1
|
Huang P, Huang Y, Dong T, Wang H, Wang M, Li X, Dong W, Yang Y, He W, Yang W. Mechanistic Insights Into GDFMD-Mediated Inhibition of Liver Fibrosis via miRNA-29b-3p Upregulation in Wilson's Disease. Mediators Inflamm 2025; 2025:2776808. [PMID: 40322065 PMCID: PMC12049248 DOI: 10.1155/mi/2776808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Accepted: 03/19/2025] [Indexed: 05/08/2025] Open
Abstract
Background: Wilson's disease (WD) is an abnormal copper metabolism disease. GanDouFuMu decoction (GDFMD) is a traditional Chinese medicine, whicn has shown good therapeutic effects in clinical treatment of WD liver fibrosis;but its regulatory mechanism is still unclear. Methods: The serum of WD patients before and after GDFMD treatment were collected, the four items of liver fibrosis were detected by ELISA. The hepatic stellate cell (HSC) activities were assesed via CCK8 assay. The mRNA levels were evaluated by qPCR. The protein levels were checked by western blot. The autophygosomes were observed by transmission electron microscope (TEM). The transdifferentiation ability of HSCs into myofibroblasts was evaluated with anti-α-SMA antibody by immunofluorescence (IF). In copper-laden rats with WD, the autophagy levels, and fibrosis level were observed by IF. Results: The four items of liver fibrosis levels were decreased. GDFMD could increase the HSCs cell activity. GDFMD could increase miRNA-29b-3p levels, which was decreased by TGF-β1. miRNA-29b-3p inhibitors could reversed the suppression response of GDFMD on the the protein expression of ULK1, beclin1, LC3, α-SMA, and Col1. GDFMD blocked the transdifferentiation of HSCs into myofibroblasts, inhibited liver fibrosis. Conclusion: GDFMD blocked the transdifferentiation of HSCs into myofibroblasts by upregulating miRNA-29b-3p, and then inhibited liver fibrosis in WD.
Collapse
Affiliation(s)
- Peng Huang
- Department of Neurology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
- Anhui University of Chinese Medicine Key Laboratory of Xin'an Medicine of the Ministry of Education, Hefei, Anhui, China
| | - Yuzhe Huang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Ting Dong
- Department of Neurology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
- Anhui University of Chinese Medicine Key Laboratory of Xin'an Medicine of the Ministry of Education, Hefei, Anhui, China
| | - Han Wang
- Department of Neurology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
- Anhui University of Chinese Medicine Key Laboratory of Xin'an Medicine of the Ministry of Education, Hefei, Anhui, China
| | - Meixia Wang
- Department of Neurology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
- Anhui University of Chinese Medicine Key Laboratory of Xin'an Medicine of the Ministry of Education, Hefei, Anhui, China
| | - Xiang Li
- Department of Neurology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
- Anhui University of Chinese Medicine Key Laboratory of Xin'an Medicine of the Ministry of Education, Hefei, Anhui, China
| | - Wei Dong
- Department of Neurology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
- Anhui University of Chinese Medicine Key Laboratory of Xin'an Medicine of the Ministry of Education, Hefei, Anhui, China
| | - Yulong Yang
- Department of Neurology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
- Anhui University of Chinese Medicine Key Laboratory of Xin'an Medicine of the Ministry of Education, Hefei, Anhui, China
| | - Wei He
- Department of Neurology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
- Anhui University of Chinese Medicine Key Laboratory of Xin'an Medicine of the Ministry of Education, Hefei, Anhui, China
| | - Wenming Yang
- Department of Neurology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
- Anhui University of Chinese Medicine Key Laboratory of Xin'an Medicine of the Ministry of Education, Hefei, Anhui, China
| |
Collapse
|
2
|
Medugorac M, Glick KM, Livun A, Lucijanic M, Galusic D, Kusec R. Gene Expression Analysis of Autophagy Markers in Primary and Secondary Myelofibrosis. J Clin Med 2025; 14:2333. [PMID: 40217782 PMCID: PMC11989297 DOI: 10.3390/jcm14072333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/19/2025] [Accepted: 03/25/2025] [Indexed: 04/14/2025] Open
Abstract
Background/Objectives: According to previous research, the process of autophagy in myeloid neoplasms has proven to be ambivalent depending on the type and stage of the disease. The aim of our work was to investigate the mechanism of autophagy in patients with primary and secondary myelofibrosis. Methods: Based on the RT-PCR method, we retrospectively analyzed the expression of Beclin-1 and LC3B-II in bone marrow cells of patients with primary and secondary myelofibrosis (74 participants) compared to the control group which had patients with lymphoma in a localized stage without bone marrow infiltration (11 participants). Results: There was no statistically significant difference in the expression of Beclin-1 and LC3B-II between patients with primary and secondary myelofibrosis and control participants. Among patients with primary myelofibrosis, higher expression of LC3B-II was statistically significantly associated with lower DIPSS. Higher Beclin-1 expression was statistically significantly associated with better patient survival. Conclusions: Our results suggest that the upregulation of autophagy genes may be associated with favorable prognosis and survival of patients with myelofibrosis.
Collapse
Affiliation(s)
- Marin Medugorac
- Division of Hematology, Department of Internal Medicine, University Hospital Centre Zagreb, 10000 Zagreb, Croatia
| | - Katarina Marija Glick
- Division of Molecular Diagnostics and Genetics, Department of Laboratory Diagnostics, University Hospital Dubrava, 10000 Zagreb, Croatia
| | - Ana Livun
- Division of Molecular Diagnostics and Genetics, Department of Laboratory Diagnostics, University Hospital Dubrava, 10000 Zagreb, Croatia
- Department of Scientific Research and Translational Medicine, University Hospital Dubrava, 10000 Zagreb, Croatia
| | - Marko Lucijanic
- Department of Scientific Research and Translational Medicine, University Hospital Dubrava, 10000 Zagreb, Croatia
- Division of Hematology, Department of Internal Medicine, University Hospital Dubrava, 10000 Zagreb, Croatia
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Davor Galusic
- Division of Hematology, Department of Internal Medicine, University Hospital Centre Split, 21000 Split, Croatia
- School of Medicine, University of Split, 21000 Split, Croatia
| | - Rajko Kusec
- Division of Molecular Diagnostics and Genetics, Department of Laboratory Diagnostics, University Hospital Dubrava, 10000 Zagreb, Croatia
- Department of Scientific Research and Translational Medicine, University Hospital Dubrava, 10000 Zagreb, Croatia
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| |
Collapse
|
3
|
Jiang Z, Huang H, Luo L, Jiang B. The Role of Autophagy on Osteogenesis of Dental Follicle Cells Under Inflammatory Microenvironment. Oral Dis 2025; 31:928-940. [PMID: 39415618 DOI: 10.1111/odi.15149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 09/05/2024] [Accepted: 09/19/2024] [Indexed: 10/19/2024]
Abstract
OBJECTIVE This study investigated the role of autophagy on osteogenesis of DFCs under inflammatory microenvironment during tooth eruption. METHODS DFCs were isolated and identified. Lipopolysaccharide (LPS) was used to construct the inflammatory microenvironment in vitro and in vivo. Cell viability was examined by CCK-8 assay. Osteogenic differentiation was evaluated by alkaline phosphatase (ALP) staining, alizarin red S (ARS) staining. The gene and protein levels were examined using qRT-PCR and western blot analysis, respectively. We observed the process of tooth eruption after local LPS injection by micro-CT and HE staining. Osteogenesis and autophagy were monitored through qRT-PCR, western blot and histological staining of specific markers. RESULTS LPS at the indicated concentrations did not produce toxic effects on DFCs, and significantly promoted the inflammatory gene expression. LPS inhibited osteogenic differentiation and activated autophagy in DFCs. Blocking autophagy with 3-MA reversed the expression of osteogenic markers in LPS-treated DFCs. Additionally, the eruption of LPS-treated teeth was accelerated and their DFs exhibited an increased expression of TNF-α and Beclin1, and decreased expression of ALP and RUNX2. CONCLUSIONS Autophagy was involved in the suppression of the DFCs osteogenesis in an LPS-induced inflammatory condition, suggesting the pivotal role of autophagy in inflammation-induced premature tooth eruption.
Collapse
Affiliation(s)
- Zhen Jiang
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Pediatric Dentistry, Stomatological Hospital and Dental School, Tongji University, Shanghai, China
| | - Haiyan Huang
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Pediatric Dentistry, Stomatological Hospital and Dental School, Tongji University, Shanghai, China
| | - Linjuan Luo
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Pediatric Dentistry, Stomatological Hospital and Dental School, Tongji University, Shanghai, China
| | - Beizhan Jiang
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Pediatric Dentistry, Stomatological Hospital and Dental School, Tongji University, Shanghai, China
| |
Collapse
|
4
|
Hoseinzadeh A, Esmaeili SA, Sahebi R, Melak AM, Mahmoudi M, Hasannia M, Baharlou R. Fate and long-lasting therapeutic effects of mesenchymal stromal/stem-like cells: mechanistic insights. Stem Cell Res Ther 2025; 16:33. [PMID: 39901306 PMCID: PMC11792531 DOI: 10.1186/s13287-025-04158-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 01/21/2025] [Indexed: 02/05/2025] Open
Abstract
A large body of evidence suggests that mesenchymal stromal cells (MSCs) are able to respond rapidly to the cytokine milieu following systemic infusion. This encounter has the potential to dictate their therapeutic efficacy (also referred to as licensing). MSCs are able to rapidly react to cellular damage by migrating to the inflamed tissue and ultimately modifying the inflammatory microenvironment. However, the limited use of MSCs in clinical practice can be attributed to a lack of understanding of the fate of MSCs in patients after administration and long term MSC-derived therapeutic activity. While the known physiological effectors of viable MSCs make a relative contribution, an innate property of MSCs as a therapeutic agent is their caspase-dependent cell death. These mechanisms may be involving the functional reprogramming of myeloid phagocytes via efferocytosis, the process by which apoptotic bodies (ABs) are identified for engulfment by both specialized and non-specialized phagocytic cells. Recent studies have provided evidence that the uptake of ABs with a distinct genetic component can induce changes in gene expression through the process of epigenetic remodeling. This phenomenon, known as 'trained immunity', has a significant impact on immunometabolism processes. It is hypothesized that the diversity of recipient cells within the inflammatory stroma adjacent to MSCs may potentially serve as a biomarker for predicting the clinical outcome of MSC treatment, while also contributing to the variable outcomes observed with MSC-based therapies. Therefore, the long-term reconstructive process of MSCs may potentially be mediated by MSC apoptosis and subsequent phagocyte-mediated efferocytosis.
Collapse
Affiliation(s)
- Akram Hoseinzadeh
- Department of Immunology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Seyed-Alireza Esmaeili
- Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reza Sahebi
- Department of Modern Sciences and Technologies, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Mahmoud Mahmoudi
- Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maliheh Hasannia
- Cancer Research Center, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Rasoul Baharlou
- Department of Immunology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran.
| |
Collapse
|
5
|
Zhang LY, Zhang K, Zhao X, Tao HP, Jia GX, Fang YG, Hou YP, Yang QE. Fetal hypoxia exposure induces Hif1a activation and autophagy in adult ovarian granulosa cells†. Biol Reprod 2024; 111:1220-1234. [PMID: 39361887 DOI: 10.1093/biolre/ioae141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 08/15/2024] [Accepted: 10/02/2024] [Indexed: 10/05/2024] Open
Abstract
Environmental hypoxia adversely impacts the reproduction of humans and animals. Previously, we showed that fetal hypoxia exposure led to granulosa cell (GC) autophagic cell death via the Foxo1/Pi3k/Akt pathway. However, the upstream regulatory mechanisms underlying GC dysfunction remain largely unexplored. Here, we tested the hypothesis that fetal hypoxia exposure altered gene expression programs in adult GCs and impaired ovarian function. We established a fetal hypoxia model in which pregnant mice were maintained in a high-plateau hypoxic environment from gestation day (E) 0-16.5 to study the impact of hypoxia exposure on the ovarian development and subsequent fertility of offspring. Compared with the unexposed control, fetal hypoxia impaired fertility by disordering ovarian function. Specifically, fetal hypoxia caused mitochondrial dysfunction, oxidant stress, and autophagy in GCs in the adult ovary. RNA sequencing analysis revealed that 437 genes were differentially expressed in the adult GCs of exposed animals. Western blotting results also revealed that fetal exposure induced high levels of hypoxia-inducible factor 1-alpha (Hif1a) expression in adult GCs. We then treated granulosa cells isolated from exposed mice with PX-478, a specific pharmacological inhibitor of Hif1a, and found that autophagy and apoptosis were effectively alleviated. Finally, by using a human ovarian granulosa-like tumor cell line (KGN) to simulate hypoxia in vitro, we showed that Hif1a regulated autophagic cell death in GCs through the Pi3k/Akt pathway. Together, these findings suggest that fetal hypoxia exposure induced persistent Hif1a expression, which impaired mitochondrial function and led to autophagic cell death in the GCs of the adult ovary.
Collapse
Affiliation(s)
- Lu-Yao Zhang
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Qinghai, China
| | - Ke Zhang
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Qinghai, China
| | - Xi Zhao
- Department of Animal Science, College of Animal Science, Xinjiang Agricultural University, Urumqi, China
| | - Hai-Ping Tao
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Qinghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Gong-Xue Jia
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Qinghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - You-Gui Fang
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Qinghai, China
| | - Yun-Peng Hou
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Qi-En Yang
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Qinghai, China
- University of Chinese Academy of Sciences, Beijing, China
- Qinghai Key Laboratory of Animal Ecological Genomics, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, Qinghai, China
| |
Collapse
|
6
|
Ryu Y, Seo JH, Kim HS, Nam YJ, Bo Noh K, Oh SH, Hwang JS, Shin YJ. COL8A2 activation enhances function of corneal endothelial cells through HIPPO signaling/mitochondria pathway. Matrix Biol 2024; 134:119-131. [PMID: 39395654 DOI: 10.1016/j.matbio.2024.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 09/24/2024] [Accepted: 10/02/2024] [Indexed: 10/14/2024]
Abstract
Corneal endothelial cells (CECs) are essential for maintaining corneal transparency and hydration through their barrier and pump functions. The COL8A2 gene encodes a component of the extracellular matrix of the cornea, which is crucial for the normal functioning of these cells. Mutations in COL8A2 are linked to corneal dystrophies, emphasizing the gene's importance in corneal health. The purpose of this research is to explore the effects of COL8A2 activation within CECs, to understand its contribution to cellular behavior and health. COL8A2 CRISPR/dCas9 activation system (aCOL8A2) was used to activate the COL8A2. In rats, wound healing and mitochondrial function were assessed after COL8A2 activation. As a result, aCOL8A2 promoted wound healing of rat corneal endothelium by increasing mitochondrial membrane potential. In cultured human CECs, proteomic analysis was performed to screen and identify the differential protein profiles between control and aCOL8A2 cells. Western blot was used to validate the differential proteins from both cells. Mitochondrial function and intracellular distribution were assessed by measuring ATP production and mitochondrial membrane potential. In cultured human CECs, aCOL8A2 increased COL8A2 and phospho-YAP levels. Transendothelial electrical resistance (TEER) was increased and actin cytoskeleton was attenuated by aCOL8A2. Gene ontology analysis revealed that the proteins were mainly involved in the regulation of folate biosynthesis, ECM-receptor interaction, cell differentiation, NADP activity and cytoskeleton. ATP production was increased, mitochondrial membrane potential was polarized and mitochondrial distribution was widespread in the aCOL8A2 group. In conclusion, aCOL8A2 induces a regulatory cascade affecting mitochondrial positioning and efficiency, mediated by alterations in the cytoskeletal architecture and the YAP signaling pathway. This sequence of events serves to bolster the functional capacities of corneal endothelial cells, including their pump and barrier functions, essential for corneal health and transparency.
