Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40.964
Filter
Add more filters

Publication year range
1.
Cell ; 187(13): 3284-3302.e23, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38843832

ABSTRACT

The cleavage of zygotes generates totipotent blastomeres. In human 8-cell blastomeres, zygotic genome activation (ZGA) occurs to initiate the ontogenesis program. However, capturing and maintaining totipotency in human cells pose significant challenges. Here, we realize culturing human totipotent blastomere-like cells (hTBLCs). We find that splicing inhibition can transiently reprogram human pluripotent stem cells into ZGA-like cells (ZLCs), which subsequently transition into stable hTBLCs after long-term passaging. Distinct from reported 8-cell-like cells (8CLCs), both ZLCs and hTBLCs widely silence pluripotent genes. Interestingly, ZLCs activate a particular group of ZGA-specific genes, and hTBLCs are enriched with pre-ZGA-specific genes. During spontaneous differentiation, hTBLCs re-enter the intermediate ZLC stage and further generate epiblast (EPI)-, primitive endoderm (PrE)-, and trophectoderm (TE)-like lineages, effectively recapitulating human pre-implantation development. Possessing both embryonic and extraembryonic developmental potency, hTBLCs can autonomously generate blastocyst-like structures in vitro without external cell signaling. In summary, our study provides key criteria and insights into human cell totipotency.


Subject(s)
Cell Differentiation , Spliceosomes , Animals , Humans , Mice , Blastocyst/metabolism , Blastocyst/cytology , Blastomeres/metabolism , Blastomeres/cytology , Cellular Reprogramming , Embryonic Development/genetics , Germ Layers/metabolism , Germ Layers/cytology , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/cytology , RNA Splicing , Spliceosomes/metabolism , Totipotent Stem Cells/metabolism , Totipotent Stem Cells/cytology , Zygote/metabolism , Cells, Cultured , Models, Molecular , Protein Structure, Tertiary , Genome, Human , Single-Cell Analysis , Growth Differentiation Factor 15/chemistry , Growth Differentiation Factor 15/genetics , Growth Differentiation Factor 15/metabolism , Epigenomics , Cell Lineage
2.
Cell ; 187(3): 712-732.e38, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38194967

ABSTRACT

Human brain development involves an orchestrated, massive neural progenitor expansion while a multi-cellular tissue architecture is established. Continuously expanding organoids can be grown directly from multiple somatic tissues, yet to date, brain organoids can solely be established from pluripotent stem cells. Here, we show that healthy human fetal brain in vitro self-organizes into organoids (FeBOs), phenocopying aspects of in vivo cellular heterogeneity and complex organization. FeBOs can be expanded over long time periods. FeBO growth requires maintenance of tissue integrity, which ensures production of a tissue-like extracellular matrix (ECM) niche, ultimately endowing FeBO expansion. FeBO lines derived from different areas of the central nervous system (CNS), including dorsal and ventral forebrain, preserve their regional identity and allow to probe aspects of positional identity. Using CRISPR-Cas9, we showcase the generation of syngeneic mutant FeBO lines for the study of brain cancer. Taken together, FeBOs constitute a complementary CNS organoid platform.


Subject(s)
Brain , Organoids , Humans , Brain/cytology , Brain/growth & development , Brain/metabolism , Central Nervous System/metabolism , Extracellular Matrix/metabolism , Pluripotent Stem Cells/metabolism , Prosencephalon/cytology , Tissue Culture Techniques , Stem Cells/metabolism , Morphogenesis
3.
Cell ; 186(10): 2092-2110.e23, 2023 05 11.
Article in English | MEDLINE | ID: mdl-37172563

ABSTRACT

The third and fourth weeks of gestation in primates are marked by several developmental milestones, including gastrulation and the formation of organ primordia. However, our understanding of this period is limited due to restricted access to in vivo embryos. To address this gap, we developed an embedded 3D culture system that allows for the extended ex utero culture of cynomolgus monkey embryos for up to 25 days post-fertilization. Morphological, histological, and single-cell RNA-sequencing analyses demonstrate that ex utero cultured monkey embryos largely recapitulated key events of in vivo development. With this platform, we were able to delineate lineage trajectories and genetic programs involved in neural induction, lateral plate mesoderm differentiation, yolk sac hematopoiesis, primitive gut, and primordial germ-cell-like cell development in monkeys. Our embedded 3D culture system provides a robust and reproducible platform for growing monkey embryos from blastocysts to early organogenesis and studying primate embryogenesis ex utero.


