Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
2.
Nat Commun ; 14(1): 5922, 2023 09 22.
Article in English | MEDLINE | ID: mdl-37739965

ABSTRACT

Alzheimer's disease (AD) is characterized by toxic protein accumulation in the brain. Ubiquitination is essential for protein clearance in cells, making altered ubiquitin signaling crucial in AD development. A defective variant, ubiquitin B + 1 (UBB+1), created by a non-hereditary RNA frameshift mutation, is found in all AD patient brains post-mortem. We now detect UBB+1 in human brains during early AD stages. Our study employs a 3D neural culture platform derived from human neural progenitors, demonstrating that UBB+1 alone induces extracellular amyloid-ß (Aß) deposits and insoluble hyperphosphorylated tau aggregates. UBB+1 competes with ubiquitin for binding to the deubiquitinating enzyme UCHL1, leading to elevated levels of amyloid precursor protein (APP), secreted Aß peptides, and Aß build-up. Crucially, silencing UBB+1 expression impedes the emergence of AD hallmarks in this model system. Our findings highlight the significance of ubiquitin signalling as a variable contributing to AD pathology and present a nonclinical platform for testing potential therapeutics.


Subject(s)
Alzheimer Disease , Humans , Alzheimer Disease/genetics , Signal Transduction , Amyloid beta-Peptides , Amyloid beta-Protein Precursor/genetics , Cell Culture Techniques, Three Dimensional
3.
Nat Commun ; 13(1): 4628, 2022 08 08.
Article in English | MEDLINE | ID: mdl-35941116

ABSTRACT

The presence of distinct stem cells that maintain the interfollicular epidermis is highly debated. Here, we report a population of keratinocytes, marked by Thy1, in the basal layer of the interfollicular epidermis. We find that epidermal cells expressing differential levels of Thy1 display distinct transcriptional signatures. Thy1+ keratinocytes do not express T cell markers, express a unique transcriptional profile, cycle significantly slower than basal epidermal progenitors and display significant expansion potential in vitro. Multicolor lineage tracing analyses and mathematical modeling reveal that Thy1+ basal keratinocytes do not compete neutrally alike interfollicular progenitors and contribute long-term to both epidermal replenishment and wound repair. Importantly, ablation of Thy1+ cells strongly impairs these processes, thus indicating the non-redundant function of Thy1+ stem cells in the epidermis. Collectively, these results reveal a distinct stem cell population that plays a critical role in epidermal homeostasis and repair.


Subject(s)
Epidermal Cells , Stem Cells , Animals , Cell Differentiation/physiology , Epidermis/metabolism , Keratinocytes/metabolism , Mice , Stem Cells/metabolism
4.
Nat Cell Biol ; 24(7): 1049-1063, 2022 07.
Article in English | MEDLINE | ID: mdl-35798842

ABSTRACT

Anchored cells of the basal epidermis constantly undergo proliferation in an overcrowded environment. An important regulator of epidermal proliferation is YAP, which can be controlled by both cell-matrix and cell-cell interactions. Here, we report that THY1, a GPI-anchored protein, inhibits epidermal YAP activity through converging molecular mechanisms. THY1 deficiency leads to increased adhesion by activating the integrin-ß1-SRC module. Notably, regardless of high cellular densities, the absence of THY1 leads to the dissociation of an adherens junction complex that enables the release and translocation of YAP. Due to increased YAP-dependent proliferation, Thy1-/- mice display enhanced wound repair and hair follicle regeneration. Taken together, our work reveals THY1 as a crucial regulator of cell-matrix and cell-cell interactions that controls YAP activity in skin homeostasis and regeneration.


Subject(s)
Adaptor Proteins, Signal Transducing , Cell Cycle Proteins , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Proliferation , Epidermis/metabolism , Homeostasis , Mice , Skin/metabolism
5.
Dev Cell ; 57(12): 1442-1452, 2022 06 20.
Article in English | MEDLINE | ID: mdl-35700729

ABSTRACT

In various placental mammals, the bidirectional exchange of cells during pregnancy can lead to the acquisition of genetically unique cells that can persist in both mother and child for decades. Over the years, it has become increasingly clear that this phenomenon, termed fetomaternal microchimerism may play key roles in a number of biological processes. In this perspective, we explore the concept of fetomaternal microchimerism and outline how fetal microchimeric cells are detected and immunologically tolerated within the maternal setting. Moreover, we discuss undertakings in the field that hint at the significant plasticity of fetal microchimeric cells and their potential roles in promoting maternal wound healing. Finally, we explore the multifaceted roles of fetal microchimeric cells in cancer development and progression. A deeper understanding of fetomaternal chimerism in healthy and diseased states will be key toward developing more efficient anti-cancer treatments and regenerative therapies.