Collapse
Affiliation(s)
- Yunkyoung Ryu
- Department of Ophthalmology, Hallym University Medical Center, Hallym University College of Medicine, Seoul, Republic of Korea; Hallym BioEyeTech Research Center, Hallym University College of Medicine, Seoul, Republic of Korea
| | - Je Hyun Seo
- Veterans Medical Research Institute, Veterans Health Service Medical Center, Seoul, Republic of Korea
| | - Hak Su Kim
- Veterans Medical Research Institute, Veterans Health Service Medical Center, Seoul, Republic of Korea
| | - Youn Joo Nam
- Department of Ophthalmology, Hallym University Medical Center, Hallym University College of Medicine, Seoul, Republic of Korea; Hallym BioEyeTech Research Center, Hallym University College of Medicine, Seoul, Republic of Korea
| | - Kyung Bo Noh
- Department of Ophthalmology, Hallym University Medical Center, Hallym University College of Medicine, Seoul, Republic of Korea; Hallym BioEyeTech Research Center, Hallym University College of Medicine, Seoul, Republic of Korea
| | - Sun-Hee Oh
- Department of Ophthalmology, Hallym University Medical Center, Hallym University College of Medicine, Seoul, Republic of Korea; Hallym BioEyeTech Research Center, Hallym University College of Medicine, Seoul, Republic of Korea
| | - Jin Sun Hwang
- Department of Ophthalmology, Hallym University Medical Center, Hallym University College of Medicine, Seoul, Republic of Korea
| | - Young Joo Shin
- Department of Ophthalmology, Hallym University Medical Center, Hallym University College of Medicine, Seoul, Republic of Korea; Hallym BioEyeTech Research Center, Hallym University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
7
|
Su J, Wang Y, Yao J, Sun L, Zhao C, Liu L, Zhang L. Systemic knockout of Tmem175 results in aberrant differentiation but no effect on hematopoietic reconstitution. Stem Cell Res 2024; 79:103469. [PMID: 38878670 DOI: 10.1016/j.scr.2024.103469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 06/06/2024] [Accepted: 06/09/2024] [Indexed: 08/04/2024] Open
Abstract
Lysosomes play crucial roles in regulating cell metabolism, and K+ channels are critical for controlling various aspects of lysosomal function. Additionally, lysosomal activity is essential for maintaining the quiescence of hematopoietic stem cells (HSCs) under both steady-state and stress conditions. Tmem175 is a lysosomal potassium channel protein. To further investigate the role of K+ channels in HSCs, our study employed knockout mice to examine the function of Tmem175. Our research findings demonstrate that the deletion of Tmem175 does not disrupt the functionality of HSCs in both stable and stressed conditions, including irradiation and intraperitoneal 5-FU injections. However, we did observe that the absence of Tmem175 impairs the long-term differentiation capacity of HSCs into myeloid differentiated subpopulation cells(In this paper, it is referred to simply as M cells)in HSC transplantation test, while promoting their differentiation into T cells. This suggests that Tmem175 plays a role in the lineage differentiation of HSCs without being essential for their self-renewal or long-term regenerative capabilities.
Collapse
Affiliation(s)
- Jingjing Su
- Key Laboratory of Molecular Pharmacology and Translational Medicine and Department of Pharmacology, College of Pharmacy, Shandong Second Medical University, Weifang , 261053, China
| | - Yue Wang
- Center for Metabolic Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Jiyuan Yao
- Center for Metabolic Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Leimin Sun
- Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310016, China
| | - Chunzhen Zhao
- Key Laboratory of Molecular Pharmacology and Translational Medicine and Department of Pharmacology, College of Pharmacy, Shandong Second Medical University, Weifang , 261053, China.
| | - Leiming Liu
- Key Laboratory of Artificial Organs and Computational Medicine, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou, Zhejiang, 310015, China.
| | - Lingling Zhang
- Center for Metabolic Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China.
| |
Collapse
|
8
|
Kovale L, Singh MK, Kim J, Ha J. Role of Autophagy and AMPK in Cancer Stem Cells: Therapeutic Opportunities and Obstacles in Cancer. Int J Mol Sci 2024; 25:8647. [PMID: 39201332 PMCID: PMC11354724 DOI: 10.3390/ijms25168647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/30/2024] [Accepted: 08/07/2024] [Indexed: 09/02/2024] Open
Abstract
Cancer stem cells represent a resilient subset within the tumor microenvironment capable of differentiation, regeneration, and resistance to chemotherapeutic agents, often using dormancy as a shield. Their unique properties, including drug resistance and metastatic potential, pose challenges for effective targeting. These cells exploit certain metabolic processes for their maintenance and survival. One of these processes is autophagy, which generally helps in energy homeostasis but when hijacked by CSCs can help maintain their stemness. Thus, it is often referred as an Achilles heel in CSCs, as certain cancers tend to depend on autophagy for survival. Autophagy, while crucial for maintaining stemness in cancer stem cells (CSCs), can also serve as a vulnerability in certain contexts, making it a complex target for therapy. Regulators of autophagy like AMPK (5' adenosine monophosphate-activated protein kinase) also play a crucial role in maintaining CSCs stemness by helping CSCs in metabolic reprogramming in harsh environments. The purpose of this review is to elucidate the interplay between autophagy and AMPK in CSCs, highlighting the challenges in targeting autophagy and discussing therapeutic strategies to overcome these limitations. This review focuses on previous research on autophagy and its regulators in cancer biology, particularly in CSCs, addresses the remaining unanswered questions, and potential targets for therapy are also brought to attention.
Collapse
Affiliation(s)
- Lochana Kovale
- Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (L.K.); (M.K.S.)
| | - Manish Kumar Singh
- Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (L.K.); (M.K.S.)
| | - Joungmok Kim
- Department of Oral Biochemistry and Molecular Biology, College of Dentistry, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Joohun Ha
- Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (L.K.); (M.K.S.)
| |
Collapse
|
9
|
Xu X, Wang J, Xia Y, Yin Y, Zhu T, Chen F, Hai C. Autophagy, a double-edged sword for oral tissue regeneration. J Adv Res 2024; 59:141-159. [PMID: 37356803 PMCID: PMC11081970 DOI: 10.1016/j.jare.2023.06.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 06/10/2023] [Accepted: 06/20/2023] [Indexed: 06/27/2023] Open
Abstract
BACKGROUND Oral health is of fundamental importance to maintain systemic health in humans. Stem cell-based oral tissue regeneration is a promising strategy to achieve the recovery of impaired oral tissue. As a highly conserved process of lysosomal degradation, autophagy induction regulates stem cell function physiologically and pathologically. Autophagy activation can serve as a cytoprotective mechanism in stressful environments, while insufficient or over-activation may also lead to cell function dysregulation and cell death. AIM OF REVIEW This review focuses on the effects of autophagy on stem cell function and oral tissue regeneration, with particular emphasis on diverse roles of autophagy in different oral tissues, including periodontal tissue, bone tissue, dentin pulp tissue, oral mucosa, salivary gland, maxillofacial muscle, temporomandibular joint, etc. Additionally, this review introduces the molecular mechanisms involved in autophagy during the regeneration of different parts of oral tissue, and how autophagy can be regulated by small molecule drugs, biomaterials, exosomes/RNAs or other specific treatments. Finally, this review discusses new perspectives for autophagy manipulation and oral tissue regeneration. KEY SCIENTIFIC CONCEPTS OF REVIEW Overall, this review emphasizes the contribution of autophagy to oral tissue regeneration and highlights the possible approaches for regulating autophagy to promote the regeneration of human oral tissue.
Collapse
Affiliation(s)
- Xinyue Xu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases and Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, PR China; Shaanxi Key Lab of Free Radical Biology and Medicine, Fourth Military Medical University, Xi'an, PR China
| | - Jia Wang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases and Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, PR China
| | - Yunlong Xia
- Shaanxi Key Lab of Free Radical Biology and Medicine, Fourth Military Medical University, Xi'an, PR China; Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, PR China
| | - Yuan Yin
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases and Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, PR China
| | - Tianxiao Zhu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases and Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, PR China; Shaanxi Key Lab of Free Radical Biology and Medicine, Fourth Military Medical University, Xi'an, PR China
| | - Faming Chen
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases and Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, PR China
| | - Chunxu Hai
- Shaanxi Key Lab of Free Radical Biology and Medicine, Fourth Military Medical University, Xi'an, PR China.
| |
Collapse
|
10
|
He Q, Zheng Q, Liu Y, Miao Y, Zhang Y, Xu T, Bai S, Zhao X, Yang X, Xu Z. High-Salt Diet Causes Defective Oocyte Maturation and Embryonic Development to Impair Female Fertility in Mice. Mol Nutr Food Res 2023; 67:e2300401. [PMID: 37863820 DOI: 10.1002/mnfr.202300401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 09/05/2023] [Indexed: 10/22/2023]
Abstract
SCOPE High salinity has been reported to induce many human disorders in tissues and organs to interfere with their normal physiological functions. However, it is unknown how salinity affects the development of female germ cells. This study suggests that a high-salt diet (HSD) may weaken oocyte quality to impair female fertility in mice and investigates the underlying mechanisms. METHODS AND RESULTS C57BL/6 female mice are fed with a regular diet (Control) or a high-salt diet (HSD). Oocyte maturation, fertilization rate, embryonic development, and female fertility are evaluated. In addition, the spindle organization, actin polymerization, and kinetochore-microtubule attachment of oocytes are examined in both groups. Moreover, single-cell transcriptome data are used to demonstrate how HSD alters the transcript levels of genes. The observations confirm that HSD leads to female subfertility due to the deterioration of oocyte and embryo quality. The mechanism underlying reveals HSD compromises the oocytes' autophagy, apoptosis level, and mitochondrial function. CONCLUSION The work illustrates that a high concentration of salt diet results in oocyte meiotic arrest, fertilization failure, and early developmental defection that embryos undergo to reduce female fertility in mice by perturbing the level of autophagy and apoptosis, mitochondrial function in oocytes.
Collapse
Affiliation(s)
- Qinyuan He
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
- Department of Obstetrics and Gynecology, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210003, China
| | - Qiutong Zheng
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
- Maternal and Child Health Care Hospital of Wuxi, Wuxi, Jiangsu, 214002, China
| | - Yanping Liu
- Center of Reproduction and Genetics, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, 215006, China
| | - Yilong Miao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, China
| | - Yumeng Zhang
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Ting Xu
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Shufen Bai
- Department of Obstetrics and Gynecology, Nanjing Pukou District Hospital of Traditional Chinese Medicine, Nanjing, Jiangsu, 210000, China
| | - Xia Zhao
- Department of Reproductive Medicine, Zhongda Hospital Affiliated to Southeast University, Nanjing, Jiangsu, 210009, China
| | - Xiaojun Yang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Zhice Xu
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
- Maternal and Child Health Care Hospital of Wuxi, Wuxi, Jiangsu, 214002, China
| |
Collapse
|
11
|
Puri D, Kelkar A, Gaurishankar B, Subramanyam D. Balance between autophagy and cell death is maintained by Polycomb-mediated regulation during stem cell differentiation. FEBS J 2023; 290:1625-1644. [PMID: 36380631 DOI: 10.1111/febs.16656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 08/23/2022] [Accepted: 10/17/2022] [Indexed: 11/17/2022]
Abstract
Autophagy is a conserved cytoprotective process, aberrations in which lead to numerous degenerative disorders. While the cytoplasmic components of autophagy have been extensively studied, the epigenetic regulation of autophagy genes, especially in stem cells, is less understood. Deciphering the epigenetic regulation of autophagy genes becomes increasingly relevant given the therapeutic benefits of small-molecule epigenetic inhibitors in novel treatment modalities. We observe that, during retinoic acid-mediated differentiation of mouse embryonic stem cells (mESCs), autophagy is induced, and identify the Polycomb group histone methyl transferase EZH2 as a regulator of this process. In mESCs, EZH2 represses several autophagy genes, including the autophagy regulator DNA damage-regulated autophagy modulator protein 1 (Dram1). EZH2 facilitates the formation of a bivalent chromatin domain at the Dram1 promoter, allowing gene expression and autophagy induction during differentiation while retaining the repressive H3K27me3 mark. EZH2 inhibition leads to loss of the bivalent domain, with consequent 'hyper-expression' of Dram1, accompanied by extensive cell death. This study shows that Polycomb group proteins help maintain a balance between autophagy and cell death during stem cell differentiation, in part, by regulating the expression of the Dram1 gene.
Collapse
Affiliation(s)
- Deepika Puri
- National Centre for Cell Science, SP Pune University, India
| | - Aparna Kelkar
- National Centre for Cell Science, SP Pune University, India
| | | | | |
Collapse
|
12
|
Xing Y, Liu C, Zhou L, Li Y, Wu D. Osteogenic effects of rapamycin on bone marrow mesenchymal stem cells via inducing autophagy. J Orthop Surg Res 2023; 18:129. [PMID: 36814286 PMCID: PMC9945701 DOI: 10.1186/s13018-023-03616-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 02/14/2023] [Indexed: 02/24/2023] Open
Abstract
BACKGROUND While autophagy is essential for stem cells' self-renewal and differentiation, its effect on bone marrow mesenchymal stem cells (BMSCs) remains unclear. This study aimed to investigate the interaction between autophagy and osteogenic differentiation using rapamycin (RAPA), a classical autophagy agonist with osteo-regulatory effects. METHODS Rat BMSC's autophagy was analyzed after osteoinduction (0, 7, 14, and 21 d) by western blotting, immunofluorescence, and real-time quantitative polymerase chain reaction (RT-qPCR). In addition, we evaluated osteogenic differentiation using alizarin red staining, alkaline phosphatase assays, and RT-qPCR/Western blotting quantification of bone sialoprotein, type 1 collagen, alkaline phosphatase, osteopontin, and Runt-related transcription factor 2 mRNA and protein levels. RESULTS The BMSC's basal autophagy level gradually decreased during osteogenic differentiation with a decrease in BECN1 level and the lipidated (LC3-II) to unlipidated (LC3-I) microtubule-associated protein 1 light chain 3 ratio and an increase in the expression of selective autophagic target p62. In contrast, it increased with increasing RAPA concentration. Furthermore, while 2 nM RAPA promoted BMSC osteogenic differentiation on days 7 and 14, 5 nM RAPA inhibited osteogenesis on days 14 and 21. Inhibition of autophagy by the inhibitor 3-methyladenine could impair RAPA's osteogenesis-enhancing effect on BMSCs. CONCLUSIONS The BMSC's basal autophagy level decreased over time during osteogenic differentiation. However, an appropriate RAPA concentration promoted BMSC osteogenic differentiation via autophagy activation.
Collapse
Affiliation(s)
- Yifeng Xing
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Institute of Stomatology & Research Center of Dental and Craniofacial Implants, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Chaowei Liu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Lin Zhou
- Institute of Stomatology & Research Center of Dental and Craniofacial Implants, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Yan Li
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Institute of Stomatology & Research Center of Dental and Craniofacial Implants, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Dong Wu
- Institute of Stomatology & Research Center of Dental and Craniofacial Implants, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
13
|
Hasan KMM, Haque MA. Autophagy and Its Lineage-Specific Roles in the Hematopoietic System. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:8257217. [PMID: 37180758 PMCID: PMC10171987 DOI: 10.1155/2023/8257217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 02/26/2023] [Accepted: 03/17/2023] [Indexed: 05/16/2023]
Abstract
Autophagy is a dynamic process that regulates the selective and nonselective degradation of cytoplasmic components, such as damaged organelles and protein aggregates inside lysosomes to maintain tissue homeostasis. Different types of autophagy including macroautophagy, microautophagy, and chaperon-mediated autophagy (CMA) have been implicated in a variety of pathological conditions, such as cancer, aging, neurodegeneration, and developmental disorders. Furthermore, the molecular mechanism and biological functions of autophagy have been extensively studied in vertebrate hematopoiesis and human blood malignancies. In recent years, the hematopoietic lineage-specific roles of different autophagy-related (ATG) genes have gained more attention. The evolution of gene-editing technology and the easy access nature of hematopoietic stem cells (HSCs), hematopoietic progenitors, and precursor cells have facilitated the autophagy research to better understand how ATG genes function in the hematopoietic system. Taking advantage of the gene-editing platform, this review has summarized the roles of different ATGs at the hematopoietic cell level, their dysregulation, and pathological consequences throughout hematopoiesis.