Subject(s)
Embryo, Mammalian , Embryonic Development , Animals , Macaca fascicularis , Blastocyst , Organogenesis , Primates
4.
Cell ; 186(10): 2078-2091.e18, 2023 05 11.
Article in English | MEDLINE | ID: mdl-37172562

ABSTRACT

Neural tube (NT) defects arise from abnormal neurulation and result in the most common birth defects worldwide. Yet, mechanisms of primate neurulation remain largely unknown due to prohibitions on human embryo research and limitations of available model systems. Here, we establish a three-dimensional (3D) prolonged in vitro culture (pIVC) system supporting cynomolgus monkey embryo development from 7 to 25 days post-fertilization. Through single-cell multi-omics analyses, we demonstrate that pIVC embryos form three germ layers, including primordial germ cells, and establish proper DNA methylation and chromatin accessibility through advanced gastrulation stages. In addition, pIVC embryo immunofluorescence confirms neural crest formation, NT closure, and neural progenitor regionalization. Finally, we demonstrate that the transcriptional profiles and morphogenetics of pIVC embryos resemble key features of similarly staged in vivo cynomolgus and human embryos. This work therefore describes a system to study non-human primate embryogenesis through advanced gastrulation and early neurulation.


Subject(s)
Neural Tube Defects , Neurulation , Tissue Culture Techniques , Animals , Humans , Blastocyst , Embryo, Mammalian , Embryonic Development , Macaca fascicularis , Neural Tube Defects/genetics , Neural Tube Defects/pathology , Tissue Culture Techniques/methods
5.
Cell ; 184(5): 1156-1170.e14, 2021 03 04.
Article in English | MEDLINE | ID: mdl-33539781

ABSTRACT

Cultivated rice varieties are all diploid, and polyploidization of rice has long been desired because of its advantages in genome buffering, vigorousness, and environmental robustness. However, a workable route remains elusive. Here, we describe a practical strategy, namely de novo domestication of wild allotetraploid rice. By screening allotetraploid wild rice inventory, we identified one genotype of Oryza alta (CCDD), polyploid rice 1 (PPR1), and established two important resources for its de novo domestication: (1) an efficient tissue culture, transformation, and genome editing system and (2) a high-quality genome assembly discriminated into two subgenomes of 12 chromosomes apiece. With these resources, we show that six agronomically important traits could be rapidly improved by editing O. alta homologs of the genes controlling these traits in diploid rice. Our results demonstrate the possibility that de novo domesticated allotetraploid rice can be developed into a new staple cereal to strengthen world food security.


Subject(s)
Crops, Agricultural/genetics , Domestication , Oryza/genetics , CRISPR-Cas Systems , Food Security , Gene Editing , Genetic Variation , Genome, Plant , Oryza/classification , Polyploidy
6.
Cell ; 183(5): 1420-1435.e21, 2020 11 25.
Article in English | MEDLINE | ID: mdl-33159857

ABSTRACT

Gastroenteropancreatic (GEP) neuroendocrine neoplasm (NEN) that consists of neuroendocrine tumor and neuroendocrine carcinoma (NEC) is a lethal but under-investigated disease owing to its rarity. To fill the scarcity of clinically relevant models of GEP-NEN, we here established 25 lines of NEN organoids and performed their comprehensive molecular characterization. GEP-NEN organoids recapitulated pathohistological and functional phenotypes of the original tumors. Whole-genome sequencing revealed frequent genetic alterations in TP53 and RB1 in GEP-NECs, and characteristic chromosome-wide loss of heterozygosity in GEP-NENs. Transcriptome analysis identified molecular subtypes that are distinguished by the expression of distinct transcription factors. GEP-NEN organoids gained independence from the stem cell niche irrespective of genetic mutations. Compound knockout of TP53 and RB1, together with overexpression of key transcription factors, conferred on the normal colonic epithelium phenotypes that are compatible with GEP-NEN biology. Altogether, our study not only provides genetic understanding of GEP-NEN, but also connects its genetics and biological phenotypes.