Subject(s)
Chimerism , Neoplasms , Animals , Child , Female , Fetus , Humans , Mammals , Maternal-Fetal Exchange , Neoplasms/genetics , Placenta , Pregnancy , Wound Healing
6.
Nat Commun ; 12(1): 6572, 2021 11 12.
Article in English | MEDLINE | ID: mdl-34772930

ABSTRACT

Damaged or superfluous cells are typically eliminated by apoptosis. Although apoptosis is a cell-autonomous process, apoptotic cells communicate with their environment in different ways. Here we describe a mechanism whereby cells under apoptotic stress can promote survival of neighbouring cells. We find that upon apoptotic stress, cells release the growth factor FGF2, leading to MEK-ERK-dependent transcriptional upregulation of pro-survival BCL-2 proteins in a non-cell autonomous manner. This transient upregulation of pro-survival BCL-2 proteins protects neighbouring cells from apoptosis. Accordingly, we find in certain cancer types a correlation between FGF-signalling, BCL-2 expression and worse prognosis. In vivo, upregulation of MCL-1 occurs in an FGF-dependent manner during skin repair, which regulates healing dynamics. Importantly, either co-treatment with FGF-receptor inhibitors or removal of apoptotic stress restores apoptotic sensitivity to cytotoxic therapy and delays wound healing. These data reveal a pathway by which cells under apoptotic stress can increase resistance to cell death in surrounding cells. Beyond mediating cytotoxic drug resistance, this process also provides a potential link between tissue damage and repair.


Subject(s)
Apoptosis/drug effects , Cell Death/drug effects , Signal Transduction/drug effects , Animals , Fibroblast Growth Factor 2/metabolism , Gene Expression Regulation, Neoplastic , HEK293 Cells , HeLa Cells , Humans , Mice , Mice, Inbred C57BL , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Up-Regulation/drug effects , Wound Healing
7.
Dev Cell ; 56(13): 1900-1916.e5, 2021 07 12.
Article in English | MEDLINE | ID: mdl-34197726

ABSTRACT

Stem cells (SCs) play a key role in homeostasis and repair. While many studies have focused on SC self-renewal and differentiation, little is known regarding the molecular mechanism regulating SC elimination and compensation upon loss. Here, we report that Caspase-9 deletion in hair follicle SCs (HFSCs) attenuates the apoptotic cascade, resulting in significant temporal delays. Surprisingly, Casp9-deficient HFSCs accumulate high levels of cleaved caspase-3 and are improperly cleared due to an essential caspase-3/caspase-9 feedforward loop. These SCs are retained in an apoptotic-engaged state, serving as mitogenic signaling centers by continuously releasing Wnt3 and instructing proliferation. Investigating the underlying mechanism, we reveal a caspase-3/Dusp8/p38 module responsible for Wnt3 induction, which operates in both normal and Casp9-deleted HFSCs. Notably, Casp9-deleted mice display accelerated wound repair and de novo hair follicle regeneration. Taken together, we demonstrate that apoptotic cells represent a dynamic SC niche, from which emanating signals drive SC proliferation and tissue regeneration.


Subject(s)
Caspase 3/genetics , Caspase 9/genetics , Dual-Specificity Phosphatases/genetics , Regeneration/genetics , Wnt3 Protein/genetics , Animals , Apoptosis/genetics , Cell Differentiation/genetics , Cell Proliferation/genetics , Cell Self Renewal/genetics , Hair Follicle/growth & development , Hair Follicle/metabolism , MAP Kinase Signaling System/genetics , Mice , Stem Cell Niche/genetics , Stem Cells/metabolism , Wound Healing/genetics
8.
Cancer Res ; 81(8): 2116-2127, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33574090