Collapse
Affiliation(s)
- Kazi Md Mahmudul Hasan
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
- Department of Biotechnology and Genetic Engineering, Islamic University, Kushtia 7003, Bangladesh
- Department of Neurology, David Geffen School of Medicine, The University of California, 710 Westwood Plaza, Los Angeles, CA 90095, USA
| | - Md Anwarul Haque
- Department of Biotechnology and Genetic Engineering, Islamic University, Kushtia 7003, Bangladesh
| |
Collapse
|
14
|
Xiong Y, Chen T, Chen L, Cai R. Gold Nanoparticles Coated with SH-PEG-NH 2 and Loaded with Ziyuglycoside I for Promoting Autophagy in Hematopoietic Stem Cells. Int J Nanomedicine 2023; 18:1347-1362. [PMID: 36974074 PMCID: PMC10039662 DOI: 10.2147/ijn.s399568] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 03/14/2023] [Indexed: 03/29/2023] Open
Abstract
Introduction Radiotherapy and chemotherapy are the fundamental causes of myelosuppression in cancer patients, which usually induce a serious hematopoietic system toxicity, causing the hemocytes and immunity decline of patients. Ziyuglycoside I (ZgI), an active ingredient isolated from traditional Chinese medicine Sanguisorba officinalis L, has been demonstrated to increase the leucocytes and protect hematopoietic stem cells, which is related to its promotion of autophagy in hematopoietic stem cells. Methods In the present study, we formulated the SH-PEG-NH2-coated gold nanoparticles loading ZgI (ZgI-AuNPs) with a enhanced autophagy promotion in hematopoietic stem cells. ZgI-AuNPs were prepared by HAuCl4-sodium citrate reduction method, and the synthesis of ZgI-AuNPs was validated by XRD, FT-IR, DSC, and TEM findings. Furthermore, the drug loading rate and the release of ZgI were evaluated, and the ZgI-AuNPs' effects on autophagy and immunofluorescence staining for LC3B were tested. Finally, the effect of ZgI-AuNPs on the autophagy and hematopoietic ability of HSCs in vivo was also carried out. Results The prepared ZgI-AuNPs have an irregular cubic crystal structure by TEM observation, and the average particle size was 340 ± 16.5 nm determined by DLS. The XRD, FT-IR and DSC detection showed that the ZgI had been well loaded in AuNPs, and the AuNPs can load the ZgI at a content of 160.63 ± 1.35 μg·mg-1. Meanwhile, the AuNPs can reduce the drug release rate of ZgI. Importantly, the ZgI-AuNPs enhanced autophagy of HSCs both in vitro and in vivo. At the same time, the gold nanoparticles enhance the hematopoietic effect of ZgI on mice HSCs. Conclusion Our research suggests that SH-PEG-NH2-coated gold nanoparticles loading ZgI has potential application in myelosuppression therapy.
Collapse
Affiliation(s)
- Yongai Xiong
- Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, 563000, People’s Republic of China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, 563000, People’s Republic of China
- Correspondence: Yongai Xiong, Email
| | - Tingting Chen
- Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, 563000, People’s Republic of China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, 563000, People’s Republic of China
| | - Lei Chen
- Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, 563000, People’s Republic of China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, 563000, People’s Republic of China
| | - Rongshan Cai
- Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, 563000, People’s Republic of China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, 563000, People’s Republic of China
| |
Collapse
|
15
|
Ding Y, Zhao J, Xu X, Zuo Q, Zhang Y, Jin K, Han W, Li B. Inhibition of Autophagy Maintains ESC Pluripotency and Inhibits Primordial Germ Cell Formation in Chickens. Stem Cells Int 2023; 2023:4956871. [PMID: 37056458 PMCID: PMC10089774 DOI: 10.1155/2023/4956871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 02/28/2023] [Accepted: 03/04/2023] [Indexed: 04/15/2023] Open
Abstract
Autophagy plays an important role in the pluripotency and differentiation of stem cells. Transcriptome data showed that the autophagy genes MAP1LC3A and MAP1LC3B were significantly upregulated in primordial germ cells (PGCs). The Kyoto Encyclopedia of Genes and Genome (KEGG) results showed that the lysosome signaling pathway, which is related to autophagy, was significantly enriched in PGCs. Quantitative RT-PCR, western blotting, and transmission electron microscopy (TEM) results showed that autophagy was expressed in both embryonic stem cells (ESCs) and PGCs but was significantly activated in PGCs. To explore the role of autophagy in the differentiation of chicken ESCs into PGCs, autophagy was activated and inhibited using rapamycin and bafilomycin A1, respectively. Results of qRT-PCR, flow cytometry, and indirect immunofluorescence showed that the efficiency of PGC formation significantly decreased after autophagy inhibition. Our results showed, for the first time, that autophagy plays an indispensable role in the formation of chicken PGCs, which lays the foundation for studying the mechanism of autophagy in chicken PGCs and in bird gene editing and the rescue of endangered birds.
Collapse
Affiliation(s)
- Ying Ding
- Key Laboratory of Animal Genetics, Breeding and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Juanjuan Zhao
- Key Laboratory of Animal Genetics, Breeding and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Xianshuai Xu
- Key Laboratory of Animal Genetics, Breeding and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Qisheng Zuo
- Key Laboratory of Animal Genetics, Breeding and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Yani Zhang
- Key Laboratory of Animal Genetics, Breeding and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Kai Jin
- Key Laboratory of Animal Genetics, Breeding and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Wei Han
- Poultry Research Institute, Chinese Academy of Agricultural Science/Jiangsu Institute of Poultry Science, Yangzhou 225009, China
| | - Bichun Li
- Key Laboratory of Animal Genetics, Breeding and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| |
Collapse
|
16
|
Autophagy in Hematological Malignancies. Cancers (Basel) 2022; 14:cancers14205072. [PMID: 36291856 PMCID: PMC9600546 DOI: 10.3390/cancers14205072] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 10/10/2022] [Accepted: 10/13/2022] [Indexed: 11/29/2022] Open
Abstract
Simple Summary Autophagy is a dynamic and tightly regulated process that seems to have dual effects in cancer. In some contexts, it can induce carcinogenesis and promote cancer cell survival, whereas in others, it acts preventing tumor cell growth and tumor progression. Thus, autophagy functions seem to strictly depend on cancer ontogenesis, progression, and type. Here, we will dive into the current knowledge of autophagy in hematological malignancies and will highlight the main genetic components involved in each cancer type. Abstract Autophagy is a highly conserved metabolic pathway via which unwanted intracellular materials, such as unfolded proteins or damaged organelles, are digested. It is activated in response to conditions of oxidative stress or starvation, and is essential for the maintenance of cellular homeostasis and other vital functions, such as differentiation, cell death, and the cell cycle. Therefore, autophagy plays an important role in the initiation and progression of tumors, including hematological malignancies, where damaged autophagy during hematopoiesis can cause malignant transformation and increase cell proliferation. Over the last decade, the importance of autophagy in response to standard pharmacological treatment of hematological tumors has been observed, revealing completely opposite roles depending on the tumor type and stage. Thus, autophagy can promote tumor survival by attenuating the cellular damage caused by drugs and/or stabilizing oncogenic proteins, but can also have an antitumoral effect due to autophagic cell death. Therefore, autophagy-based strategies must depend on the context to create specific and safe combination therapies that could contribute to improved clinical outcomes. In this review, we describe the process of autophagy and its role on hematopoiesis, and we highlight recent research investigating its role as a potential therapeutic target in hematological malignancies. The findings suggest that genetic variants within autophagy-related genes modulate the risk of developing hemopathies, as well as patient survival.
Collapse
|
17
|
Zhai C, Zhang N, Wang J, Cao M, Luan J, Liu H, Zhang Q, Zhu Y, Xue Y, Li S. Activation of Autophagy Induces Monocrotaline-Induced Pulmonary Arterial Hypertension by FOXM1-Mediated FAK Phosphorylation. Lung 2022; 200:619-631. [PMID: 36107242 DOI: 10.1007/s00408-022-00569-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 09/01/2022] [Indexed: 11/28/2022]
Abstract
PURPOSE It has been shown that activation of autophagy promotes the development of pulmonary arterial hypertension (PAH). Meanwhile, forkhead box M1 (FOXM1) has been found to induce autophagy in several types of cancer. However, it is still unclear whether FOXM1 mediates autophagy activation in PAH, and detailed mechanisms responsible for these processes are indefinite. METHOD PAH was induced by a single intraperitoneal injection of monocrotaline (MCT) to rats. The right ventricle systolic pressure (RVSP), right ventricular hypertrophy index (RVHI), percentage of medial wall thickness (%MT), α-smooth muscle actin (α-SMA) staining, and Ki67 staining were performed to evaluate the development of PAH. The protein levels of FOXM1, phospho-focal adhesion kinase (p-FAK), FAK, and LC3B were determined by immunoblotting or immunohistochemistry. RESULTS FOXM1 protein level and FAK activity were significantly increased in MCT-induced PAH rats, this was accompanied with the activation of autophagy. Pharmacological inhibition of FOXM1 or FAK suppressed MCT-induced autophagy activation, decreased RVSP, RVHI and %MT in MCT-induced PAH rats, and inhibited the proliferation of pulmonary arterial smooth muscle cells and pulmonary vessel muscularization in MCT-induced PAH rats. CONCLUSION FOXM1 promotes the development of PAH by inducing FAK phosphorylation and subsequent activation of autophagy in MCT-treated rats.
Collapse
Affiliation(s)
- Cui Zhai
- Shaanxi Key Laboratory of Brain Disorders, Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, 710021, Shaanxi, People's Republic of China
| | - Nana Zhang
- Center for Regenerative and Reconstructive Medicine, Med-X Institute of the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Jian Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No.277, West Yanta Road, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Meng Cao
- Shaanxi Key Laboratory of Brain Disorders, Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, 710021, Shaanxi, People's Republic of China
| | - Jing Luan
- Shaanxi Key Laboratory of Brain Disorders, Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, 710021, Shaanxi, People's Republic of China
| | - Huan Liu
- Shaanxi Key Laboratory of Brain Disorders, Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, 710021, Shaanxi, People's Republic of China
| | - Qianqian Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No.277, West Yanta Road, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Yanting Zhu
- Center of Nephropathy and Hemodialysis, Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi, People's Republic of China
| | - Yuxin Xue
- Shaanxi Key Laboratory of Brain Disorders, Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, 710021, Shaanxi, People's Republic of China
| | - Shaojun Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No.277, West Yanta Road, Xi'an, 710061, Shaanxi, People's Republic of China.
| |
Collapse
|
18
|
Zhang Y, Liu W, Yuan W, Cai Z, Ye G, Zheng G, Xu C, Wang X, Zeng C, Mi R, Feng P, Chen F, Wu Y, Shen H, Wang P. Impairment of APPL1/Myoferlin facilitates adipogenic differentiation of mesenchymal stem cells by blocking autophagy flux in osteoporosis. Cell Mol Life Sci 2022; 79:488. [PMID: 35984564 PMCID: PMC9391247 DOI: 10.1007/s00018-022-04511-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 07/21/2022] [Accepted: 08/02/2022] [Indexed: 11/30/2022]
Abstract
An imbalance of human mesenchymal stem cells (hMSCs) adipogenic and osteogenic differentiation is crucial in the pathogenesis of osteoporosis, and elucidation of the underlying mechanism is urgently needed. APPL1, an adaptor protein of the adiponectin receptor, was recently shown to be closely related to bone mass. However, the role of APPL1 in the imbalance of hMSC differentiation in osteoporosis is unclear. Therefore, we aimed to explore the mechanisms by which APPL1 alters hMSCs adipogenic differentiation in osteoporosis. Here, we found that APPL1 expression was downregulated in elderly patients with osteoporosis and in mouse osteoporosis model. APPL1 negatively regulated hMSC adipogenic differentiation in vivo and in vitro. Mechanistically, by enhancing ubiquitination-mediated Myoferlin degradation, downregulated APPL1 expression increased the risk of lysosome dysfunction during hMSCs adipogenic differentiation. Lysosomal dysfunction inhibited autophagy flux by suppressing autophagosome degradation and promoted hMSC differentiation towards the adipocyte lineage. Our findings suggest that APPL1/Myoferlin downregulation promoted hMSCs adipogenic differentiation by inhibiting autophagy flux, further impairing the balance of hMSCs adipogenic and osteogenic differentiation in osteoporosis; the APPL1/ Myoferlin axis may be a promising diagnostic and therapeutic target for osteoporosis.
Collapse
Affiliation(s)
- Yunhui Zhang
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-Sen University, 3025# Shennan Road, Shenzhen, 518000, People's Republic of China
| | - Wenjie Liu
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-Sen University, 3025# Shennan Road, Shenzhen, 518000, People's Republic of China
| | - Weiquan Yuan
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-Sen University, 3025# Shennan Road, Shenzhen, 518000, People's Republic of China
| | - Zhaopeng Cai
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-Sen University, 3025# Shennan Road, Shenzhen, 518000, People's Republic of China
| | - Guiwen Ye
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-Sen University, 3025# Shennan Road, Shenzhen, 518000, People's Republic of China
| | - Guan Zheng
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-Sen University, 3025# Shennan Road, Shenzhen, 518000, People's Republic of China
| | - Chenhao Xu
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-Sen University, 3025# Shennan Road, Shenzhen, 518000, People's Republic of China
| | - Xinglang Wang
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-Sen University, 3025# Shennan Road, Shenzhen, 518000, People's Republic of China
| | - Chenying Zeng
- Center for Biotherapy, The Eighth Affiliated Hospital, Sun Yat-Sen University, 3025# Shennan Road, Shenzhen, 518000, People's Republic of China
| | - Rujia Mi
- Center for Biotherapy, The Eighth Affiliated Hospital, Sun Yat-Sen University, 3025# Shennan Road, Shenzhen, 518000, People's Republic of China
| | - Pei Feng
- Center for Biotherapy, The Eighth Affiliated Hospital, Sun Yat-Sen University, 3025# Shennan Road, Shenzhen, 518000, People's Republic of China
| | - Fenglei Chen
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-Sen University, 3025# Shennan Road, Shenzhen, 518000, People's Republic of China
| | - Yanfeng Wu
- Center for Biotherapy, The Eighth Affiliated Hospital, Sun Yat-Sen University, 3025# Shennan Road, Shenzhen, 518000, People's Republic of China.
| | - Huiyong Shen
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-Sen University, 3025# Shennan Road, Shenzhen, 518000, People's Republic of China.
| | - Peng Wang
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-Sen University, 3025# Shennan Road, Shenzhen, 518000, People's Republic of China.
| |
Collapse
|
19
|
Yin Y, Tian BM, Li X, Yu YC, Deng DK, Sun LJ, Qu HL, Wu RX, Xu XY, Sun HH, An Y, He XT, Chen FM. Gold nanoparticles targeting the autophagy-lysosome system to combat the inflammation-compromised osteogenic potential of periodontal ligament stem cells: From mechanism to therapy. Biomaterials 2022; 288:121743. [PMID: 36030103 DOI: 10.1016/j.biomaterials.2022.121743] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 08/02/2022] [Accepted: 08/09/2022] [Indexed: 12/09/2022]
Abstract
Although substantial data indicate that the osteogenic potential of periodontal ligament stem cells (PDLSCs) is compromised under inflammatory conditions, the underlying mechanism remains largely unexplored. In this study, we found that both the autophagy levels and autophagic flux levels were decreased in PDLSCs incubated under inflammatory conditions (I-PDLSCs). Based on the increased expression of LC3 II (at an autophagy level) and decreased accumulation of LC3 II (at an autophagic flux level) in I-PDLSCs, we speculated that the disruption of I-PDLSC autophagy arose from dysfunction of the cellular autophagy-lysosome system. Subsequently, our hypothesis was demonstrated by inhibited autophagosome-lysosome fusion, damaged lysosomal function, and suppressed activation of transcription factor EB (TFEB, a master regulator of the autophagy-lysosome system) in I-PDLSCs and verified by TFEB overexpression in I-PDLSCs. We found that gold nanoparticle (Au NP) treatment rescued the osteogenic potential of I-PDLSCs by restoring the inflammation-compromised autophagy-lysosome system. In this context, Au NP ceased to be effective when TFEB was knocked down in PDLSCs. Our data demonstrate the crucial role of the autophagy-lysosome system in cellular osteogenesis under inflammatory conditions and suggest a new target for rescuing inflammation-induced cell dysfunction using nanomaterials to aid cell biology and tissue regeneration.