Subject(s)
Biological Specimen Banks , Neuroendocrine Tumors/pathology , Organoids/pathology , Animals , Chromosomes, Human/genetics , Genotype , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Intestinal Neoplasms/genetics , Intestinal Neoplasms/pathology , Male , Mice , Models, Genetic , Mutation/genetics , Neuroendocrine Tumors/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Phenotype , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Transcriptome/genetics , Whole Genome Sequencing
7.
Cell ; 175(6): 1607-1619.e15, 2018 11 29.
Article in English | MEDLINE | ID: mdl-30500539

ABSTRACT

In the healthy adult liver, most hepatocytes proliferate minimally. However, upon physical or chemical injury to the liver, hepatocytes proliferate extensively in vivo under the direction of multiple extracellular cues, including Wnt and pro-inflammatory signals. Currently, liver organoids can be generated readily in vitro from bile-duct epithelial cells, but not hepatocytes. Here, we show that TNFα, an injury-induced inflammatory cytokine, promotes the expansion of hepatocytes in 3D culture and enables serial passaging and long-term culture for more than 6 months. Single-cell RNA sequencing reveals broad expression of hepatocyte markers. Strikingly, in vitro-expanded hepatocytes engrafted, and significantly repopulated, the injured livers of Fah-/- mice. We anticipate that tissue repair signals can be harnessed to promote the expansion of otherwise hard-to-culture cell-types, with broad implications.


Subject(s)
Antigens, Differentiation/biosynthesis , Cell Culture Techniques , Cell Proliferation/drug effects , Hepatocytes/metabolism , Spheroids, Cellular/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Animals , Cell Line, Transformed , Hep G2 Cells , Hepatocytes/transplantation , Human Umbilical Vein Endothelial Cells , Humans , Liver/injuries , Liver/metabolism , Mice, Knockout , Spheroids, Cellular/transplantation , Time Factors
8.
Cell ; 172(3): 578-589.e17, 2018 01 25.
Article in English | MEDLINE | ID: mdl-29373830

ABSTRACT

KRASG12C was recently identified to be potentially druggable by allele-specific covalent targeting of Cys-12 in vicinity to an inducible allosteric switch II pocket (S-IIP). Success of this approach requires active cycling of KRASG12C between its active-GTP and inactive-GDP conformations as accessibility of the S-IIP is restricted only to the GDP-bound state. This strategy proved feasible for inhibiting mutant KRAS in vitro; however, it is uncertain whether this approach would translate to in vivo. Here, we describe structure-based design and identification of ARS-1620, a covalent compound with high potency and selectivity for KRASG12C. ARS-1620 achieves rapid and sustained in vivo target occupancy to induce tumor regression. We use ARS-1620 to dissect oncogenic KRAS dependency and demonstrate that monolayer culture formats significantly underestimate KRAS dependency in vivo. This study provides in vivo evidence that mutant KRAS can be selectively targeted and reveals ARS-1620 as representing a new generation of KRASG12C-specific inhibitors with promising therapeutic potential.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms, Experimental/drug therapy , Piperazines/pharmacology , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Quinazolines/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Cell Proliferation/drug effects , Cells, Cultured , Female , HCT116 Cells , HEK293 Cells , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Docking Simulation , Mutation , Piperazines/chemistry , Piperazines/therapeutic use , Protein Binding , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Quinazolines/chemistry , Quinazolines/therapeutic use
9.
Cell ; 169(2): 258-272.e17, 2017 Apr 06.
Article in English | MEDLINE | ID: mdl-28388410

