias/patología , Nucleótidos de Purina/metabolismo , Humanos , Inosina/metabolismo , Hipoxantina/metabolismo , Ratones Endogámicos C57BL , Adenina/metabolismo , Línea Celular Tumoral , Femenino
12.
Cell ; 187(12): 2907-2918, 2024 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-38848676

RESUMEN

Cancer is a disease that stems from a fundamental liability inherent to multicellular life forms in which an individual cell is capable of reneging on the interests of the collective organism. Although cancer is commonly described as an evolutionary process, a less appreciated aspect of tumorigenesis may be the constraints imposed by the organism's developmental programs. Recent work from single-cell transcriptomic analyses across a range of cancer types has revealed the recurrence, plasticity, and co-option of distinct cellular states among cancer cell populations. Here, we note that across diverse cancer types, the observed cell states are proximate within the developmental hierarchy of the cell of origin. We thus posit a model by which cancer cell states are directly constrained by the organism's "developmental map." According to this model, a population of cancer cells traverses the developmental map, thereby generating a heterogeneous set of states whose interactions underpin emergent tumor behavior.


Asunto(s)
Modelos Biológicos , Neoplasias , Animales , Humanos , Carcinogénesis/patología , Carcinogénesis/genética , Neoplasias/patología , Neoplasias/genética , Neoplasias/metabolismo , Análisis de la Célula Individual , Transcriptoma/genética , Células Madre Neoplásicas/patología
13.
Cell ; 187(10): 2375-2392.e33, 2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38653238

RESUMEN

Lysine lactylation is a post-translational modification that links cellular metabolism to protein function. Here, we find that AARS1 functions as a lactate sensor that mediates global lysine lacylation in tumor cells. AARS1 binds to lactate and catalyzes the formation of lactate-AMP, followed by transfer of lactate to the lysince acceptor residue. Proteomics studies reveal a large number of AARS1 targets, including p53 where lysine 120 and lysine 139 in the DNA binding domain are lactylated. Generation and utilization of p53 variants carrying constitutively lactylated lysine residues revealed that AARS1 lactylation of p53 hinders its liquid-liquid phase separation, DNA binding, and transcriptional activation. AARS1 expression and p53 lacylation correlate with poor prognosis among cancer patients carrying wild type p53. ß-alanine disrupts lactate binding to AARS1, reduces p53 lacylation, and mitigates tumorigenesis in animal models. We propose that AARS1 contributes to tumorigenesis by coupling tumor cell metabolism to proteome alteration.


Asunto(s)
Carcinogénesis , Ácido Láctico , Proteína p53 Supresora de Tumor , Animales , Femenino , Humanos , Ratones , Carcinogénesis/metabolismo , Carcinogénesis/genética , Línea Celular Tumoral , Ácido Láctico/metabolismo , Lisina/metabolismo , Neoplasias/metabolismo , Neoplasias/genética , Procesamiento Proteico-Postraduccional , Proteína p53 Supresora de Tumor/metabolismo , Masculino
14.
Cell ; 187(13): 3409-3426.e24, 2024 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-38744281

RESUMEN

Alterations in extracellular matrix (ECM) architecture and stiffness represent hallmarks of cancer. Whether the biomechanical property of ECM impacts the functionality of tumor-reactive CD8+ T cells remains largely unknown. Here, we reveal that the transcription factor (TF) Osr2 integrates biomechanical signaling and facilitates the terminal exhaustion of tumor-reactive CD8+ T cells. Osr2 expression is selectively induced in the terminally exhausted tumor-specific CD8+ T cell subset by coupled T cell receptor (TCR) signaling and biomechanical stress mediated by the Piezo1/calcium/CREB axis. Consistently, depletion of Osr2 alleviates the exhaustion of tumor-specific CD8+ T cells or CAR-T cells, whereas forced Osr2 expression aggravates their exhaustion in solid tumor models. Mechanistically, Osr2 recruits HDAC3 to rewire the epigenetic program for suppressing cytotoxic gene expression and promoting CD8+ T cell exhaustion. Thus, our results unravel Osr2 functions as a biomechanical checkpoint to exacerbate CD8+ T cell exhaustion and could be targeted to potentiate cancer immunotherapy.