Collapse
Affiliation(s)
- Yuan Yin
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Clinical Research Center for Oral Diseases, Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Bei-Min Tian
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Clinical Research Center for Oral Diseases, Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Xuan Li
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Clinical Research Center for Oral Diseases, Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Yao-Cheng Yu
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Dao-Kun Deng
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Clinical Research Center for Oral Diseases, Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Li-Juan Sun
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Clinical Research Center for Oral Diseases, Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Hong-Lei Qu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Clinical Research Center for Oral Diseases, Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Rui-Xin Wu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Clinical Research Center for Oral Diseases, Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Xin-Yue Xu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Clinical Research Center for Oral Diseases, Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Hai-Hua Sun
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Clinical Research Center for Oral Diseases, Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Ying An
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Clinical Research Center for Oral Diseases, Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Xiao-Tao He
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Clinical Research Center for Oral Diseases, Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.
| | - Fa-Ming Chen
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Clinical Research Center for Oral Diseases, Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.
| |
Collapse
|
20
|
Park SY, Cho HS, Chung KH, Lee BN, Kim SH, Kim WJ, Jung JY. Inactivation of PI3K/Akt promotes the odontoblastic differentiation and suppresses the stemness with autophagic flux in dental pulp cells. J Dent Sci 2022; 17:145-154. [PMID: 35028032 PMCID: PMC8739242 DOI: 10.1016/j.jds.2021.05.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/06/2021] [Indexed: 12/16/2022] Open
Abstract
Background/purpose Autophagy is involved in controlling differentiation of various cell types. The present study aimed to investigate the mechanism related to autophagy in regulating odontogenic differentiation of dental pulp cells. Materials and methods Human dental pulp cells (HDPCs) were cultured in differentiation inductive medium (DM) and odontoblastic differentiation and mineralization were evaluated by alkaline phosphatase (ALP) staining and Alizarin red S staining, respectively. Tooth cavity preparation was made on the mesial surface of lower first molars in rat. The expression of autophagy-related signal molecules was detected using Western blot analysis and Immunohistochemistry. Results HDPCs cultured in DM showed increased autophagic flux and declined phosphorylation of phosphoinositide 3-kinases (PI3K), protein kinase B (Akt), and mTOR. Dentin matrix protein-1 (DMP-1) and dentin sialoprotein (DSP), markers of odontoblastic differentiation, were upregulated and autophagic activation showing increased LC3-II and decreased p62 levels was observed during odontogenic differentiation of HDPCs. However, PI3K blocker 3-methyladenine (3MA), lentiviral shLC3 and Akt activator SC79 attenuated the expression of LC3II as well as DMP-1, ALP activity and mineralization enhanced in HDPCs under DM condition. In addition, 3MA, shLC3 and SC79 recovered the expression of pluripotency factor CD146, Oct4 and Nanog downregulated in DM condition. In rat tooth cavity preparation model, the expression of LC3B and DMP-1 was elevated near odontoblast-dentin layer during reparative dentin formation, whereas 3MA significantly reduced the expression of LC3B and DMP-1. Conclusion These findings indicated autophagy promotes the odontogenic differentiation of dental pulp cells modulating stemness via PI3K/Akt inactivation and the repair of pulp.
Collapse
Affiliation(s)
- Sam Young Park
- Department of Oral Physiology, Dental Science Research Institute, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| | - Heui Seung Cho
- Department of Oral Physiology, Dental Science Research Institute, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| | - Kyung Hwun Chung
- Dental Research Institute, School of Dentistry, Seoul National University, Seoul, Republic of Korea
| | - Bin Na Lee
- Department of Operative Dentistry, Dental Science Research Institute, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| | - Sun Hun Kim
- Department of Oral Anatomy, Dental Science Research Institute, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| | - Won Jae Kim
- Department of Oral Physiology, Dental Science Research Institute, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| | - Ji Yeon Jung
- Department of Oral Physiology, Dental Science Research Institute, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| |
Collapse
|
21
|
Extracellular IL-37 promotes osteogenic and odontogenic differentiation of human dental pulp stem cells via autophagy. Exp Cell Res 2021; 407:112780. [PMID: 34411610 DOI: 10.1016/j.yexcr.2021.112780] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 08/02/2021] [Accepted: 08/07/2021] [Indexed: 01/08/2023]
Abstract
The osteogenic and odontogenic differentiation of dental pulp stem cells (DPSCs) contribute to restoration and regeneration of dental tissue. Previous study indicated that interleukin-37 (IL-37) was an anti-inflammatory factor that affected other pro-inflammatory signals. The aim of this study was to explore the effects of IL-37 on the differentiation of DPSCs. DPSCs were cultured in growth medium with different concentrations of IL-37. We selected the optimal concentration for the following experiments by alkaline phosphatase (ALP) activity analysis, quantitative reverse-transcription polymerase chain reaction (qRT-PCR) and Western blot. Cell counting kit assay (CCK-8) and 5-Ethynyl-2'-Deoxyuridine (EdU) assay were conducted to assess the effects of IL-37 on the proliferation of DPSCs. ALP activity assay and staining, alizarin red S (ARS) staining, qRT-PCR, Western blot as well as immunofluorescence staining were conducted to assess differentiation ability of DPSCs. Western blot, immunofluorescence staining and transmission electron microscopy (TEM) were utilized to examine cell autophagy. Results showed that IL-37 enhanced the osteogenic and odontogenic differentiation ability of DPSCs with no significant influence on the proliferation of DPSCs. Autophagy in DPSCs was activated by IL-37. Activation of autophagy enhanced osteogenesis and odontogenesis of DPSCs, whereas inhibition of autophagy suppressed DPSCs osteogenic and odontogenic differentiation. In conclusion, IL-37 increased osteogenic and odontogenic differentiation via autophagy.
Collapse
|
22
|
Gunay B, Goncu E. Role of autophagy in midgut stem cells of silkworm Bombyx mori, during larval-pupal metamorphosis. ARCHIVES OF INSECT BIOCHEMISTRY AND PHYSIOLOGY 2021; 108:e21832. [PMID: 34250644 DOI: 10.1002/arch.21832] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 06/24/2021] [Accepted: 06/25/2021] [Indexed: 06/13/2023]
Abstract
Autophagy is a critical mechanism for the self-renewal, proliferation, and differentiation of stem cells. Bombyx mori midgut has stem cells that play a role in the regeneration of the larval epithelium in larval stages and the formation of the pupal midgut epithelium during larval-pupal metamorphosis. In this study, the role of the autophagy mechanism in midgut stem cells during the formation of the pupal midgut was investigated. For this purpose, two different doses of autophagy inhibitor chloroquine were administered to B. mori larvae on days 7 and 8 of the fifth larval stage. Morphological changes during the formation process of the pupal epithelium, expression levels of autophagy-related genes Atg8 and Atg12 in stem cells, and the amounts of lysosomal enzyme acid phosphatase were determined after the application. The obtained findings were evaluated in comparison with the control groups. Abnormalities in the formation of the pupal midgut after inhibition of autophagy showed the significance of the autophagy mechanism during this period.
Collapse
Affiliation(s)
- Busra Gunay
- Department of Biology, Faculty of Science, Ege University, Izmir, Turkey
| | - Ebru Goncu
- Department of Biology, Faculty of Science, Ege University, Izmir, Turkey
| |
Collapse
|
23
|
Calcium Signaling Regulates Autophagy and Apoptosis. Cells 2021; 10:cells10082125. [PMID: 34440894 PMCID: PMC8394685 DOI: 10.3390/cells10082125] [Citation(s) in RCA: 132] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/10/2021] [Accepted: 08/17/2021] [Indexed: 02/07/2023] Open
Abstract
Calcium (Ca2+) functions as a second messenger that is critical in regulating fundamental physiological functions such as cell growth/development, cell survival, neuronal development and/or the maintenance of cellular functions. The coordination among various proteins/pumps/Ca2+ channels and Ca2+ storage in various organelles is critical in maintaining cytosolic Ca2+ levels that provide the spatial resolution needed for cellular homeostasis. An important regulatory aspect of Ca2+ homeostasis is a store operated Ca2+ entry (SOCE) mechanism that is activated by the depletion of Ca2+ from internal ER stores and has gained much attention for influencing functions in both excitable and non-excitable cells. Ca2+ has been shown to regulate opposing functions such as autophagy, that promote cell survival; on the other hand, Ca2+ also regulates programmed cell death processes such as apoptosis. The functional significance of the TRP/Orai channels has been elaborately studied; however, information on how they can modulate opposing functions and modulate function in excitable and non-excitable cells is limited. Importantly, perturbations in SOCE have been implicated in a spectrum of pathological neurodegenerative conditions. The critical role of autophagy machinery in the pathogenesis of neurodegenerative diseases such as Alzheimer’s, Parkinson’s, and Huntington’s diseases, would presumably unveil avenues for plausible therapeutic interventions for these diseases. We thus review the role of SOCE-regulated Ca2+ signaling in modulating these diverse functions in stem cell, immune regulation and neuromodulation.
Collapse
|
24
|
Stergiou IE, Kapsogeorgou EK. Autophagy and Metabolism in Normal and Malignant Hematopoiesis. Int J Mol Sci 2021; 22:8540. [PMID: 34445246 PMCID: PMC8395194 DOI: 10.3390/ijms22168540] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/03/2021] [Accepted: 08/05/2021] [Indexed: 02/07/2023] Open
Abstract
The hematopoietic system relies on regulation of both metabolism and autophagy to maintain its homeostasis, ensuring the self-renewal and multipotent differentiation potential of hematopoietic stem cells (HSCs). HSCs display a distinct metabolic profile from that of their differentiated progeny, while metabolic rewiring from glycolysis to oxidative phosphorylation (OXPHOS) has been shown to be crucial for effective hematopoietic differentiation. Autophagy-mediated regulation of metabolism modulates the distinct characteristics of quiescent and differentiating hematopoietic cells. In particular, mitophagy determines the cellular mitochondrial content, thus modifying the level of OXPHOS at the different differentiation stages of hematopoietic cells, while, at the same time, it ensures the building blocks and energy for differentiation. Aberrations in both the metabolic status and regulation of the autophagic machinery are implicated in the development of hematologic malignancies, especially in leukemogenesis. In this review, we aim to investigate the role of metabolism and autophagy, as well as their interconnections, in normal and malignant hematopoiesis.
Collapse
Affiliation(s)
| | - Efstathia K. Kapsogeorgou
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| |
Collapse
|
25
|
Zhang X, Xie Q, Ye Z, Li Y, Che Z, Huang M, Zeng J. Mesenchymal Stem Cells and Tuberculosis: Clinical Challenges and Opportunities. Front Immunol 2021; 12:695278. [PMID: 34367155 PMCID: PMC8340780 DOI: 10.3389/fimmu.2021.695278] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 06/30/2021] [Indexed: 12/22/2022] Open
Abstract
Tuberculosis (TB) is one of the communicable diseases caused by Mycobacterium tuberculosis (Mtb) infection, affecting nearly one-third of the world's population. However, because the pathogenesis of TB is still not fully understood and the development of anti-TB drug is slow, TB remains a global public health problem. In recent years, with the gradual discovery and confirmation of the immunomodulatory properties of mesenchymal stem cells (MSCs), more and more studies, including our team's research, have shown that MSCs seem to be closely related to the growth status of Mtb and the occurrence and development of TB, which is expected to bring new hope for the clinical treatment of TB. This article reviews the relationship between MSCs and the occurrence and development of TB and the potential application of MSCs in the treatment of TB.
Collapse
Affiliation(s)
- Xueying Zhang
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Qi Xie
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, China
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan, China
| | - Ziyu Ye
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, China
| | - Yanyun Li
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Zhengping Che
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Mingyuan Huang
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, China
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan, China
| | - Jincheng Zeng
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, China
- Key Laboratory of Medical Bioactive Molecular Research for Department of Education of Guangdong Province, School of Basic Medicine, Guangdong Medical University, Dongguan, China
- Collaborative Innovation Center for Antitumor Active Substance Research and Development, School of Basic Medicine, Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
26
|
Aqdas M, Singh S, Amir M, Maurya SK, Pahari S, Agrewala JN. Cumulative Signaling Through NOD-2 and TLR-4 Eliminates the Mycobacterium Tuberculosis Concealed Inside the Mesenchymal Stem Cells. Front Cell Infect Microbiol 2021; 11:669168. [PMID: 34307192 PMCID: PMC8294323 DOI: 10.3389/fcimb.2021.669168] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 06/21/2021] [Indexed: 01/27/2023] Open
Abstract
For a long time, tuberculosis (TB) has been inflicting mankind with the highest morbidity and mortality. Although the current treatment is extremely potent, a few bacilli can still hide inside the host mesenchymal stem cells (MSC). The functional capabilities of MSCs are known to be modulated by TLRs, NOD-2, and RIG-1 signaling. Therefore, we hypothesize that modulating the MSC activity through TLR-4 and NOD-2 can be an attractive immunotherapeutic strategy to eliminate the Mtb hiding inside these cells. In our current study, we observed that MSC stimulated through TLR-4 and NOD-2 (N2.T4) i) activated MSC and augmented the secretion of pro-inflammatory cytokines; ii) co-localized Mtb in the lysosomes; iii) induced autophagy; iv) enhanced NF-κB activity via p38 MAPK signaling pathway; and v) significantly reduced the intracellular survival of Mtb in the MSC. Overall, the results suggest that the triggering through N2.T4 can be a future method of immunotherapy to eliminate the Mtb concealed inside the MSC.
Collapse
Affiliation(s)
- Mohammad Aqdas
- Division of Cell Biology and Immunology, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Sanpreet Singh
- Division of Cell Biology and Immunology, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Mohammed Amir
- Division of Cell Biology and Immunology, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Sudeep Kumar Maurya
- Division of Cell Biology and Immunology, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Susanta Pahari
- Division of Cell Biology and Immunology, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Javed Naim Agrewala
- Division of Cell Biology and Immunology, CSIR-Institute of Microbial Technology, Chandigarh, India.,Immunology Laboratory, Center for Biomedical Engineering, Indian Institute of Technology, Ropar, India
| |
Collapse
|
27
|
Talukdar S, Das SK, Emdad L, Fisher PB. Autophagy and senescence: Insights from normal and cancer stem cells. Adv Cancer Res 2021; 150:147-208. [PMID: 33858596 DOI: 10.1016/bs.acr.2021.01.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Autophagy is a fundamental cellular process, which allows cells to adapt to metabolic stress through the degradation and recycling of intracellular components to generate macromolecular precursors and produce energy. Autophagy is also critical in maintaining cellular/tissue homeostasis, as well preserving immunity and preventing human disease. Deregulation of autophagic processes is associated with cancer, neurodegeneration, muscle and heart disease, infectious diseases and aging. Research on a variety of stem cell types establish that autophagy plays critical roles in normal and cancer stem cell quiescence, activation, differentiation, and self-renewal. Considering its critical function in regulating the metabolic state of stem cells, autophagy plays a dual role in the regulation of normal and cancer stem cell senescence, and cellular responses to various therapeutic strategies. The relationships between autophagy, senescence, dormancy and apoptosis frequently focus on responses to various forms of stress. These are interrelated processes that profoundly affect normal and abnormal human physiology that require further elucidation in cancer stem cells. This review provides a current perspective on autophagy and senescence in both normal and cancer stem cells.
Collapse
Affiliation(s)
- Sarmistha Talukdar
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - Swadesh K Das
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - Luni Emdad
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - Paul B Fisher
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States.
| |
Collapse
|
28
|
Tong M, Zheng Q, Liu M, Chen L, Lin YH, Tang SG, Zhu YM. 5-methoxytryptophan alleviates liver fibrosis by modulating FOXO3a/miR-21/ATG5 signaling pathway mediated autophagy. Cell Cycle 2021; 20:676-688. [PMID: 33734029 DOI: 10.1080/15384101.2021.1897241] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Liver fibrosis is a critical health issue in the world due to its rapidly increasing prevalence. It is of great demand to develop effective drugs for the treatment of liver fibrosis. 5-methoxytryptophan (5-MTP) has been reported to play an important role in anti-inflammatory, anti-cancer, myocardial-protective effects. However, the anti-fibrotic effect of 5-MTP is never covered in liver. Here, we investigated anti-fibrotic effects of 5-MTP on liver fibrosis and its underlying mechanism. In vitro, 5-MTP treatment could inhibit TGF-β1-induced elevated levels of collagen I, collagen III, fibronectin and α-smooth muscle actin (SMA) by stimulating autophagy process. Mechanically, the expression of FOXO3a was enhanced by 5-MTP and then repressed the level of miR-21, eventually leading to a restoration of autophagy-related gene ATG5. Furthermore, rescue experiments showed 5-MTP could activate autophagy process and suppress the activation of LX-2 cells by regulating FOXO3a/miR-21/ATG5 pathway. Consistently, 5-MTP significantly attenuated CCl4-induced hepatic fibrosis in rat model. In conclusion, our research discovered that 5-MTP effectively alleviated liver fibrosis in vitro and in vivo, which provided new insights into the application of 5-MTP for liver fibrosis.
Collapse
Affiliation(s)
- Ming Tong
- Department of Infectious Diseases, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha Hunan Province, P.R. China.,College of Life Sciences, Hunan Normal University, Changsha Hunan Province (The First Affiliated Hospital of Hunan Normal University),P.R. China
| | - Qing Zheng
- Department of Geriatrics, Hunan Provincial People's Hospital(The First Affiliated Hospital of Hunan Normal University), Changsha Hunan Province,P.R. China
| | - Meng Liu
- Department of Infectious Diseases, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha Hunan Province, P.R. China
| | - Liang Chen
- Department of Infectious Diseases, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha Hunan Province, P.R. China
| | - Yi-He Lin
- Department of Infectious Diseases, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha Hunan Province, P.R. China
| | - Shi-Gang Tang
- Department of Infectious Diseases, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha Hunan Province, P.R. China
| | - Yi-Min Zhu
- College of Life Sciences, Hunan Normal University, Changsha Hunan Province (The First Affiliated Hospital of Hunan Normal University),P.R. China.,Institute of Emergency Medicine, Hunan Provincial Key Laboratory of Emergency and Critical Care Metabonomics, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha Hunan Province, P.R. China
| |
Collapse
|
29
|
Valek L, Tegeder I. Nucleoredoxin Knockdown in SH-SY5Y Cells Promotes Cell Renewal. Antioxidants (Basel) 2021; 10:antiox10030449. [PMID: 33805811 PMCID: PMC7999887 DOI: 10.3390/antiox10030449] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 03/04/2021] [Accepted: 03/09/2021] [Indexed: 01/13/2023] Open
Abstract
Nucleoredoxin (NXN) is a redox regulator of Disheveled and thereby of WNT signaling. Deficiency in mice leads to cranial dysmorphisms and defects of heart, brain, and bone, suggesting defects of cell fate determination. We used shRNA-mediated knockdown of NXN in SH-SY5Y neuroblastoma cells to study its impact on neuronal cells. We expected that shNXN cells would easily succumb to redox stress, but there were no differences in viability on stimulation with hydrogen peroxide. Instead, the proliferation of naïve shNXN cells was increased with a higher rate of mitotic cells in cell cycle analyses. In addition, basal respiratory rates were higher, whereas the relative change in oxygen consumption upon mitochondrial stressors was similar to control cells. shNXN cells had an increased expression of redox-sensitive heat shock proteins, Hsc70/HSPA8 and HSP90, and autophagy markers suggested an increase in autophagosome formation upon stimulation with bafilomycin and higher flux under low dose rapamycin. A high rate of self-renewal, autophagy, and upregulation of redox-sensitive chaperones appears to be an attractive anti-aging combination if it were to occur in neurons in vivo for which SH-SY5Y cells are a model.
Collapse
|
30
|
Phan THG, Paliogiannis P, Nasrallah GK, Giordo R, Eid AH, Fois AG, Zinellu A, Mangoni AA, Pintus G. Emerging cellular and molecular determinants of idiopathic pulmonary fibrosis. Cell Mol Life Sci 2021; 78:2031-2057. [PMID: 33201251 PMCID: PMC7669490 DOI: 10.1007/s00018-020-03693-7] [Citation(s) in RCA: 216] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 10/08/2020] [Accepted: 10/28/2020] [Indexed: 12/17/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF), the most common form of idiopathic interstitial pneumonia, is a progressive, irreversible, and typically lethal disease characterized by an abnormal fibrotic response involving vast areas of the lungs. Given the poor knowledge of the mechanisms underpinning IPF onset and progression, a better understanding of the cellular processes and molecular pathways involved is essential for the development of effective therapies, currently lacking. Besides a number of established IPF-associated risk factors, such as cigarette smoking, environmental factors, comorbidities, and viral infections, several other processes have been linked with this devastating disease. Apoptosis, senescence, epithelial-mesenchymal transition, endothelial-mesenchymal transition, and epithelial cell migration have been shown to play a key role in IPF-associated tissue remodeling. Moreover, molecules, such as chemokines, cytokines, growth factors, adenosine, glycosaminoglycans, non-coding RNAs, and cellular processes including oxidative stress, mitochondrial dysfunction, endoplasmic reticulum stress, hypoxia, and alternative polyadenylation have been linked with IPF development. Importantly, strategies targeting these processes have been investigated to modulate abnormal cellular phenotypes and maintain tissue homeostasis in the lung. This review provides an update regarding the emerging cellular and molecular mechanisms involved in the onset and progression of IPF.
Collapse
Affiliation(s)
- Thị Hằng Giang Phan
- Department of Immunology and Pathophysiology, University of Medicine and Pharmacy, Hue University, Hue City, Vietnam
| | - Panagiotis Paliogiannis
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, 07100, Sassari, Italy
| | - Gheyath K Nasrallah
- Department of Biomedical Sciences, College of Health Sciences Member of QU Health, Qatar University, P.O. Box 2713, Doha, Qatar.
- Biomedical Research Center Qatar University, P.O Box 2713, Doha, Qatar.
| | - Roberta Giordo
- Department of Medical Laboratory Sciences, College of Health Sciences, and Sharjah Institute for Medical Research, University of Sharjah, University City Rd, Sharjah, 27272, United Arab Emirates
| | - Ali Hussein Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, PO Box 2713, Doha, Qatar
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, PO Box 2713, Doha, Qatar
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, PO Box 11-0236, Beirut, Lebanon
| | - Alessandro Giuseppe Fois
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, 07100, Sassari, Italy
| | - Angelo Zinellu
- Department of Biomedical Sciences, University of Sassari, 07100, Sassari, Italy
| | - Arduino Aleksander Mangoni
- Department of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Adelaide, Australia.
| | - Gianfranco Pintus
- Department of Medical Laboratory Sciences, College of Health Sciences, and Sharjah Institute for Medical Research, University of Sharjah, University City Rd, Sharjah, 27272, United Arab Emirates.
- Department of Biomedical Sciences, University of Sassari, 07100, Sassari, Italy.
| |
Collapse
|
31
|
Pańczyszyn E, Jaśko M, Miłek O, Niedziela M, Męcik-Kronenberg T, Hoang-Bujnowicz A, Zięba M, Adamus G, Kowalczuk M, Osyczka AM, Tylko G. Gellan gum hydrogels cross-linked with carbodiimide stimulates vacuolation of human tooth-derived stem cells in vitro. Toxicol In Vitro 2021; 73:105111. [PMID: 33588021 DOI: 10.1016/j.tiv.2021.105111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 01/18/2021] [Accepted: 02/11/2021] [Indexed: 10/22/2022]
Abstract
The natural polysaccharides are promising compounds for applications in regenerative medicine. Gellan gum (GG) is the bacteria-derived polysaccharide widely used in food industry. Simple modifications of its chemical properties make GG superior for the development of biocompatible hydrogels. Beside reversible cationic integration of GG chains, more efficient binding is accomplished with 1-ethyl-3-[3-(dimethylamino)propyl]carbodiimide (EDC). However, the side-products of polymer cross-linking might affect viability and differentiation of stem cells introduced into the hydrogels. We found that O-acylisourea (EDU) stimulates autophagy-based vacuolation in both periodontal ligament and dental pulp stem cells. 24-h treatment of cells with GG extracts cross-linked with 15 mM EDC developed large cytoplasmic vacuoles. Freshly prepared EDU (2-6 mM) but not 15 mM EDC solutions initiated vacuole development with concomitant reduction of cell viability/metabolism. Most of the vacuoles stained with acridine orange displayed highly acidic environment further confirmed by flow cytometric analysis. Western blot of the LC3 autophagy marker followed by a transmission electron microscopy indicated the process is autophagy-dependent. We propose that the high reactivity of EDU with intracellular components initiates autophagy, although the targets of EDU remain unknown. Nevertheless, a burst release of EDU from GG hydrogels might modulate negatively cellular processes and final effectiveness of tissue regeneration.
Collapse
Affiliation(s)
- Elżbieta Pańczyszyn
- Department of Cell Biology and Imaging, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Gronostajowa 9, 30-387 Kraków, Poland.
| | - Marta Jaśko
- Department of Cell Biology and Imaging, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Gronostajowa 9, 30-387 Kraków, Poland.
| | - Oliwia Miłek
- Department of Cell Biology and Imaging, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Gronostajowa 9, 30-387 Kraków, Poland.
| | - Matylda Niedziela
- Department of Cell Biology and Imaging, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Gronostajowa 9, 30-387 Kraków, Poland
| | - Tomasz Męcik-Kronenberg
- Department of Pathomorphology, Faculty of Medical Sciences, Medical University of Silesia, 3 Maja 13, 41-800 Zabrze, Poland.
| | - Agnieszka Hoang-Bujnowicz
- Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland.
| | - Magdalena Zięba
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, Curie-Skłodowskiej 34, 41-819 Zabrze, Poland.
| | - Grażyna Adamus
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, Curie-Skłodowskiej 34, 41-819 Zabrze, Poland.
| | - Marek Kowalczuk
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, Curie-Skłodowskiej 34, 41-819 Zabrze, Poland.
| | - Anna M Osyczka
- Department of Cell Biology and Imaging, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Gronostajowa 9, 30-387 Kraków, Poland.
| | - Grzegorz Tylko
- Department of Cell Biology and Imaging, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Gronostajowa 9, 30-387 Kraków, Poland.
| |
Collapse
|
32
|
Agas D, Sabbieti MG. Archetypal autophagic players through new lenses for bone marrow stem/mature cells regulation. J Cell Physiol 2021; 236:6101-6114. [PMID: 33492700 DOI: 10.1002/jcp.30296] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 01/04/2021] [Accepted: 01/11/2021] [Indexed: 12/11/2022]
Abstract
The bone marrow landscape consists of specialized and stem/progenitor cells, which coordinate important tissue-related and systemic physiological features. Within the marrow cavity, stem/progenitor and differentiated hematopoietic and skeletal cells congregate into dynamic functional assemblies throughout specific anatomical regions, termed niches. There is a need for better understanding of the bone marrow microareas, through exploration of the intramural physical and molecular interactions of the distinctive cell populations. The elective liaisons established among the mesenchymal/stromal stem cell and hematopoietic stem cell lineage trees play a key role in orchestrating the stem/mature cell behavior and customized hierarchies within bone marrow cell populations. Recently, the autophagic apparatus has been discovered to be an important feature of bone marrow homeostasis. Autophagy-related factors involved in the labyrinthic and highly dynamic bone marrow workshop redesign the niche framework by coordinating the operational schedule of pluripotent stem and mature cells. The following report summarizes the most recent breakthroughs in our understanding of the intramural relationships between bone marrow cells and key autophagic mediators. Doubtless, the consideration of the autophagy-related and unrelated functions of main players, such as p62, Atg7, Atg5, and Beclin-1 remains a compelling task to thoroughly understand the complex relations between the heterogenic cell types that populate bone marrow.
Collapse
Affiliation(s)
- Dimitrios Agas
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Macerata, Italy
| | - Maria Giovanna Sabbieti
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Macerata, Italy
| |
Collapse
|
33
|
He Y, Ma M, Yan Y, Chen C, Luo H, Lei W. Combined pre-conditioning with salidroside and hypoxia improves proliferation, migration and stress tolerance of adipose-derived stem cells. J Cell Mol Med 2020; 24:9958-9971. [PMID: 32767741 PMCID: PMC7520330 DOI: 10.1111/jcmm.15598] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 06/10/2020] [Accepted: 06/15/2020] [Indexed: 12/18/2022] Open
Abstract
Oxidative stress after ischaemia impairs the function of transplanted stem cells. Increasing evidence has suggested that either salidroside (SAL) or hypoxia regulates growth of stem cells. However, the role of SAL in regulating function of hypoxia‐pre–conditioned stem cells remains elusive. Thus, this study aimed to determine the effect of SAL and hypoxia pre‐conditionings on the proliferation, migration and tolerance against oxidative stress in rat adipose‐derived stem cells (rASCs). rASCs treated with SAL under normoxia (20% O2) or hypoxia (5% O2) were analysed for the cell viability, proliferation, migration and resistance against H2O2‐induced oxidative stress. In addition, the activation of Akt, Erk1/2, LC3, NF‐κB and apoptosis‐associated pathways was assayed by Western blot. The results showed that SAL and hypoxia treatments synergistically enhanced the viability (fold) and proliferation of rASCs under non‐stressed conditions in association with increased autophagic flux and activation of Akt, Erk1/2 and LC3. H2O2‐induced oxidative stress, cytotoxicity, apoptosis, autophagic cell death and NF‐κB activation were inhibited by SAL or hypoxia, and further attenuated by the combined SAL and hypoxia pre‐treatment. The SAL and hypoxia pre‐treatment also enhanced the proliferation and migration of rASCs under oxidative stress in association with Akt and Erk1/2 activation; however, the combined pre‐treatment exhibited a more profound enhancement in the migration than proliferation. Our data suggest that SAL combined with hypoxia pre‐conditioning may enhance the therapeutic capacity of ASCs in post‐ischaemic repair.
Collapse
Affiliation(s)
- Yuan He
- Laboratory of Cardiovascular Diseases, Guangdong Medical University, Zhanjiang, China
| | - Mudi Ma
- Laboratory of Cardiovascular Diseases, Guangdong Medical University, Zhanjiang, China.,Cardiovascular Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Yiguang Yan
- Cardiovascular Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Can Chen
- Laboratory of Cardiovascular Diseases, Guangdong Medical University, Zhanjiang, China
| | - Hui Luo
- Southern Marine Science and Engineering Guangdong Laboratory-Zhanjiang, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China
| | - Wei Lei
- Laboratory of Cardiovascular Diseases, Guangdong Medical University, Zhanjiang, China.,Cardiovascular Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.,Southern Marine Science and Engineering Guangdong Laboratory-Zhanjiang, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
34
|
Approaches and Technologies in Male Fertility Preservation. Int J Mol Sci 2020; 21:ijms21155471. [PMID: 32751826 PMCID: PMC7432867 DOI: 10.3390/ijms21155471] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 07/21/2020] [Accepted: 07/29/2020] [Indexed: 12/12/2022] Open
Abstract
Male fertility preservation is required when treatment with an aggressive chemo-/-radiotherapy, which may lead to irreversible sterility. Due to new and efficient protocols of cancer treatments, surviving rates are more than 80%. Thus, these patients are looking forward to family life and fathering their own biological children after treatments. Whereas adult men can cryopreserve their sperm for future use in assistance reproductive technologies (ART), this is not an option in prepubertal boys who cannot produce sperm at this age. In this review, we summarize the different technologies for male fertility preservation with emphasize on prepubertal, which have already been examined and/or demonstrated in vivo and/or in vitro using animal models and, in some cases, using human tissues. We discuss the limitation of these technologies for use in human fertility preservation. This update review can assist physicians and patients who are scheduled for aggressive chemo-/radiotherapy, specifically prepubertal males and their parents who need to know about the risks of the treatment on their future fertility and the possible present option of fertility preservation.
Collapse
|
35
|
Zhai C, Feng W, Shi W, Wang J, Zhang Q, Yan X, Wang Q, Li S, Liu L, Pan Y, Zhu Y, Chai L, Li C, Liu P, Chen Y, Li M. Sphingosine-1-phosphate promotes pulmonary artery smooth muscle cells proliferation by stimulating autophagy-mediated E-cadherin/CDH1 down-regulation. Eur J Pharmacol 2020; 884:173302. [PMID: 32659302 DOI: 10.1016/j.ejphar.2020.173302] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 06/18/2020] [Accepted: 06/19/2020] [Indexed: 02/07/2023]
Abstract
It has been shown that sphingosine-1-phosphate (S1P) is elevated in patients with pulmonary arterial hypertension (PAH) and promotes the proliferation of pulmonary artery smooth muscle cells (PASMCs). Meanwhile, S1P has been found to induce the activation of autophagy in several types of human diseases including cancers. However, it is still unclear whether activation of autophagy mediates S1P-induced PASMCs proliferation, and detailed mechanisms responsible for these processes are indefinite. The aims of this study are to address these issues. S1P dose- and time-dependently reduced the expression of E-cadherin/CDH1 and stimulated PASMCs proliferation; this was accompanied with the elevation of TNF receptor-associated factor 2 (TRAF2), up-regulation and ubiquitination of BECN1 and the activation of autophagy. Prior silencing TRAF2 or BECN1 using siRNA or pre-incubation of cells with autophagy inhibitor chloroquine phosphate (CQ) suppressed S1P-induced autophagy activation and subsequent CDH1 degradation and further PASMCs proliferation. Taken together, our study indicates that S1P promotes the activation of autophagy by accelerating TRAF2-mediated BECN1 up-regulation and ubiquitination, which in turn results in CDH1 reduction and contributes to PASMCs proliferation.
Collapse
Affiliation(s)
- Cui Zhai
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, PR China
| | - Wei Feng
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, PR China
| | - Wenhua Shi
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, PR China
| | - Jian Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, PR China
| | - Qianqian Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, PR China
| | - Xin Yan
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, PR China
| | - Qingting Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, PR China
| | - Shaojun Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, PR China
| | - Lu Liu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, PR China
| | - Yilin Pan
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, PR China
| | - Yanting Zhu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, PR China
| | - Limin Chai
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, PR China
| | - Cong Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, PR China
| | - Pengtao Liu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, PR China
| | - Yuqian Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, PR China
| | - Manxiang Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, PR China.
| |
Collapse
|
36
|
Oatley M, Bölükbası ÖV, Svensson V, Shvartsman M, Ganter K, Zirngibl K, Pavlovich PV, Milchevskaya V, Foteva V, Natarajan KN, Baying B, Benes V, Patil KR, Teichmann SA, Lancrin C. Single-cell transcriptomics identifies CD44 as a marker and regulator of endothelial to haematopoietic transition. Nat Commun 2020; 11:586. [PMID: 31996681 PMCID: PMC6989687 DOI: 10.1038/s41467-019-14171-5] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 12/18/2019] [Indexed: 12/13/2022] Open
Abstract
The endothelial to haematopoietic transition (EHT) is the process whereby haemogenic endothelium differentiates into haematopoietic stem and progenitor cells (HSPCs). The intermediary steps of this process are unclear, in particular the identity of endothelial cells that give rise to HSPCs is unknown. Using single-cell transcriptome analysis and antibody screening, we identify CD44 as a marker of EHT enabling us to isolate robustly the different stages of EHT in the aorta-gonad-mesonephros (AGM) region. This allows us to provide a detailed phenotypical and transcriptional profile of CD44-positive arterial endothelial cells from which HSPCs emerge. They are characterized with high expression of genes related to Notch signalling, TGFbeta/BMP antagonists, a downregulation of genes related to glycolysis and the TCA cycle, and a lower rate of cell cycle. Moreover, we demonstrate that by inhibiting the interaction between CD44 and its ligand hyaluronan, we can block EHT, identifying an additional regulator of HSPC development. The endothelial to haematopoietic transition (EHT) is the process where haemogenic endothelium differentiates into haematopoietic stem and progenitor cells (HSPCs). Here the authors use single cell transcriptomics and antibody screening to identify CD44 as a marker of EHT that is required for EHT and HSPC development.
Collapse
Affiliation(s)
- Morgan Oatley
- European Molecular Biology Laboratory, EMBL Rome - Epigenetics and Neurobiology Unit, via E. Ramarini 32, 00015, Monterotondo, Italy
| | - Özge Vargel Bölükbası
- European Molecular Biology Laboratory, EMBL Rome - Epigenetics and Neurobiology Unit, via E. Ramarini 32, 00015, Monterotondo, Italy.,Stem Cell and Regenerative Biology Department, Harvard University, 7 Divinity Avenue, Cambridge, MA, 02138, USA
| | - Valentine Svensson
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, UK.,European Molecular Biology Laboratory, EMBL-EBI, Wellcome Genome Campus, Hinxton, Cambridgeshire, CB10 1SD, UK.,Pachter Lab, Division of Biology and Biological Engineering, California Institute of Technology, 1200 East California Boulevard, Pasadena, CA, USA
| | - Maya Shvartsman
- European Molecular Biology Laboratory, EMBL Rome - Epigenetics and Neurobiology Unit, via E. Ramarini 32, 00015, Monterotondo, Italy
| | - Kerstin Ganter
- European Molecular Biology Laboratory, EMBL Rome - Epigenetics and Neurobiology Unit, via E. Ramarini 32, 00015, Monterotondo, Italy
| | - Katharina Zirngibl
- European Molecular Biology Laboratory, Structural and Computational Biology Unit, Meyerhofstrasse 1, 69117, Heidelberg, Germany
| | - Polina V Pavlovich
- European Molecular Biology Laboratory, EMBL Rome - Epigenetics and Neurobiology Unit, via E. Ramarini 32, 00015, Monterotondo, Italy.,Moscow Institute of Physics and Technology, Institutskii Per. 9, Moscow Region, Dolgoprudny, 141700, Russia.,Max Planck Institute of Immunobiology and Epigenetics, Stübeweg 51, D-79108, Freiburg, Germany
| | - Vladislava Milchevskaya
- European Molecular Biology Laboratory, Structural and Computational Biology Unit, Meyerhofstrasse 1, 69117, Heidelberg, Germany.,Institut für Medizinische Statistik und Bioinformatik, Bachemer Strasse 86, 50931, Köln, Germany
| | - Vladimira Foteva
- European Molecular Biology Laboratory, EMBL Rome - Epigenetics and Neurobiology Unit, via E. Ramarini 32, 00015, Monterotondo, Italy
| | - Kedar N Natarajan
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, UK.,Department of Biochemistry and Molecular Biology, The University of Southern Denmark, Danish Institute for Advanced Study, Campusvej 55, 5230, Odense M, Denmark
| | - Bianka Baying
- European Molecular Biology Laboratory, Genomics Core Facility, Meyerhofstrasse 1, 69117, Heidelberg, Germany
| | - Vladimir Benes
- European Molecular Biology Laboratory, Genomics Core Facility, Meyerhofstrasse 1, 69117, Heidelberg, Germany
| | - Kiran R Patil
- European Molecular Biology Laboratory, Structural and Computational Biology Unit, Meyerhofstrasse 1, 69117, Heidelberg, Germany
| | - Sarah A Teichmann
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Christophe Lancrin
- European Molecular Biology Laboratory, EMBL Rome - Epigenetics and Neurobiology Unit, via E. Ramarini 32, 00015, Monterotondo, Italy.
| |
Collapse
|
37
|
Kaplan N, Wang J, Wray B, Patel P, Yang W, Peng H, Lavker RM. Single-Cell RNA Transcriptome Helps Define the Limbal/Corneal Epithelial Stem/Early Transit Amplifying Cells and How Autophagy Affects This Population. Invest Ophthalmol Vis Sci 2019; 60:3570-3583. [PMID: 31419300 PMCID: PMC6701873 DOI: 10.1167/iovs.19-27656] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Purpose Single-cell RNA-sequencing (scRNA-seq) was used to interrogate the relatively rare stem (SC) and early transit amplifying (TA) cell populations in limbal/corneal epithelia from wild-type and autophagy-compromised mice. Methods We conducted scRNA-seq on ocular anterior segmental tissue from wild-type and beclin 1–deficient (beclin1+/−) mice, using a 10X Gemomics pipeline. Cell populations were distinguished by t-distributed stochastic neighbor embedding. Seurat analysis was conducted to compare gene expression profiles between these two groups of mice. Differential protein expression patterns were validated by immunofluorescence staining and immunoblotting. Results Unbiased clustering detected 10 distinct populations: three clusters of mesenchymal and seven clusters of epithelial cells, based on their unique molecular signatures. A discrete group of mesenchymal cells expressed genes associated with corneal stromal SCs. We identified three limbal/corneal epithelial cell subpopulations designated as stem/early TA, mature TA, and differentiated corneal epithelial cells. Thioredoxin-interacting protein and PDZ-binding kinase (PBK) were identified as novel regulators of stem/early TA cell quiescence. PBK arrested corneal epithelial cells in G2/M phase of the cell cycle. Beclin1+/− mice displayed a decrease in proliferation-associated (Ki67, Lrig1) and stress-response (H2ax) genes. The most increased gene in beclin1+/− mice was transcription factor ATF3, which negatively regulates limbal epithelial cell proliferation. Conclusions Establishment of a comprehensive atlas of genes expressed by stromal and epithelial cells from limbus and cornea forms the foundation for unraveling regulatory networks among these distinct tissues. Similarly, scRNA-seq profiling of the anterior segmental epithelia from wild-type and autophagy-deficient mice provides new insights into how autophagy influences proliferation in these tissues.
Collapse
Affiliation(s)
- Nihal Kaplan
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States
| | - Junyi Wang
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States.,Department of Ophthalmology, Ophthalmology and Visual Science Key Lab of PLA, Chinese PLA General Hospital, Beijing, China
| | - Brian Wray
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States
| | - Priyam Patel
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States
| | - Wending Yang
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States
| | - Han Peng
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States
| | - Robert M Lavker
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States
| |
Collapse
|
38
|
Hu C, Liang X, Fang S, Xu L, Gong M, Wang Y, Bi Y, Hong S, He Y. ATRA induces the differentiation of hepatic progenitor cells by upregulating microRNA-200a. In Vitro Cell Dev Biol Anim 2019; 55:713-722. [PMID: 31515718 DOI: 10.1007/s11626-019-00390-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Accepted: 07/21/2019] [Indexed: 02/07/2023]
Abstract
Hepatic progenitor cells (HPCs) are potential seed cells for hepatocyte transplantation treatment of liver diseases. ATRA can induce the differentiation and mature function of hepatic progenitor cells, but the mechanism is still poorly understood. Here, by using microRNA array to analyze the expression profiles of microRNA (miR), we found that miR-200 family molecules in HPCs were upregulated after ATRA treatment, especially miR-200a-3p, 200c-3p, and 141-3p. ATRA induction could downregulate the expression of hepatic stem markers Oct4 and AFP, and improve the expression of hepatic markers ALB, CK18, and TAT, and the activity of ALB-GLuc, as well as indocyanine green uptake and glycogen storage function of HPCs. These above effects of ATRA on HPC differentiation were almost inhibited by blocking of miR-200a-3p, but not miR-200c-3p and 141-3p using antagomir. Cell autophagy is associated with ATRA regulation in HPCs, compared with control group, the expression of LC3 and Beclin1 increased in ATRA-treated HPCs, and orange and red fluorescent spot, which represents autophagy flow, also enhanced after ATRA treatment. However, ATRA-induced cell autophagy level was inhibited in antagomir-200a-3p+ATRA-treated cells. Therefore, the present study indicates that antagomir-200a-3p is related to ATRA-induced hepatic differentiation of HPCs through regulating cell autophagy, supporting the possible use of ATRA as a key inducer in HPC-based therapy of liver diseases.
Collapse
Affiliation(s)
- Chaoqun Hu
- Stem Cell Biology and Therapy Laboratory, Department of Pediatric Surgery, Ministry of Education Key Laboratory of Child Development and Disorders, The Children's Hospital of Chongqing Medical University, Building 5, Room 501, 136 Zhongshan Er Road, Chongqing, 400014, People's Republic of China
| | - Xiaohua Liang
- Stem Cell Biology and Therapy Laboratory, Department of Pediatric Surgery, Ministry of Education Key Laboratory of Child Development and Disorders, The Children's Hospital of Chongqing Medical University, Building 5, Room 501, 136 Zhongshan Er Road, Chongqing, 400014, People's Republic of China
| | - Shuyu Fang
- Stem Cell Biology and Therapy Laboratory, Department of Pediatric Surgery, Ministry of Education Key Laboratory of Child Development and Disorders, The Children's Hospital of Chongqing Medical University, Building 5, Room 501, 136 Zhongshan Er Road, Chongqing, 400014, People's Republic of China
| | - Lei Xu
- Department of Microbiology, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Mengjia Gong
- Stem Cell Biology and Therapy Laboratory, Department of Pediatric Surgery, Ministry of Education Key Laboratory of Child Development and Disorders, The Children's Hospital of Chongqing Medical University, Building 5, Room 501, 136 Zhongshan Er Road, Chongqing, 400014, People's Republic of China
| | - Yi Wang
- Stem Cell Biology and Therapy Laboratory, Department of Pediatric Surgery, Ministry of Education Key Laboratory of Child Development and Disorders, The Children's Hospital of Chongqing Medical University, Building 5, Room 501, 136 Zhongshan Er Road, Chongqing, 400014, People's Republic of China
| | - Yang Bi
- Stem Cell Biology and Therapy Laboratory, Department of Pediatric Surgery, Ministry of Education Key Laboratory of Child Development and Disorders, The Children's Hospital of Chongqing Medical University, Building 5, Room 501, 136 Zhongshan Er Road, Chongqing, 400014, People's Republic of China
| | - Siqi Hong
- Stem Cell Biology and Therapy Laboratory, Department of Pediatric Surgery, Ministry of Education Key Laboratory of Child Development and Disorders, The Children's Hospital of Chongqing Medical University, Building 5, Room 501, 136 Zhongshan Er Road, Chongqing, 400014, People's Republic of China.
| | - Yun He
- Stem Cell Biology and Therapy Laboratory, Department of Pediatric Surgery, Ministry of Education Key Laboratory of Child Development and Disorders, The Children's Hospital of Chongqing Medical University, Building 5, Room 501, 136 Zhongshan Er Road, Chongqing, 400014, People's Republic of China.
| |
Collapse
|
39
|
Lee IH. Mechanisms and disease implications of sirtuin-mediated autophagic regulation. Exp Mol Med 2019; 51:1-11. [PMID: 31492861 PMCID: PMC6802627 DOI: 10.1038/s12276-019-0302-7] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 05/03/2019] [Accepted: 05/13/2019] [Indexed: 12/27/2022] Open
Abstract
Accumulating evidence has indicated that sirtuins are key components of diverse physiological processes, including metabolism and aging. Sirtuins confer protection from a wide array of metabolic and age-related diseases, such as cancer, cardiovascular and neurodegenerative diseases. Recent studies have also suggested that sirtuins regulate autophagy, a protective cellular process for homeostatic maintenance in response to environmental stresses. Here, we describe various biological and pathophysiological processes regulated by sirtuin-mediated autophagy, focusing on cancer, heart, and liver diseases, as well as stem cell biology. This review also emphasizes key molecular mechanisms by which sirtuins regulate autophagy. Finally, we discuss novel insights into how new therapeutics targeting sirtuin and autophagy may potentially lead to effective strategies to combat aging and aging-related diseases.
Collapse
Affiliation(s)
- In Hye Lee
- Department of Life Science, Ewha Womans University, Seoul, South Korea.
| |
Collapse
|
40
|
Batatinha HAP, Diniz TA, de Souza Teixeira AA, Krüger K, Rosa-Neto JC. Regulation of autophagy as a therapy for immunosenescence-driven cancer and neurodegenerative diseases: The role of exercise. J Cell Physiol 2019; 234:14883-14895. [PMID: 30756377 DOI: 10.1002/jcp.28318] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 12/26/2018] [Accepted: 01/10/2019] [Indexed: 01/24/2023]
Abstract
Aging is one of the risk factors for the development of low-grade inflammation morbidities, such as several types of cancer and neurodegenerative diseases, due to changes in the metabolism, hormonal secretion, and immunosenescence. The senescence of the immune system leads to improper control of infections and tissue damage increasing age-related diseases. One of the mechanisms that maintain cellular homeostasis is autophagy, a cell-survival mechanism, and it has been proposed as one of the most powerful antiaging therapies. Regular exercise can reestablish autophagy, probably through AMP-activated protein kinase activation, and help in reducing the age-related senescence diseases. Therefore, in this study, we discuss the effects of exercise training in immunosenescence and autophagy, preventing the two main age-related disease, cancer and neurodegeneration.
Collapse
Affiliation(s)
| | - Tiego Aparecido Diniz
- Department of Cell and Developmental Biology, University of São Paulo, São Paulo, São Paulo, Brazil
| | | | - Karsten Krüger
- Department Exercise and Health, Institute of Sports Science, Leibniz University Hannover, Hannover, Germany
| | - Jose Cesar Rosa-Neto
- Department of Cell and Developmental Biology, University of São Paulo, São Paulo, São Paulo, Brazil
| |
Collapse
|
41
|
Shi L, Li B, Zhang B, Zhen C, Zhou J, Tang S. Mouse embryonic palatal mesenchymal cells maintain stemness through the PTEN-Akt-mTOR autophagic pathway. Stem Cell Res Ther 2019; 10:217. [PMID: 31358051 PMCID: PMC6664599 DOI: 10.1186/s13287-019-1340-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 06/21/2019] [Accepted: 07/14/2019] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Both genetic and environmental factors are implicated in the pathogenesis of cleft palate. However, the molecular and cellular mechanisms that regulate the development of palatal shelves, which are composed of mesenchymal cells, have not yet been fully elucidated. This study aimed to determine the stemness and multilineage differentiation potential of mouse embryonic palatal mesenchyme (MEPM) cells in palatal shelves and to explore the underlying regulatory mechanism associated with cleft palate formation. METHODS Palatal shelves excised from mice models were cultured in vitro to ascertain whether MEPM are stem cells through immunofluorescence and flow cytometry. The osteogenic, adipogenic, and chondrogenic differentiation potential of MEPM cells were also determined to characterize MEPM stemness. In addition, the role of the PTEN-Akt-mTOR autophagic pathway was investigated using quantitative RT-PCR, Western blotting, and transmission electron microscopy. RESULTS MEPM cells in culture exhibited cell surface marker expression profiles similar to that of mouse bone marrow stem cells and exhibited positive staining for vimentin (mesodermal marker), nestin (ectodermal marker), PDGFRα, Efnb1, Osr2, and Meox2 (MEPM cells markers). In addition, exposure to PDGFA stimulated chemotaxis of MEPM cells. MEPM cells exhibited stronger potential for osteogenic differentiation as compared to that for adipogenic and chondrogenic differentiation. Undifferentiated MEPM cells displayed a high concentration of autophagosomes, which disappeared after differentiation (at passage four), indicating the involvement of PTEN-Akt-mTOR signaling. CONCLUSIONS Our findings suggest that MEPM cells are ectomesenchymal stem cells with a strong osteogenic differentiation potential and that maintenance of their stemness via PTEN/AKT/mTOR autophagic signaling prevents cleft palate development.
Collapse
Affiliation(s)
- Lungang Shi
- Department of Plastic Surgery and Burn Center, the Second Affiliated Hospital of Shantou University Medical College, North Dongxia Road, Shantou, 515041 Guangdong China
| | - Binchen Li
- Shantou University Medical College, No. 22 Xinling road, Shantou, 515041 Guangdong China
| | - Binna Zhang
- Center for Translational Medicine, the Second Affiliated Hospital of Shantou University Medical College, North Dongxia Road, Shantou, 515041 Guangdong China
| | - Congyuan Zhen
- Shantou University Medical College, No. 22 Xinling road, Shantou, 515041 Guangdong China
| | - Jianda Zhou
- Department of Plastic Surgery, Third Xiangya Hospital, Central South University, Changsha, 410013 Hunan China
| | - Shijie Tang
- Department of Plastic Surgery and Burn Center, the Second Affiliated Hospital of Shantou University Medical College, North Dongxia Road, Shantou, 515041 Guangdong China
| |
Collapse
|
42
|
Magalhães-Novais S, Bermejo-Millo JC, Loureiro R, Mesquita KA, Domingues MR, Maciel E, Melo T, Baldeiras I, Erickson JR, Holy J, Potes Y, Coto-Montes A, Oliveira PJ, Vega-Naredo I. Cell quality control mechanisms maintain stemness and differentiation potential of P19 embryonic carcinoma cells. Autophagy 2019; 16:313-333. [PMID: 30990357 DOI: 10.1080/15548627.2019.1607694] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Given the relatively long life of stem cells (SCs), efficient mechanisms of quality control to balance cell survival and resistance to external and internal stress are required. Our objective was to test the relevance of cell quality control mechanisms for SCs maintenance, differentiation and resistance to cell death. We compared cell quality control in P19 stem cells (P19SCs) before and after differentiation (P19dCs). Differentiation of P19SCs resulted in alterations in parameters involved in cell survival and protein homeostasis, including the redox system, cardiolipin and lipid profiles, unfolded protein response, ubiquitin-proteasome and lysosomal systems, and signaling pathways controlling cell growth. In addition, P19SCs pluripotency was correlated with stronger antioxidant protection, modulation of apoptosis, and activation of macroautophagy, which all contributed to preserve SCs quality by increasing the threshold for cell death activation. Furthermore, our findings identify critical roles for the PI3K-AKT-MTOR pathway, as well as autophagic flux and apoptosis regulation in the maintenance of P19SCs pluripotency and differentiation potential.Abbreviations: 3-MA: 3-methyladenine; AKT/protein kinase B: thymoma viral proto-oncogene; AKT1: thymoma viral proto-oncogene 1; ATG: AuTophaGy-related; ATF6: activating transcription factor 6; BAX: BCL2-associated X protein; BBC3/PUMA: BCL2 binding component 3; BCL2: B cell leukemia/lymphoma 2; BNIP3L: BCL2/adenovirus E1B interacting protein 3-like; CASP3: caspase 3; CASP8: caspase 8; CASP9: caspase 9; CL: cardiolipin; CTSB: cathepsin B; CTSD: cathepsin D; DDIT3/CHOP: DNA-damage inducible transcript 3; DNM1L/DRP1: dynamin 1-like; DRAM1: DNA-damage regulated autophagy modulator 1; EIF2AK3/PERK: eukaryotic translation initiation factor 2 alpha kinase 3; EIF2S1/eIF2α: eukaryotic translation initiation factor 2, subunit alpha; ERN1/IRE1α: endoplasmic reticulum to nucleus signaling 1; ESCs: embryonic stem cells; KRT8/TROMA-1: cytokeratin 8; LAMP2A: lysosomal-associated membrane protein 2A; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; MTOR: mechanistic target of rapamycin kinase; NANOG: Nanog homeobox; NAO: 10-N-nonyl acridine orange; NFE2L2/NRF2: nuclear factor, erythroid derived 2, like 2; OPA1: OPA1, mitochondrial dynamin like GTPase; P19dCs: P19 differentiated cells; P19SCs: P19 stem cells; POU5F1/OCT4: POU domain, class 5, transcription factor 1; PtdIns3K: phosphatidylinositol 3-kinase; RA: retinoic acid; ROS: reactive oxygen species; RPS6KB1/p70S6K: ribosomal protein S6 kinase, polypeptide 1; SCs: stem cells; SOD: superoxide dismutase; SHC1-1/p66SHC: src homology 2 domain-containing transforming protein C1, 66 kDa isoform; SOX2: SRY (sex determining region Y)-box 2; SQSTM1/p62: sequestosome 1; SPTAN1/αII-spectrin: spectrin alpha, non-erythrocytic 1; TOMM20: translocase of outer mitochondrial membrane 20; TRP53/p53: transformation related protein 53; TUBB3/betaIII-tubulin: tubulin, beta 3 class III; UPR: unfolded protein response; UPS: ubiquitin-proteasome system.
Collapse
Affiliation(s)
| | - Juan C Bermejo-Millo
- Department of Morphology and Cell Biology, University of Oviedo, Oviedo, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Rute Loureiro
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Cantanhede, Portugal
| | - Katia A Mesquita
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Cantanhede, Portugal
| | - M Rosário Domingues
- Mass Spectrometry Centre, Department of Chemistry & QOPNA, University of Aveiro, Aveiro, Portugal
| | - Elisabete Maciel
- Mass Spectrometry Centre, Department of Chemistry & QOPNA, University of Aveiro, Aveiro, Portugal.,Department of Biology & CESAM, University of Aveiro, Aveiro, Portugal
| | - Tânia Melo
- Mass Spectrometry Centre, Department of Chemistry & QOPNA, University of Aveiro, Aveiro, Portugal
| | - Inês Baldeiras
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Cantanhede, Portugal.,School of Medicine, University of Coimbra, Coimbra, Portugal
| | - Jenna R Erickson
- Department of Biomedical Sciences, University of Minnesota-Duluth, Duluth, MN, USA
| | - Jon Holy
- Department of Biomedical Sciences, University of Minnesota-Duluth, Duluth, MN, USA
| | - Yaiza Potes
- Department of Morphology and Cell Biology, University of Oviedo, Oviedo, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Ana Coto-Montes
- Department of Morphology and Cell Biology, University of Oviedo, Oviedo, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Paulo J Oliveira
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Cantanhede, Portugal
| | - Ignacio Vega-Naredo
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Cantanhede, Portugal.,Department of Morphology and Cell Biology, University of Oviedo, Oviedo, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| |
Collapse
|
43
|
Zhang M, Liu F, Zhou P, Wang Q, Xu C, Li Y, Bian L, Liu Y, Zhou J, Wang F, Yao Y, Fang Y, Li D. The MTOR signaling pathway regulates macrophage differentiation from mouse myeloid progenitors by inhibiting autophagy. Autophagy 2019; 15:1150-1162. [PMID: 30724690 DOI: 10.1080/15548627.2019.1578040] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Understanding of the mechanism for myeloid differentiation provides important insights into the hematopoietic developmental processes. By using an ESC-derived myeloid progenitor cell model, we found that CSF2/GM-CSF triggered macrophage differentiation and activation of the MTOR signaling pathway. Activation or inhibition of the MTOR signaling enhanced or attenuated macrophage differentiation, respectively, suggesting a critical function. We further showed that macroautophagy/autophagy was inhibited with the addition of CSF2. Furthermore, pharmacological inhibition and genetic modification of autophagy enhanced macrophage differentiation and rescued the inhibitory effect on differentiation caused by MTOR inhibition. Thus, the MTOR signaling pathway regulates macrophage differentiation of myeloid progenitors by inhibiting autophagy. Our results provide new insights into the mechanisms for myeloid differentiation and may prove useful for therapeutic applications of hematopoietic and myeloid progenitor cells. Abbreviations: 2-DG: 2-deoxy-D-glucose; ADGRE1/F4/80: adhesion G protein-coupled receptor E1; BM: bone marrow; CQ: chloroquine; ECAR: extracellular acidification rate; ESC: embryonic stem cell; CSF2/GM-CSF: colony stimulating factor 2; CSF3/G-CSF: colony stimulating factor 3; HPC: hematopoietic progenitor cell; ITGAM/CD11b: integrin alpha M; LPS: lipopolysaccharide; MFI: median fluorescence intensity; MTOR: mechanistic target of rapamycin kinase; RPS6KB1/p70S6K1: ribosomal protein S6 kinase, polypeptide 1; shRNA: short hairpin RNA; SQSTM1/p62: sequestosome 1.
Collapse
Affiliation(s)
- Meichao Zhang
- a Department of Radiation Oncology, Shanghai Ninth People's Hospital , Shanghai Jiaotong University School of Medicine , Shanghai , China
| | - Furao Liu
- a Department of Radiation Oncology, Shanghai Ninth People's Hospital , Shanghai Jiaotong University School of Medicine , Shanghai , China
| | - Pingting Zhou
- a Department of Radiation Oncology, Shanghai Ninth People's Hospital , Shanghai Jiaotong University School of Medicine , Shanghai , China
| | - Qian Wang
- b Department of Oncology, Shanghai Ninth People's Hospital , Shanghai Jiaotong University School of Medicine , Shanghai , China
| | - Ci Xu
- a Department of Radiation Oncology, Shanghai Ninth People's Hospital , Shanghai Jiaotong University School of Medicine , Shanghai , China
| | - Yanyan Li
- a Department of Radiation Oncology, Shanghai Ninth People's Hospital , Shanghai Jiaotong University School of Medicine , Shanghai , China
| | - Lei Bian
- a Department of Radiation Oncology, Shanghai Ninth People's Hospital , Shanghai Jiaotong University School of Medicine , Shanghai , China
| | - Yuanhua Liu
- c Department of Chemotherapy , Nanjing Medical University Affiliated Cancer Hospital, Cancer Institute of Jiangsu Province , Nanjing , Jiangsu , China
| | - Jiaxi Zhou
- d State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital , Chinese Academy of Medical Sciences & Peking Union Medical College , Tianjin , China
| | - Fei Wang
- e Department of Cell and Developmental Biology , University of Illinois at Urbana-Champaign , Urbana , IL , USA
| | - Yuan Yao
- a Department of Radiation Oncology, Shanghai Ninth People's Hospital , Shanghai Jiaotong University School of Medicine , Shanghai , China
| | - Yong Fang
- f Department of Burns and Plastic Surgery, Shanghai Ninth People's Hospital , Shanghai JiaoTong University School of Medicine , Shanghai , China
| | - Dong Li
- a Department of Radiation Oncology, Shanghai Ninth People's Hospital , Shanghai Jiaotong University School of Medicine , Shanghai , China
| |
Collapse
|
44
|
Autophagy in Development and Differentiation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1206:469-487. [PMID: 31776999 DOI: 10.1007/978-981-15-0602-4_22] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Autophagy is crucial in the differentiation and development of both mammals and invertebrates, as a rapid response to environmental and hormonal stimuli. Autophagy is also important for intracellular renewal, maintaining the health of terminally differentiated cells. Studies of Drosophila, Caenorhabditis elegans, and other species revealed abnormal autophagy lead to developmental and differential abnormality, including those in salivary glands and midgut development, protein aggregation, removal of apoptotic cell corpses, and development of dauer and synapse. Autophagy also participates in the development of mammalian embryos before implantation into the uterus, adaption to the nascent hunger environment, blood cells production, and cell differentiation in adipogenesis. Autophagy found in various stem cells, like hematopoietic stem cells, bone marrow mesenchymal stem cells and neural stem cells (NSCs), is tightly associated with their self-renewal, directed differentiation, and senescence.