ABSTRACT

A complex interplay of environmental factors impacts the metabolism of human cells, but neither traditional culture media nor mouse plasma mimic the metabolite composition of human plasma. Here, we developed a culture medium with polar metabolite concentrations comparable to those of human plasma (human plasma-like medium [HPLM]). Culture in HPLM, relative to that in traditional media, had widespread effects on cellular metabolism, including on the metabolome, redox state, and glucose utilization. Among the most prominent was an inhibition of de novo pyrimidine synthesis-an effect traced to uric acid, which is 10-fold higher in the blood of humans than of mice and other non-primates. We find that uric acid directly inhibits uridine monophosphate synthase (UMPS) and consequently reduces the sensitivity of cancer cells to the chemotherapeutic agent 5-fluorouracil. Thus, media that better recapitulates the composition of human plasma reveals unforeseen metabolic wiring and regulation, suggesting that HPLM should be of broad utility.


Subject(s)
Culture Media/chemistry , Multienzyme Complexes/antagonists & inhibitors , Orotate Phosphoribosyltransferase/antagonists & inhibitors , Orotidine-5'-Phosphate Decarboxylase/antagonists & inhibitors , Uric Acid/metabolism , Aged , Animals , Cell Culture Techniques , Cell Line, Tumor , Fluorouracil/pharmacology , Glucose/metabolism , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Male , Mice , Middle Aged , Multienzyme Complexes/chemistry , Orotate Phosphoribosyltransferase/chemistry , Orotidine-5'-Phosphate Decarboxylase/chemistry , Protein Domains , Pyrimidines/biosynthesis
10.
Physiol Rev ; 103(4): 2623-2677, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37171807

ABSTRACT

Mammalian eggs (oocytes) are formed during fetal life and establish associations with somatic cells to form primordial follicles that create a store of germ cells (the primordial pool). The size of this pool is influenced by key events during the formation of germ cells and by factors that influence the subsequent activation of follicle growth. These regulatory pathways must ensure that the reserve of oocytes within primordial follicles in humans lasts for up to 50 years, yet only approximately 0.1% will ever be ovulated with the rest undergoing degeneration. This review outlines the mechanisms and regulatory pathways that govern the processes of oocyte and follicle formation and later growth, within the ovarian stroma, through to ovulation with particular reference to human oocytes/follicles. In addition, the effects of aging on female reproductive capacity through changes in oocyte number and quality are emphasized, with both the cellular mechanisms and clinical implications discussed. Finally, the details of current developments in culture systems that support all stages of follicle growth to generate mature oocytes in vitro and emerging prospects for making new oocytes from stem cells are outlined.


Subject(s)
Oocytes , Ovarian Follicle , Animals , Humans , Female , Oocytes/physiology , Ovarian Follicle/metabolism , Ovary/metabolism , Oogenesis/physiology , Mammals/physiology , Aging
11.
Annu Rev Genet ; 55: 661-681, 2021 11 23.
Article in English | MEDLINE | ID: mdl-34546796

ABSTRACT

Plants exhibit remarkable lineage plasticity, allowing them to regenerate organs that differ from their respective origins. Such developmental plasticity is dependent on the activity of pluripotent founder cells or stem cells residing in meristems. At the shoot apical meristem (SAM), the constant flow of cells requires continuing cell specification governed by a complex genetic network, with the WUSCHEL transcription factor and phytohormone cytokinin at its core. In this review, I discuss some intriguing recent discoveries that expose new principles and mechanisms of patterning and cell specification acting both at the SAM and prior to meristem organogenesis during shoot regeneration. I also highlight unanswered questions and future challenges in the study of SAM and meristem regeneration. Finally, I put forward a model describing stochastic events mediated by epigenetic factors to explain how the gene regulatory network might be initiated at the onset of shoot regeneration.