Asunto(s)
Linfocitos T CD8-positivos , Factores de Transcripción , Animales , Femenino , Humanos , Ratones , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Línea Celular Tumoral , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Matriz Extracelular/metabolismo , Histona Desacetilasas/metabolismo , Ratones Endogámicos C57BL , Neoplasias/inmunología , Neoplasias/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal , Agotamiento de Células T , Factores de Transcripción/metabolismo , Microambiente Tumoral , Estrés Mecánico
15.
Cell ; 187(10): 2536-2556.e30, 2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38653237

RESUMEN

Cysteine-focused chemical proteomic platforms have accelerated the clinical development of covalent inhibitors for a wide range of targets in cancer. However, how different oncogenic contexts influence cysteine targeting remains unknown. To address this question, we have developed "DrugMap," an atlas of cysteine ligandability compiled across 416 cancer cell lines. We unexpectedly find that cysteine ligandability varies across cancer cell lines, and we attribute this to differences in cellular redox states, protein conformational changes, and genetic mutations. Leveraging these findings, we identify actionable cysteines in NF-κB1 and SOX10 and develop corresponding covalent ligands that block the activity of these transcription factors. We demonstrate that the NF-κB1 probe blocks DNA binding, whereas the SOX10 ligand increases SOX10-SOX10 interactions and disrupts melanoma transcriptional signaling. Our findings reveal heterogeneity in cysteine ligandability across cancers, pinpoint cell-intrinsic features driving cysteine targeting, and illustrate the use of covalent probes to disrupt oncogenic transcription-factor activity.


Asunto(s)
Cisteína , Neoplasias , Animales , Humanos , Ratones , Línea Celular Tumoral , Cisteína/metabolismo , Cisteína/química , Ligandos , Melanoma/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , FN-kappa B/química , FN-kappa B/metabolismo , Oxidación-Reducción , Transducción de Señal , Factores de Transcripción SOXE/química , Factores de Transcripción SOXE/metabolismo
16.
Cell ; 187(16): 4389-4407.e15, 2024 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-38917788

RESUMEN

Fewer than 200 proteins are targeted by cancer drugs approved by the Food and Drug Administration (FDA). We integrate Clinical Proteomic Tumor Analysis Consortium (CPTAC) proteogenomics data from 1,043 patients across 10 cancer types with additional public datasets to identify potential therapeutic targets. Pan-cancer analysis of 2,863 druggable proteins reveals a wide abundance range and identifies biological factors that affect mRNA-protein correlation. Integration of proteomic data from tumors and genetic screen data from cell lines identifies protein overexpression- or hyperactivation-driven druggable dependencies, enabling accurate predictions of effective drug targets. Proteogenomic identification of synthetic lethality provides a strategy to target tumor suppressor gene loss. Combining proteogenomic analysis and MHC binding prediction prioritizes mutant KRAS peptides as promising public neoantigens. Computational identification of shared tumor-associated antigens followed by experimental confirmation nominates peptides as immunotherapy targets. These analyses, summarized at https://targets.linkedomics.org, form a comprehensive landscape of protein and peptide targets for companion diagnostics, drug repurposing, and therapy development.


Asunto(s)
Neoplasias , Proteogenómica , Humanos , Proteogenómica/métodos , Neoplasias/genética , Neoplasias/tratamiento farmacológico , Neoplasias/terapia , Neoplasias/metabolismo , Terapia Molecular Dirigida , Inmunoterapia/métodos , Antígenos de Neoplasias/metabolismo , Antígenos de Neoplasias/genética , Línea Celular Tumoral , Antineoplásicos/uso terapéutico , Antineoplásicos/farmacología , Péptidos/metabolismo , Proteómica , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo
17.
Cell ; 186(8): 1670-1688, 2023 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-36858045

RESUMEN

The uptake and metabolism of nutrients support fundamental cellular process from bioenergetics to biomass production and cell fate regulation. While many studies of cell metabolism focus on cancer cells, the principles of metabolism elucidated in cancer cells apply to a wide range of mammalian cells. The goal of this review is to discuss how the field of cancer metabolism provides a framework for revealing principles of cell metabolism and for dissecting the metabolic networks that allow cells to meet their specific demands. Understanding context-specific metabolic preferences and liabilities will unlock new approaches to target cancer cells to improve patient care.


