Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 310
Filter
1.
Respir Res ; 25(1): 284, 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39026235

ABSTRACT

Idiopathic pulmonary fibrosis is a lethal, progressive, and irreversible condition that has become a significant focus of medical research due to its increasing incidence. This rising trend presents substantial challenges for patients, healthcare providers, and researchers. Despite the escalating burden of pulmonary fibrosis, the available therapeutic options remain limited. Currently, the United States Food and Drug Administration has approved two drugs for the treatment of pulmonary fibrosis-nintedanib and pirfenidone. However, their therapeutic effectiveness is limited, and they cannot reverse the fibrosis process. Additionally, these drugs are associated with significant side effects. Myofibroblasts play a central role in the pathophysiology of pulmonary fibrosis, significantly contributing to its progression. Consequently, strategies aimed at inhibiting myofibroblast differentiation or promoting their dedifferentiation hold promise as effective treatments. This review examines the regulation of myofibroblast dedifferentiation, exploring various signaling pathways, regulatory targets, and potential pharmaceutical interventions that could provide new directions for therapeutic development.


Subject(s)
Cell Dedifferentiation , Myofibroblasts , Humans , Myofibroblasts/pathology , Myofibroblasts/metabolism , Myofibroblasts/drug effects , Cell Dedifferentiation/drug effects , Cell Dedifferentiation/physiology , Animals , Idiopathic Pulmonary Fibrosis/pathology , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/drug therapy , Signal Transduction/physiology , Antifibrotic Agents/therapeutic use , Antifibrotic Agents/pharmacology , Pulmonary Fibrosis/pathology , Pulmonary Fibrosis/metabolism
2.
Front Endocrinol (Lausanne) ; 15: 1414447, 2024.
Article in English | MEDLINE | ID: mdl-38915897

ABSTRACT

Type 2 diabetes (T2D) is a polygenic metabolic disorder characterized by insulin resistance in peripheral tissues and impaired insulin secretion by the pancreas. While the decline in insulin production and secretion was previously attributed to apoptosis of insulin-producing ß-cells, recent studies indicate that ß-cell apoptosis rates are relatively low in diabetes. Instead, ß-cells primarily undergo dedifferentiation, a process where they lose their specialized identity and transition into non-functional endocrine progenitor-like cells, ultimately leading to ß-cell failure. The underlying mechanisms driving ß-cell dedifferentiation remain elusive due to the intricate interplay of genetic factors and cellular stress. Understanding these mechanisms holds the potential to inform innovative therapeutic approaches aimed at reversing ß-cell dedifferentiation in T2D. This review explores the proposed drivers of ß-cell dedifferentiation leading to ß-cell failure, and discusses current interventions capable of reversing this process, thus restoring ß-cell identity and function.


Subject(s)
Cell Dedifferentiation , Diabetes Mellitus, Type 2 , Insulin-Secreting Cells , Humans , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/physiology , Insulin-Secreting Cells/cytology , Cell Dedifferentiation/physiology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Animals , Cell Differentiation/physiology , Apoptosis/physiology , Insulin Secretion/physiology
3.
Front Endocrinol (Lausanne) ; 15: 1427723, 2024.
Article in English | MEDLINE | ID: mdl-38904049

ABSTRACT

The pathogeneses of type 1 and type 2 diabetes involve the progressive loss of functional beta cell mass, primarily attributed to cellular demise and/or dedifferentiation. While the scientific community has devoted significant attention to unraveling beta cell dedifferentiation in type 2 diabetes, its significance in type 1 diabetes remains relatively unexplored. This perspective article critically analyzes the existing evidence for beta cell dedifferentiation in type 1 diabetes, emphasizing its potential to reduce beta cell autoimmunity. Drawing from recent advancements in both human studies and animal models, we present beta cell identity as a promising target for managing type 1 diabetes. We posit that a better understanding of the mechanisms of beta cell dedifferentiation in type 1 diabetes is key to pioneering interventions that balance beta cell function and immunogenicity.


Subject(s)
Cell Dedifferentiation , Diabetes Mellitus, Type 1 , Insulin-Secreting Cells , Animals , Humans , Autoimmunity , Cell Dedifferentiation/physiology , Diabetes Mellitus, Type 1/pathology , Insulin-Secreting Cells/pathology , Insulin-Secreting Cells/physiology
4.
Medicine (Baltimore) ; 103(13): e37595, 2024 Mar 29.
Article in English | MEDLINE | ID: mdl-38552064