ABSTRACT

Targeting cancer stem cells (CSC) can serve as an effective approach toward limiting resistance to therapies. While basal-like (triple-negative) breast cancers encompass cells with CSC features, rational therapies remain poorly established. We show here that the receptor tyrosine kinase Met promotes YAP activity in basal-like breast cancer and find enhanced YAP activity within the CSC population. Interfering with YAP activity delayed basal-like cancer formation, prevented luminal to basal transdifferentiation, and reduced CSC. YAP knockout mammary glands revealed a decrease in ß-catenin target genes, suggesting that YAP is required for nuclear ß-catenin activity. Mechanistically, nuclear YAP interacted with ß-catenin and TEAD4 at gene regulatory elements. Proteomic patient data revealed an upregulation of the YAP signature in basal-like breast cancers. Our findings demonstrate that in basal-like breast cancers, ß-catenin activity is dependent on YAP signaling and controls the CSC program. These findings suggest that targeting the YAP/TEAD4/ß-catenin complex offers a potential therapeutic strategy for eradicating CSCs in basal-like breast cancers. SIGNIFICANCE: These findings show that YAP cooperates with ß-catenin in basal-like breast cancer to regulate CSCs and that targeting this interaction may be a novel CSC therapy for patients with basal-like breast cancer. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/8/2116/F1.large.jpg.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Neoplastic Stem Cells/metabolism , Proto-Oncogene Proteins c-met/metabolism , Triple Negative Breast Neoplasms/metabolism , beta Catenin/metabolism , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/genetics , Animals , Carcinogenesis , Cell Line, Tumor , Cell Transdifferentiation , DNA-Binding Proteins/metabolism , Female , Gene Expression Regulation, Neoplastic , Heterografts , Humans , Mammary Glands, Animal/cytology , Mammary Glands, Animal/metabolism , Mice , Muscle Proteins/metabolism , Neoplastic Stem Cells/pathology , Proteomics , TEA Domain Transcription Factors , Transcription Factors/metabolism , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/prevention & control , Triple Negative Breast Neoplasms/therapy , Up-Regulation , Wnt Proteins/metabolism , YAP-Signaling Proteins , beta Catenin/genetics
9.
Cancer Discov ; 11(2): 245-265, 2021 02.
Article in English | MEDLINE | ID: mdl-33462123

ABSTRACT

Cell suicide pathways, termed regulated cell death (RCD), play a critical role in organismal development, homeostasis, and pathogenesis. Here, we provide an overview of key RCD modalities, namely apoptosis, entosis, necroptosis, pyroptosis, and ferroptosis. We explore how various RCD modules serve as a defense mechanism against the emergence of cancer as well as the manner in which they can be exploited to drive oncogenesis. Furthermore, we outline current therapeutic agents that activate RCD and consider novel RCD-based strategies for tumor elimination. SIGNIFICANCE: A variety of antitumor therapeutics eliminate cancer cells by harnessing the devastating potential of cellular suicide pathways, emphasizing the critical importance of RCD in battling cancer. This review supplies a mechanistic perspective of distinct RCD modalities and explores the important role they play in tumorigenesis. We discuss how RCD modules serve as a double-edged sword as well as novel approaches aimed at selectively manipulating RCD for tumor eradication.


Subject(s)
Neoplasms/pathology , Regulated Cell Death , Humans
10.
Nat Commun ; 10(1): 2348, 2019 05 28.
Article in English | MEDLINE | ID: mdl-31138796

ABSTRACT

Most studies on the skin focus primarily on the hair follicle and interfollicular epidermis, whereas little is known regarding the homeostasis of the sebaceous gland (SG). The SG has been proposed to be replenished by different pools of hair follicle stem cells and cells that resides in the SG base, marked by Blimp1. Here, we demonstrate that single Blimp1+ cells isolated from mice have the potential to generate SG organoids in vitro. Mimicking SG homeostasis, the outer layer of these organoids is composed of proliferating cells that migrate inward, undergo terminal differentiation and generating lipid-filled sebocytes. Performing confocal microscopy and mass-spectrometry, we report that these organoids exhibit known markers and a lipidomic profile similar to SGs in vivo. Furthermore, we identify a role for c-Myc in sebocyte proliferation and differentiation, and determine that SG organoids can serve as a platform for studying initial stages of acne vulgaris, making this a useful platform to identify potential therapeutic targets.


Subject(s)
Cell Differentiation , Cell Proliferation , Organoids/metabolism , Positive Regulatory Domain I-Binding Factor 1/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Sebaceous Glands/metabolism , Animals , Epidermis/metabolism , Epidermis/ultrastructure , In Vitro Techniques , Lipid Metabolism , Mass Spectrometry , Mice , Microscopy, Confocal , Organoids/ultrastructure , Sebaceous Glands/ultrastructure , Stem Cells/metabolism , Tissue Culture Techniques
11.
J Cell Death ; 12: 1179066019836967, 2019.
Article in English | MEDLINE | ID: mdl-30983862

ABSTRACT

Although much is known regarding intestinal stem cell (ISC) self-renewal and differentiation, the specific mechanisms used for their elimination is unclear. We recently discovered that the pro-apoptotic protein ARTS, a Septin4 isoform, interacts with X-linked inhibitor of apoptosis (XIAP) in the ISC niche to regulate stem cell survival during intestinal homeostasis and regeneration. These findings point to an intriguing avenue of translational research, examining how manipulation of stem cell apoptosis through the ARTS/XIAP module can affect stem-cell-dependent processes.

12.
Nat Commun ; 9(1): 4582, 2018 11 02.
Article in English | MEDLINE | ID: mdl-30389919

ABSTRACT

Stem cells (SCs) play a pivotal role in fueling homeostasis and regeneration. While much focus has been given to self-renewal and differentiation pathways regulating SC fate, little is known regarding the specific mechanisms utilized for their elimination. Here, we report that the pro-apoptotic protein ARTS (a Septin4 isoform) is highly expressed in cells comprising the intestinal SC niche and that its deletion protects Lgr5+ and Paneth cells from undergoing apoptotic cell death. As a result, the Sept4/ARTS-/- crypt displays augmented proliferation and, in culture, generates massive cystic-like organoids due to enhanced Wnt/ß-catenin signaling. Importantly, Sept4/ARTS-/- mice exhibit resistance against intestinal damage in a manner dependent upon Lgr5+ SCs. Finally, we show that ARTS interacts with XIAP in intestinal crypt cells and that deletion of XIAP can abrogate Sept4/ARTS-/--dependent phenotypes. Our results indicate that intestinal SCs utilize specific apoptotic proteins for their elimination, representing a unique target for regenerative medicine.


Subject(s)
Apoptosis , Intestines/cytology , Regeneration , Septins/metabolism , Stem Cell Niche , Animals , Cell Proliferation , Cytoprotection , Gene Deletion , Mice, Inbred C57BL , Wnt Signaling Pathway , Wounds and Injuries/pathology , X-Linked Inhibitor of Apoptosis Protein/metabolism
13.
Mol Cell ; 70(4): 573-587.e4, 2018 05 17.
Article in English | MEDLINE | ID: mdl-29775577

ABSTRACT

Apoptosis culminates in the activation of caspase-3, which plays an important role in implementing the cell death program. Here, we reveal a non-apoptotic role of caspase-3 as a key regulator of cell proliferation and organ size. Caspase-3 is specifically activated in the proliferating cells of the sebaceous gland, but does not instruct cell elimination. Deletion or chemical inhibition of caspase-3 diminishes cell proliferation, decreases cell number and reduces sebaceous gland size in vivo. Exploring the underlying mechanism, we demonstrate that α-catenin is cleaved by caspase-3, thus facilitating the activation and nuclear translocation of yes-associated protein (YAP), a vital regulator of organ size. Accordingly, activation of caspase-3 leads to YAP-dependent organ size augmentation. Finally, we show that X-linked inhibitor of apoptosis protein (XIAP) serves as an endogenous feedback antagonist for the caspase-3/YAP signaling module. Taken together, we report here a molecular mechanism wherein the apoptotic machinery is refocused to regulate cell proliferation and orchestrate organ size.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Caspase 3/physiology , Cell Proliferation , Feedback, Physiological , Inhibitor of Apoptosis Proteins/physiology , Phosphoproteins/metabolism , RNA Splicing Factors/physiology , alpha Catenin/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Apoptosis , Cell Cycle Proteins , Female , Male , Mice , Mice, Knockout , Organ Size , Phosphoproteins/genetics , Protein Transport , YAP-Signaling Proteins , alpha Catenin/genetics
14.
Drug Resist Updat ; 28: 1-12, 2016 09.
Article in English | MEDLINE | ID: mdl-27620951

ABSTRACT

Over the past two decades cancer stem cells (CSCs) have emerged as essential players in the pathogenesis of cancer, with the capacity to initiate, maintain and repopulate different tumors. Within the tumor bulk, CSCs represent a small subpopulation, bestowed with the capacity to self-renew and yield heterogeneous lineages of cancer cells. In many scenarios, CSCs exhibit increased resistance toward irradiation and chemotherapy, and given their spectacular ability to replenish the tumor, they constitute a substantial therapeutic challenge. In this review, we provide a brief overview of the concept of CSCs and the experimental methodology utilized for identifying and isolating these unique cells. We discuss how CSCs are regulated within the tumor microenvironment as well as the role they portray in seeding fresh tumors. Finally, we explore the mechanisms that enable CSCs to evade modern therapeutic approaches and the possible strategies that can be utilized to prevent CSCs from resurrecting the disease.


Subject(s)
Biomarkers, Tumor/genetics , Cell Lineage/genetics , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Neoplasms/pathology , Neoplastic Stem Cells/pathology , Animals , Antigens, CD/genetics , Antigens, CD/immunology , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/immunology , Cell Lineage/drug effects , Cell Lineage/immunology , Disease Progression , Drug Resistance, Neoplasm/immunology , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/immunology , Humans , Mice , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/immunology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/immunology , Signal Transduction , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...