Subject(s)
Arabidopsis Proteins , Meristem , Arabidopsis Proteins/genetics , Gene Expression Regulation, Plant/genetics , Gene Regulatory Networks , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Meristem/genetics , Meristem/metabolism , Plant Shoots/genetics , Plant Shoots/metabolism , Regeneration/genetics
12.
EMBO J ; 43(11): 2127-2165, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38580776

ABSTRACT

The in vitro oxygen microenvironment profoundly affects the capacity of cell cultures to model physiological and pathophysiological states. Cell culture is often considered to be hyperoxic, but pericellular oxygen levels, which are affected by oxygen diffusivity and consumption, are rarely reported. Here, we provide evidence that several cell types in culture actually experience local hypoxia, with important implications for cell metabolism and function. We focused initially on adipocytes, as adipose tissue hypoxia is frequently observed in obesity and precedes diminished adipocyte function. Under standard conditions, cultured adipocytes are highly glycolytic and exhibit a transcriptional profile indicative of physiological hypoxia. Increasing pericellular oxygen diverted glucose flux toward mitochondria, lowered HIF1α activity, and resulted in widespread transcriptional rewiring. Functionally, adipocytes increased adipokine secretion and sensitivity to insulin and lipolytic stimuli, recapitulating a healthier adipocyte model. The functional benefits of increasing pericellular oxygen were also observed in macrophages, hPSC-derived hepatocytes and cardiac organoids. Our findings demonstrate that oxygen is limiting in many terminally-differentiated cell types, and that considering pericellular oxygen improves the quality, reproducibility and translatability of culture models.


Subject(s)
Adipocytes , Cell Differentiation , Oxygen , Oxygen/metabolism , Adipocytes/metabolism , Adipocytes/cytology , Humans , Cell Culture Techniques/methods , Animals , Glycolysis , Hepatocytes/metabolism , Cell Hypoxia , Mitochondria/metabolism , Mice , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Cells, Cultured , Glucose/metabolism , Macrophages/metabolism
13.
Mol Cell ; 80(5): 828-844.e6, 2020 12 03.
Article in English | MEDLINE | ID: mdl-33128871

ABSTRACT

Cancer-associated mutations that stabilize NRF2, an oxidant defense transcription factor, are predicted to promote tumor development. Here, utilizing 3D cancer spheroid models coupled with CRISPR-Cas9 screens, we investigate the molecular pathogenesis mediated by NRF2 hyperactivation. NRF2 hyperactivation was necessary for proliferation and survival in lung tumor spheroids. Antioxidant treatment rescued survival but not proliferation, suggesting the presence of distinct mechanisms. CRISPR screens revealed that spheroids are differentially dependent on the mammalian target of rapamycin (mTOR) for proliferation and the lipid peroxidase GPX4 for protection from ferroptosis of inner, matrix-deprived cells. Ferroptosis inhibitors blocked death from NRF2 downregulation, demonstrating a critical role of NRF2 in protecting matrix-deprived cells from ferroptosis. Interestingly, proteomics analyses show global enrichment of selenoproteins, including GPX4, by NRF2 downregulation, and targeting NRF2 and GPX4 killed spheroids overall. These results illustrate the value of spheroid culture in revealing environmental or spatial differential dependencies on NRF2 and reveal exploitable vulnerabilities of NRF2-hyperactivated tumors.


Subject(s)
CRISPR-Cas Systems , Cell Culture Techniques , Cell Proliferation , Ferroptosis , NF-E2-Related Factor 2/metabolism , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Spheroids, Cellular/metabolism , A549 Cells , Humans , NF-E2-Related Factor 2/genetics , Neoplasm Proteins/genetics , Neoplasms/genetics , Neoplasms/pathology , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Spheroids, Cellular/pathology , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
14.
Trends Biochem Sci ; 48(2): 103-105, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36114088

ABSTRACT

The metabolism plays a fundamental role in cellular signaling pathways, but commonly used cell culture media do not reflect physiological metabolite concentrations. The metabolic control hub mammalian target of rapamycin complex 1 (mTORC1) kinase is an illuminating example that it is about time to advance our cell culture to become more physiological and relevant.


Subject(s)
Signal Transduction , TOR Serine-Threonine Kinases , TOR Serine-Threonine Kinases/metabolism , Signal Transduction/physiology , Multiprotein Complexes/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Cell Culture Techniques
15.
EMBO J ; 42(23): e113955, 2023 Dec 01.
Article in English | MEDLINE | ID: mdl-37850882

ABSTRACT

Recent studies have reported the differentiation of pluripotent cells into oocytes in vitro. However, the developmental competence of in vitro-generated oocytes remains low. Here, we perform a comprehensive comparison of mouse germ cell development in vitro over all culture steps versus in vivo with the goal to understand mechanisms underlying poor oocyte quality. We show that the in vitro differentiation of primordial germ cells to growing oocytes and subsequent follicle growth is critical for competence for preimplantation development. Systematic transcriptome analysis of single oocytes that were subjected to different culture steps identifies genes that are normally upregulated during oocyte growth to be susceptible for misregulation during in vitro oogenesis. Many misregulated genes are Polycomb targets. Deregulation of Polycomb repression is therefore a key cause and the earliest defect known in in vitro oocyte differentiation. Conversely, structurally normal in vitro-derived oocytes fail at zygotic genome activation and show abnormal acquisition of 5-hydroxymethylcytosine on maternal chromosomes. Our data identify epigenetic regulation at an early stage of oogenesis limiting developmental competence and suggest opportunities for future improvements.


Subject(s)
Epigenesis, Genetic , Oocytes , Female , Animals , Mice , Ovarian Follicle , Oogenesis/genetics , Germ Cells
16.
Trends Immunol ; 45(1): 32-47, 2024 01.
Article in English | MEDLINE | ID: mdl-38135599

ABSTRACT

The human immune system is a complex network of coordinated components that are crucial for health and disease. Animal models, commonly used to study immunomodulatory agents, are limited by species-specific differences, low throughput, and ethical concerns. In contrast, in vitro modeling of human immune responses can enable species- and population-specific mechanistic studies and translational development within the same study participant. Translational accuracy of in vitro models is enhanced by accounting for genetic, epigenetic, and demographic features such as age, sex, and comorbidity. This review explores various human in vitro immune models, considers evidence that they may resemble human in vivo responses, and assesses their potential to accelerate and de-risk vaccine discovery and development.


Subject(s)
Vaccination , Vaccines , Animals , Humans , Immunity
17.
Proc Natl Acad Sci U S A ; 121(25): e2322872121, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38857405

ABSTRACT

Despite an abundance of support for culturally inclusive learning environments, there is little consensus regarding how to change educational contexts to effectively and sustainably foster cultural inclusion. To address this gap, we report findings from a research-practice partnership that leveraged the Culture Cycle Framework (CCF) to expand educators' praxis to include both independent and interdependent models of self. Most U.S. schools validate independent cultural models (i.e., those that prioritize individuality, uniqueness, and personal agency) and overlook interdependent models (i.e., those that prioritize connectedness, relationality, and collective well-being), which are more common among students from marginalized racial and socioeconomic backgrounds. Using a quasi-experimental longitudinal design, we trained school leadership to integrate ideas about cultural inclusion (i.e., validating the importance of both independent and interdependent cultural models) into school-wide flagship practices. We assessed downstream indicators of culture change by surveying teachers and students across the district and found that a) leadership-level training enhanced school-wide beliefs about cultural inclusion, b) teachers' endorsement of culturally inclusive beliefs predicted their use of culturally inclusive practices, and c) teachers' use of culturally inclusive practices predicted enhanced psychosocial and academic outcomes among students. This research represents a comprehensive culture change effort using the CCF and illustrates a means of fostering inclusion-focused educational culture change and assessing downstream consequences of culture change initiatives.


Subject(s)
Leadership , Humans , Schools , School Teachers/psychology , Female , Male , Students/psychology , Cultural Diversity , Culture
18.
Proc Natl Acad Sci U S A ; 121(26): e2319175121, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38885385