Collapse
|
45
|
Hassanpour M, Rezabakhsh A, Pezeshkian M, Rahbarghazi R, Nouri M. Distinct role of autophagy on angiogenesis: highlights on the effect of autophagy in endothelial lineage and progenitor cells. Stem Cell Res Ther 2018; 9:305. [PMID: 30409213 PMCID: PMC6225658 DOI: 10.1186/s13287-018-1060-5] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Autophagy plays a critical role in the dynamic growth of each cell through different conditions. It seems that this intracellular mechanism acts as a two-edged sword against the numerous cell insults. Previously, autophagy was described in the context of cell activity and behavior, but little knowledge exists related to the role of autophagy in endothelial cells, progenitors, and stem cells biology from different tissues. Angiogenic behavior of endothelial lineage and various stem cells are touted as an inevitable feature in the restoration of different damaged tissues and organs. This capacity was found to be dictated by autophagy signaling pathway. This review article highlights the fundamental role of cell autophagic response in endothelial cells function, stem cells dynamic, and differentiation rate. It seems that elucidation of the mechanisms related to pro- and/or anti-angiogenic potential of autophagy inside endothelial cells and stem cells could help us to modulate stem cell therapeutic feature post-transplantation.
Collapse
Affiliation(s)
- Mehdi Hassanpour
- Department of Clinical Biochemistry and Laboratory Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Stem Cell Research Center, Tabriz University of Medical Sciences, Imam Reza St., Golgasht St., Tabriz, 5166614756 Iran
- Stem Cell And Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aysa Rezabakhsh
- Emergency Medicine Research Team, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Masoud Pezeshkian
- Department of Applied Drug Research, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Imam Reza St., Golgasht St., Tabriz, 5166614756 Iran
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Nouri
- Department of Clinical Biochemistry and Laboratory Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Stem Cell Research Center, Tabriz University of Medical Sciences, Imam Reza St., Golgasht St., Tabriz, 5166614756 Iran
| |
Collapse
|
46
|
Yun CW, Lee SH. The Roles of Autophagy in Cancer. Int J Mol Sci 2018; 19:ijms19113466. [PMID: 30400561 PMCID: PMC6274804 DOI: 10.3390/ijms19113466] [Citation(s) in RCA: 674] [Impact Index Per Article: 96.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 10/29/2018] [Accepted: 11/02/2018] [Indexed: 12/12/2022] Open
Abstract
Autophagy is an intracellular degradative process that occurs under several stressful conditions, including organelle damage, the presence of abnormal proteins, and nutrient deprivation. The mechanism of autophagy initiates the formation of autophagosomes that capture degraded components and then fuse with lysosomes to recycle these components. The modulation of autophagy plays dual roles in tumor suppression and promotion in many cancers. In addition, autophagy regulates the properties of cancer stem-cells by contributing to the maintenance of stemness, the induction of recurrence, and the development of resistance to anticancer reagents. Although some autophagy modulators, such as rapamycin and chloroquine, are used to regulate autophagy in anticancer therapy, since this process also plays roles in both tumor suppression and promotion, the precise mechanism of autophagy in cancer requires further study. In this review, we will summarize the mechanism of autophagy under stressful conditions and its roles in tumor suppression and promotion in cancer and in cancer stem-cells. Furthermore, we discuss how autophagy is a promising potential therapeutic target in cancer treatment.
Collapse
Affiliation(s)
- Chul Won Yun
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul 04401, Korea.
| | - Sang Hun Lee
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul 04401, Korea.
- Department of Biochemistry, Soonchunhyang University College of Medicine, Cheonan 31538, Korea.
| |
Collapse
|
47
|
Hasani S, Boroujeni ME, Aliaghaei A, Baghai K, Rostami A. Dopaminergic induction of human adipose-derived mesenchymal stem cells is accompanied by transcriptional activation of autophagy. Cell Biol Int 2018; 42:1688-1694. [DOI: 10.1002/cbin.11056] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Accepted: 09/16/2018] [Indexed: 12/11/2022]
Affiliation(s)
- Sanaz Hasani
- Faculty of Medical Biotechnology; Department of Stem Cells and Regenerative Medicine; National Institute of Genetic Engineering and Biotechnology; Tehran Iran
| | - Mahdi Eskandarian Boroujeni
- Faculty of Medical Biotechnology; Department of Stem Cells and Regenerative Medicine; National Institute of Genetic Engineering and Biotechnology; Tehran Iran
| | - Abbas Aliaghaei
- Cell Biology and Anatomical Sciences; School of Medicine; Shahid Beheshti University of Medical Sciences; Tehran Iran
| | - Kaveh Baghai
- Basic and Molecular Epidemiology of Gastrointestinal Disorder Research center; Research institute for Gastroenterology and Liver Diseases; Shahid Beheshti University of Medical Sciences; Tehran Iran
| | - Amin Rostami
- Gastroenterology and Liver Disease Research Center; Research institute for Gastroenterology and Liver Diseases; Shahid Beheshti University of Medical Sciences; Tehran Iran
| |
Collapse
|
48
|
Zecchini S, Giovarelli M, Perrotta C, Morisi F, Touvier T, Di Renzo I, Moscheni C, Bassi MT, Cervia D, Sandri M, Clementi E, De Palma C. Autophagy controls neonatal myogenesis by regulating the GH-IGF1 system through a NFE2L2- and DDIT3-mediated mechanism. Autophagy 2018; 15:58-77. [PMID: 30081710 DOI: 10.1080/15548627.2018.1507439] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Macroautophagy/autophagy is emerging as an important process in adult muscle stem cells functions: it regulates metabolic reprogramming during activation from a quiescent state, maintains stemness and prevents senescence. We now show that autophagy is specifically required for neonatal myogenesis and muscle development. Specific deletion of Atg7 in PAX7+ (paired box 7) precursors led in mice to a dwarf phenotype, with an effect restricted to the neonatal phase of muscle development. Atg7 knockdown suppressed neonatal satellite cell (nSC) proliferation and differentiation, downregulating the GH-IGF1 functions. When we disrupted autophagy, NFE2L2/NRF2 (nuclear factor, erythroid 2 like 2) accumulated in muscle and nSCs and negatively modulated DDIT3/CHOP (DNA-damage inducible transcript 3) expression. Lower levels of DDIT3 were responsible for reduced GHR expression leading to impaired local production of IGF1. Our results conclusively identify a novel autophagy-dependent pathway that regulates nSC behavior and indicate that autophagy is required for skeletal muscle development in the neonatal phase. Abbreviations: AKT/protein kinase B: Thymoma viral proto-oncogene; ASCs: adult stem cells; ATF4: activating transcription factor 4; ATG7: autophagy related 7; BAT: brown adipose tissue; BMP: bone morphogenetic protein; CEBPB: CCAAT/enhancer binding protein (C/EBP), beta; CSA: cross sectional area; CTNNB1: catenin (cadherin associated protein), beta 1; DDIT3: DNA-damage inducible transcript 3; DM: differentiation medium; E: embryonic stage; EIF2AK3/PERK; EIF4EBP1: eukaryotic translation initiation factor 2 alpha kinase 3; eukaryotic translation initiation factor 4E binding protein 1; ER: endoplasmic reticulum; FGF21: fibroblast growth factor 21; GH: growth hormone; GHR: growth hormone receptor; HSCs: hematopoietic stem cells; IGF1: insulin-like growth factor 1; ITGAM: integrin alpha M; KEAP1: kelch-like ECH-associated protein 1; LY6A/Sca-1; MAP1LC3: lymphocyte antigen 6 complex, locus A; microtubule-associated protein 1 light chain 3; MAPK1/ERK2: mitogen-activated protein kinase 1; MAPK3/ERK1: mitogen-activated protein kinase 3; miRNAs: microRNAs; MSCs: mesenchymal stem cells; MTOR: mechanistic target of rapamycin kinase; mtUPR: mitochondrial unfolded protein response; MYF5: myogenic factor 5; MYH: myosin, heavy polypeptide; MYOD1: myogenic differentiation 1; MYOG: myogenin; NFE2L2: nuclear factor, erythroid derived 2, like 2; nSC: neonatal satellite cells; NSCs: neuronal stem cells; P: postnatal day; PAX7: paired box 7; PECAM1: platelet/endothelial cell adhesion molecule 1; PPARG: peroxisome proliferator activated receptor gamma; PTPRC: protein tyrosine phosphatase, receptor type, C; ROS: reactive oxygen species; RPS6: ribosomal protein S6; SCs: adult satellite cells; SQSTM1: sequestosome 1; STAT5: signal transducer and activator of transcription 5; TGFB1: transforming growth factor beta 1; WAT: white adipose tissue; WT: wild type.
Collapse
Affiliation(s)
- Silvia Zecchini
- a Unit of Clinical Pharmacology , University Hospital "Luigi Sacco"-ASST Fatebenefratelli Sacco , Milano , Italy
| | - Matteo Giovarelli
- b Department of Biomedical and Clinical Sciences "Luigi Sacco" , Università degli Studi di Milano , Milano , Italy
| | - Cristiana Perrotta
- b Department of Biomedical and Clinical Sciences "Luigi Sacco" , Università degli Studi di Milano , Milano , Italy
| | - Federica Morisi
- c Division of Genetics and Cell Biology , IRCCS Ospedale San Raffaele , Milano , Italy
| | - Thierry Touvier
- d Biology of Myelin Unit, Division of Genetics and Cell Biology , IRCCS Ospedale San Raffaele , Milano , Italy
| | - Ilaria Di Renzo
- b Department of Biomedical and Clinical Sciences "Luigi Sacco" , Università degli Studi di Milano , Milano , Italy
| | - Claudia Moscheni
- b Department of Biomedical and Clinical Sciences "Luigi Sacco" , Università degli Studi di Milano , Milano , Italy
| | - Maria Teresa Bassi
- e Laboratory of Molecular Biology , IRCCS Eugenio Medea , Bosisio Parini , Italy
| | - Davide Cervia
- f Department for Innovation in Biological, Agro-food and Forest systems , Università degli Studi della Tuscia , Viterbo , Italy
| | - Marco Sandri
- g Department of Biomedical Science , University of Padova , Padova , Italy.,h Laboratory of Molecular Biology , Venetian Institute of Molecular Medicine , Padova , Italy
| | - Emilio Clementi
- e Laboratory of Molecular Biology , IRCCS Eugenio Medea , Bosisio Parini , Italy.,i Department of Biomedical and Clinical Sciences "Luigi Sacco" , Università degli Studi di Milano , Milano , Italy
| | - Clara De Palma
- a Unit of Clinical Pharmacology , University Hospital "Luigi Sacco"-ASST Fatebenefratelli Sacco , Milano , Italy.,b Department of Biomedical and Clinical Sciences "Luigi Sacco" , Università degli Studi di Milano , Milano , Italy
| |
Collapse
|
49
|
Wei C, Chen C, Cheng Y, Zhu L, Wang Y, Luo C, He Y, Yang Z, Ji Z. Ailanthone induces autophagic and apoptotic cell death in human promyelocytic leukemia HL-60 cells. Oncol Lett 2018; 16:3569-3576. [PMID: 30127963 PMCID: PMC6096173 DOI: 10.3892/ol.2018.9101] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 07/03/2018] [Indexed: 12/11/2022] Open
Abstract
Ailanthone, which is extracted from the traditional Chinese medicinal plant Ailanthus altissima, has been thoroughly demonstrated to have anti-tumor, anti-HIV, anti-inflammatory, anti-malarial, anti-allergic and anti-microbial activities. However, the anti-proliferative effects of ailanthone on HL-60 cells and potential mechanisms underlying those effects have not been reported. In the present study, we demonstrated the potent cytotoxicity of ailanthone against HL-60 cells. Annexin V-APC/7-ADD staining assay indicated that ailanthone increased the number of apoptotic cells in a dose-dependent manner. PI staining showed that ailanthone increased the percentage of G0/G1-phase cells in a dose-dependent manner. Acridine orange staining suggested that ailanthone induced the formation of acidic vesicular organelles in HL-60 cells and pretreatment with BaF-A1 could attenuate this process. Western blotting showed that ailanthone up-regulated the protein expression levels of beclin-1 and LC3-II and down-regulated those of LC3-I and p62 in a dose-dependent manner. Use of BaF-A1 showed that the anti-proliferative effects of ailanthone on HL-60 cells may be partly attributable to the induction of autophagy-mediated apoptosis by MTT assay and annexin V-APC/7-ADD staining assay.
Collapse
Affiliation(s)
- Cheng Wei
- The Cancer Center, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui 241001, P.R. China
| | - Chuanrong Chen
- Department of Oncology, Wuhu No. 2 People's Hospital, Wuhu, Anhui 241001, P.R. China
| | - Yuxin Cheng
- Department of Oncology, Wannan Medical College, Wuhu, Anhui 241003, P.R. China
| | - Lin Zhu
- Department of Oncology, Wannan Medical College, Wuhu, Anhui 241003, P.R. China
| | - Yu Wang
- Department of Oncology, Wannan Medical College, Wuhu, Anhui 241003, P.R. China
| | - Can Luo
- Department of Oncology, Wannan Medical College, Wuhu, Anhui 241003, P.R. China
| | - Yang He
- The Cancer Center, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui 241001, P.R. China
| | - Zhiming Yang
- The Cancer Center, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui 241001, P.R. China
| | - Zhaoning Ji
- The Cancer Center, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui 241001, P.R. China
| |
Collapse
|
50
|
Ozkan S, Isildar B, Oncul M, Baslar Z, Kaleli S, Koyuturk M. Ultrastructural analysis of human umbilical cord derived MSCs at undifferentiated stage and during osteogenic and adipogenic differentiation. Ultrastruct Pathol 2018; 42:199-210. [PMID: 29624114 DOI: 10.1080/01913123.2018.1453905] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Mesenchymal stem cells (MSCs) are considered as an important tool for regenerative medicine and experimental treatments. Unveiling the ultrastructural changes during the differentiation of MSCs might help us to understand the nature of the process and to develop novel therapeutic approaches. For this purpose, human umbilical cord (hUC) was chosen as MSC source. In the first place, MSCs were isolated from sub-amniotic, intervascular and perivascular areas of hUC by enzymatic and tissue explant method to determine the most favorable region of hUC and technique for further processing. Therefore, microscopic and growth kinetics analyses showed that there was no clear difference in the morphologies and proliferation rates among the hUC-MSC groups. Flow cytometric analysis showed that CD44 and CD90 MSC markers were highly expressed, while CD34 and CD45 hematopoietic stem cells markers were expressed at low degree. Because our preliminary results showed that there was no conspicuous superiority among the hUC-MSCs groups, whole UC was utilized as a source, and tissue explant method was applied to isolate MSCs for further differentiation analysis. At the 1st and 3rd week of osteogenic and adipogenic differentiation, ultrastructural analysis showed an increase in the number of secondary lysosomes in comparison with the undifferentiated status. Increase in the mitochondrial content was also detected at the 1st week of adipogenic differentiation. Consequently, ultrastructural changes including increase in the number of mitochondria and secondary lysosomes during the adipogenic and osteogenic differentiation could be attributed to the switch in energy metabolism of the MSCs and increment in the lysosomal activity respectively.
Collapse
Affiliation(s)
- Serbay Ozkan
- a Department of Histology and Embryology, Cerrahpasa Medical Faculty , Istanbul University , Istanbul , Turkey
| | - Basak Isildar
- a Department of Histology and Embryology, Cerrahpasa Medical Faculty , Istanbul University , Istanbul , Turkey
| | - Mahmut Oncul
- b Department of Obstetrics and Gynecology, Cerrahpasa Medical Faculty , Istanbul University , Istanbul , Turkey
| | - Zafer Baslar
- c Division of Hematology, Department of Internal Medicine, Cerrahpasa Medical Faculty , Istanbul University , Istanbul , Turkey
| | - Semih Kaleli
- b Department of Obstetrics and Gynecology, Cerrahpasa Medical Faculty , Istanbul University , Istanbul , Turkey
| | - Meral Koyuturk
- a Department of Histology and Embryology, Cerrahpasa Medical Faculty , Istanbul University , Istanbul , Turkey
| |
Collapse
|