Asunto(s)
Células , Redes y Vías Metabólicas , Neoplasias , Animales , Humanos , Fenómenos Fisiológicos Celulares , Metabolismo Energético , Mamíferos , Neoplasias/metabolismo , Células/metabolismo
18.
Cell ; 186(18): 3945-3967.e26, 2023 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-37582358

RESUMEN

Post-translational modifications (PTMs) play key roles in regulating cell signaling and physiology in both normal and cancer cells. Advances in mass spectrometry enable high-throughput, accurate, and sensitive measurement of PTM levels to better understand their role, prevalence, and crosstalk. Here, we analyze the largest collection of proteogenomics data from 1,110 patients with PTM profiles across 11 cancer types (10 from the National Cancer Institute's Clinical Proteomic Tumor Analysis Consortium [CPTAC]). Our study reveals pan-cancer patterns of changes in protein acetylation and phosphorylation involved in hallmark cancer processes. These patterns revealed subsets of tumors, from different cancer types, including those with dysregulated DNA repair driven by phosphorylation, altered metabolic regulation associated with immune response driven by acetylation, affected kinase specificity by crosstalk between acetylation and phosphorylation, and modified histone regulation. Overall, this resource highlights the rich biology governed by PTMs and exposes potential new therapeutic avenues.


Asunto(s)
Neoplasias , Procesamiento Proteico-Postraduccional , Proteómica , Humanos , Acetilación , Histonas/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Fosforilación , Proteómica/métodos
19.
Cell ; 186(8): 1541-1563, 2023 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-37059064

RESUMEN

Recent identification of oncogenic cells within healthy tissues and the prevalence of indolent cancers found incidentally at autopsies reveal a greater complexity in tumor initiation than previously appreciated. The human body contains roughly 40 trillion cells of 200 different types that are organized within a complex three-dimensional matrix, necessitating exquisite mechanisms to restrain aberrant outgrowth of malignant cells that have the capacity to kill the host. Understanding how this defense is overcome to trigger tumorigenesis and why cancer is so extraordinarily rare at the cellular level is vital to future prevention therapies. In this review, we discuss how early initiated cells are protected from further tumorigenesis and the non-mutagenic pathways by which cancer risk factors promote tumor growth. By nature, the absence of permanent genomic alterations potentially renders these tumor-promoting mechanisms clinically targetable. Finally, we consider existing strategies for early cancer interception with perspectives on the next steps for molecular cancer prevention.


Asunto(s)
Neoplasias , Humanos , Carcinogénesis , Transformación Celular Neoplásica , Genómica/métodos , Neoplasias/epidemiología , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Factores de Riesgo
20.
Cell ; 186(9): 1824-1845, 2023 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-37116469

RESUMEN

Cachexia, a systemic wasting condition, is considered a late consequence of diseases, including cancer, organ failure, or infections, and contributes to significant morbidity and mortality. The induction process and mechanistic progression of cachexia are incompletely understood. Refocusing academic efforts away from advanced cachexia to the etiology of cachexia may enable discoveries of new therapeutic approaches. Here, we review drivers, mechanisms, organismal predispositions, evidence for multi-organ interaction, model systems, clinical research, trials, and care provision from early onset to late cachexia. Evidence is emerging that distinct inflammatory, metabolic, and neuro-modulatory drivers can initiate processes that ultimately converge on advanced cachexia.


Asunto(s)
Caquexia , Humanos , Caquexia/tratamiento farmacológico , Caquexia/etiología , Caquexia/metabolismo , Caquexia/patología , Músculo Esquelético/metabolismo , Neoplasias/complicaciones , Neoplasias/metabolismo , Neoplasias/patología , Infecciones/complicaciones , Infecciones/patología , Insuficiencia Multiorgánica/complicaciones , Insuficiencia Multiorgánica/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA