Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Cell Death Dis ; 15(2): 121, 2024 02 08.
Article in English | MEDLINE | ID: mdl-38331954

ABSTRACT

Mutation in CUL4B gene is one of the most common causes for X-linked intellectual disability (XLID). CUL4B is the scaffold protein in CUL4B-RING ubiquitin ligase (CRL4B) complex. While the roles of CUL4B in cancer progression and some developmental processes like adipogenesis, osteogenesis, and spermatogenesis have been studied, the mechanisms underlying the neurological disorders in patients with CUL4B mutations are poorly understood. Here, using 2D neuronal culture and cerebral organoids generated from the patient-derived induced pluripotent stem cells and their isogenic controls, we demonstrate that CUL4B is required to prevent premature cell cycle exit and precocious neuronal differentiation of neural progenitor cells. Moreover, loss-of-function mutations of CUL4B lead to increased synapse formation and enhanced neuronal excitability. Mechanistically, CRL4B complex represses transcription of PPP2R2B and PPP2R2C genes, which encode two isoforms of the regulatory subunit of protein phosphatase 2 A (PP2A) complex, through catalyzing monoubiquitination of H2AK119 in their promoter regions. CUL4B mutations result in upregulated PP2A activity, which causes inhibition of AKT and ERK, leading to premature cell cycle exit. Activation of AKT and ERK or inhibition of PP2A activity in CUL4B mutant organoids rescues the neurogenesis defect. Our work unveils an essential role of CUL4B in human cortical development.


Subject(s)
Protein Phosphatase 2 , Proto-Oncogene Proteins c-akt , Male , Humans , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Protein Phosphatase 2/genetics , Cullin Proteins/genetics , Cullin Proteins/metabolism , Mutation/genetics , Neurogenesis/genetics
2.
Oncogene ; 42(42): 3113-3126, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37653114

ABSTRACT

Lung cancer is the leading cause of cancer-related death worldwide. KRAS mutations are the most common oncogenic alterations found in lung cancer. Unfortunately, treating KRAS-mutant lung adenocarcinoma (ADC) remains a major oncotherapeutic challenge. Here, we used both autochthonous and transplantable KRAS-mutant tumor models to investigate the role of tumor-derived CUL4B in KRAS-driven lung cancers. We showed that knockout or knockdown of CUL4B promotes lung ADC growth and progression in both models. Mechanistically, CUL4B directly binds to the promoter of Cxcl2 and epigenetically represses its transcription. CUL4B deletion increases the expression of CXCL2, which binds to CXCR2 on myeloid-derived suppressor cells (MDSCs) and promotes their migration to the tumor microenvironment. Targeting of MDSCs significantly delayed the growth of CUL4B knockdown KRAS-mutant tumors. Collectively, our study provides mechanistic insights into the novel tumor suppressor-like functions of CUL4B in regulating KRAS-driven lung tumor development.

3.
Bone Res ; 11(1): 29, 2023 Jun 02.
Article in English | MEDLINE | ID: mdl-37268647

ABSTRACT

Dysregulated lineage commitment of mesenchymal stem cells (MSCs) contributes to impaired bone formation and an imbalance between adipogenesis and osteogenesis during skeletal aging and osteoporosis. The intrinsic cellular mechanism that regulates MSC commitment remains unclear. Here, we identified Cullin 4B (CUL4B) as a critical regulator of MSC commitment. CUL4B is expressed in bone marrow MSCs (BMSCs) and downregulated with aging in mice and humans. Conditional knockout of Cul4b in MSCs resulted in impaired postnatal skeletal development with low bone mass and reduced bone formation. Moreover, depletion of CUL4B in MSCs aggravated bone loss and marrow adipose accumulation during natural aging or after ovariectomy. In addition, CUL4B deficiency in MSCs reduced bone strength. Mechanistically, CUL4B promoted osteogenesis and inhibited adipogenesis of MSCs by repressing KLF4 and C/EBPδ expression, respectively. The CUL4B complex directly bound to Klf4 and Cebpd and epigenetically repressed their transcription. Collectively, this study reveals CUL4B-mediated epigenetic regulation of the osteogenic or adipogenic commitment of MSCs, which has therapeutic implications in osteoporosis.

4.
Cell Death Dis ; 14(6): 388, 2023 06 30.
Article in English | MEDLINE | ID: mdl-37391410

ABSTRACT

Chemotherapy is a common strategy to treat cancer. However, acquired resistance and metastasis are the major obstacles to successful treatment. Anastasis is a process by which cells survive executioner caspase activation when facing apoptotic stress. Here we demonstrate that colorectal cancer cells can undergo anastasis after transient exposure to chemotherapeutic drugs. Using a lineage tracing system to label and isolate cells that have experienced executioner caspase activation in response to drug treatment, we show that anastasis grants colorectal cancer cells enhanced migration, metastasis, and chemoresistance. Mechanistically, treatment with chemotherapeutic drugs induces upregulated expression of cIAP2 and activation of NFκB, which are required for cells to survive executioner caspase activation. The elevated cIAP2/NFκB signaling persists in anastatic cancer cells to promote migration and chemoresistance. Our study unveils that cIAP2/NFκB-dependent anastasis promotes acquired resistance and metastasis after chemotherapy.


Subject(s)
Cell Death Reversal , Colorectal Neoplasms , Humans , Drug Resistance, Neoplasm , NF-kappa B , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Caspases
5.
Oncogenesis ; 12(1): 34, 2023 Jun 24.
Article in English | MEDLINE | ID: mdl-37355711

ABSTRACT

Cancer relapse and metastasis are major obstacles for effective treatment. One important mechanism to eliminate cancer cells is to induce apoptosis. Activation of executioner caspases is the key step in apoptosis and was considered "a point of no return". However, in recent years, accumulating evidence has demonstrated that cells can survive executioner caspase activation in response to apoptotic stimuli through a process named anastasis. Here we show that breast cancer cells that have survived through anastasis (anastatic cells) after exposure to chemotherapeutic drugs acquire enhanced proliferation and migration. Mechanistically, cadherin 12 (CDH12) is persistently upregulated in anastatic cells and promotes breast cancer malignancy via activation of ERK and CREB. Moreover, we demonstrate that executioner caspase activation induced by chemotherapeutic drugs results in loss of DNA methylation and repressive histone modifications in the CDH12 promoter region, leading to increased CDH12 expression. Our work unveils the mechanism underlying anastasis-induced enhancement in breast cancer malignancy, offering new therapeutic targets for preventing post-chemotherapy cancer relapse and metastasis.

6.
Cell Rep ; 42(6): 112550, 2023 06 27.
Article in English | MEDLINE | ID: mdl-37224018

ABSTRACT

Diabetic kidney disease (DKD) is the most prevalent chronic kidney disease. Macrophage infiltration in the kidney is critical for the progression of DKD. However, the underlying mechanism is far from clear. Cullin 4B (CUL4B) is the scaffold protein in CUL4B-RING E3 ligase complexes. Previous studies have shown that depletion of CUL4B in macrophages aggravates lipopolysaccharide-induced peritonitis and septic shock. In this study, using two mouse models for DKD, we demonstrate that myeloid deficiency of CUL4B alleviates diabetes-induced renal injury and fibrosis. In vivo and in vitro analyses reveal that loss of CUL4B suppresses migration, adhesion, and renal infiltration of macrophages. Mechanistically, we show that high glucose upregulates CUL4B in macrophages. CUL4B represses expression of miR-194-5p, which leads to elevated integrin α9 (ITGA9), promoting migration and adhesion. Our study suggests the CUL4B/miR-194-5p/ITGA9 axis as an important regulator for macrophage infiltration in diabetic kidneys.


Subject(s)
Diabetes Mellitus , Diabetic Nephropathies , MicroRNAs , Animals , Mice , Cullin Proteins/genetics , Cullin Proteins/metabolism , Diabetes Mellitus/metabolism , Diabetic Nephropathies/genetics , Diabetic Nephropathies/metabolism , Integrin alpha Chains/metabolism , Macrophages/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism
7.
Cell Death Differ ; 30(6): 1488-1502, 2023 06.
Article in English | MEDLINE | ID: mdl-37024604

ABSTRACT

CD4+ T helper (Th) cell differentiation is regulated by lineage-specific expression of transcription factors, which is tightly associated with epigenetic modifications, including histone acetylation and methylation. However, the factors regulating histone modifications involved in Th cell differentiation remain largely unknown. We herein demonstrated a critical role of Cullin 4B (CUL4B) in restricting Th1 and Th2 cell differentiation. CUL4B, which is assembled into the CUL4B-RING E3 ligase (CRL4B) complex, participates in various physiological and developmental processes through epigenetic repression of transcription. Depletion of Cul4b in CD4+ T cells enhanced Th1 and Th2 cell differentiation. In vivo, an aggravated Th2 response caused by the absence of CUL4B was observed in a murine asthma model. Mechanistically, the CRL4B complex promoted monoubiquitination at H2AK119 (H2AK119ub1) and polycomb repressive complex 2 (PRC2)-mediated trimethylation at H3K27 (H3K27me3) at Tbx21 and Maf and consequently repressed their expression during Th cell differentiation. Our study suggests that CRL4B complex-mediated H2AK119ub1 deposition functions to prevent the aberrant expression of Th1 and Th2 lineage-specific genes.


Subject(s)
Epigenesis, Genetic , Polycomb Repressive Complex 2 , Animals , Mice , Ubiquitination , Methylation , Cell Differentiation , Polycomb Repressive Complex 2/metabolism
8.
J Assist Reprod Genet ; 40(1): 75-81, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36471203

ABSTRACT

PURPOSE: We aimed to identify pathogenic variants in a female patient with primary infertility and recurrent failure of in vitro fertilization with zygotic cleavage failure. METHODS: The genomic DNA from the affected individual was subjected to whole-exome sequencing and the variant was confirmed by Sanger sequencing. The functional effect of the identified variant was further investigated in 293 T cells. RESULTS: We identified a novel homozygous deletion in BTG4 (c.580_616del) in the affected individual. The deletion results in frameshift and replacement of the last 29 residues (aa195-223) with 66 random amino acids. The mutated amino acid residues are highly conserved among mammalian species. Co-immunoprecipitation in 293 T cells showed that the mutation abolished the interaction between BTG4 and PABPN1L. CONCLUSION: This study conforms previous studies and expands the mutational spectrum of BTG4. Our findings prove the functional importance of the C-terminal of BTG4. BTG4 is a potential diagnostic and therapeutic target for patients suffering from zygotic cleavage failure.


Subject(s)
Infertility, Female , Animals , Female , Humans , Cell Cycle Proteins/genetics , Fertilization in Vitro/methods , Homozygote , Infertility, Female/genetics , Infertility, Female/pathology , Mammals , Mutation/genetics , Poly(A)-Binding Proteins/genetics , Sequence Deletion
9.
Mol Metab ; 56: 101423, 2022 02.
Article in English | MEDLINE | ID: mdl-34954108

ABSTRACT

OBJECTIVE: Genome-wide association studies identified ORMDL3 as an obesity-related gene, and its expression was negatively correlated with body mass index. However, the precise biological roles of ORMDL3 in obesity and lipid metabolism remain uncharacterized. Here, we investigate the function of ORMDL3 in adipose tissue thermogenesis and high fat diet (HFD)-induced insulin resistance. METHODS: Ormdl3-deficient (Ormdl3-/-) mice were employed to delineate the function of ORMDL3 in brown adipose tissue (BAT) thermogenesis and white adipose tissue (WAT) browning. Glucose and lipid homeostasis in Ormdl3-/- mice fed a HFD were assessed. The lipid composition in adipose tissue was evaluated by mass spectrometry. Primary adipocytes in culture were used to determine the mechanism by which ORMDL3 regulates white adipose browning. RESULTS: BAT thermogenesis and WAT browning were significantly impaired in Ormdl3-/- mice upon cold exposure or administration with the ß3 adrenergic agonist. In addition, compared to WT mice, Ormdl3-/- mice displayed increased weight gain and insulin resistance in response to HFD. The induction of uncoupling protein 1 (UCP1), a marker of thermogenesis, was attenuated in primary adipocytes derived from Ormdl3-/- mice. Importantly, ceramide levels were elevated in the adipose tissue of Ormdl3-/- mice. In addition, the reduction in thermogenesis and increase in body weight caused by Ormdl3 deficiency could be rescued by inhibiting the production of ceramides. CONCLUSION: Our findings suggest that ORMDL3 contributes to the regulation of BAT thermogenesis, WAT browning, and insulin resistance.


Subject(s)
Adipose Tissue, Brown , Ceramides , Insulin Resistance , Membrane Proteins , Thermogenesis , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Animals , Ceramides/biosynthesis , Energy Metabolism , Genome-Wide Association Study , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout
10.
Life (Basel) ; 11(8)2021 Aug 20.
Article in English | MEDLINE | ID: mdl-34440599

ABSTRACT

Aging is characterized by a time dependent impairment of physiological function and increased susceptibility to death. It is the major risk factor for neurodegeneration. Neurodegenerative disorders including Alzheimer's disease (AD) and Parkinson's disease (PD) are the main causes of dementia in the old population. Gut microbiota is a community of microorganisms colonized in the gastrointestinal (GI) tract. The alteration of gut microbiota has been proved to be associated with aging and aging related neurodegeneration. Drosophila is a powerful tool to study microbiota-mediated physiological and pathological functions. Here, we summarize the recent advances using Drosophila as model organisms to clarify the molecular mechanisms and develop a therapeutic method targeting microbiota in aging and aging-related neurodegenerative disorders.

11.
Cancer Biol Med ; 2021 May 09.
Article in English | MEDLINE | ID: mdl-33969670

ABSTRACT

OBJECTIVE: Hepatitis B virus (HBV) infection is a major public health problem worldwide. However, the regulatory mechanisms underlying HBV replication remain unclear. Cullin 4B-RING ubiquitin E3 ligase (CRL4B) is involved in regulating diverse physiological and pathophysiological processes. In our study, we aimed to explain the role of CUL4B in HBV infection. METHODS: Cul4b transgenic mice or conditional knockout mice, as well as liver cell lines with CUL4B overexpression or knockdown, were used to assess the role of CUL4B in HBV replication. Immunoprecipitation assays and immunofluorescence staining were performed to study the interaction between CUL4B and HBx. Cycloheximide chase assays and in vivo ubiquitination assays were performed to evaluate the half-life and the ubiquitination status of HBx. RESULTS: The hydrodynamics-based hepatitis B model in Cul4b transgenic or conditional knockout mice indicated that CUL4B promoted HBV replication (P < 0.05). Moreover, the overexpression or knockdown system in human liver cell lines validated that CUL4B increased HBV replication in an HBx-dependent manner. Importantly, immunoprecipitation assays and immunofluorescence staining showed an interaction between CUL4B and HBx. Furthermore, CUL4B upregulated HBx protein levels by inhibiting HBx ubiquitination and proteasomal degradation (P < 0.05). Finally, a positive correlation between CUL4B expression and HBV pgRNA level was observed in liver tissues from HBV-positive patients and HBV transgenic mice. CONCLUSIONS: CUL4B enhances HBV replication by interacting with HBx and disrupting its ubiquitin-dependent proteasomal degradation. CUL4B may therefore be a potential target for anti-HBV therapy.

12.
J Pathol ; 254(2): 185-198, 2021 06.
Article in English | MEDLINE | ID: mdl-33638154

ABSTRACT

Tamoxifen (TAM) resistance is a significant clinical challenge in endocrine therapies for estrogen receptor (ER)-positive breast cancer patients. Cullin 4B (CUL4B), which acts as a scaffold protein in CUL4B-RING ubiquitin ligase complexes (CRL4B), is frequently overexpressed in cancer and represses tumor suppressors through diverse epigenetic mechanisms. However, the role and the underlying mechanisms of CUL4B in regulating drug resistance remain unknown. Here, we showed that CUL4B promotes TAM resistance in breast cancer cells through a miR-32-5p/ER-α36 axis. We found that upregulation of CUL4B correlated with decreased TAM sensitivity of breast cancer cells, and knockdown of CUL4B or expression of a dominant-negative CUL4B mutant restored the response to TAM in TAM-resistant MCF7-TAMR and T47D-TAMR cells. Mechanistically, we demonstrated that CUL4B renders breast cancer cells TAM-resistant by upregulating ER-α36 expression, which was mediated by downregulation of miR-32-5p. We further showed that CRL4B epigenetically represses the transcription of miR-32-5p by catalyzing monoubiquitination at H2AK119 and coordinating with PRC2 and HDAC complexes to promote trimethylation at H3K27 at the promoter of miR-32-5p. Pharmacologic or genetic inhibition of CRL4B/PRC2/HDAC complexes significantly increased TAM sensitivity in breast cancer cells in vitro and in vivo. Taken together, our findings thus establish a critical role for the CUL4B-miR-32-5p-ER-α36 axis in the regulation of TAM resistance and have important therapeutic implications for combined application of TAM and the inhibitors of CRL4B/PRC2/HDAC complex in breast cancer treatment. © 2021 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/pathology , Cullin Proteins/metabolism , Estrogen Receptor alpha/genetics , MicroRNAs/genetics , Tamoxifen/pharmacology , Animals , Breast Neoplasms/drug therapy , Cell Line, Tumor , Cullin Proteins/genetics , Drug Resistance, Neoplasm , Estrogen Receptor alpha/metabolism , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , Mice , Mutation , Transcription Factors/genetics , Transcription Factors/metabolism , Ubiquitination , Xenograft Model Antitumor Assays
13.
Cell Mol Immunol ; 18(2): 339-349, 2021 02.
Article in English | MEDLINE | ID: mdl-31729464

ABSTRACT

Toll-like receptors (TLRs) play critical roles in innate immunity and inflammation. The molecular mechanisms by which TLR signaling is fine-tuned remain to be completely elucidated. Cullin 4B (CUL4B), which assembles the CUL4B-RING E3 ligase complex (CRL4B), has been shown to regulate diverse developmental and physiological processes by catalyzing monoubiquitination for histone modification or polyubiquitination for proteasomal degradation. Here, we identified the role of CUL4B as an intrinsic negative regulator of the TLR-triggered inflammatory response. Deletion of CUL4B in macrophages increased the production of proinflammatory cytokines and decreased anti-inflammatory cytokine IL-10 production in response to pathogens that activate TLR3, TLR4, or TLR2. Myeloid cell-specific Cul4b knockout mice were more susceptible to septic shock when challenged with lipopolysaccharide, polyinosinic-polycytidylic acid or Salmonella typhimurium infection. We further demonstrated that enhanced TLR-induced inflammatory responses in the absence of CUL4B were mediated by increased GSK3ß activity. Suppression of GSK3ß activity efficiently blocked the TLR-triggered increase in proinflammatory cytokine production and attenuated TLR-triggered death in Cul4b mutant mice. Mechanistically, CUL4B was found to negatively regulate TLR-triggered signaling by epigenetically repressing the transcription of Pten, thus maintaining the anti-inflammatory PI3K-AKT-GSK3ß pathway. The upregulation of PTEN caused by CUL4B deletion led to uncontrolled GSK3ß activity and excessive inflammatory immune responses. Thus, our findings indicate that CUL4B functions to restrict TLR-triggered inflammatory responses through regulating the AKT-GSK3ß pathway.


Subject(s)
Cullin Proteins/physiology , Endotoxemia/pathology , Inflammation/pathology , Macrophages/immunology , PTEN Phosphohydrolase/antagonists & inhibitors , Shock, Septic/pathology , Toll-Like Receptors/metabolism , Animals , Endotoxemia/etiology , Endotoxemia/metabolism , Female , Inflammation/etiology , Inflammation/metabolism , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Salmonella typhimurium/physiology , Shock, Septic/etiology , Shock, Septic/metabolism
14.
Oncogene ; 39(17): 3588-3603, 2020 04.
Article in English | MEDLINE | ID: mdl-32127645

ABSTRACT

CUL4B, which acts as a scaffold protein in CUL4B-RING ubiquitin ligase (CRL4B) complexes, participates in a variety of biological processes. Previous studies have shown that CUL4B is often overexpressed and exhibits oncogenic activities in a variety of solid tumors. However, the roles and the underlying mechanisms of CUL4B in bladder cancer (BC) were poorly understood. Here, we showed that CUL4B levels were overexpressed and positively correlated with the malignancy of BC, and CUL4B could confer BC cells increased motility, invasiveness, stemness, and chemoresistance. The PIK3CA/AKT pathway was identified as a critical downstream mediator of CUL4B-driven oncogenicity in BC cells. Furthermore, we demonstrated that CRL4B epigenetically repressed the transcription of miR-372/373, via catalyzing monoubiquitination of H2AK119 at the gene cluster encoding miR-372/373, leading to upregulation of PIK3CA and activation of AKT. Our findings thus establish a critical role for the CUL4B-miR-372/373-PIK3CA/AKT axis in the pathogenesis of BC and have important prognostic and therapeutic implications in BC.


Subject(s)
Class I Phosphatidylinositol 3-Kinases/metabolism , Cullin Proteins/metabolism , MicroRNAs/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Neoplasm/metabolism , Signal Transduction , Urinary Bladder Neoplasms/metabolism , Animals , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases/genetics , Cullin Proteins/genetics , Epigenesis, Genetic , Female , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , Neoplasm Metastasis , Proto-Oncogene Proteins c-akt/genetics , RNA, Neoplasm/genetics , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology
15.
Oncogenesis ; 9(2): 20, 2020 Feb 13.
Article in English | MEDLINE | ID: mdl-32054830

ABSTRACT

Given that colorectal cancer stem cells (CCSCs) play key roles in the tumor dormancy, metastasis, and relapse, targeting CCSCs is a promising strategy in cancer therapy. Here, we aimed to identify the new regulators of CCSCs and found that Cullin 4B (CUL4B), which possesses oncogenic properties in multiple solid tumors, drives the development and metastasis of colon cancer by sustaining cancer stem-like features. Elevated expression of CUL4B was confirmed in colon tumors and was associated with poor overall survival. Inhibition of CUL4B in cancer cell lines and patient-derived tumor organoids led to reduced sphere formation, proliferation and metastasis capacity. Mechanistically, CUL4B coordinates with PRC2 complex to repress miR34a expression, thus upregulates oncogenes including MYCN and NOTCH1, which are targeted by miR34a. Furthermore, we found that elevated CUL4B expression is associated with miR34a downregulation and upregulation of miR34a target genes in colon cancer specimens. Collectively, our findings demonstrate that CUL4B functions to repress miR34a in maintaining cancer stemness in CRC and provides a potential therapeutic target.

16.
Biomed Res Int ; 2020: 1626378, 2020.
Article in English | MEDLINE | ID: mdl-33524082

ABSTRACT

Alport syndrome (AS) is a hereditary nephropathy which is characterized by molecular abnormalities in collagen IV. Here, we report compound mutations of the COL4A3 gene including a novel allele identified in a patient with Alport syndrome. The patient was a 25-year-old Chinese woman. She has a history of proteinuria and hematuria with cleft lip and palate. The pathologic results were consistent with Alport syndrome. The patient received ACEI treatment but did not respond well to the treatment. Sequencing results revealed that the patient carried two heterozygous mutations in the COL4A3 gene, including a known mutation (c.4243G>C, p.G1415R), which was inherited from her father, and a previously undescribed allele (c.4216G>A, p.G1406R) inherited from her mother. To date, at least 294 different variants of COL4A3 have been reported according to the Human Gene Mutation Database (HGMD). Identification of c.4216G>A as a new AS-related mutation may contribute to both genetic diagnosis of AS and further functional study of COL4A3.


Subject(s)
Alleles , Autoantigens/genetics , Collagen Type IV/genetics , Mutation/genetics , Nephritis, Hereditary/genetics , Adult , Autoantigens/chemistry , Base Sequence , Biopsy , Child, Preschool , Collagen Type IV/chemistry , Conserved Sequence/genetics , Female , Humans , Kidney/pathology , Kidney/ultrastructure , Male , Nephritis, Hereditary/pathology , Pedigree , Exome Sequencing
17.
Biomaterials ; 225: 119539, 2019 12.
Article in English | MEDLINE | ID: mdl-31622821

ABSTRACT

Photostimulation has been widely used in neuromodulation. However, existing optogenetics techniques require genetic alternation of the targeted cell or tissue. Here, we report that neural stem cells (NSCs) constitutionally express blue/red light-sensitive photoreceptors. The proliferation and regulation of NSCs to neuronal or glial cells are wavelength-specific. Our results showed a 4.3-fold increase in proliferation and 2.7-fold increase in astrocyte differentiation for cells under low-power blue monochromatic light exposure (455 nm, 300 µW/cm2). The melanopsin (Opn4)/transient receptor potential channel 6 (TRPC6) non-visual opsin serves as a key photoreceptor response to blue light irradiation. Two-dimensional gel electrophoresis coupled with mass spectrometry further highlighted the Jun activation domain-binding protein 1 (Jab1) as a novel and specific modulator in phototransduction pathways induced by blue light exposure. Quiescent adult NSCs reside in specific regions of the mammalian brain. Therefore, we showed that melanopsin/TRPC6 expressed in these regions and blue light stimulation through optical fibers could directly stimulate the NSCs in vivo. Upconversion nanoparticles (UCNPs) converted deep-penetrating near-infrared (NIR) light into specific wavelengths of visible light. Accordingly, we demonstrated that UCNP-mediated NIR light could be used to modulate in vivo NSC differentiation in a less invasive manner. In the future, this light-triggered system of NSCs will enable nongenetic and noninvasive neuromodulation with therapeutic potential for central nervous system diseases.


Subject(s)
Cell Differentiation , Neural Stem Cells/cytology , Neuroglia/cytology , Neurons/cytology , Optogenetics , Animals , Cell Differentiation/radiation effects , Cell Proliferation/radiation effects , Cell Self Renewal/radiation effects , Infrared Rays , Light Signal Transduction/radiation effects , Mice, Inbred C57BL , Nanoparticles/chemistry , Neural Stem Cells/radiation effects , Neuroglia/radiation effects , Neurons/radiation effects
18.
Oncogene ; 38(30): 5860-5872, 2019 07.
Article in English | MEDLINE | ID: mdl-31235785

ABSTRACT

Cancer progression depends on a tumor-supportive microenvironment. Myeloid-derived suppressor cells (MDSCs) represent key cellular components in tumor microenvironment and have been demonstrated to facilitate tumor progression by restricting host immune responses and by sustaining the malignancy of cancer cells. CUL4B, which assembles the CUL4B-RING E3 ligase complex (CRL4B), possesses a potent oncogenic property in cancer cells by epigenetically inactivating many tumor suppressors. However, CUL4B in hematopoietic cells exerts tumor-suppressive effect by restricting the accumulation and function of MDSCs. How CUL4B regulates the function of MDSCs is not fully characterized. In the present study, we demonstrate that the enhanced growth and metastasis of transplanted tumor cells in hematopoietic or myeloid cell-specific Cul4b knockout recipient mice is mediated by increased production of IL-6 in MDSCs. CUL4B complex epigenetically represses IL-6 transcription in myeloid cells. The IL-6 produced by MDSCs renders cancer cells stem cell-like properties by activating IL-6/STAT3 signaling. This crosstalk was effectively blocked either by blocking IL-6 in MDSCs or by inhibition of STAT3 activation in tumor cells. These findings provide a new mechanistic insight into the cancer-promoting property of MDSCs.


Subject(s)
Cullin Proteins/genetics , Interleukin-6/metabolism , Melanoma, Experimental/pathology , Myeloid-Derived Suppressor Cells/metabolism , Up-Regulation , Animals , Cell Line, Tumor , Melanoma, Experimental/metabolism , Mice , Mice, Knockout , STAT3 Transcription Factor/metabolism , Tumor Microenvironment
19.
Future Microbiol ; 13: 1117-1128, 2018 08.
Article in English | MEDLINE | ID: mdl-30043649

ABSTRACT

AIM: The aim of present study is to investigate the relationship between gut microbiota and Alzheimer's disease (AD) using Drosophila model. MATERIALS & METHODS: The microbiota was characterized by Illumina sequencing of 16S rRNA gene. Gas chromatography-mass spectrometer was performed to measure the level of short-chain fatty acids (SCFAs), metabolites of the commensal microbiota. RESULTS: The diversity of the gut microbiota increased in AD Drosophila. As the most enriched bacteria at genus level, the proportions of Acetobacter and Lactobacillus decreased dramatically. Acetate was the most abundant SCFA derived from the dysregulated microbiota and markedly downregulated in AD Drosophila. CONCLUSION: Our study on Drosophila model suggests that dysregulation of gut microbiota may participate in AD pathogenesis by influencing SCFA level.


Subject(s)
Acetates/metabolism , Bacteria/metabolism , Drosophila/microbiology , Gastrointestinal Microbiome/physiology , Acetates/analysis , Alzheimer Disease/metabolism , Alzheimer Disease/microbiology , Animals , Bacteria/classification , Bacteria/genetics , Biodiversity , Disease Models, Animal , Gastrointestinal Microbiome/genetics , Metabolome/genetics , Phenotype , RNA, Ribosomal, 16S/genetics
20.
J Clin Invest ; 127(7): 2631-2646, 2017 Jun 30.
Article in English | MEDLINE | ID: mdl-28604389

ABSTRACT

Somatostatin secreted by pancreatic δ cells mediates important paracrine interactions in Langerhans islets, including maintenance of glucose metabolism through the control of reciprocal insulin and glucagon secretion. Disruption of this circuit contributes to the development of diabetes. However, the precise mechanisms that control somatostatin secretion from islets remain elusive. Here, we found that a super-complex comprising the cullin 4B-RING E3 ligase (CRL4B) and polycomb repressive complex 2 (PRC2) epigenetically regulates somatostatin secretion in islets. Constitutive ablation of CUL4B, the core component of the CRL4B-PRC2 complex, in δ cells impaired glucose tolerance and decreased insulin secretion through enhanced somatostatin release. Moreover, mechanistic studies showed that the CRL4B-PRC2 complex, under the control of the δ cell-specific transcription factor hematopoietically expressed homeobox (HHEX), determines the levels of intracellular calcium and cAMP through histone posttranslational modifications, thereby altering expression of the Cav1.2 calcium channel and adenylyl cyclase 6 (AC6) and modulating somatostatin secretion. In response to high glucose levels or urocortin 3 (UCN3) stimulation, increased expression of cullin 4B (CUL4B) and the PRC2 subunit histone-lysine N-methyltransferase EZH2 and reciprocal decreases in Cav1.2 and AC6 expression were found to regulate somatostatin secretion. Our results reveal an epigenetic regulatory mechanism of δ cell paracrine interactions in which CRL4B-PRC2 complexes, Cav1.2, and AC6 expression fine-tune somatostatin secretion and facilitate glucose homeostasis in pancreatic islets.


Subject(s)
Cullin Proteins/metabolism , Insulin/metabolism , Multienzyme Complexes/metabolism , Paracrine Communication , Somatostatin-Secreting Cells/metabolism , Somatostatin/metabolism , Adenylyl Cyclases/genetics , Adenylyl Cyclases/metabolism , Animals , Calcium/metabolism , Calcium Channels, L-Type/genetics , Calcium Channels, L-Type/metabolism , Cullin Proteins/genetics , Cyclic AMP/metabolism , Epigenesis, Genetic , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Insulin/genetics , Insulin Secretion , Mice , Mice, Knockout , Multienzyme Complexes/genetics , Somatostatin/genetics , Somatostatin-Secreting Cells/cytology , Transcription Factors/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...