ABSTRACT

BACKGROUND: Skin grafting is a common method of treating damaged skin; however, surgical complications may arise in patients with poor health. Currently, no effective conservative treatment is available for extensive skin loss. Mature adipocytes, which constitute a substantial portion of adipose tissue, have recently emerged as a potential source of stemness. When de-lipidated, these cells exhibit fibroblast-like characteristics and the ability to redifferentiate, offering homogeneity and research utility as "dedifferentiated fat cells." METHODS AND RESULTS: We conducted an in vitro study to induce fibroblast-like traits in the adipose tissue by transdifferentiating mature adipocytes for skin regeneration. Human subcutaneous fat tissues were isolated and purified from mature adipocytes that underwent a transformation process over 14 days of cultivation. Microscopic analysis revealed lipid degradation over time, ultimately transforming cells into fibroblast-like forms. Flow cytometry was used to verify their characteristics, highlighting markers such as CD90 and CD105 (mesenchymal stem cell markers) and CD56 and CD106 (for detecting fibroblast characteristics). Administering dedifferentiated fat cells with transforming growth factor-ß at the identified optimal differentiation concentration of 5 ng/mL for a span of 14 days led to heightened expression of alpha smooth muscle actin and fibronectin, as evidenced by RNA and protein analysis. Meanwhile, functional validation through cell sorting demonstrated limited fibroblast marker expression in both treated and untreated cells after transdifferentiation by transforming growth factor-ß. CONCLUSION: Although challenges remain in achieving more effective transformation and definitive fibroblast differentiation, our trial could pave the way for a novel skin regeneration treatment strategy.


Subject(s)
Cell Dedifferentiation , Cell Transdifferentiation , Humans , Pilot Projects , Cell Dedifferentiation/physiology , Adipose Tissue , Adipocytes/metabolism , Cell Differentiation , Fibroblasts/metabolism , Transforming Growth Factors/metabolism , Cells, Cultured
5.
Biochem Biophys Res Commun ; 667: 104-110, 2023 07 30.
Article in English | MEDLINE | ID: mdl-37210870

ABSTRACT

Biomechanical forces are known to regulate the biological behaviors of cells. Although negative pressure has been used for wound healing, it is still unknown about its role in regulating cell plasticity. We investigated whether negative pressure could induce the dedifferentiation of hepatocytes. Using a commercial device, we found that the exposure of primary human hepatocytes to -50 mmHg quickly induced the formation of stress fibers and obviously changed cell morphology in 72 h. Moreover, the exposure of hepatocytes to -50 mmHg significantly upregulated RhoA, ROCK1, and ROCK2 in 1-6 h, and dramatically enhanced the expression of marker molecules on "stemness", such as OCT4, SOX2, KLF4, MYC, NANOG, and CD133 in 6-72 h. However, all these changes in hepatocytes induced by -50 mmHg stimulation were almost abrogated by ROCK inhibitor Y27623. Our data suggest that an appropriate force of negative pressure stimulation can effectively induce the dedifferentiation of hepatocytes via RhoA/ROCK pathway activation.


Subject(s)
Cell Dedifferentiation , Hepatocytes , rhoA GTP-Binding Protein , Humans , Hepatocytes/metabolism , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Signal Transduction , Cell Dedifferentiation/genetics , Cell Dedifferentiation/physiology
6.
Theranostics ; 12(12): 5470-5487, 2022.
Article in English | MEDLINE | ID: mdl-35910794

ABSTRACT

Rationale: Wound healing is among the most complicated physiological processes and requires the synchronization of various cell types with distinct roles to re-establish the condition of the original skin. Patients affected by peripheral neuropathies often experience failure to heal. Loss of Schwann cells (SCs), a crucial population of peripheral nervous system cells in skin, may contribute to chronic wounds. However, the role of SCs in wound healing are poorly understood. Methods: The activity of SCs was investigated by using a cell atlas of the wound healing process, which was generated by integrating single-cell RNA sequencing (scRNA-seq) libraries covering different states of mouse back skin. The results of in silico analysis were validated by in vitro cell culture and in vivo mouse model. Selective inhibitors and conditional RNAi by virus transfection were utilized to investigate the role of SCs in wound healing. Findings from mouse experiments were further verified in scRNA-seq analysis of diabetic patients. Results: Our in silico analysis revealed the heterogeneous cellular components of skin and the dynamic interactions of neural crest derived cells (NCs) with other cell types. We found that SCs dedifferentiated at an early stage of wound repair with upregulated Wnt signaling. We also identified dedifferentiated SC (dSC) defect in diabetic wounds in both mouse and human. Wnt inhibition at the wound site repressed SC dedifferentiation, leading to defective repair. Furthermore, dSCs derived TGF-ß3, which is context-dependent, promoted the migration of fibroblasts and keratinocytes. Moreover, TGF-ß3 supplementation enhanced the healing of chronic wounds in diabetic mice with impaired SCs. Conclusion: Our study thus advances the understanding of the roles of neural-derived cells in skin regeneration and suggests a potential therapeutic strategy for wound healing disorders.


Subject(s)
Cell Dedifferentiation , Diabetes Mellitus, Experimental , Peripheral Nervous System Diseases , Schwann Cells , Transforming Growth Factor beta3 , Wound Healing , Animals , Cell Dedifferentiation/genetics , Cell Dedifferentiation/physiology , Humans , Mice , Peripheral Nervous System Diseases/genetics , Schwann Cells/physiology , Skin/injuries , Skin/innervation , Transforming Growth Factor beta3/genetics , Wound Healing/genetics , Wound Healing/physiology
7.
Dev Cell ; 57(2): 166-179.e6, 2022 01 24.
Article in English | MEDLINE | ID: mdl-35016013

ABSTRACT

Loss of differentiated cells to tissue damage is a hallmark of many diseases. In slow-turnover tissues, long-lived differentiated cells can re-enter the cell cycle or transdifferentiate to another cell type to promote repair. Here, we show that in a high-turnover tissue, severe damage to the differentiated compartment induces progenitors to transiently acquire a unique transcriptional and morphological postmitotic state. We highlight this in an acute villus injury model in the mouse intestine, where we identified a population of progenitor-derived cells that covered injured villi. These atrophy-induced villus epithelial cells (aVECs) were enriched for fetal markers but were differentiated and lineage committed. We further established a role for aVECs in maintaining barrier integrity through the activation of yes-associated protein (YAP). Notably, loss of YAP activity led to impaired villus regeneration. Thus, we define a key repair mechanism involving the activation of a fetal-like program during injury-induced differentiation, a process we term "adaptive differentiation."


Subject(s)
Adaptation, Biological/physiology , Cell Dedifferentiation/physiology , Wound Healing/physiology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Cycle , Cell Cycle Proteins/metabolism , Cell Dedifferentiation/genetics , Cell Differentiation/physiology , Cell Proliferation/physiology , Epithelial Cells/metabolism , Female , Intestinal Mucosa/injuries , Intestinal Mucosa/metabolism , Male , Mice , Mice, Inbred C57BL , Models, Animal , Phosphoproteins/metabolism , Regeneration , Signal Transduction/physiology , Stem Cells/cytology , YAP-Signaling Proteins/metabolism
8.
Histopathology ; 80(1): 135-149, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34958502

ABSTRACT

Primary cutaneous and mucosal melanoma shows a wide histological spectrum. The correct diagnosis depends upon the demonstration of melanocytic differentiation by recognition of an associated in-situ component or immunohistochemical evidence of a melanocytic phenotype using conventional melanocytic markers, such as S-100, SOX10, Melan-A and HMB-45. Exceptionally, melanomas lose their melanocytic phenotype, at least focally, and show differentiation towards other lineages. Review of the literature shows that de- and trans-differentiation in melanoma is rare but probably under-recognised and under-reported. These often large and frequently ulcerated tumours affect adults and show a wide anatomical distribution, including mucosal sites, although there is a predilection for sun-damaged skin of the head and neck. Histologically, the tumours are biphasic and contain a pre-existing conventional melanoma. The de-differentiated component closely resembles atypical fibroxanthoma, both morphologically and immunohistochemically. Trans-differentiated melanoma may show rhabdomyosarcomatous or spindle cell carcinomatous features. Undifferentiated melanomas are similar tumours in which the conventional melanoma component is absent. Their diagnosis depends entirely upon the clinical context and identification of a classical melanoma driver gene mutation, i.e. BRAF V600E. The diagnosis of these rare and unusual tumours is challenging, and requires thorough tumour sampling and recognition of the background of a pre-existing but often focal conventional melanoma together with molecular analysis.


Subject(s)
Cell Dedifferentiation/physiology , Cell Differentiation/physiology , Melanoma/pathology , Skin Neoplasms/pathology , Skin/pathology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Humans , Melanoma/genetics , Melanoma/metabolism , Mutation , Skin/metabolism , Skin Neoplasms/genetics , Skin Neoplasms/metabolism
9.
Mol Neurobiol ; 59(1): 429-444, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34708329

ABSTRACT

Silent information regulator 6 (SIRT6) is a mammalian homolog of the nicotinamide adenine dinucleotide (NAD)-dependent deacetylase sirtuin family. Previous studies have been reported a pro-regenerative role of SIRT6 in central nervous system injury. However, the role of SIRT6 in peripheral nerve injury is still unknown. Given the importance and necessity of Schwann cell dedifferentiation response to peripheral nerve injury, we aim to investigate the molecular mechanism of SIRT6 steering Schwann cell dedifferentiation during Wallerian degeneration in injured peripheral nerve. Herein, we first examined the expression pattern of SIRT6 after peripheral nerve injury. Using the explants of sciatic nerve, an ex vivo model of nerve degeneration, we provided evidences indicating that SIRT6 inhibitor accelerates Schwann cell dedifferentiation as well as axonal and myelin degeneration, while SIRT6 activator attenuates this process. Moreover, in an in vitro Schwann cell dedifferentiation model, we found SIRT6 inhibitor promotes Schwann cell dedifferentiation through upregulating the expression of c-Jun. In addition, downregulation of c-Jun reverse the effects of SIRT6 inhibition on the Schwann cells dedifferentiation and axonal and myelin degeneration. In summary, we first described SIRT6 acts as a negative regulator for Schwann cells dedifferentiation during Wallerian degeneration and c-Jun worked as a direct downstream partner of SIRT6 in injured peripheral nerve.


Subject(s)
Cell Dedifferentiation/physiology , Peripheral Nerve Injuries/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Schwann Cells/metabolism , Sirtuins/metabolism , Wallerian Degeneration/metabolism , Animals , Cell Dedifferentiation/drug effects , Peripheral Nerve Injuries/pathology , Rats , Schwann Cells/drug effects , Sirtuins/antagonists & inhibitors , Wallerian Degeneration/pathology
10.
J Neuroinflammation ; 18(1): 293, 2021 Dec 17.
Article in English | MEDLINE | ID: mdl-34920725

ABSTRACT

BACKGROUND: Lysophosphatidic acid (LPA) is a pleiotropic lipid messenger that addresses at least six specific G-protein coupled receptors. Accumulating evidence indicates a significant involvement of LPA in immune cell regulation as well as Schwann cell physiology, with potential relevance for the pathophysiology of peripheral neuroinflammation. However, the role of LPA signaling in inflammatory neuropathies has remained completely undefined. Given the broad expression of LPA receptors on both Schwann cells and cells of the innate and adaptive immune system, we hypothesized that inhibition of LPA signaling may ameliorate the course of disease in experimental autoimmune neuritis (EAN). METHODS: We induced active EAN by inoculation of myelin protein 2 peptide (P255-78) in female Lewis rats. Animals received the orally available LPA receptor antagonist AM095, specifically targeting the LPA1 receptor subtype. AM095 was administered daily via oral gavage in a therapeutic regimen from 10 until 28 days post-immunization (dpi). Analyses were based on clinical testing, hemogram profiles, immunohistochemistry and morphometric assessment of myelination. RESULTS: Lewis rats treated with AM095 displayed a significant improvement in clinical scores, most notably during the remission phase. Cellular infiltration of sciatic nerve was only discretely affected by AM095. Hemogram profiles indicated no impact on circulating leukocytes. However, sciatic nerve immunohistochemistry revealed a reduction in the number of Schwann cells expressing the dedifferentiation marker Sox2 paralleled by a corresponding increase in differentiating Sox10-positive Schwann cells. In line with this, morphometric analysis of sciatic nerve semi-thin sections identified a significant increase in large-caliber myelinated axons at 28 dpi. Myelin thickness was unaffected by AM095. CONCLUSION: Thus, LPA1 signaling may present a novel therapeutic target for the treatment of inflammatory neuropathies, potentially affecting regenerative responses in the peripheral nerve by modulating Schwann cell differentiation.


Subject(s)
Cell Dedifferentiation/physiology , Neuritis, Autoimmune, Experimental/immunology , Receptors, Lysophosphatidic Acid/immunology , Schwann Cells/immunology , Signal Transduction/physiology , Animals , Biphenyl Compounds/pharmacology , Biphenyl Compounds/therapeutic use , Cell Dedifferentiation/drug effects , Female , Isoxazoles/pharmacology , Isoxazoles/therapeutic use , Neuritis, Autoimmune, Experimental/drug therapy , Neuritis, Autoimmune, Experimental/metabolism , Rats , Rats, Inbred Lew , Receptors, Lysophosphatidic Acid/antagonists & inhibitors , Receptors, Lysophosphatidic Acid/metabolism , Schwann Cells/drug effects , Schwann Cells/metabolism , Signal Transduction/drug effects
11.
Life Sci ; 287: 120126, 2021 Dec 15.
Article in English | MEDLINE | ID: mdl-34758295

ABSTRACT

AIMS: Astrocytes re-acquire stem cell potential upon inflammation, thereby becoming a promising source of cells for regenerative medicine. Nanog is an essential transcription factor to maintain the characteristics of stem cells. We aimed to investigate the role of Nanog in astrocyte dedifferentiation. MAIN METHODS: TNF-α was used to induce the dedifferentiation of primary rat spinal cord astrocytes. The expression of immature markers CD44 and Musashi-1 was detected by qRT-PCR and immunofluorescence. The Nanog gene is knocked down by small interference RNA. Nanog expression was measured by qRT-PCR and western blotting. BAY 11-7082 was used to suppress NF-κB signals in astrocytes. NF-κB signaling was evaluated by Western blotting. KEY FINDINGS: Our results showed that TNF-α promoted the re-expression of CD44 and Musashi-1 in astrocytes. Dedifferentiated astrocytes could be induced to differentiate into oligodendrocyte lineage cells indicating that the astrocytes had pluripotency. In addition, TNF-α treatment activated NF-κB signaling pathway and up-regulated Nanog. Knockdown of Nanog reversed the increase of CD44 and Musashi-1 induced by TNF-α without affecting the activation of NF-κB signaling. Importantly, blocking NF-κB signaling by BAY 11-7082 inhibited the expression of immature markers suggesting that TNF-α induces dedifferentiation of astrocytes through the NF-κB signaling pathway. BAY 11-7082 could also inhibit the expression of Nanog, which indicated that Nanog was regulated by NF-κB signaling pathway. SIGNIFICANCE: These findings indicate that activation of the NF-κB signaling pathway through TNF-α leads to astrocytes dedifferentiation via Nanog. These results expand our understanding of the mechanism of astrocytes dedifferentiation.


Subject(s)
Astrocytes/metabolism , Cell Dedifferentiation/physiology , NF-kappa B/metabolism , Nanog Homeobox Protein/biosynthesis , Spinal Cord/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Animals , Astrocytes/drug effects , Cell Dedifferentiation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Female , Male , Rats , Signal Transduction/drug effects , Signal Transduction/physiology , Spinal Cord/cytology , Spinal Cord/drug effects , Up-Regulation/drug effects , Up-Regulation/physiology
12.
Int J Mol Sci ; 22(19)2021 Sep 22.
Article in English | MEDLINE | ID: mdl-34638532

ABSTRACT

Bone metastases from prostate cancer (PCa) result from a complex cross-talk between PCa cells and osteoblasts (OB). Thus, targeting this interplay has become an attractive strategy to interfere with PCa bone dissemination. The agents currently used in clinical trials have proved ineffective, boosting research to identify additional mechanisms that may be involved in this two-directional talk. Here, we investigated whether and how 5-hydro-5-methylimidazolone (MG-H1), a specific methylglyoxal (MG)-derived advanced glycation end product (AGE), was a novel player in the dialogue between PCa and OB to drive PCa bone metastases. Conditioned medium from osteotropic PC3 PCa cells, pre-treated or not with a specific MG scavenger, was administrated to human primary OB and cell morphology, mesenchymal trans-differentiation, pro-osteogenic determinants, PCa-specific molecules, and migration/invasion were studied by phase-contrast microscopy, real-time PCR, western blot and specific assays, respectively. We found that PC3 cells were able to release MG-H1 that, by binding to the receptor for AGEs (RAGE) on OB, reprogrammed them into a less-differentiate phenotype, endowed with some PCa-specific molecular features and malignant properties, in a mechanism involving reactive oxidative species (ROS) production and NF-kB pathway activation. These findings provide novel insights into the mechanisms of PCa osteoblastic metastases and foster in vivo research toward new therapeutic strategies interfering with PCa/OB cross-talk.


Subject(s)
Bone Neoplasms/secondary , Cell Dedifferentiation/physiology , Imidazoles/metabolism , Ornithine/analogs & derivatives , Osteoblasts/cytology , Prostatic Neoplasms/pathology , Antigens, Neoplasm/metabolism , Bone and Bones/pathology , Cell Line, Tumor , Cell Movement/physiology , Culture Media, Conditioned/pharmacology , Humans , Male , Mitogen-Activated Protein Kinases/metabolism , Ornithine/metabolism , PC-3 Cells , Prostate/pathology , Reactive Oxygen Species/metabolism
13.
Dev Biol ; 480: 105-113, 2021 12.
Article in English | MEDLINE | ID: mdl-34481794

ABSTRACT

Regeneration is a key developmental process by which organisms recover vital tissue and organ components following injury or disease. A growing interest is focused on the elucidation and characterization of the molecular mechanisms involved in these regenerative processes. We have now analyzed the possible role of the Wnt/ß-catenin pathway on the regeneration of the intestine in the sea cucumber Holothuria glaberrima. For this we have studied the expression in vivo of Wnt-associated genes and have implemented the use of Dicer-substrate interference RNA (DsiRNA) to knockdown the expression of ß-catenin transcript on gut rudiment explants. Neither cell dedifferentiation nor apoptosis were affected by the reduction of ß-catenin transcripts in the gut rudiment explants. Yet, the number of proliferating cells decreased significantly following the interference, suggesting that the Wnt/ß-catenin signaling pathway plays a significant role in cell proliferation, but not in cell dedifferentiation nor apoptosis during the regeneration of the intestine. The development of the in vitro RNAi protocol is a significant step in analyzing specific gene functions involved in echinoderm regeneration.


Subject(s)
Intestines/metabolism , Muscle, Skeletal/physiology , Wnt Signaling Pathway/physiology , Animals , Apoptosis/physiology , Cell Dedifferentiation/physiology , Cell Proliferation/genetics , Holothuria/metabolism , Holothuria/physiology , Intestines/growth & development , Muscle, Skeletal/metabolism , Regeneration/physiology , Sea Cucumbers/metabolism , Sea Cucumbers/physiology , Wnt Proteins/metabolism , Wnt Signaling Pathway/genetics , beta Catenin/metabolism
14.
Eur J Endocrinol ; 185(4): 565-576, 2021 Sep 01.
Article in English | MEDLINE | ID: mdl-34374650

ABSTRACT

OBJECTIVE: Progressive beta-cell dysfunction is a hallmark of type 2 diabetes (T2D). Increasing evidence indicates that over-stimulating proinsulin synthesis causes proinsulin misfolding and impairs insulin maturation and storage in db/db mice. However, defective insulin maturation in patients with T2D remains unknown. METHODS: We examined intra-islet and intra-cellular distributions of proinsulin and insulin and proinsulin to insulin ratio in the islets of patients with T2D. The expression of transcription factor NKX6.1 and dedifferentiation marker ALDH1A3, as well as glucagon, were detected by immunofluorescence. RESULTS: We identified a novel subgroup of beta cells expressing only proinsulin but not insulin. Importantly, significantly increased proinsulin positive and insulin negative (PI+/INS-) cells were evident in T2D, and this increase was strongly correlated with levels of hemoglobin A1C (HbA1c) in T2D and prediabetes. The percentages of beta cells expressing prohormone convertase 1/3 and carboxypeptidase E were not reduced. Indeed, while proinsulin displayed a higher degree of co-localization with the golgi markers GM130/TGN46 in control beta cells, it appeared to be more diffused within the cytoplasm and less co-localized with GM130/TGN46 in PI+/INS- cells. Furthermore, the key functional transcription factor NKX6.1 markedly decreased in the islets of T2D, especially in the cells with PI+/INS-. The decreased NKX6.1+/PI+/INS+ was strongly correlated with levels of HbA1c in T2D. Almost all PI+/INS- cells showed absence of NKX6.1. Moreover, the percentages of PI+/INS- cells expressing ALDH1A3 were elevated along with an increased acquisition of glucagon immunostaining. CONCLUSION: Our data demonstrate defective insulin maturation in patients with T2D.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Proinsulin/metabolism , Protein Processing, Post-Translational/physiology , Adult , Aldehyde Oxidoreductases/metabolism , Case-Control Studies , Cell Dedifferentiation/physiology , China , Diabetes Mellitus, Type 2/pathology , Female , Glucagon/metabolism , Homeodomain Proteins/metabolism , Humans , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Insulin-Secreting Cells/physiology , Male , Middle Aged , Prediabetic State/metabolism , Prediabetic State/pathology
15.
Exp Biol Med (Maywood) ; 246(24): 2618-2629, 2021 12.
Article in English | MEDLINE | ID: mdl-34424090

ABSTRACT

Non-keratinizing nasopharyngeal carcinoma, the major subtype of nasopharyngeal carcinoma, is characterized by low differentiation and a close relation to Epstein-Barr virus infection, which indicates a link between Epstein-Barr virus oncogenesis and loss of differentiation, and raises our interest in investigating the involvement of Epstein-Barr virus in nasopharyngeal carcinoma dedifferentiation. Our previous study showed abundant expression of an Epstein-Barr virus-encoded microRNA, BART10-3p, in nasopharyngeal carcinoma tissues, but the association between BART10-3p and nasopharyngeal carcinoma differentiation remains unknown. Here, we examined the expression and prognostic value of BART10-3p, and undertook bioinformatics analysis and functional assays to investigate the influence of BART10-3p on nasopharyngeal carcinoma differentiation and proliferation and the underpinning mechanism. Microarray analysis identified BART10-3p as the most significantly upregulated Epstein-Barr virus-encoded microRNA in nasopharyngeal carcinoma tissues and the upregulation was confirmed in two public datasets. The expression of BART10-3p was an independent unfavorable prognosticator in nasopharyngeal carcinoma and its integration with the clinical stage showed improved prognosis predictive performance. Bioinformatics analysis suggested a potential role of BART10-3p in tumor differentiation and progression. Functional assays demonstrated that BART10-3p could promote nasopharyngeal carcinoma cell dedifferentiation, epithelial-mesenchymal transition, and proliferation in vitro, and tumorigenicity in vivo. Mechanistically, BART10-3p directly targeted the 3'UTR of ALK7 and suppressed its expression. Reconstitution of ALK7 rescued BART10-3p-induced malignant phenotypes. Overall, our study demonstrates that BART10-3p promotes dedifferentiation and proliferation of nasopharyngeal carcinoma by targeting ALK7, suggesting a promising therapeutic opportunity to reverse the malignant phenotypes of nasopharyngeal carcinoma.


Subject(s)
Activin Receptors, Type I/metabolism , Epstein-Barr Virus Infections/virology , MicroRNAs/metabolism , Nasopharyngeal Carcinoma/pathology , Nasopharyngeal Neoplasms/pathology , RNA, Viral/metabolism , Cell Dedifferentiation/physiology , Cell Proliferation/physiology , Gene Expression Regulation, Neoplastic/physiology , Herpesvirus 4, Human , Humans , Nasopharyngeal Carcinoma/metabolism , Nasopharyngeal Carcinoma/virology , Nasopharyngeal Neoplasms/metabolism , Nasopharyngeal Neoplasms/virology , Tumor Cells, Cultured
16.
Diabetes ; 70(9): 2058-2066, 2021 09.
Article in English | MEDLINE | ID: mdl-34417264

ABSTRACT

Loss of mature ß-cell function and identity, or ß-cell dedifferentiation, is seen in both type 1 and type 2 diabetes. Two competing models explain ß-cell dedifferentiation in diabetes. In the first model, ß-cells dedifferentiate in the reverse order of their developmental ontogeny. This model predicts that dedifferentiated ß-cells resemble ß-cell progenitors. In the second model, ß-cell dedifferentiation depends on the type of diabetogenic stress. This model, which we call the "Anna Karenina" model, predicts that in each type of diabetes, ß-cells dedifferentiate in their own way, depending on how their mature identity is disrupted by any particular diabetogenic stress. We directly tested the two models using a ß-cell-specific lineage-tracing system coupled with RNA sequencing in mice. We constructed a multidimensional map of ß-cell transcriptional trajectories during the normal course of ß-cell postnatal development and during their dedifferentiation in models of both type 1 diabetes (NOD) and type 2 diabetes (BTBR-Lepob/ob ). Using this unbiased approach, we show here that despite some similarities between immature and dedifferentiated ß-cells, ß-cell dedifferentiation in the two mouse models is not a reversal of developmental ontogeny and is different between different types of diabetes.


Subject(s)
Cell Dedifferentiation/physiology , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 2/pathology , Insulin-Secreting Cells/pathology , Animals , Cell Lineage/physiology , Mice
17.
Neurobiol Aging ; 106: 80-94, 2021 10.
Article in English | MEDLINE | ID: mdl-34256190

ABSTRACT

Healthy aging is accompanied by reduced cognitive control and widespread alterations in the underlying brain networks; but the extent to which large-scale functional networks in older age show reduced specificity across different domains of cognitive control is unclear. Here we use cov-STATIS (a multi-table multivariate technique) to examine similarity of functional connectivity during different domains of cognitive control-inhibition, initiation, shifting, and working memory-across the adult lifespan. We report two major findings: (1) Functional connectivity patterns during initiation, inhibition, and shifting were more similar in older ages, particularly for control and default networks, a pattern consistent with dedifferentiation of the neural correlates associated with cognitive control; and (2) Networks exhibited age-related reconfiguration such that frontal, default, and dorsal attention networks were more integrated whereas sub-networks of somato-motor system were more segregated in older age. Together these findings offer new evidence for dedifferentiation and reconfiguration of functional connectivity underlying different aspects of cognitive control in normal aging.


Subject(s)
Brain/physiology , Cell Dedifferentiation/physiology , Cognition/physiology , Healthy Aging/physiology , Longevity/physiology , Nerve Net/physiology , Adult , Aged , Aged, 80 and over , Aging/physiology , Aging/psychology , Brain/diagnostic imaging , Female , Healthy Aging/psychology , Humans , Magnetic Resonance Imaging/methods , Male , Middle Aged , Nerve Net/diagnostic imaging , Psychomotor Performance/physiology , Young Adult
18.
Nutrients ; 13(5)2021 May 10.
Article in English | MEDLINE | ID: mdl-34068827

ABSTRACT

Diabetes is a metabolic disease characterized by hyperglycemia. Over 90% of patients with diabetes have type 2 diabetes. Pancreatic ß-cells are endocrine cells that produce and secrete insulin, an essential endocrine hormone that regulates blood glucose levels. Deficits in ß-cell function and mass play key roles in the onset and progression of type 2 diabetes. Apoptosis has been considered as the main contributor of ß-cell dysfunction and decrease in ß-cell mass for a long time. However, recent studies suggest that ß-cell failure occurs mainly due to increased ß-cell dedifferentiation rather than limited ß-cell proliferation or increased ß-cell death. In this review, we summarize the current advances in the understanding of the pancreatic ß-cell dedifferentiation process including potential mechanisms. A better understanding of ß-cell dedifferentiation process will help to identify novel therapeutic targets to prevent and/or reverse ß-cell loss in type 2 diabetes.


Subject(s)
Cell Dedifferentiation/physiology , Diabetes Mellitus, Type 2 , Insulin-Secreting Cells/physiology , Animals , Apoptosis , Cell Proliferation , Cytokines , Diabetes Mellitus, Type 2/pathology , Endoplasmic Reticulum Stress , Humans , Inflammation , Insulin/metabolism , MicroRNAs/metabolism , Oxidative Stress , RNA, Long Noncoding
19.
PLoS One ; 16(5): e0251054, 2021.
Article in English | MEDLINE | ID: mdl-33951105

ABSTRACT

In contrast to mammals, lower vertebrates are capable of extraordinary myocardial regeneration thanks to the ability of their cardiomyocytes to undergo transient dedifferentiation and proliferation. Somatic cells can be temporarily reprogrammed to a proliferative, dedifferentiated state through forced expression of Oct3/4, Sox2, Klf4 and c-Myc (OSKM). Here, we aimed to induce transient reprogramming of mammalian cardiomyocytes in vitro utilising an OSKM-encoding non-integrating vector. Reprogramming factor expression in postnatal rat and mouse cardiomyocytes triggered rapid but limited cell dedifferentiation. Concomitantly, a significant increase in cell viability, cell cycle related gene expression and Ki67 positive cells was observed consistent with an enhanced cell cycle activation. The transient nature of this partial reprogramming was confirmed as cardiomyocyte-specific cell morphology, gene expression and contractile activity were spontaneously recovered by day 15 after viral transduction. This study provides the first evidence that adenoviral OSKM delivery can induce partial reprogramming of postnatal cardiomyocytes. Therefore, adenoviral mediated transient reprogramming could be a novel and feasible strategy to recapitulate the regenerative mechanisms of lower vertebrates.


Subject(s)
Cell Dedifferentiation/physiology , Cellular Reprogramming/physiology , Myocytes, Cardiac/physiology , Animals , Cell Cycle/physiology , Cell Survival/physiology , Cells, Cultured , Gene Expression/physiology , Ki-67 Antigen/metabolism , Kruppel-Like Factor 4 , Mammals/metabolism , Mammals/physiology , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/metabolism , Rats , Rats, Sprague-Dawley
20.
Adv Anat Pathol ; 28(3): 107-118, 2021 May 01.
Article in English | MEDLINE | ID: mdl-33825717

ABSTRACT

High-grade transformation (HGT) or dedifferentiation has been described in a variety of salivary gland carcinomas, including acinic cell carcinoma, secretory carcinoma, adenoid cystic carcinoma, epithelial-myoepithelial carcinoma, polymorphous adenocarcinoma, low-grade mucoepidermoid carcinoma, and hyalinizing clear cell carcinoma. High-grade (HG) transformed tumors are composed of a conventional low-grade component characterized by specific microscopic and immunohistochemical features for the given entity, intermingled with or juxtaposed to areas of HG morphology. This is usually either poorly differentiated adenocarcinoma, carcinoma not otherwise specified, or undifferentiated carcinoma, in which the original line of differentiation is lost. The HG component is composed of solid nests of anaplastic cells with large vesicular pleomorphic nuclei, prominent nucleoli, and abundant cytoplasm. Frequent mitoses and extensive necrosis may be present. The Ki-67 labeling index is consistently higher in the HG component. The molecular genetic mechanisms responsible for HGT of salivary gland carcinomas are largely unknown, though p53 inactivation and human epidermal growth factor receptor 2 overexpression and/or gene amplification have been demonstrated in the HG component in a few examples, the frequency varies for each histologic type. Salivary gland carcinomas with HGT are more aggressive than conventional carcinomas, with a higher local recurrence rate and a poorer prognosis. They have a high propensity for cervical lymph node metastasis suggesting a need for a wider resection and neck dissection. HGT of salivary gland carcinoma can occur either at initial presentation or less commonly at the time of recurrence, sometimes following postoperative radiotherapy. The potential for HGT in almost any type of salivary gland carcinoma warrants a thorough sampling of all salivary gland malignancies to prevent oversight of a HG component.


Subject(s)
Carcinoma/pathology , Cell Dedifferentiation/physiology , Cell Transformation, Neoplastic/pathology , Salivary Gland Neoplasms/pathology , Salivary Glands/pathology , Biomarkers, Tumor/genetics , Carcinoma/genetics , Cell Transformation, Neoplastic/genetics , Humans , Receptor, ErbB-2/genetics , Salivary Gland Neoplasms/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...