ABSTRACT

Cumulative culture, the accumulation of modifications, innovations, and improvements over generations through social learning, is a key determinant of the behavioral diversity across Homo sapiens populations and their ability to adapt to varied ecological habitats. Generations of improvements, modifications, and lucky errors allow humans to use technologies and know-how well beyond what a single naive individual could invent independently within their lifetime. The human dependence on cumulative culture may have shaped the evolution of biological and behavioral traits in the hominin lineage, including brain size, body size, life history, sociality, subsistence, and ecological niche expansion. Yet, we do not know when, in the human career, our ancestors began to depend on cumulative culture. Here, we show that hominins likely relied on a derived form of cumulative culture by at least ~600 kya, a result in line with a growing body of existing evidence. We analyzed the complexity of stone tool manufacturing sequences over the last 3.3 My of the archaeological record. We then compare these to the achievable complexity without cumulative culture, which we estimate using nonhuman primate technologies and stone tool manufacturing experiments. We find that archaeological technologies become significantly more complex than expected in the absence of cumulative culture only after ~600 kya.


Subject(s)
Archaeology , Hominidae , Animals , Humans , Cultural Evolution , Tool Use Behavior , Biological Evolution , Fossils , Technology , History, Ancient
19.
Proc Natl Acad Sci U S A ; 121(11): e2316500121, 2024 Mar 12.
Article in English | MEDLINE | ID: mdl-38442157

ABSTRACT

Evaluating the ability of cytotoxic T lymphocytes (CTLs) to eliminate tumor cells is crucial, for instance, to predict the efficiency of cell therapy in personalized medicine. However, the destruction of a tumor by CTLs involves CTL migration in the extra-tumoral environment, accumulation on the tumor, antigen recognition, and cooperation in killing the cancer cells. Therefore, identifying the limiting steps in this complex process requires spatio-temporal measurements of different cellular events over long periods. Here, we use a cancer-on-a-chip platform to evaluate the impact of adenomatous polyposis coli (APC) mutation on CTL migration and cytotoxicity against 3D tumor spheroids. The APC mutated CTLs are found to have a reduced ability to destroy tumor spheroids compared with control cells, even though APC mutants migrate in the extra-tumoral space and accumulate on the spheroids as efficiently as control cells. Once in contact with the tumor however, mutated CTLs display reduced engagement with the cancer cells, as measured by a metric that distinguishes different modes of CTL migration. Realigning the CTL trajectories around localized killing cascades reveals that all CTLs transition to high engagement in the 2 h preceding the cascades, which confirms that the low engagement is the cause of reduced cytotoxicity. Beyond the study of APC mutations, this platform offers a robust way to compare cytotoxic cell efficiency of even closely related cell types, by relying on a multiscale cytometry approach to disentangle complex interactions and to identify the steps that limit the tumor destruction.


Subject(s)
Adenomatous Polyposis Coli , Neoplasms , Humans , Neoplasms/genetics , T-Lymphocytes, Cytotoxic , Mutation , Lab-On-A-Chip Devices
20.
EMBO J ; 41(18): e110815, 2022 09 15.
Article in English | MEDLINE | ID: mdl-35912849

ABSTRACT

In vitro oogenesis is key to elucidating the mechanism of human female germ-cell development and its anomalies. Accordingly, pluripotent stem cells have been induced into primordial germ cell-like cells and into oogonia with epigenetic reprogramming, yet further reconstitutions remain a challenge. Here, we demonstrate ex vivo reconstitution of fetal oocyte development in both humans and cynomolgus monkeys (Macaca fascicularis). With an optimized culture of fetal ovary reaggregates over three months, human and monkey oogonia enter and complete the first meiotic prophase to differentiate into diplotene oocytes that form primordial follicles, the source for oogenesis in adults. The cytological and transcriptomic progressions of fetal oocyte development in vitro closely recapitulate those in vivo. A comparison of single-cell transcriptomes among humans, monkeys, and mice unravels primate-specific and conserved programs driving fetal oocyte development, the former including a distinct transcriptomic transformation upon oogonia-to-oocyte transition and the latter including two active X chromosomes with little X-chromosome upregulation. Our study provides a critical step forward for realizing human in vitro oogenesis and uncovers salient characteristics of fetal oocyte development in primates.


Subject(s)
Meiosis , Oogenesis , Animals , Female , Humans , Macaca fascicularis , Mice , Oocytes , Oogenesis/physiology , Ovary
SELECTION OF CITATIONS
SEARCH DETAIL